Next Article in Journal
Biomarkers in Heart Failure: A Review and a Wish
Previous Article in Journal
Vacuolated Marrow Cytopenias from Copper Deficiency to UBA1-Mutant VEXAS: Molecular Landscape, Systematic Review, and Cost-Efficient Diagnostic Algorithm
Previous Article in Special Issue
Modification in Structures of Active Compounds in Anticancer Mitochondria-Targeted Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Redefining Chemoresistance: Natural Bioactives as Molecular Modulators at the Cancer–Tumor Microenvironment Interface

by
Claudia Reytor-González
1,
Emilia Jiménez-Flores
1,
Natalí González
2 and
Daniel Simancas-Racines
1,*
1
Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito 170527, Ecuador
2
Universidad UTE, Facultad de Odontología, Quito 170527, Ecuador
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(16), 8037; https://doi.org/10.3390/ijms26168037 (registering DOI)
Submission received: 20 July 2025 / Revised: 14 August 2025 / Accepted: 15 August 2025 / Published: 20 August 2025

Abstract

Therapeutic resistance remains a critical barrier in effective cancer treatment, contributing to disease recurrence, progression, and reduced patient survival. In recent years, natural bioactive compounds have emerged as promising adjuncts in oncology due to their ability to modulate multiple biological processes involved in resistance. This review explores current evidence on the role of natural compounds in influencing cancer cell behavior and their interactions with the tumor microenvironment. By organizing these compounds into chemical families, we provide a structured overview of their potential to enhance the efficacy of standard chemotherapy and reduce resistance-related mechanisms. We also highlight innovative strategies, including combination therapies and advanced drug delivery systems, that aim to improve their clinical applicability. Overall, this work underscores the relevance of integrating natural bioactives into modern cancer therapy and calls for further translational research to bridge preclinical findings with clinical implementation.

1. Introduction

Therapeutic resistance remains one of the most pressing challenges in modern oncology, representing a critical barrier to the long-term success of cancer treatments [1]. Despite significant advances in drug development, molecular diagnostics, and targeted therapies, resistance to chemotherapy continues to limit patient survival and quality of life [2,3]. In both solid and hematologic malignancies, tumor cells frequently adapt to cytotoxic stress through a myriad of mechanisms, ultimately rendering standard chemotherapeutic regimens ineffective [4]. These mechanisms include increased drug efflux, enhanced deoxyribonucleic acid (DNA) repair capacity, apoptosis evasion, metabolic reprogramming, and epigenetic alterations [5]. However, recent insights suggest that the development of chemoresistance is not solely an intrinsic property of cancer cells, but also profoundly influenced by the complex and dynamic ecosystem in which these cells reside, the tumor microenvironment (TME) [6].
The TME, a highly heterogeneous and interactive network composed of stromal cells, immune cells, extracellular matrix (ECM) components, cytokines, and growth factors, plays a pivotal role in shaping cancer progression and therapeutic response [7]. This microenvironment not only facilitates tumor growth and invasion but also actively participates in the development of resistance through paracrine signaling, immune modulation, and the induction of phenotypic plasticity in tumor cells [8]. The bidirectional crosstalk between cancer cells and the TME fosters a milieu conducive to survival under therapeutic pressure, thereby contributing to both primary and acquired chemoresistance [9].
Considering the limitations of conventional monotherapies and the inherent complexity of resistance pathways, there is a growing interest in agents capable of simultaneously targeting multiple molecular and cellular processes [10]. Natural bioactive compounds, secondary metabolites derived from plants, fungi, bacteria, and marine organisms, have emerged as promising candidates in this regard [11,12,13,14]. Unlike synthetic drugs, which often act on a single target or pathway, many natural compounds possess broad-spectrum biological activities that enable them to modulate a wide array of signaling cascades implicated in cancer pathogenesis and treatment resistance [15].
Notably, several natural bioactives such as curcumin, resveratrol, quercetin, epigallocatechin gallate (EGCG), and sulforaphane have demonstrated the ability to interfere with key regulators of chemoresistance, including nuclear factor-kappa B (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), signal transducer and activator of transcription (STAT) 3, and hypoxia-inducible factors (HIFs) [16]. These compounds also exhibit anti-inflammatory, antioxidant, anti-angiogenic, and immunomodulatory properties, positioning them as ideal candidates for disrupting the protective mechanisms conferred by the TME [17]. In preclinical studies, many of these bioactives have been shown to sensitize resistant tumor cells to chemotherapeutic agents, reverse multidrug resistance phenotypes, and attenuate the pro-tumorigenic effects of stromal elements [18,19,20].
Given their pleiotropic nature, relatively low toxicity, and potential for synergistic interactions with standard therapies, natural bioactives are increasingly being investigated as adjuncts to existing cancer treatment protocols [21]. However, despite promising in vitro and in vivo findings, their clinical translation has been hampered by issues related to bioavailability, pharmacokinetics, and standardization [22]. Nevertheless, advancements in formulation technologies, such as nanoencapsulation and targeted delivery systems, are helping to overcome these limitations, opening new avenues for the integration of natural compounds into evidence-based oncology [23,24,25].
This review aims to synthesize current knowledge on the molecular mechanisms through which natural bioactive compounds influence chemoresistance, with a particular focus on their interactions at the cancer–TME interface. By exploring how these agents modulate key signaling pathways, cellular cross-talk, and environmental stress responses, we seek to provide a comprehensive framework for understanding their potential as multifaceted therapeutic tools. Ultimately, redefining chemoresistance through the lens of natural bioactives may offer novel strategies for improving treatment outcomes and overcoming one of the most formidable obstacles in cancer therapy.

2. Key Molecular Mechanisms Underlying Therapeutic Resistance

Tumor drug resistance arises from a complex interplay of cellular adaptations that enable cancer cells to survive even under aggressive therapeutic pressures. One of the most pivotal strategies employed by malignant cells is the evasion of apoptosis. Under normal physiological conditions, chemotherapeutic agents aim to induce programmed cell death via the intrinsic mitochondrial pathway. This involves mitochondrial outer membrane permeabilization, which initiates cytochrome c release and caspase cascade activation, ultimately leading to apoptosis. However, in resistant tumors, this pathway is frequently disrupted. Cancer cells often overexpress anti-apoptotic genes and proteins, which inhibit mitochondrial outer membrane permeabilization and prevent the apoptotic process. Simultaneously, key pro-apoptotic proteins are downregulated or mutated, effectively shifting the balance toward cell survival.
Oncogenes contribute further to this apoptotic evasion. A prime example is Astrocyte Elevated Gene 1, which drives resistance by promoting the translation of multidrug resistance protein 1 (MDR1) mRNA, thereby enhancing efflux of chemotherapeutics such as doxorubicin. This oncogene also activates adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling, initiating autophagy as a protective mechanism and mediating resistance to doxorubicin and 5-fluorouracil. Furthermore, it impairs retinoic acid receptor function and upregulates MET receptor tyrosine kinase and Aldehyde Dehydrogenase 3 Family Member A1, reinforcing resistance through multiple molecular axes [26]. Another oncogene implicated in chemoresistance is AXL receptor tyrosine kinase, frequently overexpressed in resistant tumors. It facilitates resistance through its interaction with downstream receptor tyrosine kinase (RTK) signaling pathways, including Epidermal Growth Factor Receptor (EGFR), human epidermal growth factor receptor (HER) 2, and 3, thus contributing to enhanced cell survival and adaptation [27].
Survival pathways activated downstream of RTKs further entrench resistance. The PI3K/AKT/mTOR and Mitogen-Activated Protein Kinase (MAPK) signaling cascades are central to these processes. PI3K, upon activation, catalyzes the formation of Phosphatidylinositol-3,4,5-trisphosphate, which recruits and activates AKT via phosphorylation by 3-phosphoinositide-dependent protein kinase-1 and mTORC (mammalian target of rapamycin complex) 2. Activated AKT promotes cell survival by inactivating pro-apoptotic targets such as B-cell lymphoma (BCL) 2-Associated agonist of cell death and caspase-9, while simultaneously activating mTORC1, which enhances protein synthesis and cellular proliferation. In parallel, the MAPK pathway activates nuclear transcription factors like E-twenty-six-like transcription factor 1 and cellular oncogene fos, leading to upregulation of cyclin D1 and anti-apoptotic proteins, including BCL-2 and myeloid cell leukemia (MCL) -1. The Janus kinase (JAK)/STAT pathway complements this network. Activated STAT3 and STAT5 transcriptionally induce anti-apoptotic and immunosuppressive genes, facilitating both tumor survival and immune evasion. NF-κB, another key signaling axis, is activated through IκB kinase degradation and supports tumor persistence through the transcription of pro-survival and pro-inflammatory genes. These pathways are not isolated; extensive crosstalk among them ensures signal redundancy and robustness, complicating therapeutic efforts. PI3K/AKT can amplify NF-κB signaling, while MAPK and JAK/STAT share transcriptional targets with overlapping biological outcomes [28].
In tandem with these survival networks, dysregulated autophagy plays a substantial role in resistance. Autophagy, a lysosome-mediated degradation process, allows cells to recycle damaged components and generate metabolic substrates necessary for energy production and redox balance. In cancer, autophagy is frequently upregulated under therapeutic stress, enabling cells to avert apoptosis and sustain their bioenergetic demands. Autophagy initiation is controlled by the Unc-51-like autophagy activating kinase 1 complex, inhibited by mTORC1 under nutrient-rich conditions. Inhibition of mTORC1, via starvation or targeted drugs, releases this complex, initiating autophagosome formation with the help of Vacuolar Protein Sorting 34 and Beclin-1. While mTOR inhibition may impede proliferation, it simultaneously triggers autophagy, creating a paradox in treatment strategies. Extracellular signal-regulated kinases (ERKs) 1 and 2 in the MAPK pathway also contribute by modulating autophagy-related proteins and transcription factors, reinforcing cell survival under stress [29].
One of the most prominent and well-characterized mechanisms of drug resistance involves increased drug efflux via ATP-binding cassette (ABC) transporters (Figure 1). Among these, P-glycoprotein (P-gp)/ATP-binding cassette sub-family B member 1 (ABCB1) is the most well-known and frequently implicated in multidrug resistance (MDR). P-gp plays a physiological role in protecting tissues by pumping out xenobiotics and toxins, but in tumors, its overexpression contributes to chemotherapy failure. Notably, its regulation is influenced by transcriptional inputs from PI3K/AKT and NF-κB signaling pathways. Additionally, epigenetic alterations, such as hypermethylation of the ABCB1 promoter, can paradoxically increase P-gp expression [30].
Other transporters such as ABCG2 and ABCC1 also participate in MDR, handling a broad spectrum of anticancer drugs including anthracyclines, taxanes, and platinum compounds [31]. ABCG2, for instance, acts as a xenobiotic pump with a significant role in effluxing chemotherapeutics in resistant cancers [32]. These ABC proteins are not only expressed on the plasma membrane, but also in intracellular vesicles, and they critically influence the pharmacokinetics of anticancer agents in humans. Importantly, they are also found in extracellular vesicles, which participate in drug expulsion and the horizontal transfer of resistance phenotypes to other cells in the TME [33]. Across solid tumors, overexpression of ABC transporters, particularly P-gp, remains one of the most consistent features of acquired drug resistance [30].
DNA repair mechanisms and epigenetic modifications further enhance resistance (Figure 1). DNA methyltransferases (DNMTs) catalyze the addition of methyl groups to cytosine residues in cytosine–phosphate–Guanine islands, silencing the expression of tumor suppressor genes such as tumor protein p53 and pro-apoptotic effectors like BAX. Meanwhile, histone deacetylases (HDACs) promote chromatin compaction and gene repression, counteracted by histone acetyltransferases that facilitate gene transcription through chromatin relaxation. These changes are not static; they are dynamically regulated by oncogenic signaling pathways and tumor-derived environmental cues. Moreover, non-coding RNAs, including microRNAs and long non-coding RNAs, contribute significantly to resistance by targeting genes involved in apoptosis, drug metabolism, and DNA repair. Dysregulation in the miRNA biogenesis machinery, such as aberrant activity of Drosha and Dicer, further disrupts microRNA profiles, exacerbating resistance. Notably, some microRNAs have been shown to increase P-gp expression, linking epigenetic regulation directly to drug efflux mechanisms [34,35].
Beyond genetic and epigenetic factors, metabolic reprogramming underlies another layer of tumor adaptability. Cancer cells frequently adopt aerobic glycolysis, a phenomenon known as the Warburg effect, in which glucose is converted into lactate despite sufficient oxygen availability, suggesting the beneficial effects of adopting a low-sugar diet [36]. This metabolic shift supports biosynthetic processes and ATP production needed for rapid proliferation. Additionally, mitochondrial adaptations enhance redox balance and bioenergetic flexibility, promoting cell survival even under nutrient-deprived and hypoxic conditions [37].
This schematic outlines key cellular and molecular mechanisms underlying drug resistance, including tumor microenvironment changes, epigenetic alterations, epithelial-to-mesenchymal transition (EMT), enhanced drug efflux, and increased DNA repair. Drug efflux transporters use ATP hydrolysis to expel chemotherapeutic agents (30), while upregulated DNA repair systems enable cancer cells to survive genotoxic therapies such as platinum-based drugs and topoisomerase inhibitors [30,38,39]. Abbreviations—ABC: ATP binding cassette; EGFR: epidermal growth factor receptor; ATP: adenosine triphosphate; DNA: deoxyribonucleic acid; RTK: receptor tyrosine kinases; CAF: cancer-associated fibroblast; TAM: tumor-associated macrophage; and HER 2: human epidermal growth factor receptor 2.

3. The Tumor Microenvironment as a Co-Regulator of Resistance

The TME is increasingly recognized as a pivotal co-regulator of cancer progression and therapeutic resistance. Composing a heterogeneous network of malignant cells, stromal components, immune infiltrates, and ECM, the TME forms a dynamic ecosystem that profoundly influences tumor biology (Figure 2) [40].
At the core of tumor heterogeneity lies the presence of cancer stem cells (CSCs), a subpopulation endowed with self-renewal, differentiation capacity, and tumor-initiating potential [41]. These cells orchestrate tumor relapse and drug resistance through intrinsic features such as quiescence, enhanced DNA damage response, and overexpression of ABC transporters [39]. Molecularly, CSCs engage signaling pathways including wingless-type MMTV integration site family (Wnt)/β-catenin, Notch, Hedgehog, PI3K/AKT, and NF-κB, which promote survival, stemness, and inflammatory microenvironments conducive to therapy evasion [42]. Physiologically, CSCs often reside in specialized hypoxic niches within the tumor, where HIFs, primarily HIF-1α, mediate metabolic reprogramming to glycolysis and autophagy, supporting CSC maintenance and resistance [42]. Hypoxic stress also induces EMT, enhancing CSC invasiveness and metastatic potential [43]. Together, these molecular and environmental adaptations empower CSCs to survive cytotoxic assaults and contribute substantially to intratumoral heterogeneity and therapeutic failure [43].
Tumor-associated macrophages (TAMs) represent a key immunosuppressive cell population within the TME, arising from circulating monocytes recruited and polarized by tumor-derived signals (Figure 2) [44]. Recent studies have elucidated mechanisms underlying TAM-mediated resistance, including the polarization of macrophages via signaling pathways such as STAT3/IL-10 and PI3K/AKT, which enhance the efficacy of immune checkpoint blockade when modulated [45,46,47]. Moreover, TAMs interplay with adipocytes within the TME, wherein adipocyte-secreted inflammatory cytokines activate NF-κB signaling in macrophages, linking obesity-associated inflammation to endocrine therapy resistance [48,49]. Effective management of obesity is essential, as the chronic inflammation associated with this condition has been implicated in the development or aggravation of various diseases, including cancer [50,51,52,53,54,55].
Cancer-associated fibroblasts (CAFs), the predominant stromal cell type in many solid tumors, originate from activated fibroblasts influenced by tumor-secreted cytokines such as transforming growth factor beta (TGF-β), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF) 2, and can also derive from mesenchymal stem cells or other cell types via transdifferentiation [56]. These cells modulate tumor progression and resistance through diverse mechanisms, including remodeling the ECM, secreting growth factors and cytokines, and mediating metabolic crosstalk [57]. CAFs enhance CSC survival by activating pathways such as Wnt/β-catenin, NF-κB, and STAT3, thus promoting stemness and resistance to agents like oxaliplatin, 5-fluorouracil, gemcitabine, and cisplatin in colorectal, gastric, and pancreatic cancers [58,59,60]. Exosomes secreted by CAFs carry microRNAs and proteins that modulate tumor cell gene expression, induce autophagy, and facilitate drug resistance [61,62,63,64,65,66]. CAFs also regulate immune suppression by recruiting immunosuppressive cells and inducing their polarization via cytokines such as IL-6, TGF-β, and Insulin-like Growth Factor Binding Protein 7, further blunting anti-tumor immunity [57].
A notable CAF function is their involvement in promoting resistance to radiotherapy [67]. They do so by secreting protective cytokines (e.g., IL-6, TGF-β), facilitating autophagy in cancer cells, and direct cell contact, activating survival pathways such as β1 integrin and Focal Adhesion Kinase (FAK) signaling [42]. CAFs are intrinsically radioresistant, partly due to defects in p53/p21 and high expression of Survivin, and radiotherapy itself can induce a secretory phenotype in CAFs characterized by pro-tumorigenic cytokines like IL-6, IL-8, and osteopontin that enhance tumor proliferation and immunosuppression post-irradiation [57]. Additionally, CAFs upregulate immune checkpoint molecules in cancers such as colorectal and lung adenocarcinoma, promoting immune escape [68,69]. They also contribute to ECM remodeling and aberrant angiogenesis, limiting immune cell infiltration and fostering an immunosuppressive milieu [70,71,72,73].
Endothelial cells and cancer-associated adipocytes (CAAs), though less abundant than CAFs, are critical in shaping tumor progression and therapy resistance. Tumor endothelial cells facilitate aberrant neovascularization and vasculogenesis, which can exacerbate hypoxia and promote resistance by limiting T cell infiltration [57]. Vascular endothelial growth factor (VEGF), secreted by endothelial and tumor cells, is central to angiogenesis and suppresses dendritic cell maturation, reducing antigen presentation and enhancing programmed cell death 1 ligand 1 (PD-L1) expression on immune cells, thereby impairing effective T cell responses [74]. ECM remodeling is exacerbated by hypoxia and HIF-1α signaling, which upregulate collagen and fibrosis-associated genes, further reinforcing an immunosuppressive and pro-metastatic niche [75]. Enzymes like lysyl oxidase cross-link ECM fibers, increasing tumor stiffness and acting as a physical barrier to therapeutics [76]. Additionally, aberrant ECM contributes to metabolic stress and activates integrin-FAK signaling pathways in tumor cells, promoting survival and therapy resistance [77].
Hypoxia, a hallmark of solid tumors driven by inadequate vascularization relative to rapid tumor growth, activates HIFs, mainly HIF-1α, which orchestrate transcriptional programs that regulate angiogenesis, metabolic adaptation, immune modulation, and ECM remodeling [76]. Stabilized HIF-1α induces proangiogenic factors such as VEGF, PDGF, FGF, angiopoietins, and matrix metalloproteinases (MMPs), driving the formation of abnormal, leaky vasculature that facilitates invasion, metastasis, and therapy resistance [78,79]. Additionally, HIF-1α modulates metabolic reprogramming toward glycolysis, generating acidic microenvironments rich in lactate that suppress immune effector cells while promoting regulatory populations like regulatory T cells and myeloid-derived suppressor cells (MDSCs) [42]. Reactive oxygen species (ROS) and endoplasmic reticulum stress further regulate HIF signaling, adding layers of complexity to tumor adaptation [80]. The temporal “HIF switch” from HIF-1α in acute hypoxia to HIF-2α in chronic states enables sustained vascular maturation and energy balance, reinforcing tumor progression [81]. Hypoxia also induces immune checkpoints such as PD-L1, Human Leukocyte Antigen-G, CD47, and V-domain Ig Suppressor of T cell Activation, which inhibit T cell and macrophage function and facilitate immune evasion [42].
Collectively, the interplay among CSCs, TAMs, CAFs, endothelial cells, CAAs, and the ECM under hypoxic and immunosuppressive conditions drives tumor heterogeneity, immune escape, angiogenesis, and resistance to diverse therapies [82]. These reciprocal interactions and adaptive mechanisms underscore the complexity of targeting the TME to overcome resistance. While targeting single components like immune checkpoints or angiogenesis has yielded limited success, integrative strategies that consider the multifactorial nature of TME regulation are essential for improving clinical outcomes. Emerging technologies such as single-cell RNA sequencing hold promise for unraveling stromal-immune crosstalk and identifying novel therapeutic targets [57].
Lastly, emerging evidence highlights the role of gut microbiota in oncogenesis and therapy resistance, particularly in colorectal cancer [83]. Dysbiosis, characterized by the predominance of colibactin-producing Escherichia coli, induces DNA damage, ROS generation, and chromosomal instability, contributing to tumor progression and chemoresistance. Targeting such microbial factors may represent novel adjunctive strategies in cancer treatment [84,85].
The TME drives tumor heterogeneity, immune escape, and resistance to chemotherapy, radiotherapy, and immunotherapy. Stromal fibroblasts, immune cells, ECM components, and vasculature activate pro-survival signaling in tumor cells, remodel the ECM to hinder drug delivery, and promote VEGF-independent vascularization. CAAs secrete IL-6, tumor necrosis factor alpha (TNF-α), and insulin-like growth factor-1, stimulating proliferative and survival pathways, metabolic reprogramming, and chronic inflammation through the autotaxin–lysophosphatidic acid pathway. Tumor cells evade immune surveillance by downregulating tumor-associated antigens and major histocompatibility complex molecules, overexpressing immune checkpoint ligands, shedding antigens, and releasing immunosuppressive mediators such as TGF-β, IL-10, and PD-L1, resulting in cytotoxic T lymphocyte exhaustion and tumor persistence. TAMs further contribute to tumor progression, metastasis, angiogenesis, invasion, and CSC expansion through cytokine and growth factor release. CCL8 recruits monocytes, induces CSF-1 production to maintain TAM survival and proliferation, and binds SIGLEC1 to enhance tumor cell motility; inhibition of the CSF-1/CSF-1 receptor axis reduces angiogenesis. IL-1 facilitates metastatic cell recruitment and colonization. Due to their plasticity, TAMs exhibit distinct phenotypes: M1-like TAMs mediate tumoricidal, pro-inflammatory effects, whereas M2-like TAMs promote tumor growth, invasion, angiogenesis, and immune suppression via IL-10, TGF-β, VEGF, and PD-L1. M2-like TAMs also enhance therapeutic resistance by protecting tumor cells from immune destruction, inducing abnormal vasculature, and inhibiting CTL activity [86,87,88,89,90,91,92,93,94,95,96,97,98,99]. Abbreviations—TME: tumor microenvironment; ECM: extracellular matrix; CAAs: cancer-associated adipocytes; MHC: major histocompatibility complex; CCL8: C-C motif chemokine ligand 8; MDSC: myeloid-derived suppressor cell; TGF-β: transforming growth factor beta; Treg: regulatory T cell; PD-L1: programmed cell death 1 ligand 1; VEGF: vascular endothelial growth factor; TAM: tumor-associated macrophage; SIGLEC1: sialic acid-binding immunoglobulin-type lectin 1; IL-1: interleukin-1; IL-6: interleukin-6; IL-10: interleukin-10; TNF– α: tumor necrosis factor-alpha; IGF-1: insulin-like growth factor-1; EGF: epidermal growth factor; PDGFβ: platelet-derived growth factor beta; CSC: cancer stem cell; CSF1: colony-stimulating factor-1; CSF1R: colony-stimulating factor 1 receptor; CTL: cytotoxic T lymphocytes; HIF-1α: hypoxia-inducible factor 1-alpha; and VEGF-A: vascular endothelial growth factor A.

4. Natural Bioactive Compounds with Functional Evidence

Since the interconnected mechanisms described above form a strong barrier to effective cancer treatment, there is growing interest in combination therapies that target multiple pathways, such as autophagy, epigenetic alterations, drug efflux, and dysbiosis, to overcome resistance and improve patient outcomes [100]. Emerging nutritional strategies, including calorie restriction and ketogenic or plant-based diets (i.e., Mediterranean diet), aim to disrupt cancer metabolism and counteract resistance, supporting their use as valuable adjuncts in oncology and therapeutic approaches for other conditions [101,102,103,104,105,106,107,108,109]. These effects are largely attributed to the rich content of bioactive compounds in such diets [110,111], including polyphenols, alkaloids, terpenoids, diterpenes, bioactive lipids, marine-derived metabolites and anthraquinones (Figure 3), which modulate key pathways such as inflammation, oxidative stress, DNA repair, and apoptosis (Figure 4) [112,113,114].

4.1. Polyphenols

Polyphenols represent a broad spectrum of plant-based secondary metabolites recognized for their ability to regulate oxidative balance, immune responses, and carcinogenic pathways [137]. Beyond their well-established antioxidant roles, these compounds modulate inflammation, promote apoptosis, and influence epigenetic and transcriptional landscapes. Their biological activities are often mediated through signaling cascades involving NF-κB, STAT3, PI3K/Akt, and MAPK, which are frequently dysregulated in cancer [138,139,140].
Curcumin, a non-flavonoid polyphenol from Curcuma longa, exerts potent anti-inflammatory and anticancer effects through diverse molecular targets. It inhibits NF-κB signaling by stabilizing the inhibitor of nuclear factor kappa B alpha and blocking the nuclear translocation of NF-κB subunits, thereby downregulating pro-inflammatory mediators such as IL-1β and Cyclooxygenase (COX)-2 [141]. In breast cancer cells, it reduces p100/p52 expression, limiting proliferation and invasion [142]. Additionally, curcumin disrupts STAT3 activation by preventing its phosphorylation and DNA-binding activity, suppressing the expression of survival genes like Mcl-1, X-linked inhibitor of apoptosis protein gene, and PD-L1 [143,144,145]. It also enhances antitumor immunity by increasing CD8+ T cell infiltration and modulating the TME [146].
On an epigenetic level, curcumin inhibits DNMTs and HDACs, alters histone modifications, and restores tumor suppressor gene expression [147,148,149]. It also regulates a broad array of non-coding RNAs, particularly microRNAs and long non-coding RNAs, which influence cancer cell proliferation, invasion, metastasis, and apoptosis in several tumor types, including breast, gastric, ovarian, and colorectal cancers (MING). Through this modulation, curcumin can reverse chemoresistance, enhancing the efficacy of conventional anticancer therapies [150].
Resveratrol, another polyphenol found in grapes and berries, is known for its capacity to inhibit cancer progression by disrupting NF-κB-mediated transcription of inflammatory and anti-apoptotic genes, including COX-2, Bcl-2, and MMP-9 [151,152,153]. It also targets STAT3 and ERK pathways, downregulating IL-6 production and delaying tumor growth in pancreatic models [154]. In cholangiocarcinoma, inhibition of STAT3 is associated with enhanced autophagy via BECLIN-1 [155]. Additionally, resveratrol hinders EMT and cancer stemness by downregulating CD44, CD133, and SLUG through modulation of PI3K/Akt and Smad signaling [124,125,156,157]. On the epigenetic level, it activates Sirtuin 1 (SIRT1) gene, reshapes histone acetylation patterns, alters DNA methylation, and modulates microRNAs, collectively reprogramming gene expression linked to cell cycle control and apoptosis [158,159].
EGCG, the primary catechin in green tea, inhibits tumor-promoting pathways by blocking NF-κB and STAT3 activation, thus reducing the transcription of IL-6, MMP-9, and CD44 [160,161,162]. EGCG also suppresses the PI3K/Akt axis, leading to activation of p53, p21, Bax, and caspases, while downregulating anti-apoptotic proteins like Bcl-2 [126]. Importantly, EGCG exerts significant epigenetic control by inhibiting DNMTs and HDACs, reactivating silenced tumor suppressors, and regulating EMT-associated microRNAs [163,164,165]. Furthermore, it alters the tumor immune landscape by targeting MDSCs through modulation of the Arginase 1 (Arg-1)/inducible nitric oxide synthase/STAT3 axis and related signaling pathways [166].

4.2. Alkaloids

Alkaloids like berberine (BBR) and vincristine are known for their strong anticancer properties, primarily due to their ability to modulate key intracellular signaling networks that govern angiogenesis inhibition, mitochondrial stability, cell cycle progression, and mechanisms of multidrug resistance [167]. BBR, a naturally occurring benzylisoquinoline alkaloid with a characteristic yellow crystalline appearance, is present in various parts of medicinal plants, including roots, bark, leaves, and rhizomes [168]. On a mechanistic level, BBR initiates apoptosis and mitochondrial impairment by elevating intracellular ROS, prompting cytochrome c release, and activating caspase-3 through the upregulation of Growth Arrest and DNA Damage-inducible protein 153 (GADD153) and the involvement of the PI3K/Akt/mTOR and AMPK–p53 axes [169]. In cancers such as cervical, thyroid, and glioblastoma, it further promotes intrinsic apoptotic signaling, increases intracellular calcium levels, and shifts the Bax/Bcl-2 ratio in favor of apoptosis [170].
BBR also plays a crucial role in halting cell cycle progression at multiple checkpoints (G0/G1, G1, and G2/M), largely by increasing the expression of cyclin-dependent kinase inhibitors CIP1/p21 and Kip1/p27, while downregulating cyclins A and D1 and cyclin-dependent kinases 1, 2, 4, and 6 [171]. These effects are reinforced by the suppression of murine double minute 2, which allows for enhanced stabilization and activation of the p53 protein, along with the regulation of specific microRNAs (miR-23a and miR-214-3p) and interactions with calmodulin [172]. Moreover, BBR has been shown to potentiate the effects of multiple chemotherapeutic agents, including cisplatin, doxorubicin, 5-fluorouracil, niraparib, icotinib, and Osimertinib, and to increase tumor sensitivity to radiotherapy [173]. One major contributor to these effects is its ability to downregulate the expression of the MDR1 gene, which encodes the P-gp efflux pump, a key factor in chemotherapy resistance [174]. Notably, in the setting of Candida albicans infection, BBR takes advantage of a pH-dependent reversal in Mdr1p transporter function, typically an efflux mechanism, to enable its own intracellular accumulation, offering a unique antifungal strategy [175].
Vincristine and vinblastine, two Vinca alkaloids derived from Catharanthus roseus, exhibit strong cytotoxic effects by interfering with microtubule formation, thereby impairing mitotic spindle assembly and inducing cell cycle arrest at the G2/M transition via cyclin B1 accumulation [176,177]. This disruption ultimately leads to apoptosis through mitochondrial pathways, involving c-Jun N-terminal kinase-mediated activation of pro-apoptotic proteins such as Bax and Bak, mitochondrial outer membrane permeabilization, and caspase-3 and -9 cleavage [127]. Beyond their antimitotic actions, these compounds influence additional signaling pathways, such as NF-κB signaling by facilitating inhibitor of nuclear factor kappa B alpha degradation, enabling nuclear translocation of NF-κB and transcriptional control over genes related to cell death. Furthermore, vincristine regulates a variety of microRNAs, including miR-155, miR-122, miR-1179, and miR-222-3p, which target critical survival pathways such as PI3K/Akt, MAPK, and JAK/STAT, thereby promoting apoptosis and overcoming resistance in cancer cells [178].

4.3. Terpenoids and Diterpenes

Among natural bioactive compounds, terpenoids such as andrographolide and paclitaxel have demonstrated promising therapeutic effects in resistance models, particularly through their roles in autophagy modulation and microtubule dynamics. Andrographolide, a diterpenoid lactone extracted from Andrographis paniculata, exhibits diverse pharmacological properties, including anti-inflammatory, anti-diabetic, and anticancer activities [179]. Mechanistically, it interferes with pro-survival autophagic signaling by inhibiting the PI3K/AKT/mTOR pathway and activating the c-Jun N-terminal kinase cascade, thereby reducing HIF-1α expression, a key regulator of tumor progression under hypoxia [180]. This action is associated with mitochondrial dysfunction, as shown by increased p53 expression, an elevated Bax/Bcl-2 ratio, and enhanced radiosensitivity in tumor cells [181]. Furthermore, andrographolide exerts anti-angiogenic and anti-inflammatory effects by downregulating NF-κB, Erk1/2, nicotinamide adenine dinucleotide phosphate oxidase, ROS, and P38 MAPK, while concurrently activating activator protein-1 [182]. Its ability to inhibit oncogenic pathways such as NF-κB, HIF-1, and JAK/STAT has been observed across various cancer types, including lung, breast, ovarian, and colorectal malignancies [182,183,184,185].
Paclitaxel, another diterpenoid of natural origin, was first isolated from the bark of Taxus brevifolia and is known for its unique mechanism of stabilizing microtubules [128]. By binding to the β-tubulin subunit and promoting M-loop conformational changes, it enhances lateral protofilament interactions and induces mitotic arrest [186,187]. Additionally, paclitaxel triggers autophagy in various cancers by increasing expression of Beclin-1, Autophagy protein 5, and microtubule-associated protein 1 light chain 3, contributing to both apoptotic and caspase-independent cell death pathways [188,189]. Due to sustainability concerns, alternative production approaches, including biosynthesis and tissue culture, are being actively explored [190].

4.4. Bioactive Lipids and Marine-Derived Metabolites

Marine-derived bioactive compounds such as fucoxanthin (FX) and kahalalide F (KF) demonstrate significant promise in modulating cellular responses to mitochondrial dysfunction and non-canonical apoptosis [129]. FX, a carotenoid abundant in brown algae, protects cells from mitochondrial stress by preserving membrane potential, enhancing ATP synthesis, and reducing ROS overproduction through the activation of signaling cascades such as Akt/glycogen synthase kinase 3β/Tyrosine-protein kinase Fyn and Nuclear Factor Erythroid 2-Related Factor 2/Heme Oxygenase-1 [191]. FX counteracts toxin-induced mitochondrial dysfunction, such as that caused by Ochratoxin A, by restoring the activity of respiratory complexes I and III, upregulating mitochondrial genes (ND1, ND5, CO-I), and activating mitochondrial biogenesis via peroxisome proliferator-activated receptor gamma coactivator 1-α and nuclear respiratory factor 1 [192]. Its metabolite fucoxanthinol exhibits superior efficacy in restoring antioxidant defenses and mitophagy markers such as PTEN-induced kinase 1 and Parkin, attenuating both ROS and intrinsic apoptotic signaling via caspase-9, -3, and Bax/Bcl-2 modulation [130,192]. Additionally, FX induces apoptosis-inducing factor-dependent, caspase-independent apoptosis, modulates mitochondrial dynamics, and mitigates endoplasmic reticulum stress-related apoptosis via GADD153/glucose-regulated protein 78 suppression [130,193].
KF, a cyclic depsipeptide originally isolated from Bryopsis spp., induces rapid mitochondrial membrane depolarization, lysosomal permeabilization, and endoplasmic reticulum dilation, culminating in oncosis-like, caspase-independent cell death [194,195]. Unlike traditional apoptosis, KF disrupts cellular architecture via swelling, blebbing, and lipid bilayer dissolution, possibly mediated by HER3 interference and AKT pathway suppression [196]. Its selectivity for tumor cells and independence from MDR1 or BCL2 pathways highlight its therapeutic potential in apoptosis-resistant cancers [197,198].
Building on the anticancer effects of marine-derived compounds, Nortopsentins, bis-indolyl alkaloids from marine sponges, demonstrate potent cytotoxic and anti-inflammatory activities. These compounds and their derivatives are active against a broad spectrum of cancer cell lines, including lung, breast, colon, ovarian, pancreatic, liver, bladder, prostate, melanoma, renal, sarcoma, and uterine cancers [131]. Their bis-indole scaffold, often containing an imidazole moiety, allows them to target key signaling pathways by inhibiting cyclin-dependent kinase 1, inducing cell cycle arrest at G0/G1 or G2/M phases, and triggering apoptosis through nuclear condensation and membrane blebbing. In colorectal cancer stem cells, adaptive activation of CD44v6-mediated Wnt signaling can occur, but co-treatment with checkpoint Kinase 1 inhibitors like rabusertib disrupts this pathway and promotes apoptosis in both resistant and non-resistant cell populations, highlighting the versatility of Nortopsentin derivatives in targeting multiple oncogenic mechanisms [194].

4.5. Anthraquinones and Cobalamins

Anthraquinones, naturally occurring compounds in medicinal plants, exhibit neuroprotective, anti-inflammatory, anticancer, hepatoprotective, and anti-aging activities, largely through modulation of ROS [132]. They inhibit tumor growth by targeting key molecular pathways and proteins, including kinases, topoisomerases, telomerases, matrix metalloproteinases, and G-quadruplex DNA, disrupting processes critical for cancer cell survival. Certain derivatives, such as 1,3-dihydroxy-9,10-anthraquinones, induce G2/M arrest and apoptosis in cancer cells, while tetracyclic anthraquinone–pyridine hybrids show potent activity against both drug-sensitive and multidrug-resistant leukemia cells, suggesting potential to overcome chemoresistance [133]. Safety concerns persist, as compounds like rhein and emodin have been linked to hepatotoxicity, reproductive toxicity, and developmental defects, and chronic laxative use has been associated with melanosis coli and rare irreversible tissue changes [199]. Complementary strategies, such as cobalamins (C63H88CoN14O14P), which are vitamin B12 derivatives, can enhance anticancer efficacy by selectively targeting tumor cells and improving delivery; for instance, their use in phthalocyanine-based photodynamic therapy reduces dark toxicity and increases therapeutic performance [200,201].

5. Emerging Strategies for Therapeutic Combination

Despite notable advancements in cancer therapies, conventional treatments continue to face significant challenges, including nonspecific cytotoxicity, inadequate tumor targeting, and the recurrent development of resistance to drugs [202]. These limitations often lead to suboptimal therapeutic outcomes, relapse, and treatment failure. To address these concerns, growing attention has been given to integrating natural bioactive compounds, particularly those sourced from plants and marine organisms, into standard cancer therapies [203]. These molecules are recognized for their multitargeting abilities and generally exhibit lower toxicity compared to synthetic drugs [202,204,205]. They can influence immune mechanisms, promoting programmed cell death, hindering tumor cell proliferation, and improving tumor sensitivity to conventional chemotherapy. When used in combination with standard anticancer drugs, they often generate synergistic effects that can mitigate toxicity and help overcome multidrug resistance [206].
Numerous natural compounds have demonstrated enhanced anticancer efficacy when administered alongside chemotherapeutics (Table 1). For instance, EGCG intensifies the effects of cisplatin and paclitaxel by increasing DNA damage and lowering oxidative stress levels [207,208]. Curcumin, widely known for its anti-inflammatory and antineoplastic effects, acts through mechanisms including NF-κB inhibition. Nonetheless, its antioxidant properties may counteract ROS-dependent drugs such as doxorubicin [209]. Even so, curcumin remains effective when co-administered with drugs like paclitaxel, melphalan, and prednisone, especially in gastric cancer and multiple myeloma, owing to its ability to block NF-κB signaling [210]. In fact, adherence to the Mediterranean diet has been associated with protective effects against gastric cancer, largely attributed to the bioactive compounds present in plant-based foods [211].
Several additional compounds work through complementary biological pathways. For example, gambogic acid and solamargine have been shown to increase the impact of cisplatin in non-small-cell lung cancer (NSCLC) by repressing Hedgehog signaling. Similarly, sulforaphane and gefitinib exert comparable effects on this pathway [212]. Hederagenin enhances cancer therapy outcomes by modulating autophagy markers such as microtubule-associated protein light chain 3 I and II. Resveratrol, sourced from grapes, bolsters the therapeutic action of paclitaxel by inhibiting anti-apoptotic proteins like Bcl-2 and survivin through mTOR pathway suppression. Flavonoids such as apigenin boost cisplatin-induced apoptosis and reduce cancer stem cell fractions via p53-mediated mechanisms [213]. Other compounds, including dehydrobruceine B, intensify the effectiveness of platinum-based chemotherapy by inducing mitochondrial oxidative stress [214,215]. Notably, curcumin also exhibits protective effects on healthy tissues during cisplatin treatment in NSCLC, supporting its dual role as a sensitizer and a cytoprotective agent [216].
Despite these promising results, many natural compounds face substantial pharmacokinetic challenges. Their clinical application is hindered by issues like low aqueous solubility, instability in physiological environments, limited absorption, and short systemic circulation, which collectively reduce therapeutic efficacy and necessitate higher, potentially toxic, dosages [217]. To address these drawbacks, a variety of advanced drug delivery technologies have been introduced, such as nanocarriers, liposomes, and targeting systems [218].
Nanoparticles (NPs), generally sized between 1 and 100 nanometers, offer numerous benefits. Their high surface area and small dimensions support improved drug encapsulation, sustained release, and preferential accumulation in tumor tissues via the enhanced permeability and retention effect. Furthermore, they can be surface-modified with targeting molecules, such as antibodies, peptides, or folate, to bind tumor-associated receptors like HER 2, EGFR, the transferrin receptor, and αvβ3 integrins, enhancing targeted delivery and minimizing off-target toxicity [219].
The use of environmentally friendly NPs derived from marine organisms further expands therapeutic possibilities. For example, silver NPs produced from the green alga Caulerpa taxifolia prompt apoptosis in lung cancer cells through membrane damage. Similarly, jellyfish-derived gold NPs inhibit the activity of oncogenic kinases like AKT and ERK in HeLa cells. Other cases include copper oxide nanoparticles synthesized by Rhodotorula mucilaginosa and chitin–silver hybrids from shrimp shells, which promote apoptosis through Bax and caspase activation while suppressing anti-apoptotic proteins like Bcl-2 and Bcl-xL (JEONG). These marine-based nanocarriers offer both inherent anticancer properties and enhanced drug transport functions.
Liposomes, built from lipid bilayers, represent another key nanocarrier with clinical relevance. They protect encapsulated drugs from degradation and can carry both hydrophobic and hydrophilic agents. PEGylation, modification with polyethylene glycol, prolongs systemic circulation by reducing immune detection, improving drug accumulation at the tumor site [220]. A prominent example is PEGylated liposomal doxorubicin, which demonstrates reduced heart toxicity compared to unencapsulated doxorubicin. Innovations in liposome design have led to tumor-specific targeting. For instance, liposomes conjugated with antibodies against melanoma-associated antigen MAGE-A1 have shown improved delivery of doxorubicin in experimental models [221]. FF-10832, a PEGylated liposomal version of gemcitabine, has shown favorable pharmacokinetics, including prolonged half-life, better tumor uptake, and reduced hematologic side effects [220]. Stimuli-responsive delivery systems represent another frontier. These “smart” carriers are engineered to respond to tumor microenvironmental cues such as low pH, redox gradients, or specific enzymes. pH-sensitive liposomes and enzyme-cleavable polymeric NPs allow precise drug release at tumor sites while sparing healthy tissues [222].
Beyond liposomes, solid lipid Nps, nanoemulsions, and dendrimers have been developed to enhance oral bioavailability and avoid first-pass metabolism. These systems shield active agents from enzymatic degradation and improve uptake by optimizing parameters such as particle size, surface charge, and lipid content [217,222]. Additionally, modifying drug molecules into prodrugs or optimizing their chemical structure can further improve metabolic stability. Emerging studies have also highlighted the role of gut microbiota in modulating drug metabolism and absorption. Microbial enzymes can modify drug molecules through hydrolysis, reduction, or deconjugation, influencing therapeutic activity before systemic circulation begins [223]. The effects of these microbial transformations can vary based on diet, age, antibiotic use, and host genetics, complicating pharmacokinetic predictability. Tailoring treatment strategies according to microbiome profiles could significantly enhance personalized medicine approaches.
Nanotechnology has also transformed immunotherapy by improving the delivery of checkpoint inhibitors, vaccines, and immune modulators [224]. Liposomes and nanoparticles have been utilized to transport agents like anti-programmed cell death protein-1 (PD-1), anti-PD-L1, and anti-cytotoxic T-lymphocyte-associated protein-4, improving their tumor localization and enhancing therapeutic results [225]. Targeted nanocarriers equipped with antibodies have improved drug delivery in melanoma models, although excessive targeting affinity may increase off-target interactions [221].
Additional methods include the use of tumor-targeting peptides that bind specifically to integrins overexpressed in tumor vasculature, enhancing nanocarrier penetration and retention [219]. Multifunctional “theranostic” systems, combining therapeutic and diagnostic components, are emerging as valuable tools for real-time monitoring of treatment distribution and efficacy. These systems seek to personalize drug release both spatially and temporally, reduce off-target exposure, and adapt therapy to individual patient needs.
Finally, oral lipid-based platforms, such as self-emulsifying drug delivery systems, have demonstrated improvements in systemic bioavailability by enhancing lymphatic transport and bypassing liver metabolism. Co-delivery of chemotherapeutic agents with efflux pump blockers or modulators of the tumor microenvironment through these platforms further combats resistance by facilitating higher intracellular drug concentrations.
Table 1. Synergic effect between natural compounds and chemotherapeutic drugs.
Table 1. Synergic effect between natural compounds and chemotherapeutic drugs.
TypeNatural CompoundChemotherapy DrugTissue and OriginMechanism
PolyphenolCurcumin [226]DoxorubicinBreast, tumoralInhibits the ATPase function of ABCB4 while leaving its protein expression levels unchanged.
Resveratrol [227]5-fluorouracilColon, tumoralRegulates the TNF-β signaling cascade, promotes apoptosis, and inhibits NFκB pathway activation.
Resveratrol [228]CisplatinBasal alveolar epithelial, tumoralTriggers apoptosis by influencing autophagy-related cell death mechanisms.
Urolithin A [229]OxaliplatinColon, tumoralStabilization of p53 and activation of its downstream target genes, leading to control of the cell cycle and suppression of glycolysis.
AlkaloidNeferine and isoliensinine [230]CisplatinColon, tumoralEnhanced cellular absorption of Cisplatin and initiation of mitochondrial apoptosis through a ROS-dependent pathway.
Berberine [231]CisplatinOvarian, tumoralReduction in cell growth along with the promotion of both apoptosis and necroptosis as forms of cell death.
Emetine [232]CisplatinOvarian, tumoralDecreased cell survival capacity.
Tetrandrine [233]CisplatinBreast, tumoralInitiation of apoptosis through a mechanism mediated by ROS.
Piperlongumine [234]DoxorubicinProstate, tumoralInhibitory effect on cell proliferation and promotion of apoptosis, marked by increased levels of cleaved poly (ADP-ribose) polymerase and caspase-3 proteins.
Piperlongumine [235]PaclitaxelIntestinal, tumoralCell death triggered by ROS.
TerpenoidOridonin [236]CisplatinBronchial epithelium cell, tumoralApoptosis initiation via activation of autophagosomes through the AMPK/Akt/mTOR signaling pathway.
Borneol [237]DoxorubicinGlioma cell, tumoralBorneol increases the cellular absorption of doxorubicin and stimulates the generation of ROS.
Vielanin k [238]DoxorubicinBreast and mammary cell, tumoral and non-tumoralTriggering of endoplasmic reticulum stress and mitochondrial apoptosis through the IRE1α-TRAF2-JNK signaling pathway.
Vielanin P [239]DoxorubicinBreast and myelegenous leukemia cell, tumoralPromotion of doxorubicin buildup by decreasing MRP1 expression through the PI3K/Nrf2 signaling pathway.
Ginkgolide B [240]GemcitabinePancreatic cell, tumoralInhibition of NFκB activity combined with an enhancement of antiproliferative effects.
Pachymic acid and dehydrotumulosic acid [241]Doxorubicin and CisplatinLiver, breast, and lung cells, tumoralBlockage of P-glycoprotein (Pgp) activity leading to increased doxorubicin accumulation and amplification of its biological effects.

6. Current Challenges and Future Perspectives

Although bioactive compounds such as curcumin, resveratrol, and EGCG have demonstrated promising synergy with conventional chemotherapeutics in preclinical studies, translating these results to clinical efficacy remains a major hurdle. Pharmacokinetic differences between animal models and humans frequently lead to discrepancies in drug absorption, distribution, metabolism, and excretion, resulting in limited clinical success [242,243]. Curcumin, in particular, suffers from poor bioavailability and rapid metabolic clearance in humans, which severely restricts its systemic exposure despite encouraging in vitro outcomes [244,245]. Consequently, updated translational approaches, such as patient-derived xenografts, 3D tumor organoids, and early-phase pharmacokinetic profiling, are increasingly advocated to better simulate human biology and optimize dosing strategies [246,247,248]. Ochoa et al. [134] performed a study where they examined the combination of curcumin and resveratrol with chemotherapy in colorectal and breast cancer and exhibited improved tolerability but did not achieve statistically significant efficacy, largely due to suboptimal dosing and bioavailability challenges.
Another critical barrier lies in the standardization and reproducibility of botanical extracts. Unlike synthetic drugs, natural products are inherently complex mixtures whose chemical profiles can vary based on geographic origin, harvest season, and extraction protocols [249]. This variability undermines consistency in experimental results and complicates regulatory approval [250,251]. To address this, the ConPhyMP guidelines have been introduced to promote standardized practices in botanical authentication, chromatographic techniques (including fingerprinting), and quantification of key marker compounds [252]. Despite widespread recognition of these standards, many research laboratories still lack the capacity or standardized protocols to implement them fully, resulting in ongoing discrepancies between batches and studies.
Technological advances in nanocarrier design, including NPs and liposomes, hold great promise for overcoming pharmacokinetic limitations [253]. Novel platforms such as erythrocyte membrane-coated NPs and glycoprotein-targeted mesoporous silica systems have demonstrated improved drug stability, evasion of immune surveillance, and controlled release in tumor-specific environments (i.e., pH-sensitive or enzyme-triggered), showing efficacy in lung cancer models [24,245,254,255,256]. Liposomal formulations incorporating curcumin and paclitaxel via inhalation have likewise induced selective apoptosis in NSCLC models while sparing normal lung tissue [257]. These developments illustrate how nanotechnology can enhance both the delivery and therapeutic index of bioactive–chemotherapy combinations [258,259].
Combining natural compounds with immunotherapy is another emerging frontier [260]. Certain phytochemicals act as immune checkpoint modulators [261]; for example, apigenin downregulates PD-L1 expression and berberine promotes PD-L1 degradation, thereby enhancing cytotoxic T-cell activity [254]. EGCG has been shown to prevent chemotherapy-induced upregulation of PD-L1 via the JAK2/STAT1 signaling axis, preserving effector CD8+ T-cell function in models of resistant lung cancer [262]. Moreover, co-administration of apigenin with anti-PD-1 therapy in murine Lewis lung carcinoma models enhanced T-cell infiltration and tumor regression, suggesting that phytochemicals may effectively augment the immune effects of checkpoint inhibitors [254].
Personalized oncology further expands the potential of these combined strategies by tailoring treatments based on individual molecular and microbiome profiles [263,264]. The gut microbiota plays a pivotal role in modifying drug metabolism and immune responses; microbial enzymes can convert certain phytochemicals into active or inactive metabolites, influencing therapeutic efficacy and toxicity [265,266]. Consequently, microbiome-aware drug formulation, incorporating probiotics, prebiotics (i.e., by nutrition), or microbiome-preserving nanoparticles, could improve bioavailability and reduce variability across patient populations [267,268]. Similarly, tumor profiling that includes PD-L1 status, microsatellite instability, and tumor mutational burden could guide the selection of optimized bioactive–immunotherapy combinations, supporting more personalized and potentially effective regimens [269,270,271].
Furthermore, theranostic nanoparticle systems that co-encapsulate diagnostic imaging agents and therapeutic payloads offer real-time monitoring of drug distribution and response [272]. These smart carriers can adjust release kinetics based on imaging feedback and pharmacodynamic markers, forming the basis of closed-loop therapeutic systems that personalize dosing and minimize off-target toxicity [273,274].
Taken together, the current challenges and future perspectives in integrating natural bioactives into cancer therapy can be summarized by three key priorities: 1) enhancing translational predictability via advanced human-relevant models and robust pharmacokinetic data; 2) ensuring reproducibility and regulatory compliance through standardized botanical preparations like those recommended by ConPhyMP; and 3) advancing precision therapy using smart nanocarriers capable of delivering synergistic bioactive and chemotherapeutic agents in an immunologically informed, patient-specific manner [275]. These comprehensive strategies have the potential to transform preclinical findings into clinically impactful treatments.

7. Conclusions

In the context of growing therapeutic resistance, natural bioactive compounds have demonstrated substantial potential to influence both intrinsic and microenvironment-driven mechanisms that allow cancer cells to survive, adapt, and evade treatment. Their multitarget nature, encompassing effects on cell death regulation, stress responses, immune modulation, and stromal interactions, positions them as ideal candidates for complementary therapy. Unlike single-target synthetic agents, these compounds can act on various levels simultaneously, addressing the complexity of resistance in a more holistic way.
This review has highlighted key chemical families, such as polyphenols, alkaloids, terpenoids, and marine-derived metabolites, that show functional relevance in resistance models. The integration of these agents with conventional chemotherapy, supported by advances in delivery systems and pharmacokinetic improvements, may help overcome the limitations of current treatment protocols. However, to move from experimental promise to clinical reality, several challenges remain. These include the need for standardized extracts, consistent bioavailability, and robust clinical trial data.
Ultimately, harnessing the therapeutic potential of natural bioactives requires a transdisciplinary approach. Collaboration between molecular researchers, pharmacologists, clinicians, and nutriologists is essential to unlock new therapeutic pathways and provide more personalized, effective, and less toxic options for cancer patients. Natural bioactives do not replace conventional therapy, but they may be key allies in redefining resistance and improving long-term outcomes.

Author Contributions

Conceptualization, C.R.-G. and D.S.-R.; methodology, C.R.-G.; writing—original draft preparation, C.R.-G. and D.S.-R.; writing—review and editing, C.R.-G., E.J.-F., N.G. and D.S.-R.; visualization, E.J.-F.; supervision, D.S.-R. All authors have read and agreed to the published version of the manuscript.

Funding

Universidad UTE will fund the publication fee for this article.

Acknowledgments

The authors are grateful to Universidad UTE for their support.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABCATP-binding cassette
ABCB1ATP-binding cassette sub-family B member 1
Aktprotein kinase B
AMPKadenosine monophosphate-activated protein kinase
Arg-1arginase 1
ATPadenosine triphosphate
BAKBcl-2 antagonist/killer 1
BAXBcl-2-associated X protein
BCLB-cell lymphoma
CAAscancer-associated adipocyte
CAFscancer-associated fibroblasts
CSCscancer stem cells
CTLcytotoxic T lymphocyte
DNMTsDNA methyltransferases
ECMextracellular matrix
EGCGepigallocatechin gallate
EGFepidermal growth factor
EGFRepidermal growth factor receptor
EMTepithelial-to-mesenchymal transition
ERKextracellular signal-regulated kinase
FAKfocal adhesion kinase
FGFfibroblast growth factor
FXfucoxanthin
GADD153growth arrest and DNA damage-inducible protein 153
HDACshistone deacetylases
HERhuman epidermal growth factor receptor
HIFshypoxia-inducible factors
IGF-1insulin-like growth factor-1
ILinterleukin
iNOSinducible nitric oxide synthase
JAKJanus kinase
KFkahalalide F
MAPKmitogen-activated protein kinase
MCLmyeloid cell leukemia
MDRmultidrug resistance
MDR1multidrug resistance protein 1
MDSCsmyeloid-derived suppressor cells
MHCmajor histocompatibility complex
MMPsmatrix metalloproteinases
mTORmammalian target of rapamycin
mTORcmammalian target of rapamycin complex
NPsnanoparticles
NF-κBnuclear factor-kappa B
NSCLCnon-small-cell lung cancer
P-gpP-glycoprotein
PD-1programmed cell death protein 1
PD-L1programmed cell death 1 ligand 1
PDGFplatelet-derived growth factor
PI3Kphosphoinositide 3-kinase
Tregsregulatory T cells
ROSreactive oxygen species
RTKreceptor tyrosine kinase
STATsignal transducer and activator of transcription
TAMstumor-associated macrophages
TGF-βtransforming growth factor beta
TMEtumor microenvironment
TNF-α tumor necrosis factor alpha
TNF-β tumor necrosis factor beta
VEGFvascular endothelial growth factor
Wntwingless-type MMTV integration site family

References

  1. Dakal, T.C.; Sharma, N.K.; Sharma, A. Editorial: Revisiting the challenges and opportunities in cancer drug resistance. Front. Mol. Biosci. 2024, 11, 1497754. [Google Scholar] [CrossRef]
  2. Shaham, S.H.; Vij, P.; Tripathi, M.K. Advances in Targeted and Chemotherapeutic Strategies for Colorectal Cancer: Current Insights and Future Directions. Biomedicines 2025, 13, 642. [Google Scholar] [CrossRef] [PubMed]
  3. Rosendo-Chalma, P.; Díaz-Landy, E.N.; Antonio-Véjar, V.; Ortiz Tejedor, J.G.; Reytor-González, C.; Simancas-Racines, D.; Bigoni-Ordóñez, G.D. Endometriosis: Challenges in Clinical Molecular Diagnostics and Treatment. Int. J. Mol. Sci. 2025, 26, 3979. [Google Scholar] [CrossRef]
  4. Cortesi, M.; Rossino, G.; Chakrabarty, A.; Rossi, D. Editorial: Tumor adaptation to cellular stresses: Mechanisms, biomarkers and therapeutic opportunities. Front. Med. 2023, 10, 1268976. [Google Scholar] [CrossRef]
  5. Oršolić, N.; Jazvinšćak Jembrek, M. Potential Strategies for Overcoming Drug Resistance Pathways Using Propolis and Its Polyphenolic/Flavonoid Compounds in Combination with Chemotherapy and Radiotherapy. Nutrients 2024, 16, 3741. [Google Scholar] [CrossRef]
  6. Fatima, S. Tumor Microenvironment: A Complex Landscape of Cancer Development and Drug Resistance. Cureus 2025, 17, e82090. [Google Scholar] [CrossRef]
  7. Yun, H.; Dong, F.; Wei, X.; Yan, X.; Zhang, R.; Zhang, X.; Wang, Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol. Rep. 2024, 53, 14. [Google Scholar] [CrossRef] [PubMed]
  8. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef]
  9. Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15. [Google Scholar] [CrossRef]
  10. Haque, A.; Brazeau, D.; Amin, A.R. Perspectives on natural compounds in chemoprevention and treatment of cancer: An update with new promising compounds. Eur. J. Cancer 2021, 149, 165–183. [Google Scholar] [CrossRef]
  11. Abuzahrah, S.S.; Elbehairi, S.E.I.; Bakhsh, T.; Atwa, A.; Juaid, N.; Mekky, R.H. Marine-derived secondary metabolites in oncology: A comprehensive review. Curr. Res. Biotechnol. 2025, 10, 100300. [Google Scholar] [CrossRef]
  12. Pongen, Y.L.; Thirumurugan, D.; Ramasubburayan, R.; Prakash, S. Harnessing actinobacteria potential for cancer prevention and treatment. Microb. Pathog. 2023, 183, 106324. [Google Scholar] [CrossRef] [PubMed]
  13. Asma, S.T.; Acaroz, U.; Imre, K.; Morar, A.; Shah, S.R.A.; Hussain, S.Z.; Arslan-Acaroz, D.; Demirbas, H.; Hajrulai-Musliu, Z.; Istanbullugil, F.R.; et al. Natural Products/Bioactive Compounds as a Source of Anticancer Drugs. Cancers 2022, 14, 6203. [Google Scholar] [CrossRef] [PubMed]
  14. Kaliaperumal, K.; Salendra, L.; Liu, Y.; Ju, Z.; Sahu, S.K.; Elumalai, S.; Subramanian, K.; Alotaibi, N.M.; Alshammari, N.; Saeed, M.; et al. Isolation of anticancer bioactive secondary metabolites from the sponge-derived endophytic fungi Penicillium sp. and in-silico computational docking approach. Front. Microbiol. 2023, 14, 1216928. [Google Scholar] [CrossRef]
  15. Sun, L.; Zhou, W.; Zhang, H.; Guo, Q.; Yang, W.; Li, B.; Sun, Z.; Gao, S.; Cui, R. Modulation of Multiple Signaling Pathways of the Plant-Derived Natural Products in Cancer. Front. Oncol. 2019, 9, 1153. [Google Scholar] [CrossRef]
  16. Sharifi-Rad, J.; Ozleyen, A.; Boyunegmez Tumer, T.; Oluwaseun Adetunji, C.; El Omari, N.; Balahbib, A.; Taheri, Y.; Bouyahya, A.; Martorell, M.; Martins, N.; et al. Natural Products and Synthetic Analogs as a Source of Antitumor Drugs. Biomolecules 2019, 9, 679. [Google Scholar] [CrossRef]
  17. Ahmed, K.R.; Rahman, M.d.M.; Islam, M.d.N.; Fahim, M.d.M.H.; Rahman, M.A.; Kim, B. Antioxidants activities of phytochemicals perspective modulation of autophagy and apoptosis to treating cancer. Biomed. Pharmacother. 2024, 174, 116497. [Google Scholar] [CrossRef]
  18. de Oliveira Júnior, R.G.; Christiane Adrielly, A.F.; da Silva Almeida, J.R.G.; Grougnet, R.; Thiéry, V.; Picot, L. Sensitization of tumor cells to chemotherapy by natural products: A systematic review of preclinical data and molecular mechanisms. Fitoterapia 2018, 129, 383–400. [Google Scholar] [CrossRef]
  19. Talib, W.H.; Alsayed, A.R.; Barakat, M.; Abu-Taha, M.I.; Mahmod, A.I. Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2021, 9, 1353. [Google Scholar] [CrossRef]
  20. Zou, J.-Y.; Chen, Q.-L.; Luo, X.-C.; Damdinjav, D.; Abdelmohsen, U.R.; Li, H.-Y.; Battulga, T.; Chen, H.-B.; Wang, Y.-Q.; Zhang, J.-Y. Natural products reverse cancer multidrug resistance. Front. Pharmacol. 2024, 15, 1348076. [Google Scholar] [CrossRef]
  21. Cruz-Martins, N. Advances in Plants-Derived Bioactives for Cancer Treatment. Cells 2023, 12, 1112. [Google Scholar] [CrossRef]
  22. Yadav, J.; El Hassani, M.; Sodhi, J.; Lauschke, V.M.; Hartman, J.H.; Russell, L.E. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab. Rev. 2021, 53, 207–233. [Google Scholar] [CrossRef] [PubMed]
  23. Cheng, X.; Xie, Q.; Sun, Y. Advances in nanomaterial-based targeted drug delivery systems. Front. Bioeng. Biotechnol. 2023, 11, 1177151. [Google Scholar] [CrossRef] [PubMed]
  24. Islam, S.; Ahmed, M.M.S.; Islam, M.A.; Hossain, N.; Chowdhury, M.A. Advances in nanoparticles in targeted drug delivery—A review. Results Surf. Interfaces. 2025, 19, 100529. [Google Scholar] [CrossRef]
  25. Ammar, M.M.; Ali, R.; Abd Elaziz, N.A.; Habib, H.; Abbas, F.M.; Yassin, M.T.; Maniah, K.; Abdelaziz, R. Nanotechnology in oncology: Advances in biosynthesis, drug delivery, and theranostics. Discov. Oncol. 2025, 16, 1172. [Google Scholar] [CrossRef]
  26. Manna, D.; Sarkar, D. Multifunctional Role of Astrocyte Elevated Gene-1 (AEG-1) in Cancer: Focus on Drug Resistance. Cancers 2021, 13, 1792. [Google Scholar] [CrossRef]
  27. Wium, M.; Ajayi-Smith, A.F.; Paccez, J.D.; Zerbini, L.F. The Role of the Receptor Tyrosine Kinase Axl in Carcinogenesis and Development of Therapeutic Resistance: An Overview of Molecular Mechanisms and Future Applications. Cancers 2021, 13, 1521. [Google Scholar] [CrossRef]
  28. Li, X.; Huang, X.; Chang, M.; Lin, R.; Zhang, J.; Lu, Y. Updates on altered signaling pathways in tumor drug resistance. Vis. Cancer Med. 2024, 5, 6. [Google Scholar] [CrossRef]
  29. Garg, P.; Malhotra, J.; Kulkarni, P.; Horne, D.; Salgia, R.; Singhal, S.S. Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells. Cancers 2024, 16, 2478. [Google Scholar] [CrossRef]
  30. Xue, X.; Liang, X.-J. Overcoming drug efflux-based multidrug resistance in cancer with nanotechnology. Chin. J. Cancer 2012, 31, 100–109. [Google Scholar] [CrossRef]
  31. Honeywell, R.J.; Kathmann, I.; Giovannetti, E.; Tibaldi, C.; Smit, E.F.; Rovithi, M.N.; Verheul, H.M.W.; Peters, G.J. Epithelial Transfer of the Tyrosine Kinase Inhibitors Erlotinib, Gefitinib, Afatinib, Crizotinib, Sorafenib, Sunitinib, and Dasatinib: Implications for Clinical Resistance. Cancers 2020, 12, 3322. [Google Scholar] [CrossRef]
  32. Szczygieł, M.; Markiewicz, M.; Szafraniec, M.J.; Hojda, A.; Fiedor, L.; Urbanska, K. Systemic Mobilization of Breast Cancer Resistance Protein in Response to Oncogenic Stress. Cancers 2022, 14, 313. [Google Scholar] [CrossRef] [PubMed]
  33. Fontana, F.; Anselmi, M.; Limonta, P. Molecular Mechanisms of Cancer Drug Resistance: Emerging Biomarkers and Promising Targets to Overcome Tumor Progression. Cancers 2022, 14, 1614. [Google Scholar] [CrossRef] [PubMed]
  34. Doldi, V.; El Bezawy, R.; Zaffaroni, N. MicroRNAs as Epigenetic Determinants of Treatment Response and Potential Therapeutic Targets in Prostate Cancer. Cancers 2021, 13, 2380. [Google Scholar] [CrossRef] [PubMed]
  35. Seborova, K.; Vaclavikova, R.; Rob, L.; Soucek, P.; Vodicka, P. Non-Coding RNAs as Biomarkers of Tumor Progression and Metastatic Spread in Epithelial Ovarian Cancer. Cancers 2021, 13, 1839. [Google Scholar] [CrossRef]
  36. Ruiz-Pozo, V.A.; Guevara-Ramírez, P.; Paz-Cruz, E.; Tamayo-Trujillo, R.; Cadena-Ullauri, S.; Frias-Toral, E.; Simancas-Racines, D.; Altuna-Roshkova, Y.; Reytor-González, C.; Zambrano, A.K. The role of the Mediterranean diet in prediabetes management and prevention: A review of molecular mechanisms and clinical outcomes. Food Agric. Immunol. 2024, 35, 2398042. [Google Scholar] [CrossRef]
  37. Kar, A.; Agarwal, S.; Singh, A.; Bajaj, A.; Dasgupta, U. Insights into molecular mechanisms of chemotherapy resistance in cancer. Transl. Oncol. 2024, 42, 101901. [Google Scholar] [CrossRef]
  38. Kinnel, B.; Singh, S.K.; Oprea-Ilies, G.; Singh, R. Targeted Therapy and Mechanisms of Drug Resistance in Breast Cancer. Cancers 2023, 15, 1320. [Google Scholar] [CrossRef]
  39. Gu, Y.; Yang, R.; Zhang, Y.; Guo, M.; Takehiro, K.; Zhan, M.; Yang, L.; Wang, H. Molecular mechanisms and therapeutic strategies in overcoming chemotherapy resistance in cancer. Mol. Biomed. 2025, 6, 2. [Google Scholar] [CrossRef]
  40. La Rocca, A.; De Gregorio, V.; Lagreca, E.; Vecchione, R.; Netti, P.A.; Imparato, G. Colorectal Cancer Bioengineered Microtissues as a Model to Replicate Tumor-ECM Crosstalk and Assess Drug Delivery Systems In Vitro. Int. J. Mol. Sci. 2023, 24, 5678. [Google Scholar] [CrossRef]
  41. Marzagalli, M.; Fontana, F.; Raimondi, M.; Limonta, P. Cancer Stem Cells—Key Players in Tumor Relapse. Cancers 2021, 13, 376. [Google Scholar] [CrossRef]
  42. Salemme, V.; Centonze, G.; Avalle, L.; Natalini, D.; Piccolantonio, A.; Arina, P.; Morellato, A.; Ala, U.; Taverna, D.; Turco, E.; et al. The role of tumor microenvironment in drug resistance: Emerging technologies to unravel breast cancer heterogeneity. Front. Oncol. 2023, 13, 1170264. [Google Scholar] [CrossRef]
  43. Proietto, M.; Crippa, M.; Damiani, C.; Pasquale, V.; Sacco, E.; Vanoni, M.; Gilardi, M. Tumor heterogeneity: Preclinical models, emerging technologies, and future applications. Front. Oncol. 2023, 13, 1164535. [Google Scholar] [CrossRef]
  44. Zhu, L.; Jiang, M.; Wang, H.; Sun, H.; Zhu, J.; Zhao, W.; Fang, Q.; Yu, J.; Chen, P.; Wu, S.; et al. A narrative review of tumor heterogeneity and challenges to tumor drug therapy. Ann. Transl. Med. 2021, 9, 1351. [Google Scholar] [CrossRef]
  45. Chen, S.; Wang, M.; Lu, T.; Liu, Y.; Hong, W.; He, X.; Cheng, Y.; Liu, J.; Wei, Y.; Wei, X. JMJD6 in tumor-associated macrophage regulates macrophage polarization and cancer progression via STAT3/IL-10 axis. Oncogene 2023, 42, 2737–2750. [Google Scholar] [CrossRef] [PubMed]
  46. Tang, B.; Zhu, J.; Wang, Y.; Chen, W.; Fang, S.; Mao, W.; Xu, Z.; Yang, Y.; Weng, Q.; Zhao, Z.; et al. Targeted xCT-mediated Ferroptosis and Protumoral Polarization of Macrophages Is Effective against HCC and Enhances the Efficacy of the Anti-PD-1/L1 Response. Adv. Sci. 2023, 10, 2203973. [Google Scholar] [CrossRef] [PubMed]
  47. Li, Y.; Shen, Z.; Chai, Z.; Zhan, Y.; Zhang, Y.; Liu, Z.; Liu, Y.; Li, Z.; Lin, M.; Zhang, Z.; et al. Targeting MS4A4A on tumour-associated macrophages restores CD8+ T-cell-mediated antitumour immunity. Gut 2023, 72, 2307–2320. [Google Scholar] [CrossRef] [PubMed]
  48. Diaz Bessone, M.I.; Gattas, M.J.; Laporte, T.; Tanaka, M.; Simian, M. The Tumor Microenvironment as a Regulator of Endocrine Resistance in Breast Cancer. Front. Endocrinol. 2019, 10, 547. [Google Scholar] [CrossRef]
  49. Reytor-González, C.; Simancas-Racines, D.; Román-Galeano, N.M.; Campuzano-Donoso, M.; Carella, A.M.; Zambrano-Villacres, R.; Marinelli, T.; Coppola, L.; Marchetti, M.; Galasso, M.; et al. Obesity and breast cancer: Exploring the nexus of chronic inflammation, metabolic dysregulation, and nutritional strategies. Food Agric. Immunol. 2025, 36, 2521270. [Google Scholar] [CrossRef]
  50. Simancas-Racines, D.; Campuzano-Donoso, M.; Román-Galeano, N.M.; Zambrano-Villacres, R.; Memoli, P.; Verde, L.; Reytor-González, C.; Carbone, L. Obesity and endometrial cancer: Biological mechanisms, nutritional strategies, and clinical perspectives. Food Agric. Immunol. 2025, 36, 2510961. [Google Scholar] [CrossRef]
  51. Sarno, G.; Reytor-González, C.; Frias-Toral, E.; Campuzano-Donoso, M.; Katsanos, C.S.; Simancas-Racines, D. Navigating the weight: The impact of obesity on gastrointestinal cancer surgery and strategies for improved outcomes. Semin. Cancer Biol. 2025, 114, 138–149. [Google Scholar] [CrossRef]
  52. Sarno, G.; Simancas-Racines, D.; Gargiulo, A.; Tedesco, A.; Iacone, B.; Reytor-González, C.; Parise-Vasco, J.M.; Angamarca Iguago, J.; Sarno, S.; Frias-Toral, E.; et al. Impact of obesity on postoperative complications in colorectal cancer surgery: A systematic review and meta-analysis. Semin. Cancer Biol. 2025, 113, 176–189. [Google Scholar] [CrossRef]
  53. Simancas-Racines, D.; Annunziata, G.; Verde, L.; Fascì-Spurio, F.; Reytor-González, C.; Muscogiuri, G.; Frias-Toral, E.; Barrea, L. Nutritional Strategies for Battling Obesity-Linked Liver Disease: The Role of Medical Nutritional Therapy in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Management. Curr. Obes. Rep. 2025, 14, 7. [Google Scholar] [CrossRef] [PubMed]
  54. Reytor-González, C.; Annunziata, G.; Campuzano-Donoso, M.; Morales-López, T.; Basantes-Tituaña, C.; Fascì-Spurio, F.; Verde, L.; Muscogiuri, G.; Barrea, L.; Frias-Toral, E.; et al. Endocrinologist’s crucial role in metabolic dysfunction-associated steatotic liver disease: A comprehensive review. Minerva Endocrinol. 2025, 50, 209–226. [Google Scholar] [CrossRef] [PubMed]
  55. Reytor-González, C.; Parise-Vasco, J.M.; González, N.; Simancas-Racines, A.; Zambrano-Villacres, R.; Zambrano, A.K.; Simancas-Racines, D. Obesity and periodontitis: A comprehensive review of their interconnected pathophysiology and clinical implications. Front. Nutr. 2024, 11, 1440216. [Google Scholar] [CrossRef] [PubMed]
  56. Kobayashi, H.; Gieniec, K.A.; Lannagan, T.R.M.; Wang, T.; Asai, N.; Mizutani, Y.; Iida, T.; Ando, R.; Thomas, E.M.; Sakai, A.; et al. The Origin and Contribution of Cancer-Associated Fibroblasts in Colorectal Carcinogenesis. Gastroenterology 2022, 162, 890–906. [Google Scholar] [CrossRef]
  57. Krisnawan, V.E.; Stanley, J.A.; Schwarz, J.K.; DeNardo, D.G. Tumor Microenvironment as a Regulator of Radiation Therapy: New Insights into Stromal-Mediated Radioresistance. Cancers 2020, 12, 2916. [Google Scholar] [CrossRef]
  58. Liang, L.; Li, W.; Li, X.; Jin, X.; Liao, Q.; Li, Y.; Zhou, Y. ‘Reverse Warburg effect’ of cancer-associated fibroblasts (Review). Int. J. Oncol. 2022, 60, 67. [Google Scholar] [CrossRef]
  59. Zhao, Q.; Huang, L.; Qin, G.; Qiao, Y.; Ren, F.; Shen, C.; Wang, S.; Liu, S.; Lian, J.; Wang, D.; et al. Cancer-associated fibroblasts induce monocytic myeloid-derived suppressor cell generation via IL-6/exosomal miR-21-activated STAT3 signaling to promote cisplatin resistance in esophageal squamous cell carcinoma. Cancer Lett. 2021, 518, 35–48. [Google Scholar] [CrossRef]
  60. Xiang, H.; Ramil, C.P.; Hai, J.; Zhang, C.; Wang, H.; Watkins, A.A.; Afshar, R.; Georgiev, P.; Sze, M.A.; Song, X.S.; et al. Cancer-Associated Fibroblasts Promote Immunosuppression by Inducing ROS-Generating Monocytic MDSCs in Lung Squamous Cell Carcinoma. Cancer Immunol. Res. 2020, 8, 436–450. [Google Scholar] [CrossRef]
  61. Wang, W.-Z.; Cao, X.; Bian, L.; Gao, Y.; Yu, M.; Li, Y.-T.; Xu, J.-G.; Wang, Y.-H.; Yang, H.-F.; You, D.-Y.; et al. Analysis of mRNA-miRNA interaction network reveals the role of CAFs-derived exosomes in the immune regulation of oral squamous cell carcinoma. BMC Cancer 2023, 23, 591. [Google Scholar] [CrossRef] [PubMed]
  62. Miaomiao, S.; Xiaoqian, W.; Yuwei, S.; Chao, C.; Chenbo, Y.; Yinghao, L.; Yichen, H.; Jiao, S.; Kuisheng, C. Cancer-associated fibroblast-derived exosome microRNA-21 promotes angiogenesis in multiple myeloma. Sci. Rep. 2023, 13, 9671. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, Z.; Shang, J.; Yang, Q.; Dai, Z.; Liang, Y.; Lai, C.; Feng, T.; Zhong, D.; Zou, H.; Sun, L.; et al. Exosomes derived from human adipose mesenchymal stem cells ameliorate hepatic fibrosis by inhibiting PI3K/Akt/mTOR pathway and remodeling choline metabolism. J. Nanobiotechnol. 2023, 21, 29. [Google Scholar] [CrossRef]
  64. Zhang, Y.; Yin, C.; Wei, C.; Xia, S.; Qiao, Z.; Zhang, X.-W.; Yu, B.; Zhou, J.; Wang, R. Exosomal miR-625-3p secreted by cancer-associated fibroblasts in colorectal cancer promotes EMT and chemotherapeutic resistance by blocking the CELF2/WWOX pathway. Pharmacol. Res. 2022, 186, 106534. [Google Scholar] [CrossRef] [PubMed]
  65. Huang, H.; Wang, Z.; Zhang, Y.; Pradhan, R.N.; Ganguly, D.; Chandra, R.; Murimwa, G.; Wright, S.; Gu, X.; Maddipati, R.; et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 2022, 40, 656–673.e7. [Google Scholar] [CrossRef]
  66. Song, M.; He, J.; Pan, Q.-Z.; Yang, J.; Zhao, J.; Zhang, Y.-J.; Huang, Y.; Tang, Y.; Wang, Q.; He, J.; et al. Cancer-Associated Fibroblast-Mediated Cellular Crosstalk Supports Hepatocellular Carcinoma Progression. Hepatology 2021, 73, 1717–1735. [Google Scholar] [CrossRef]
  67. Feng, B.; Wu, J.; Shen, B.; Jiang, F.; Feng, J. Cancer-associated fibroblasts and resistance to anticancer therapies: Status, mechanisms, and countermeasures. Cancer Cell Int. 2022, 22, 166. [Google Scholar] [CrossRef]
  68. Yu, W.; Lei, Q.; Yang, L.; Qin, G.; Liu, S.; Wang, D.; Ping, Y.; Zhang, Y. Contradictory roles of lipid metabolism in immune response within the tumor microenvironment. J. Hematol. Oncol. 2021, 14, 187. [Google Scholar] [CrossRef]
  69. Kennel, K.B.; Bozlar, M.; De Valk, A.F.; Greten, F.R. Cancer-Associated Fibroblasts in Inflammation and Antitumor Immunity. Clin. Cancer Res. 2023, 29, 1009–1016. [Google Scholar] [CrossRef]
  70. Galbo, P.M.; Zang, X.; Zheng, D. Molecular Features of Cancer-associated Fibroblast Subtypes and their Implication on Cancer Pathogenesis, Prognosis, and Immunotherapy Resistance. Clin. Cancer Res. 2021, 27, 2636–2647. [Google Scholar] [CrossRef]
  71. Glabman, R.A.; Choyke, P.L.; Sato, N. Cancer-Associated Fibroblasts: Tumorigenicity and Targeting for Cancer Therapy. Cancers 2022, 14, 3906. [Google Scholar] [CrossRef]
  72. Bhattacharjee, S.; Hamberger, F.; Ravichandra, A.; Miller, M.; Nair, A.; Affo, S.; Filliol, A.; Chin, L.; Savage, T.M.; Yin, D.; et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J. Clin. Investig. 2021, 131, e146987. [Google Scholar] [CrossRef] [PubMed]
  73. Li, X.; Sun, Z.; Peng, G.; Xiao, Y.; Guo, J.; Wu, B.; Li, X.; Zhou, W.; Li, J.; Li, Z.; et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics 2022, 12, 620–638. [Google Scholar] [CrossRef] [PubMed]
  74. Vimalraj, S. A concise review of VEGF, PDGF, FGF, Notch, angiopoietin, and HGF signalling in tumor angiogenesis with a focus on alternative approaches and future directions. Int. J. Biol. Macromol. 2022, 221, 1428–1438. [Google Scholar] [CrossRef] [PubMed]
  75. Magar, A.G.; Morya, V.K.; Kwak, M.K.; Oh, J.U.; Noh, K.C. A Molecular Perspective on HIF-1α and Angiogenic Stimulator Networks and Their Role in Solid Tumors: An Update. Int. J. Mol. Sci. 2024, 25, 3313. [Google Scholar] [CrossRef] [PubMed]
  76. Malekan, M.; Ebrahimzadeh, M.A.; Sheida, F. The role of Hypoxia-Inducible Factor-1alpha and its signaling in melanoma. Biomed. Pharmacother. 2021, 141, 111873. [Google Scholar] [CrossRef]
  77. Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2020, 6, 160. [Google Scholar] [CrossRef]
  78. Luo, Z.; Tian, M.; Yang, G.; Tan, Q.; Chen, Y.; Li, G.; Zhang, Q.; Li, Y.; Wan, P.; Wu, J. Hypoxia signaling in human health and diseases: Implications and prospects for therapeutics. Signal Transduct. Target. Ther. 2022, 7, 218. [Google Scholar] [CrossRef]
  79. Lin, S.; Chai, Y.; Zheng, X.; Xu, X. The role of HIF in angiogenesis, lymphangiogenesis, and tumor microenvironment in urological cancers. Mol. Biol. Rep. 2024, 51, 14. [Google Scholar] [CrossRef]
  80. Manuelli, V.; Pecorari, C.; Filomeni, G.; Zito, E. Regulation of redox signaling in HIF-1-dependent tumor angiogenesis. FEBS J. 2022, 289, 5413–5425. [Google Scholar] [CrossRef]
  81. Bakleh, M.Z.; Al Haj Zen, A. The Distinct Role of HIF-1α and HIF-2α in Hypoxia and Angiogenesis. Cells 2025, 14, 673. [Google Scholar] [CrossRef]
  82. Jiang, Y.; Zhang, H.; Wang, J.; Liu, Y.; Luo, T.; Hua, H. Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J. Hematol. Oncol. 2022, 15, 34. [Google Scholar] [CrossRef]
  83. Simancas-Racines, D.; Reytor-González, C.; Frias-Toral, E.; Katsanos, C.S.; Hidalgo, R. Weighty matters: Unraveling the impact of obesity on colorectal cancer and nutritional interventions. Semin. Cancer Biol. 2025, 114, 29–40. [Google Scholar] [CrossRef]
  84. Pandey, K.; Umar, S. Microbiome in drug resistance to colon cancer. Curr. Opin. Physiol. 2021, 23, 100472. [Google Scholar] [CrossRef]
  85. Veziant, J.; Villéger, R.; Barnich, N.; Bonnet, M. Gut Microbiota as Potential Biomarker and/or Therapeutic Target to Improve the Management of Cancer: Focus on Colibactin-Producing Escherichia coli in Colorectal Cancer. Cancers 2021, 13, 2215. [Google Scholar] [CrossRef] [PubMed]
  86. Belotti, D.; Pinessi, D.; Taraboletti, G. Alternative Vascularization Mechanisms in Tumor Resistance to Therapy. Cancers 2021, 13, 1912. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, X.; Liu, Y.; Qi, Y.; Huang, Y.; Hu, F.; Dong, F.; Shu, K.; Lei, T. Signal Pathways Involved in the Interaction Between Tumor-Associated Macrophages/TAMs and Glioblastoma Cells. Front. Oncol. 2022, 12, 822085. [Google Scholar] [CrossRef]
  88. Downs-Canner, S.M.; Meier, J.; Vincent, B.G.; Serody, J.S. B Cell Function in the Tumor Microenvironment. Annu. Rev. Immunol. 2022, 40, 169–193. [Google Scholar] [CrossRef]
  89. Chmiel, P.; Rychcik-Pazyrska, P.; Stec, R. Defining Tumor Microenvironment as a Possible Target for Effective GEP-NENs Immunotherapy—A Systematic Review. Cancers 2023, 15, 5232. [Google Scholar] [CrossRef]
  90. Zhao, X.; Zhu, Y.; He, Y.; Gu, W.; Zhou, Q.; Jin, B.; Chen, S.; Lin, H. Unraveling the immune evasion mechanisms in the tumor microenvironment of head and neck squamous cell carcinoma. Front. Immunol. 2025, 16, 1597202. [Google Scholar] [CrossRef]
  91. Kim, S.K.; Cho, S.W. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment. Front. Pharmacol. 2022, 13, 868695. [Google Scholar] [CrossRef] [PubMed]
  92. Ghebremedhin, A.; Athavale, D.; Zhang, Y.; Yao, X.; Balch, C.; Song, S. Tumor-Associated Macrophages as Major Immunosuppressive Cells in the Tumor Microenvironment. Cancers 2024, 16, 3410. [Google Scholar] [CrossRef] [PubMed]
  93. Shah, D.D.; Chorawala, M.R.; Raghani, N.R.; Patel, R.; Fareed, M.; Kashid, V.A.; Prajapati, B.G. Tumor microenvironment: Recent advances in understanding and its role in modulating cancer therapies. Med. Oncol. 2025, 42, 117. [Google Scholar] [CrossRef]
  94. Zhang, H.; Dai, Z.; Wu, W.; Wang, Z.; Zhang, N.; Zhang, L.; Zeng, W.-J.; Liu, Z.; Cheng, Q. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J. Exp. Clin. Cancer Res. 2021, 40, 184. [Google Scholar] [CrossRef]
  95. Wang, H.; Tian, T.; Zhang, J. Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): From Mechanism to Therapy and Prognosis. Int. J. Mol. Sci. 2021, 22, 8470. [Google Scholar] [CrossRef]
  96. Bigos, K.J.A.; Quiles, C.G.; Lunj, S.; Smith, D.J.; Krause, M.; Troost, E.G.C.; West, C.M.; Hoskin, P.; Choudhury, A. Tumour response to hypoxia: Understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front. Oncol. 2024, 14, 1331355. [Google Scholar] [CrossRef]
  97. Rømer, A.M.A.; Thorseth, M.-L.; Madsen, D.H. Immune Modulatory Properties of Collagen in Cancer. Front. Immunol. 2021, 12, 791453. [Google Scholar] [CrossRef]
  98. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  99. Zaghdoudi, S.; Decaup, E.; Belhabib, I.; Samain, R.; Cassant-Sourdy, S.; Rochotte, J.; Brunel, A.; Schlaepfer, D.; Cros, J.; Neuzillet, C.; et al. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol. Med. 2020, 12, e12010. [Google Scholar] [CrossRef]
  100. Whisner, C.M.; Athena Aktipis, C. The Role of the Microbiome in Cancer Initiation and Progression: How Microbes and Cancer Cells Utilize Excess Energy and Promote One Another’s Growth. Curr. Nutr. Rep. 2019, 8, 42–51. [Google Scholar] [CrossRef]
  101. Ilerhunmwuwa, N.P.; Abdul Khader, A.H.S.; Smith, C.; Cliff, E.R.S.; Booth, C.M.; Hottel, E.; Aziz, M.; Lee-Smith, W.; Goodman, A.; Chakraborty, R.; et al. Dietary interventions in cancer: A systematic review of all randomized controlled trials. JNCI J. Natl. Cancer Inst. 2024, 116, 1026–1034. [Google Scholar] [CrossRef] [PubMed]
  102. Martínez-Garay, C.; Djouder, N. Dietary interventions and precision nutrition in cancer therapy. Trends Mol. Med. 2023, 29, 489–511. [Google Scholar] [CrossRef] [PubMed]
  103. Muscogiuri, G.; Verde, L.; Frias-Toral, E.; Reytor-González, C.; Annunziata, G.; Proganò, M.; Savastano, S.; Simancas-Racines, D.; Colao, A.; Barrea, L. Weight loss, changes in body composition and inflammatory status after a very low-energy ketogenic therapy (VLEKT): Does gender matter? J. Transl. Med. 2024, 22, 949. [Google Scholar] [CrossRef] [PubMed]
  104. Reytor-González, C.; Zambrano, A.K.; Frias-Toral, E.; Campuzano-Donoso, M.; Simancas-Racines, D. Mediterranean diet and breast cancer: A narrative review. Medwave 2025, 25, e3027. [Google Scholar] [CrossRef]
  105. Simancas-Racines, D.; Reytor-González, C.; Zambrano, A.K.; Annunziata, G.; Carella, A.M.; Verde, L.; Frias-Toral, E.; Guerra, C.V.; Hidalgo, R. Unlocking the potential: Very-low-energy ketogenic therapy in obesity-related disorders. Food Agric. Immunol. 2025, 36, 2442368. [Google Scholar] [CrossRef]
  106. Reytor-González, C.; Simancas-Racines, D.; Campuzano-Donoso, M.; Castano Jimenez, J.; Román-Galeano, N.M.; Sarno, G.; Frias-Toral, E. Harnessing nutrition to combat MASLD: A comprehensive guide to food-based therapeutic strategies. Food Agric. Immunol. 2025, 36, 2496499. [Google Scholar] [CrossRef]
  107. Reytor-González, C.; Frias-Toral, E.; Nuñez-Vásquez, C.; Parise-Vasco, J.M.; Zambrano-Villacres, R.; Simancas-Racines, D.; Schiavo, L. Preventing and Managing Pre- and Postoperative Micronutrient Deficiencies: A Vital Component of Long-Term Success in Bariatric Surgery. Nutrients 2025, 17, 741. [Google Scholar] [CrossRef]
  108. Simancas-Racines, D.; Frias-Toral, E.; Campuzano-Donoso, M.; Ramos-Sarmiento, D.; Zambrano-Villacres, R.; Reytor-González, C.; Schiavo, L. Preoperative Nutrition in Bariatric Surgery: A Narrative Review on Enhancing Surgical Success and Patient Outcomes. Nutrients 2025, 17, 566. [Google Scholar] [CrossRef]
  109. Reytor-González, C.; Simancas-Racines, D.; Román-Galeano, N.M.; Annunziata, G.; Galasso, M.; Zambrano-Villacres, R.; Verde, L.; Muscogiuri, G.; Frias-Toral, E.; Barrea, L. Chrononutrition and Energy Balance: How Meal Timing and Circadian Rhythms Shape Weight Regulation and Metabolic Health. Nutrients 2025, 17, 2135. [Google Scholar] [CrossRef]
  110. Ubago-Guisado, E.; Rodríguez-Barranco, M.; Ching-López, A.; Petrova, D.; Molina-Montes, E.; Amiano, P.; Barricarte-Gurrea, A.; Chirlaque, M.-D.; Agudo, A.; Sánchez, M.-J. Evidence Update on the Relationship between Diet and the Most Common Cancers from the European Prospective Investigation into Cancer and Nutrition (EPIC) Study: A Systematic Review. Nutrients 2021, 13, 3582. [Google Scholar] [CrossRef]
  111. Ruban, M.; Pozhidaeva, E.; Bolotina, L.; Kaprin, A. The Role of Diet and Nutrition in Cancer Development and Management: From Molecular Mechanisms to Personalized Interventions. Foods 2025, 14, 1788. [Google Scholar] [CrossRef]
  112. Golonko, A.; Pienkowski, T.; Swislocka, R.; Orzechowska, S.; Marszalek, K.; Szczerbinski, L.; Swiergiel, A.H.; Lewandowski, W. Dietary factors and their influence on immunotherapy strategies in oncology: A comprehensive review. Cell Death Dis. 2024, 15, 254. [Google Scholar] [CrossRef]
  113. Bose, S.; Allen, A.E.; Locasale, J.W. The Molecular Link from Diet to Cancer Cell Metabolism. Mol. Cell 2020, 78, 1034–1044. [Google Scholar] [CrossRef] [PubMed]
  114. Mercier, B.D.; Tizpa, E.; Philip, E.J.; Feng, Q.; Huang, Z.; Thomas, R.M.; Pal, S.K.; Dorff, T.B.; Li, Y.R. Dietary Interventions in Cancer Treatment and Response: A Comprehensive Review. Cancers 2022, 14, 5149. [Google Scholar] [CrossRef] [PubMed]
  115. Wyer, S.; Townsend, D.M.; Ye, Z.; Kourtidis, A.; Choo, Y.-M.; Branco de Barros, A.L.; Donia, M.S.; Hamann, M.T. Recent advances and limitations in the application of kahalalides for the control of cancer. Biomed. Pharmacother. 2022, 148, 112676. [Google Scholar] [CrossRef] [PubMed]
  116. Zhai, K.; Brockmüller, A.; Kubatka, P.; Shakibaei, M.; Büsselberg, D. Curcumin’s Beneficial Effects on Neuroblastoma: Mechanisms, Challenges, and Potential Solutions. Biomolecules 2020, 10, 1469. [Google Scholar] [CrossRef] [PubMed]
  117. Augimeri, G.; Montalto, F.I.; Giordano, C.; Barone, I.; Lanzino, M.; Catalano, S.; Andò, S.; De Amicis, F.; Bonofiglio, D. Nutraceuticals in the Mediterranean Diet: Potential Avenues for Breast Cancer Treatment. Nutrients 2021, 13, 2557. [Google Scholar] [CrossRef]
  118. Granja, A.; Frias, I.; Neves, A.R.; Pinheiro, M.; Reis, S. Therapeutic Potential of Epigallocatechin Gallate Nanodelivery Systems. Biomed. Res. Int. 2017, 2017, 5813793. [Google Scholar] [CrossRef]
  119. Bhavsar, P.; Jha, L.L.; Bera, K.; Patel, S. Phytoconstituents Loaded Liposomes Fabricated Based on Box Behnken Design for Metabolic Syndrome In Vitro and In Vivo Characterization. J. Nat. Remedies 2023, 23, 1035–1052. [Google Scholar] [CrossRef]
  120. Gündoğdu, Ö.; Şen, S.; Kishali, N. Synthesis of Bicyclic Pyrrolidine Derivatives and their Photoluminescent Properties. Biomed. J. Sci. Tech. Res. 2023, 49. [Google Scholar] [CrossRef]
  121. Lu, J.; Ma, Y.; Wu, J.; Huang, H.; Wang, X.; Chen, Z.; Chen, J.; He, H.; Huang, C. A review for the neuroprotective effects of andrographolide in the central nervous system. Biomed. Pharmacother. 2019, 117, 109078. [Google Scholar] [CrossRef]
  122. Principe, D.R.; Underwood, P.W.; Korc, M.; Trevino, J.G.; Munshi, H.G.; Rana, A. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma and Barriers to Therapeutic Efficacy. Front. Oncol. 2021, 11, 688377. [Google Scholar] [CrossRef]
  123. Murthy, H.N.; Joseph, K.S.; Paek, K.Y.; Park, S.Y. Production of anthraquinones from cell and organ cultures of Morinda species. Appl. Microbiol. Biotechnol. 2023, 107, 2061–2071. [Google Scholar] [CrossRef] [PubMed]
  124. Shen, Y.A.; Lin, C.H.; Chi, W.H.; Wang, C.Y.; Hsieh, Y.T.; Wei, Y.H.; Chen, Y.J. Resveratrol Impedes the Stemness, Epithelial-Mesenchymal Transition, and Metabolic Reprogramming of Cancer Stem Cells in Nasopharyngeal Carcinoma through p53 Activation. Evid.-Based Complement. Altern. Med. 2013, 2013, 590393. [Google Scholar] [CrossRef] [PubMed]
  125. Saiful Hakim, A.R.; Chee, C.F.; Wong, T.W.; Abu Kasim, N.H.; Nasruddin, N.S.; Yazid, F. Exploring resveratrol’s inhibitory potential on lung cancer stem cells: A scoping review of mechanistic pathways across cancer models. Med. Oncol. 2025, 42, 318. [Google Scholar] [CrossRef] [PubMed]
  126. Capasso, L.; De Masi, L.; Sirignano, C.; Maresca, V.; Basile, A.; Nebbioso, A.; Rigano, D.; Bontempo, P. Epigallocatechin Gallate (EGCG): Pharmacological Properties, Biological Activities and Therapeutic Potential. Molecules 2025, 30, 654. [Google Scholar] [CrossRef]
  127. Banyal, A.; Tiwari, S.; Sharma, A.; Chanana, I.; Patel, S.K.S.; Kulshrestha, S.; Kumar, P. Vinca alkaloids as a potential cancer therapeutics: Recent update and future challenges. 3 Biotech 2023, 13, 211. [Google Scholar] [CrossRef]
  128. Bozdaganyan, M.; Fedorov, V.; Kholina, E.; Kovalenko, I.; Gudimchuk, N.; Orekhov, P. Exploring tubulin-paclitaxel binding modes through extensive molecular dynamics simulations. Sci. Rep. 2025, 15, 8378. [Google Scholar] [CrossRef]
  129. Ferdous, K.A.; Jansen, J.; Amjad, E.; Pray, E.; Bloch, R.; Benoit, A.; Callahan, M.; Park, H.-A. Mitochondrial protective potential of fucoxanthin in brain disorders. J. Nutr. Sci. 2024, 13, e21. [Google Scholar] [CrossRef]
  130. Ahmed, S.A.; Mendonca, P.; Elhag, R.; Soliman, K.F.A. Anticancer Effects of Fucoxanthin through Cell Cycle Arrest, Apoptosis Induction, Angiogenesis Inhibition, and Autophagy Modulation. Int. J. Mol. Sci. 2022, 23, 16091. [Google Scholar] [CrossRef]
  131. Pecoraro, C.; Terrana, F.; Panzeca, G.; Parrino, B.; Cascioferro, S.; Diana, P.; Giovannetti, E.; Carbone, D. Nortopsentins as Leads from Marine Organisms for Anticancer and Anti-Inflammatory Agent Development. Molecules 2023, 28, 6450. [Google Scholar] [CrossRef] [PubMed]
  132. Zhao, L.; Zheng, L. A Review on Bioactive Anthraquinone and Derivatives as the Regulators for ROS. Molecules 2023, 28, 8139. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, D.; Wang, X.-H.; Yu, X.; Cao, F.; Cai, X.; Chen, P.; Li, M.; Feng, Y.; Li, H.; Wang, X. Pharmacokinetics of Anthraquinones from Medicinal Plants. Front. Pharmacol. 2021, 12, 638993. [Google Scholar] [CrossRef] [PubMed]
  134. Ochoa-Sanchez, A.; Sahare, P.; Pathak, S.; Banerjee, A.; Estevez, M.; Duttaroy, A.K.; Luna-Bárcenas, G.; Paul, S. Evaluation of the synergistic effects of curcumin-resveratrol co-loaded biogenic silica on colorectal cancer cells. Front. Pharmacol. 2024, 15, 1341773. [Google Scholar] [CrossRef]
  135. Ciuca, M.D.; Racovita, R.C. Curcumin: Overview of Extraction Methods, Health Benefits, and Encapsulation and Delivery Using Microemulsions and Nanoemulsions. Int. J. Mol. Sci. 2023, 24, 8874. [Google Scholar] [CrossRef]
  136. Shaharudin, N.S.; Surindar Singh, G.K.; Kek, T.; Sultan, S. Targeting signaling pathways with andrographolide in cancer therapy (Review). Mol. Clin. Oncol. 2024, 21, 81. [Google Scholar] [CrossRef]
  137. Simancas-Racines, D.; Román-Galeano, N.M.; Verde, L.; Annunziata, G.; Marchetti, M.; Matos, A.; Campuzano-Donoso, M.; Reytor-González, C.; Muscogiuri, G.; Barrea, L.; et al. Targeting Cytokine Dysregulation in Psoriasis: The Role of Dietary Interventions in Modulating the Immune Response. Int. J. Mol. Sci. 2025, 26, 2895. [Google Scholar] [CrossRef]
  138. Guo, H.; Liu, Y.; Wan, T.; Song, D.; Palanisamy, C.P.; Geng, J.; Pei, J.; Özmen, S.; Abd El-Aty, A.M. Toward personalized cancer management: Role of precision nutrition–diet interventions. J. Funct. Foods 2024, 123, 106584. [Google Scholar] [CrossRef]
  139. Pisoschi, A.M.; Iordache, F.; Stanca, L.; Cimpeanu, C.; Furnaris, F.; Geicu, O.I.; Bilteanu, L.; Serban, A.I. Comprehensive and critical view on the anti-inflammatory and immunomodulatory role of natural phenolic antioxidants. Eur. J. Med. Chem. 2024, 265, 116075. [Google Scholar] [CrossRef]
  140. Jantan, I.; Haque, M.d.A.; Arshad, L.; Harikrishnan, H.; Septama, A.W.; Mohamed-Hussein, Z.-A. Dietary polyphenols suppress chronic inflammation by modulation of multiple inflammation-associated cell signaling pathways. J. Nutr. Biochem. 2021, 93, 108634. [Google Scholar] [CrossRef]
  141. Mileo, A.M.; Nisticò, P.; Miccadei, S. Polyphenols: Immunomodulatory and Therapeutic Implication in Colorectal Cancer. Front. Immunol. 2019, 10, 729. [Google Scholar] [CrossRef]
  142. Zoi, V.; Galani, V.; Lianos, G.D.; Voulgaris, S.; Kyritsis, A.P.; Alexiou, G.A. The Role of Curcumin in Cancer Treatment. Biomedicines 2021, 9, 1086. [Google Scholar] [CrossRef] [PubMed]
  143. Xu, L.; Zhang, Y.; Tian, K.; Chen, X.; Zhang, R.; Mu, X.; Wu, Y.; Wang, D.; Wang, S.; Liu, F.; et al. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. J. Exp. Clin. Cancer Res. 2018, 37, 261. [Google Scholar] [CrossRef] [PubMed]
  144. Huang, X.; Wang, Y.; Yang, W.; Dong, J.; Li, L. Regulation of dietary polyphenols on cancer cell pyroptosis and the tumor immune microenvironment. Front. Nutr. 2022, 9, 974896. [Google Scholar] [CrossRef] [PubMed]
  145. Golmohammadi, M.; Zamanian, M.Y.; Al-Ani, A.M.; Jabbar, T.L.; Kareem, A.K.; Aghaei, Z.H.; Tahernia, H.; Hjazi, A.; Jissir, S.A.; Hakimizadeh, E. Targeting STAT3 signaling pathway by curcumin and its analogues for breast cancer: A narrative review. Anim. Model. Exp. Med. 2024, 7, 853–867. [Google Scholar] [CrossRef]
  146. Kötting, C.; Hofmann, L.; Lotfi, R.; Engelhardt, D.; Laban, S.; Schuler, P.J.; Hoffmann, T.K.; Brunner, C.; Theodoraki, M.N. Immune-Stimulatory Effects of Curcumin on the Tumor Microenvironment in Head and Neck Squamous Cell Carcinoma. Cancers 2021, 13, 1335. [Google Scholar] [CrossRef]
  147. Ameer, S.F.; Mohamed, M.Y.; Elzubair, Q.A.; Sharif, E.A.M.; Ibrahim, W.N. Curcumin as a novel therapeutic candidate for cancer: Can this natural compound revolutionize cancer treatment? Front. Oncol. 2024, 14, 1438040. [Google Scholar] [CrossRef]
  148. Wu, R.; Wang, L.; Yin, R.; Hudlikar, R.; Li, S.; Kuo, H.D.; Peter, R.; Sargsyan, D.; Guo, Y.; Liu, X.; et al. Epigenetics/epigenomics and prevention by curcumin of early stages of inflammatory-driven colon cancer. Mol. Carcinog. 2020, 59, 227–236. [Google Scholar] [CrossRef]
  149. Ming, T.; Tao, Q.; Tang, S.; Zhao, H.; Yang, H.; Liu, M.; Ren, S.; Xu, H. Curcumin: An epigenetic regulator and its application in cancer. Biomed. Pharmacother. 2022, 156, 113956. [Google Scholar] [CrossRef]
  150. Cacciola, N.A.; Cuciniello, R.; Petillo, G.D.; Piccioni, M.; Filosa, S.; Crispi, S. An Overview of the Enhanced Effects of Curcumin and Chemotherapeutic Agents in Combined Cancer Treatments. Int. J. Mol. Sci. 2023, 24, 12587. [Google Scholar] [CrossRef]
  151. Qian, W.; Xiao, Q.; Wang, L.; Qin, T.; Xiao, Y.; Li, J.; Yue, Y.; Zhou, C.; Duan, W.; Ma, Q.; et al. Resveratrol slows the tumourigenesis of pancreatic cancer by inhibiting NFκB activation. Biomed. Pharmacother. 2020, 127, 110116. [Google Scholar] [CrossRef] [PubMed]
  152. Buhrmann, C.; Yazdi, M.; Bashiri Dezfouli, A.; Samani Sahraneshin, F.; Ebrahimi, S.M.; Hamidollah Ghaffari, S.; Yaghmaie, M.; Barin, A.; Shakibaei, M.; Shayan, P. Significant decrease in the viability and tumor stem cell marker expression in tumor cell lines treated with curcumin. J. Herb. Med. 2020, 22, 100339. [Google Scholar] [CrossRef]
  153. Hedayati, N.; Safari, M.H.; Milasi, Y.E.; Kahkesh, S.; Farahani, N.; Khoshnazar, S.M.; Dorostgou, Z.; Alaei, E.; Alimohammadi, M.; Rahimzadeh, P.; et al. Modulation of the PI3K/Akt signaling pathway by resveratrol in cancer: Molecular mechanisms and therapeutic opportunity. Discov. Oncol. 2025, 16, 669. [Google Scholar] [CrossRef] [PubMed]
  154. Xie, C.; Liang, C.; Wang, R.; Yi, K.; Zhou, X.; Li, X.; Chen, Y.; Miao, D.; Zhong, C.; Zhu, J. Resveratrol suppresses lung cancer by targeting cancer stem-like cells and regulating tumor microenvironment. J. Nutr. Biochem. 2023, 112, 109211. [Google Scholar] [CrossRef]
  155. Thongchot, S.; Ferraresi, A.; Vidoni, C.; Salwa, A.; Vallino, L.; Kittirat, Y.; Loilome, W.; Namwat, N.; Isidoro, C. Preclinical evidence for preventive and curative effects of resveratrol on xenograft cholangiocarcinogenesis. Cancer Lett. 2024, 582, 216589. [Google Scholar] [CrossRef]
  156. Chitcholtan, K.; Singh, M.; Tino, A.; Garrill, A.; Sykes, P. Effects of Resveratrol on In Vivo Ovarian Cancer Cells Implanted on the Chorioallantoic Membrane (CAM) of a Chicken Embryo Model. Int. J. Mol. Sci. 2024, 25, 4374. [Google Scholar] [CrossRef]
  157. Guo, K.; Feng, Y.; Zheng, X.; Sun, L.; Wasan, H.S.; Ruan, S.; Chen, Y.-J. Resveratrol and Its Analogs: Potent Agents to Reverse Epithelial-to-Mesenchymal Transition in Tumors. Front. Oncol. 2021, 11, 644134. [Google Scholar] [CrossRef]
  158. Ribeiro, E.; Vale, N. The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens. Targets 2024, 2, 307–326. [Google Scholar] [CrossRef]
  159. Wang, Z.A.; Hsu, W.; Liu, W.R. Role of SIRT1 in Epigenetics. In Handbook of Nutrition, Diet, and Epigenetics; Springer International Publishing: Cham, Switzerland, 2019; pp. 311–329. [Google Scholar] [CrossRef]
  160. Marín, V.; Burgos, V.; Pérez, R.; Maria, D.A.; Pardi, P.; Paz, C. The Potential Role of Epigallocatechin-3-Gallate (EGCG) in Breast Cancer Treatment. Int. J. Mol. Sci. 2023, 24, 10737. [Google Scholar] [CrossRef]
  161. Zhou, J.; Lin, H.; Xu, P.; Yao, L.; Xie, Q.; Mao, L.; Wang, Y. Matcha green tea prevents obesity-induced hypothalamic inflammation via suppressing the JAK2/STAT3 signaling pathway. Food Funct. 2020, 11, 8987–8995. [Google Scholar] [CrossRef]
  162. Kciuk, M.; Alam, M.; Ali, N.; Rashid, S.; Głowacka, P.; Sundaraj, R.; Celik, I.; Yahya, E.B.; Dubey, A.; Zerroug, E.; et al. Epigallocatechin-3-Gallate Therapeutic Potential in Cancer: Mechanism of Action and Clinical Implications. Molecules 2023, 28, 5246. [Google Scholar] [CrossRef]
  163. Sheng, J.; Shi, W.; Guo, H.; Long, W.; Wang, Y.; Qi, J.; Celik, I.; Yahya, E.B.; Dubey, A.; Zerroug, E.; et al. The Inhibitory Effect of (−)-Epigallocatechin-3-Gallate on Breast Cancer Progression via Reducing SCUBE2 Methylation and DNMT Activity. Molecules 2019, 24, 2899. [Google Scholar] [CrossRef]
  164. Kamboj, N.; Sharma, S.; Kumar, R. Neuroprotective insights into epigallocatechin gallate (EGCG) for neurodegenerative disorders. Explor. Neurosci. 2025, 4, 100673. [Google Scholar] [CrossRef]
  165. Bontempo, P.; Capasso, L.; De Masi, L.; Nebbioso, A.; Rigano, D. Therapeutic Potential of Natural Compounds Acting through Epigenetic Mechanisms in Cardiovascular Diseases: Current Findings and Future Directions. Nutrients 2024, 16, 2399. [Google Scholar] [CrossRef]
  166. Xu, P.; Yan, F.; Zhao, Y.; Chen, X.; Sun, S.; Wang, Y.; Ying, L. Green Tea Polyphenol EGCG Attenuates MDSCs-mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine Breast Cancer Model. Nutrients 2020, 12, 1042. [Google Scholar] [CrossRef] [PubMed]
  167. Reytor-González, C.; Simancas-Racines, D.; Jiménez-Flores, E.; Campuzano-Donoso, M.; Carella, A.M.; Coppola, L.; Marchetti, M.; Zambrano-Villacres, R.; Sarno, G. Oesophageal adenocarcinoma, obesity, and cancer: The role of nutrition in prevention and management. Food Agric. Immunol. 2025, 36, 2510951. [Google Scholar] [CrossRef]
  168. Nadalin, P.; Kim, Y.G.; Park, S.U. Recent Studies on Berberine and Its Biological and Pharmacological Activities. EXCLI J. 2023, 22, 315–328, Leibniz Research Centre for Working Environment and Human Factors. [Google Scholar] [CrossRef]
  169. Almatroodi, S.A.; Alsahli, M.A.; Rahmani, A.H. Berberine: An Important Emphasis on Its Anticancer Effects through Modulation of Various Cell Signaling Pathways. Molecules 2022, 27, 5889. [Google Scholar] [CrossRef]
  170. Habtemariam, S. Recent Advances in Berberine Inspired Anticancer Approaches: From Drug Combination to Novel Formulation Technology and Derivatization. Molecules 2020, 25, 1426. [Google Scholar] [CrossRef]
  171. Sajeev, A.; Sailo, B.; Unnikrishnan, J.; Talukdar, A.; Alqahtani, M.S.; Abbas, M.; Alqahtani, A.; Sethi, G.; Kunnumakkara, A.B. Unlocking the potential of Berberine: Advancing cancer therapy through chemosensitization and combination treatments. Cancer Lett. 2024, 597, 217019. [Google Scholar] [CrossRef]
  172. Kou, Y.; Tong, B.; Wu, W.; Liao, X.; Zhao, M. Berberine Improves Chemo-Sensitivity to Cisplatin by Enhancing Cell Apoptosis and Repressing PI3K/AKT/mTOR Signaling Pathway in Gastric Cancer. Front. Pharmacol. 2020, 11, 616251. [Google Scholar] [CrossRef]
  173. Rozwadowska, P.; Bator, P.; Razik, M.; Ramian, J.; Rybak, J.; Magiera, B.; Magiera, K.; Razik, W. Anticancer properties of berberine—Analysis of the latest reports. J. Educ. Health Sport 2024, 61, 73–86. [Google Scholar] [CrossRef]
  174. Zhang, C.; Sheng, J.; Li, G.; Zhao, L.; Wang, Y.; Yang, W.; Yao, X.; Sun, L.; Zhang, Z.; Cui, R. Effects of Berberine and Its Derivatives on Cancer: A Systems Pharmacology Review. Front. Pharmacol. 2020, 10, 1461. [Google Scholar] [CrossRef]
  175. Tong, Y.; Zhang, J.; Sun, N.; Wang, X.-M.; Wei, Q.; Zhang, Y.; Huang, R.; Pu, Y.; Dai, H.; Ren, B.; et al. Berberine reverses multidrug resistance in Candida albicans by hijacking the drug efflux pump Mdr1p. Sci. Bull. 2021, 66, 1895–1905. [Google Scholar] [CrossRef]
  176. Goswami, S.; Ali, A.; Prasad, M.E.; Singh, P. Pharmacological significance of Catharanthus roseus in cancer management: A review. Pharmacol. Res.-Mod. Chin. Medicine. 2024, 11, 100444. [Google Scholar] [CrossRef]
  177. Taub, J.W.; Buck, S.A.; Xavier, A.C.; Edwards, H.; Matherly, L.H.; Ge, Y. The evolution and history of Vinca alkaloids: From the Big Bang to the treatment of pediatric acute leukemia. Pediatr. Blood Cancer 2024, 71, e31247. [Google Scholar] [CrossRef] [PubMed]
  178. Mendonce, K.C.; Palani, N.; Rajadesingu, S.; Radhakrishnan, K.; Ayyar, M.; Priya, L.S. Pharmacological potential of bioactive compounds in Catharanthus roseus extract: A comprehensive review. Toxicol. Rep. 2025, 14, 101998. [Google Scholar] [CrossRef] [PubMed]
  179. Dai, Y.; Chen, S.-R.; Chai, L.; Zhao, J.; Wang, Y.; Wang, Y. Overview of pharmacological activities of Andrographis paniculata and its major compound andrographolide. Crit. Rev. Food Sci. Nutr. 2019, 59, S17–S29. [Google Scholar] [CrossRef]
  180. Paul, S.; Roy, D.; Pati, S.; Sa, G. The Adroitness of Andrographolide as a Natural Weapon Against Colorectal Cancer. Front. Pharmacol. 2021, 12, 731492. [Google Scholar] [CrossRef]
  181. Li, X.; Tian, R.; Liu, L.; Wang, L.; He, D.; Cao, K.; Ma, J.K.; Huang, C. Andrographolide enhanced radiosensitivity by downregulating glycolysis via the inhibition of the PI3K-Akt-mTOR signaling pathway in HCT116 colorectal cancer cells. J. Int. Med. Res. 2020, 48, 0300060520946169. [Google Scholar] [CrossRef]
  182. Tundis, R.; Patra, J.K.; Bonesi, M.; Das, S.; Nath, R.; Das Talukdar, A.; Das, G.; Loizzo, M.R. Anti-Cancer Agent: The Labdane Diterpenoid-Andrographolide. Plants 2023, 12, 1969. [Google Scholar] [CrossRef]
  183. Arsakhant, P.; Sirion, U.; Chairoungdua, A.; Suksen, K.; Piyachaturawat, P.; Suksamrarn, A.; Saeeng, R. Design and synthesis of C-12 dithiocarbamate andrographolide analogues as an anticancer agent. Bioorg. Med. Chem. Lett. 2020, 30, 127263. [Google Scholar] [CrossRef] [PubMed]
  184. Cai, W.; Li, J.; Chen, C.; Wu, J.; Li, J.; Xue, X. Design, synthesis, and anticancer evaluation of novel andrographolide derivatives bearing an α,β-unsaturated ketone moiety. Bioorg. Chem. 2021, 112, 104941. [Google Scholar] [CrossRef] [PubMed]
  185. Beesetti, S.L.; Jayadev, M.; Subhashini, G.V.; Mansour, L.; Alwasel, S.; Harrath, A.H. Andrographolide as a therapeutic agent against breast and ovarian cancers. Open Life Sci. 2019, 14, 462–469. [Google Scholar] [CrossRef] [PubMed]
  186. Vovchenko, M.; Alexandova, V.; Mustyatsa, V.; Gudimchuk, N. Study of the Microtubule-Paclitaxel Interaction in vitro and in silico. In Proceedings of the International Conference “Mathematical Biology and Bioinformatics”, Moscow, Russia, 12–19 October 2020; IMPB RAS: Moscow, Russia, 2020; Volume 8. [Google Scholar] [CrossRef]
  187. Paier, C.R.K.; Maranhão, S.S.; Carneiro, T.R.; Lima, L.M.; Rocha, D.D.; Santos, R.S.; Farias, K.M.; Moraes-Filho, M.O.; Pessoa, C. Natural products as new antimitotic compounds for anticancer drug development. Clinics 2018, 73, e813s. [Google Scholar] [CrossRef]
  188. Deng, S.; Shanmugam, M.K.; Kumar, A.P.; Yap, C.T.; Sethi, G.; Bishayee, A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer 2019, 125, 1228–1246. [Google Scholar] [CrossRef]
  189. Al-Bari, M.d.A.A.; Ito, Y.; Ahmed, S.; Radwan, N.; Ahmed, H.S.; Eid, N. Targeting Autophagy with Natural Products as a Potential Therapeutic Approach for Cancer. Int. J. Mol. Sci. 2021, 22, 9807. [Google Scholar] [CrossRef]
  190. Yang, Y.-H.; Mao, J.-W.; Tan, X.-L. Research progress on the source, production, and anti-cancer mechanisms of paclitaxel. Chin. J. Nat. Med. 2020, 18, 890–897. [Google Scholar] [CrossRef]
  191. Lee, N.; Youn, K.; Yoon, J.-H.; Lee, B.; Kim, D.H.; Jun, M. The Role of Fucoxanthin as a Potent Nrf2 Activator via Akt/GSK-3β/Fyn Axis against Amyloid-β Peptide-Induced Oxidative Damage. Antioxidants 2023, 12, 629. [Google Scholar] [CrossRef]
  192. Elmorsy, E.M.; Al Doghaither, H.A.; Al-Ghafari, A.B.; Alyamani, S.A.; Mohammed, Z.M.S.; Ebrahim, N.A.; Elshopakey, G.E.; Shabana, S.M. Through its genoprotective, mitochondrial bioenergetic modulation, and antioxidant effects, Fucoxanthin and its metabolite minimize Ochratoxin A-induced nephrotoxicity in HK-2 human kidney cells. BMC Nephrol. 2025, 26, 379. [Google Scholar] [CrossRef]
  193. Teng, C.; Wu, J.; Zhang, Z.; Wang, J.; Yang, Y.; Dong, C.; Wu, L.; Lin, Z.; Hu, Y.; Wang, J.; et al. Fucoxanthin ameliorates endoplasmic reticulum stress and inhibits apoptosis and alleviates intervertebral disc degeneration in rats by upregulating Sirt1. Phytother. Res. 2024, 38, 2114–2127. [Google Scholar] [CrossRef] [PubMed]
  194. Dalisay, D.S.; Tenebro, C.P.; Sabido, E.M.; Suarez, A.F.L.; Paderog, M.J.V.; Reyes-Salarda, R.; Saludes, J.P. Marine-Derived Anticancer Agents Targeting Apoptotic Pathways: Exploring the Depths for Novel Cancer Therapies. Mar. Drugs 2024, 22, 114. [Google Scholar] [CrossRef] [PubMed]
  195. Sasikumar, P.; Aparna, V.; Sebastian, A.T.; Muneer, A.; Prabha, B.; Vipin, C.L.; Ijinu, T.P. Clinically tested marine mollusk-derived anticancer agents: Chemico-pharmacological aspects. Stud. Nat. Prod. Chem. 2024, 83, 95–131. [Google Scholar] [CrossRef]
  196. Mayer, A.M.S.; Hall, M.L.; Lach, J.; Clifford, J.; Chandrasena, K.; Canton, C.; Kontoyianni, M.; Choo, Y.-M.; Karan, D.; Hamann, M.T. RSK1 vs. RSK2 Inhibitory Activity of the Marine β-Carboline Alkaloid Manzamine A: A Biochemical, Cervical Cancer Protein Expression, and Computational Study. Mar. Drugs 2021, 19, 506. [Google Scholar] [CrossRef]
  197. Ryu, B.; Avalon, N.; Cuau, M.; Almaliti, J.; Din, M.O.; Brennan, C.; Glukhov, E.; Knight, R.; Gerwick, L.; Gerwick, W. Cyanobacteria Join the Kahalalide Conversation: Genome and Metabolite Evidence for Structurally Related Peptides. ChemRxiv 2025, 1–25. [Google Scholar] [CrossRef]
  198. Flores-Holguín, N.; Frau, J.; Glossman-Mitnik, D. Investigating the Chemical Reactivity of Kahalalides: A Promising Source of Therapeutic Peptides from Marine Natural Products Using Conceptual Density Functional Theory. ChemistrySelect 2023, 8, e202303207. [Google Scholar] [CrossRef]
  199. Malik, M.S.; Alsantali, R.I.; Jassas, R.S.; Alsimaree, A.A.; Syed, R.; Alsharif, M.A.; Kulkarni, K.; Morad, M.; Althagafi, I.I.; Ahmed, S. A Journey of anthraquinones as anticancer agents—A systematic review of recent literature. RSC Adv. 2021, 11, 35806–35827. [Google Scholar] [CrossRef]
  200. Mantareva, V.; Iliev, I.; Sulikovska, I.; Durmuş, M.; Angelov, I. Cobalamin (Vitamin B12) in Anticancer Photodynamic Therapy with Zn(II) Phthalocyanines. Int. J. Mol. Sci. 2023, 24, 4400. [Google Scholar] [CrossRef]
  201. Tolymbekova, A.; Lezina, L. CD320 Receptor and Vitamin B12 as Potential Targets for Anti-Cancer Therapy. Int. J. Mol. Sci. 2025, 26, 5652. [Google Scholar] [CrossRef]
  202. Rachman, F.; Wibowo, J.T. Exploring Marine Rare Actinomycetes: Untapped Resources of Bioactive Compounds in Clinical Development. BIO Web Conf. 2024, 92, 02012. [Google Scholar] [CrossRef]
  203. Roy, A.; Datta, S.; Bhatia, K.S.; Bhumika Jha, P.; Prasad, R. Role of plant derived bioactive compounds against cancer. S. Afr. J. Bot. 2022, 149, 1017–1028. [Google Scholar] [CrossRef]
  204. Qiu, Y.; Chen, S.; Yu, M.; Shi, J.; Liu, J.; Li, X.; Chen, J.; Sun, X.; Huang, G.; Zheng, C. Natural Products from Marine-Derived Fungi with Anti-Inflammatory Activity. Mar. Drugs 2024, 22, 433. [Google Scholar] [CrossRef]
  205. Tamzi, N.N.; Rahman, M.M.; Das, S. Recent Advances in Marine-Derived Bioactives Towards Cancer Therapy. Int. J. Transl. Med. 2024, 4, 740–781. [Google Scholar] [CrossRef]
  206. Taylor, W.F.; Yanez, M.; Moghadam, S.E.; Moridi Farimani, M.; Soroury, S.; Ebrahimi, S.N.; Tabefam, M.; Jabbarzadeh, E. 7-epi-Clusianone, a Multi-Targeting Natural Product with Potential Chemotherapeutic, Immune-Modulating, and Anti-Angiogenic Properties. Molecules 2019, 24, 4415. [Google Scholar] [CrossRef] [PubMed]
  207. Chaachouay, N. Synergy, Additive Effects, and Antagonism of Drugs with Plant Bioactive Compounds. Drugs Drug Candidates 2025, 4, 4. [Google Scholar] [CrossRef]
  208. Cheng, Z.; Zhang, Z.; Han, Y.; Wang, J.; Wang, Y.; Chen, X.; Shao, Y.; Cheng, Y.; Zhou, W.; Lu, X.; et al. A review on anti-cancer effect of green tea catechins. J. Funct. Foods 2020, 74, 104172. [Google Scholar] [CrossRef]
  209. Ibrahim Fouad, G.; Ahmed, K.A. Curcumin Ameliorates Doxorubicin-Induced Cardiotoxicity and Hepatotoxicity Via Suppressing Oxidative Stress and Modulating iNOS, NF-κB, and TNF-α in Rats. Cardiovasc. Toxicol. 2022, 22, 152–166. [Google Scholar] [CrossRef]
  210. Kunnumakkara, A.B.; Hegde, M.; Parama, D.; Girisa, S.; Kumar, A.; Daimary, U.D.; Garodia, P.; Yenisetti, S.C.; Oommen, O.V.; Aggarwal, B.B. Role of Turmeric and Curcumin in Prevention and Treatment of Chronic Diseases: Lessons Learned from Clinical Trials. ACS Pharmacol. Transl. Sci. 2023, 6, 447–518. [Google Scholar] [CrossRef]
  211. Reytor-González, C.; Zambrano, A.K.; Montalvan, M.; Frias-Toral, E.; Simancas-Racines, A.; Simancas-Racines, D. Adherence to the Mediterranean Diet and its association with gastric cancer: Health benefits from a Planeterranean perspective. J. Transl. Med. 2024, 22, 483. [Google Scholar] [CrossRef]
  212. Boța, M.; Vlaia, L.; Jîjie, A.-R.; Marcovici, I.; Crişan, F.; Oancea, C.; Dehelean, C.A.; Mateescu, T.; Moacă, E.-A. Exploring Synergistic Interactions between Natural Compounds and Conventional Chemotherapeutic Drugs in Preclinical Models of Lung Cancer. Pharmaceuticals 2024, 17, 598. [Google Scholar] [CrossRef]
  213. Li, Y.; Chen, X.; He, W.; Xia, S.; Jiang, X.; Li, X.; Bai, J.; Li, N.; Chen, L.; Yang, B. Apigenin Enhanced Antitumor Effect of Cisplatin in Lung Cancer via Inhibition of Cancer Stem Cells. Nutr. Cancer 2021, 73, 1489–1497. [Google Scholar] [CrossRef]
  214. Yang, B.; Chen, Y.; Shi, J. Reactive Oxygen Species (ROS)-Based Nanomedicine. Chem. Rev. 2019, 119, 4881–4985. [Google Scholar] [CrossRef]
  215. Li, Y.; Li, Y.; Yao, Y.; Li, H.; Gao, C.; Sun, C.; Zhuang, J. Potential of cucurbitacin as an anticancer drug. Biomed. Pharmacother. 2023, 168, 115707. [Google Scholar] [CrossRef]
  216. Abdul Satar, N.; Ismail, M.N.; Yahaya, B.H. Synergistic Roles of Curcumin in Sensitising the Cisplatin Effect on a Cancer Stem Cell-Like Population Derived from Non-Small Cell Lung Cancer Cell Lines. Molecules 2021, 26, 1056. [Google Scholar] [CrossRef]
  217. Ioele, G.; Chieffallo, M.; Occhiuzzi, M.A.; De Luca, M.; Garofalo, A.; Ragno, G.; Grande, F. Anticancer Drugs: Recent Strategies to Improve Stability Profile, Pharmacokinetic and Pharmacodynamic Properties. Molecules 2022, 27, 5436. [Google Scholar] [CrossRef] [PubMed]
  218. Liu, B.; Zhou, H.; Tan, L.; Siu, K.T.H.; Guan, X.-Y. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct. Target. Ther. 2024, 9, 175. [Google Scholar] [CrossRef] [PubMed]
  219. Hong, L.; Li, W.; Li, Y.; Yin, S. Nanoparticle-based drug delivery systems targeting cancer cell surfaces. RSC Adv. 2023, 13, 21365–21382. [Google Scholar] [CrossRef] [PubMed]
  220. Chen, J.; Hu, S.; Sun, M.; Shi, J.; Zhang, H.; Yu, H.; Yang, Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur. J. Pharm. Sci. 2024, 193, 106688. [Google Scholar] [CrossRef]
  221. Amin, M.; Seynhaeve, A.L.B.; Sharifi, M.; Falahati, M.; ten Hagen, T.L.M. Liposomal Drug Delivery Systems for Cancer Therapy: The Rotterdam Experience. Pharmaceutics 2022, 14, 2165. [Google Scholar] [CrossRef]
  222. Ciftci, F.; Özarslan, A.C.; Kantarci, İ.C.; Yelkenci, A.; Tavukcuoglu, O.; Ghorbanpour, M. Advances in Drug Targeting, Drug Delivery, and Nanotechnology Applications: Therapeutic Significance in Cancer Treatment. Pharmaceutics 2025, 17, 121. [Google Scholar] [CrossRef]
  223. Wu, K.; Kwon, S.H.; Zhou, X.; Fuller, C.; Wang, X.; Vadgama, J.; Wu, Y. Overcoming Challenges in Small-Molecule Drug Bioavailability: A Review of Key Factors and Approaches. Int. J. Mol. Sci. 2024, 25, 13121. [Google Scholar] [CrossRef]
  224. Rastakhiz, S.; Yazdani, M.; Shariat, S.; Arab, A.; Momtazi-Borojeni, A.A.; Barati, N.; Mansourian, M.; Amin, M.; Abbasi, A.; Saberi, Z.; et al. Preparation of nanoliposomes linked to HER2/neu-derived (P5) peptide containing MPL adjuvant as vaccine against breast cancer. J. Cell Biochem. 2019, 120, 1294–1303. [Google Scholar] [CrossRef]
  225. Ding, Y.; Wang, L.; Li, H.; Miao, F.; Zhang, Z.; Hu, C.; Yu, W.; Tang, Q.; Shao, G. Application of lipid nanovesicle drug delivery system in cancer immunotherapy. J. Nanobiotechnol. 2022, 20, 214. [Google Scholar] [CrossRef] [PubMed]
  226. Wen, H.; Fu, L.; Huang, J.; Dai, Y.; Wang, B.; Xu, G.; Wu, L.; Zhou, H. Curcumin reverses doxorubicin resistance via inhibition the efflux function of ABCB4 in doxorubicin-resistant breast cancer cells. Mol. Med. Rep. 2019, 19, 5162–5168. [Google Scholar] [CrossRef]
  227. Buhrmann, C.; Shayan, P.; Brockmueller, A.; Shakibaei, M. Resveratrol Suppresses Cross-Talk between Colorectal Cancer Cells and Stromal Cells in Multicellular Tumor Microenvironment: A Bridge between In Vitro and In Vivo Tumor Microenvironment Study. Molecules 2020, 25, 4292. [Google Scholar] [CrossRef] [PubMed]
  228. Hu, S.; Li, X.; Xu, R.; Ye, L.; Kong, H.; Zeng, X.; Wang, H.; Xie, W. The synergistic effect of resveratrol in combination with cisplatin on apoptosis via modulating autophagy in A549 cells. Acta Biochim. Biophys. Sin. 2016, 48, 528–535. [Google Scholar] [CrossRef] [PubMed]
  229. Norden, E.; Heiss, E.H. Urolithin A gains in antiproliferative capacity by reducing the glycolytic potential via the p53/TIGAR axis in colon cancer cells. Carcinogenesis 2019, 40, 93–101. [Google Scholar] [CrossRef] [PubMed]
  230. Manogaran, P.; Beeraka, N.M.; Huang, C.-Y.; Vijaya Padma, V. Neferine and isoliensinine enhance ‘intracellular uptake of cisplatin’ and induce ‘ROS-mediated apoptosis’ in colorectal cancer cells—A comparative study. Food Chem. Toxicol. 2019, 132, 110652. [Google Scholar] [CrossRef] [PubMed]
  231. Liu, L.; Fan, J.; Ai, G.; Liu, J.; Luo, N.; Li, C.; Cheng, Z. Berberine in combination with cisplatin induces necroptosis and apoptosis in ovarian cancer cells. Biol. Res. 2019, 52, 37. [Google Scholar] [CrossRef]
  232. Alam, M.N.; Yu, J.Q.; Beale, P.; Huq, F. Cisplatin in combination with emetine and patulin showed dose and sequence dependent synergism against ovarian cancer. Synergy 2020, 10, 100060. [Google Scholar] [CrossRef]
  233. Bhagya, N.; Prabhu, A.; Rekha, P.D.; Chandrashekar, K.R. Combination of tetrandrine and cisplatin synergises cytotoxicity and apoptosis in triple negative breast cancer. Synergy 2020, 10, 100063. [Google Scholar] [CrossRef]
  234. Piska, K.; Koczurkiewicz, P.; Wnuk, D.; Karnas, E.; Bucki, A.; Wójcik-Pszczoła, K.; Jamrozik, M.; Michalik, M.; Kołaczkowski, M.; Pękala, E. Synergistic anticancer activity of doxorubicin and piperlongumine on DU-145 prostate cancer cells—The involvement of carbonyl reductase 1 inhibition. Chem. Biol. Interact. 2019, 300, 40–48. [Google Scholar] [CrossRef]
  235. Rawat, L.; Hegde, H.; Hoti, S.L.; Nayak, V. Piperlongumine induces ROS mediated cell death and synergizes paclitaxel in human intestinal cancer cells. Biomed. Pharmacother. 2020, 128, 110243. [Google Scholar] [CrossRef]
  236. Yang, H.; Gao, Y.; Fan, X.; Liu, X.; Peng, L.; Ci, X. Oridonin Sensitizes Cisplatin-Induced Apoptosis via AMPK/Akt/mTOR-Dependent Autophagosome Accumulation in A549 Cells. Front. Oncol. 2019, 9, 769. [Google Scholar] [CrossRef]
  237. Cao, W.; Li, Y.; Hou, Y.; Yang, M.; Fu, X.; Zhao, B.; Jiang, H.M.; Fu, X.Y. Enhanced anticancer efficiency of doxorubicin against human glioma by natural borneol through triggering ROS-mediated signal. Biomed. Pharmacother. 2019, 118, 109261. [Google Scholar] [CrossRef] [PubMed]
  238. Zhang, L.N.; Xia, Y.Z.; Zhang, C.; Zhang, H.; Luo, J.G.; Yang, L.; Kong, L.Y. Vielanin K enhances doxorubicin-induced apoptosis via activation of IRE1α-TRAF2-JNK pathway and increases mitochondrial Ca2+ influx in MCF-7 and MCF-7/MDR cells. Phytomedicine 2020, 78, 153329. [Google Scholar] [CrossRef] [PubMed]
  239. Gao, H.-L.; Xia, Y.-Z.; Zhang, Y.-L.; Yang, L.; Kong, L.-Y. Vielanin P enhances the cytotoxicity of doxorubicin via the inhibition of PI3K/Nrf2-stimulated MRP1 expression in MCF-7 and K562 DOX-resistant cell lines. Phytomedicine 2019, 58, 152885. [Google Scholar] [CrossRef]
  240. Lou, C.; Lu, H.; Ma, Z.; Liu, C.; Zhang, Y. Ginkgolide B enhances gemcitabine sensitivity in pancreatic cancer cell lines via inhibiting PAFR/NF-κB pathway. Biomed. Pharmacother. 2019, 109, 563–572. [Google Scholar] [CrossRef]
  241. Li, Y.; Li, X.; Lu, Y.; Chaurasiya, B.; Mi, G.; Shi, D.; Chen, D.; Webster, T.J.; Tu, J.; Shen, Y. Co-delivery of Poria cocos extract and doxorubicin as an ‘all-in-one’ nanocarrier to combat breast cancer multidrug resistance during chemotherapy. Nanomedicine 2020, 23, 102095. [Google Scholar] [CrossRef]
  242. Indorf, P.; Patzak, A.; Lichtenberger, F. Drug metabolism in animal models and humans: Translational aspects and chances for individual therapy. Acta Physiol. 2021, 233, e13734. [Google Scholar] [CrossRef]
  243. Li, Y.; Meng, Q.; Yang, M.; Liu, D.; Hou, X.; Tang, L.; Wang, X.; Lyu, Y.; Chen, X.; Liu, K.; et al. Current trends in drug metabolism and pharmacokinetics. Acta Pharm. Sin. B. 2019, 9, 1113–1144. [Google Scholar] [CrossRef] [PubMed]
  244. Sohn, S.-I.; Priya, A.; Balasubramaniam, B.; Muthuramalingam, P.; Sivasankar, C.; Selvaraj, A.; Valliammai, A.; Jothi, R.; Pandian, S. Biomedical Applications and Bioavailability of Curcumin—An Updated Overview. Pharmaceutics 2021, 13, 2102. [Google Scholar] [CrossRef] [PubMed]
  245. Jakobušić Brala, C.; Karković Marković, A.; Kugić, A.; Torić, J.; Barbarić, M. Combination Chemotherapy with Selected Polyphenols in Preclinical and Clinical Studies—An Update Overview. Molecules 2023, 28, 3746. [Google Scholar] [CrossRef]
  246. Corsini, N.S.; Knoblich, J.A. Human organoids: New strategies and methods for analyzing human development and disease. Cell 2022, 185, 2756–2769. [Google Scholar] [CrossRef]
  247. Chaudhary, N.; La Ferlita, A.; Choudhary, B.S.; Jog, E.; Kazi, M.; Yahya, S.; Dalwai, A.; Ostwal, V.; Singh, S.; Redkar, S.; et al. Patient-Derived Organoids and Xenografts Uncover Therapeutic Vulnerabilities in Colorectal Signet Ring Cell Carcinomas. Clin. Cancer Res. 2025, 31, 1359–1373. [Google Scholar] [CrossRef]
  248. Zhang, W.; Zheng, X. Patient-derived xenografts or organoids in the discovery of traditional and self-assembled drug for tumor immunotherapy. Front. Oncol. 2023, 13, 1122322. [Google Scholar] [CrossRef]
  249. Aliu, T.B.; Obun, F.E.; Raji, H.; Badmus, K. Safety Evaluation and Concerns of Natural Products in Traditional Medicine. AROC Pharm. Biotechnol. 2025, 5, 9–17. [Google Scholar] [CrossRef]
  250. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  251. Simoben, C.V.; Babiaka, S.B.; Moumbock, A.F.A.; Namba-Nzanguim, C.T.; Eni, D.B.; Medina-Franco, J.L.; Günther, S.; Ntie-Kang, F.; Sippl, W. Challenges in natural product-based drug discovery assisted with in silico -based methods. RSC Adv. 2023, 13, 31578–31594. [Google Scholar] [CrossRef]
  252. Heinrich, M.; Jalil, B.; Abdel-Tawab, M.; Echeverria, J.; Kulić, Ž.; McGaw, L.J.; Pezzuto, J.M.; Potterat, O.; Wang, J.-B. Best Practice in the chemical characterisation of extracts used in pharmacological and toxicological research—The ConPhyMP—Guidelines12. Front. Pharmacol. 2022, 13, 953205. [Google Scholar] [CrossRef]
  253. Chehelgerdi, M.; Chehelgerdi, M.; Allela, O.Q.B.; Pecho, R.D.C.; Jayasankar, N.; Rao, D.P.; Thamaraikani, T.; Vasanthan, M.; Viktor, P.; Lakshmaiya, N.; et al. Progressing nanotechnology to improve targeted cancer treatment: Overcoming hurdles in its clinical implementation. Mol. Cancer 2023, 22, 169. [Google Scholar] [CrossRef]
  254. Yu, L.; Jin, Y.; Song, M.; Zhao, Y.; Zhang, H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022, 14, 1589. [Google Scholar] [CrossRef] [PubMed]
  255. Sabit, H.; Pawlik, T.M.; Radwan, F.; Abdel-Hakeem, M.; Abdel-Ghany, S.; Wadan, A.H.S.; Elzawahri, M.; El-Hashash, A.; Arneth, B. Precision nanomedicine: Navigating the tumor microenvironment for enhanced cancer immunotherapy and targeted drug delivery. Mol. Cancer 2025, 24, 160. [Google Scholar] [CrossRef] [PubMed]
  256. Liu, B.; Liu, W.; Xu, M.; Zhao, T.; Zhou, B.; Zhou, R.; Zhu, Z.; Chen, X.; Bao, Z.; Wang, K.; et al. Drug delivery systems based on mesoporous silica nanoparticles for the management of hepatic diseases. Acta Pharm. Sin. B 2025, 15, 809–833. [Google Scholar] [CrossRef] [PubMed]
  257. Lee, W.-H.; Loo, C.-Y.; Traini, D.; Young, P.M. Development and Evaluation of Paclitaxel and Curcumin Dry Powder for Inhalation Lung Cancer Treatment. Pharmaceutics 2020, 13, 9. [Google Scholar] [CrossRef]
  258. Chavda, V.P.; Patel, A.B.; Mistry, K.J.; Suthar, S.F.; Wu, Z.-X.; Chen, Z.-S.; Hou, K. Nano-Drug Delivery Systems Entrapping Natural Bioactive Compounds for Cancer: Recent Progress and Future Challenges. Front. Oncol. 2022, 12, 867655. [Google Scholar] [CrossRef]
  259. Ghazal, H.; Waqar, A.; Yaseen, F.; Shahid, M.; Sultana, M.; Tariq, M.; Bashir, M.K.; Tahseen, H.; Raza, T.; Ahmad, F. Role of nanoparticles in enhancing chemotherapy efficacy for cancer treatment. Next Mater. 2024, 2, 100128. [Google Scholar] [CrossRef]
  260. Yang, L.; Han, T.; Liu, R.; Shi, S.; Luan, S.; Meng, S. Plant-derived natural compounds: A new frontier in inducing immunogenic cell death for cancer treatment. Biomed. Pharmacother. 2024, 177, 117099. [Google Scholar] [CrossRef]
  261. Lee, J.; Han, Y.; Wang, W.; Jo, H.; Kim, H.; Kim, S.; Yang, K.-M.; Kim, S.-J.; Dhanasekaran, D.N.; Song, Y.S. Phytochemicals in Cancer Immune Checkpoint Inhibitor Therapy. Biomolecules 2021, 11, 1107. [Google Scholar] [CrossRef]
  262. Ravindran Menon, D.; Li, Y.; Yamauchi, T.; Osborne, D.G.; Vaddi, P.K.; Wempe, M.F.; Zhai, Z.; Fujita, M. EGCG Inhibits Tumor Growth in Melanoma by Targeting JAK-STAT Signaling and Its Downstream PD-L1/PD-L2-PD1 Axis in Tumors and Enhancing Cytotoxic T-Cell Responses. Pharmaceuticals 2021, 14, 1081. [Google Scholar] [CrossRef]
  263. Fayyaz, A.; Haqqi, A.; Khan, R.; Irfan, M.; Khan, K.; Reiner, Ž.; Sharifi-Rad, J.; Calina, D. Revolutionizing cancer treatment: The rise of personalized immunotherapies. Discov. Oncol. 2024, 15, 756. [Google Scholar] [CrossRef]
  264. Krzyszczyk, P.; Acevedo, A.; Davidoff, E.J.; Timmins, L.M.; Marrero-Berrios, I.; Patel, M.; White, C.; Lowe, C.; Sherba, J.J.; Hartmanshenn, C.; et al. The growing role of precision and personalized medicine for cancer treatment. Technology 2018, 06, 79–100. [Google Scholar] [CrossRef]
  265. Zheng, C.-C.; Gao, L.; Sun, H.; Zhao, X.-Y.; Gao, Z.-Q.; Liu, J.; Guo, W. Advancements in enzymatic reaction-mediated microbial transformation. Heliyon 2024, 10, e38187. [Google Scholar] [CrossRef]
  266. Pant, A.; Maiti, T.K.; Mahajan, D.; Das, B. Human Gut Microbiota and Drug Metabolism. Microb. Ecol. 2023, 86, 97–111. [Google Scholar] [CrossRef]
  267. Kadam, O.; Dalai, S.; Chauhan, B.; Guru, R.R.; Mitra, S.; Raytekar, N.; Kumar, R. Nanobiotechnology Unveils the Power of Probiotics: A Comprehensive Review on the Synergistic Role of Probiotics and Advanced Nanotechnology in Enhancing Geriatric Health. Cureus 2025, 17, 1–21. [Google Scholar] [CrossRef] [PubMed]
  268. Cho, Y.S.; Han, K.; Xu, J.; Moon, J.J. Novel strategies for modulating the gut microbiome for cancer therapy. Adv. Drug Deliv. Rev. 2024, 210, 115332. [Google Scholar] [CrossRef] [PubMed]
  269. Sholl, L.M.; Hirsch, F.R.; Hwang, D.; Botling, J.; Lopez-Rios, F.; Bubendorf, L.; Mino-Kenudson, M.; Roden, A.C.; Beasley, M.B.; Borczuk, A.; et al. The Promises and Challenges of Tumor Mutation Burden as an Immunotherapy Biomarker: A Perspective from the International Association for the Study of Lung Cancer Pathology Committee. J. Thorac. Oncol. 2020, 15, 1409–1424. [Google Scholar] [CrossRef]
  270. Strickler, J.H.; Hanks, B.A.; Khasraw, M. Tumor Mutat. Burd. A Predict. Immunother. Response: Is. More Always Better? Clin. Cancer Res. 2021, 27, 1236–1241. [Google Scholar] [CrossRef]
  271. Cho, Y.A.; Lee, H.; Kim, D.G.; Kim, H.; Ha, S.Y.; Choi, Y.-L.; Jang, K.-T.; Kim, K.-M. PD-L1 Expression Is Significantly Associated with Tumor Mutation Burden and Microsatellite Instability Score. Cancers 2021, 13, 4659. [Google Scholar] [CrossRef]
  272. González-Méndez, I.; Sorroza-Martínez, K.; Cuétara-Guadarrama, F.; Vonlanthen, M.; Rivera, E. An overview of theranostic nanomedicine. In Theranostics Nanomaterials in Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2025; pp. 1–10. [Google Scholar] [CrossRef]
  273. Al-Thani, A.N.; Jan, A.G.; Abbas, M.; Geetha, M.; Sadasivuni, K.K. Nanoparticles in cancer theragnostic and drug delivery: A comprehensive review. Life Sci. 2024, 352, 122899. [Google Scholar] [CrossRef]
  274. Puccetti, M.; Pariano, M.; Schoubben, A.; Giovagnoli, S.; Ricci, M. Biologics, theranostics, and personalized medicine in drug delivery systems. Pharmacol. Res. 2024, 201, 107086. [Google Scholar] [CrossRef]
  275. Manzari-Tavakoli, A.; Babajani, A.; Tavakoli, M.M.; Safaeinejad, F.; Jafari, A. Integrating natural compounds and nanoparticle-based drug delivery systems: A novel strategy for enhanced efficacy and selectivity in cancer therapy. Cancer Med. 2024, 13, e7010. [Google Scholar] [CrossRef]
Figure 1. Pathways underlying cancer drug resistance.
Figure 1. Pathways underlying cancer drug resistance.
Ijms 26 08037 g001
Figure 2. Tumor microenvironment constituents and the contribution of tumor-associated macrophages to cancer progression.
Figure 2. Tumor microenvironment constituents and the contribution of tumor-associated macrophages to cancer progression.
Ijms 26 08037 g002
Figure 3. Chemical structures of natural compounds. This figure showcases the chemical structures of bioactive natural products, including polyphenols (yellow), alkaloids (green), terpenoids and diterpenes (light brown, bioactive lipids and marine-derived metabolites (light grey, and anthraquinones (turquoise). These compounds, sourced from terrestrial and marine environments, exhibit a wide range of biological activities such as antioxidant, anti-inflammatory, antimicrobial, and anticancer effects, reflecting the chemical diversity and therapeutic potential of natural substances [115,116,117,118,119,120,121,122,123].
Figure 3. Chemical structures of natural compounds. This figure showcases the chemical structures of bioactive natural products, including polyphenols (yellow), alkaloids (green), terpenoids and diterpenes (light brown, bioactive lipids and marine-derived metabolites (light grey, and anthraquinones (turquoise). These compounds, sourced from terrestrial and marine environments, exhibit a wide range of biological activities such as antioxidant, anti-inflammatory, antimicrobial, and anticancer effects, reflecting the chemical diversity and therapeutic potential of natural substances [115,116,117,118,119,120,121,122,123].
Ijms 26 08037 g003
Figure 4. Anticancer mechanisms and resistance pathways of natural bioactive compounds. Natural bioactives and their molecular actions, including inhibition of oncogenic signaling, modulation of inflammatory pathways, induction of apoptosis and cell cycle arrest, epigenetic regulation, activation of autophagy, and targeting of the tumor microenvironment. Several compounds also enhance therapeutic sensitivity and counteract multidrug resistance [115,124,125,126,127,128,129,130,131,132,133,134,135,136]. Green arrows indicate an enhanced effect; red arrows indicate a diminished effect. Abbreviations—Akt: protein kinase B; Arg-1: arginase-1; ATP: adenosine triphosphate; Bak: Bcl-2 homologous antagonist killer; Bax: Bcl-2-associated X protein; Bcl-2: B-cell lymphoma 2; CD44: cluster of differentiation 44; CD133: cluster of differentiation 133; CDKs: cyclin-dependent kinases; Chk1: checkpoint kinase 1; COX-2: cyclooxygenase-2; DNA: deoxyribonucleic acid; DNMT: DNA methyltransferase; EGCG: epigallocatechin gallate; Erk1/2: extracellular signal-regulated kinase ½; HDAC: histone deacetylase; HER3: human epidermal growth factor receptor 3; HIF-1α: hypoxia-inducible factor 1-alpha; IL-1β: interleukin-1 beta; IL-6: interleukin-6; iNOS: inducible nitric oxide synthase; Mcl-1: myeloid cell leukemia 1; MDR1: multidrug resistance protein 1; MDSCs: myeloid-derived suppressor cells; miRNA: microRNA; MMP: matrix metallopeptidase; mTOR: mammalian target of rapamycin; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; P-gp: P-glycoprotein; PD-L1: programmed death-ligand 1; p21: cyclin-dependent kinase inhibitor 1; p27: cyclin-dependent kinase inhibitor 1B; p53: tumor protein p53; PI3K: phosphoinositide 3-kinase; ROS: reactive oxygen species; SIRT1: sirtuin 1; SLUG: Zinc finger protein SNAI2; STAT3: signal transducer and activator of transcription 3; TME: tumor microenvironment; Wnt: wingless-related integration site; and XIAP: X-linked inhibitor of apoptosis protein.
Figure 4. Anticancer mechanisms and resistance pathways of natural bioactive compounds. Natural bioactives and their molecular actions, including inhibition of oncogenic signaling, modulation of inflammatory pathways, induction of apoptosis and cell cycle arrest, epigenetic regulation, activation of autophagy, and targeting of the tumor microenvironment. Several compounds also enhance therapeutic sensitivity and counteract multidrug resistance [115,124,125,126,127,128,129,130,131,132,133,134,135,136]. Green arrows indicate an enhanced effect; red arrows indicate a diminished effect. Abbreviations—Akt: protein kinase B; Arg-1: arginase-1; ATP: adenosine triphosphate; Bak: Bcl-2 homologous antagonist killer; Bax: Bcl-2-associated X protein; Bcl-2: B-cell lymphoma 2; CD44: cluster of differentiation 44; CD133: cluster of differentiation 133; CDKs: cyclin-dependent kinases; Chk1: checkpoint kinase 1; COX-2: cyclooxygenase-2; DNA: deoxyribonucleic acid; DNMT: DNA methyltransferase; EGCG: epigallocatechin gallate; Erk1/2: extracellular signal-regulated kinase ½; HDAC: histone deacetylase; HER3: human epidermal growth factor receptor 3; HIF-1α: hypoxia-inducible factor 1-alpha; IL-1β: interleukin-1 beta; IL-6: interleukin-6; iNOS: inducible nitric oxide synthase; Mcl-1: myeloid cell leukemia 1; MDR1: multidrug resistance protein 1; MDSCs: myeloid-derived suppressor cells; miRNA: microRNA; MMP: matrix metallopeptidase; mTOR: mammalian target of rapamycin; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; P-gp: P-glycoprotein; PD-L1: programmed death-ligand 1; p21: cyclin-dependent kinase inhibitor 1; p27: cyclin-dependent kinase inhibitor 1B; p53: tumor protein p53; PI3K: phosphoinositide 3-kinase; ROS: reactive oxygen species; SIRT1: sirtuin 1; SLUG: Zinc finger protein SNAI2; STAT3: signal transducer and activator of transcription 3; TME: tumor microenvironment; Wnt: wingless-related integration site; and XIAP: X-linked inhibitor of apoptosis protein.
Ijms 26 08037 g004
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

Reytor-González, C.; Jiménez-Flores, E.; González, N.; Simancas-Racines, D. Redefining Chemoresistance: Natural Bioactives as Molecular Modulators at the Cancer–Tumor Microenvironment Interface. Int. J. Mol. Sci. 2025, 26, 8037. https://doi.org/10.3390/ijms26168037

AMA Style

Reytor-González C, Jiménez-Flores E, González N, Simancas-Racines D. Redefining Chemoresistance: Natural Bioactives as Molecular Modulators at the Cancer–Tumor Microenvironment Interface. International Journal of Molecular Sciences. 2025; 26(16):8037. https://doi.org/10.3390/ijms26168037

Chicago/Turabian Style

Reytor-González, Claudia, Emilia Jiménez-Flores, Natalí González, and Daniel Simancas-Racines. 2025. "Redefining Chemoresistance: Natural Bioactives as Molecular Modulators at the Cancer–Tumor Microenvironment Interface" International Journal of Molecular Sciences 26, no. 16: 8037. https://doi.org/10.3390/ijms26168037

APA Style

Reytor-González, C., Jiménez-Flores, E., González, N., & Simancas-Racines, D. (2025). Redefining Chemoresistance: Natural Bioactives as Molecular Modulators at the Cancer–Tumor Microenvironment Interface. International Journal of Molecular Sciences, 26(16), 8037. https://doi.org/10.3390/ijms26168037

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

Article Metrics

Back to TopTop