ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Systematic Review

Integrative Network Pharmacology and Molecular Docking Approaches in Herbal Medicine Research. A Systematic Review of Applications, Advances, and Translational Potential

[version 1; peer review: awaiting peer review]
PUBLISHED 09 Dec 2025
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS AWAITING PEER REVIEW

This article is included in the AI in Medicine and Healthcare collection.

Abstract

Background

Network pharmacology and molecular docking have emerged as important in modern herbal drug discovery, offering systems-level understanding into phytochemical interactions and therapeutic mechanisms. This review aimed to evaluate the application of integrated network pharmacology and molecular docking in herbal medicine research

Methods

A systematic search was conducted across PubMed, Scopus, and Web of Science for studies published between 2010 and August, 2025. Eligible studies included both network pharmacology and molecular docking in medicinal plants, botanical formulations, or isolated phytochemicals. Screening and selection followed PRISMA-ScR guidelines. Data were extracted on study characteristics, computational platforms, biological validation strategies, phytochemical profiles, protein targets, and mechanistic pathways.

Results

36 studies met the eligibility criteria. Publications increased significantly after 2020, with most originating from Asia, particularly China (55.6%) and India (25.0%). Majority (61.1%) employed purely in silico pipelines, while 22.2% combined computational and in vitro validation, and 16.7% incorporated in vivo models. Flavonoids such as quercetin, kaempferol, apigenin, and luteolin accounted for most docked phytochemicals. Key molecular targets included ESR1, EGFR, AKT1, TNF, CASP3, and PTGS2, aligning predominantly with cancer, inflammatory, and metabolic pathways. Key signaling cascades PI3K-Akt, MAPK/ERK, NF-κB, and Wnt/β-catenin were consistently reported. Almost half of the studies (47.2%) integrated molecular dynamics simulations, while ADMET screening was reported in 71.4% of cases.

Conclusion

This review demonstrates that integrated network pharmacology and molecular docking have become foundational in herbal medicine research and growing with advances in computational biology. These approaches reveal multi-target mechanisms supporting the therapeutic potential of phytochemicals, yet translational progress is constrained by inconsistent pipelines. Strengthening validation frameworks, broadening phytochemical discovery beyond common flavonoids, and enhancing global research participation will be essential to accelerate clinically relevant herbal drug development.

Keywords

Network pharmacology, Molecular docking, medicinal plants, Phytochemicals, Drug discovery, Computational Biology,

1. Introduction

Herbal medicine constitutes one of the oldest therapeutic modalities and continues to represent an important component of primary health systems globally.1 Increasing global interest in phytomedicine is driven by an increase in demand for natural therapeutic agents, recognition of ethnopharmacological knowledge, and the search for more safer compounds. Despite its extensive cultural and clinical relevance, progress in herbal medicine research has historically been constrained by challenges.2 These includes the complexity of multi-component formulations, limited mechanistic understanding, and insufficient molecular validation.3

Network pharmacology has emerged as a systems-level strategy for elucidating compound–target–pathway interactions and capturing the multi-target mechanisms characteristic of botanical therapeutics. Network pharmacology enables the interrogation of synergistic effects, prediction of key therapeutic nodes, and functional enrichment analysis by modelling biological networks and disease pathways.4 This approach aligns with the principles of traditional medicine, moving beyond reductionist drug discovery paradigms and offering a framework for evaluating multi-component interventions.5

Furthermore, complementary to network-based inference, molecular docking provides atomic-level understanding into ligand–protein binding interactions. This enables validation, target prediction, and pharmacodynamic potential assessment. Docking simulations allow estimation of binding affinity, identification of key residues involved in ligand recognition and prioritization of lead phytochemicals for downstream experimentation.6 When applied in combination, network pharmacology and molecular docking constitute an integrative in silico pipeline capable of accelerating mechanistic elucidation, drug-binding affinity, and phytochemical prioritization prior to laboratory validation.7,8

In recent years, an expanding body of literature has applied these computational approaches to medicinal plants and traditional formulations, contributing to deeper mechanistic understanding and discovery of promising bioactive compounds.9 However, the field remains methodologically heterogeneous, with variation in data sources, analytical workflows, validation strategies, and reporting standards. Consolidation of current evidence is therefore essential to map research trends, evaluate methodological rigor, identify knowledge gaps, and guide translational advancement. This systematic review synthesizes studies that have jointly employed network pharmacology and molecular docking to investigate herbal medicines and phytochemicals. The review aims to: (i) characterize methodological practices and computational resources used across studies; (ii) identify key therapeutic areas and biological pathways explored; (iii) assess the extent of experimental verification supporting computational predictions. Through this synthesis, the review provides an evidence-based foundation to inform best practices and enhance the translation of herbal bioactive into clinically relevant therapeutic candidates.

2. Methods

2.1 Study design

A systematic review was conducted in accordance with PRISMA 2020 and the PRISMA extension for Scoping Reviews (PRISMA-ScR). The protocol specified the research question, eligibility criteria, information sources, search strategy, screening processes, data extraction fields, quality appraisal approach, and synthesis plan before data collection commenced.

2.2 Eligibility criteria

Studies were included if they met specific methodological and time-based criteria. Eligible papers were original, peer-reviewed research articles published between January 1, 2010 to 1st August, 2025. Each study was required to use both network pharmacology and molecular docking as part of its research approach, and the interventions had to involve herbal medicines, traditional plant formulations, or isolated phytochemicals. Studies also needed to report measurable scientific outputs such as compound–target interactions, protein network results, pathway or enrichment analysis, or docking scores showing ligand–receptor binding. For consistency and accurate interpretation, only studies published in English were considered. Studies were excluded if they did not use both network pharmacology and molecular docking together. Review articles, systematic reviews, meta-analyses, commentaries, letters, conference abstracts, and unpublished work were not included. Research focusing only on synthetic drugs without any herbal component was excluded. In addition, non-English publications and grey literature, such as theses and institutional reports, were not considered in this review.

2.3 Information sources and search strategy

A comprehensive literature search was conducted on 1st August 2025 to identify eligible studies. Three major scientific databases were searched including PubMed, Scopus, and Web of Science. A total of 83 records were retrieved across the three databases (PubMed = 26, Scopus = 10, Web of Science = 47). All articles were retrieved as CSV file and imported into Rayyan for systematic screening, duplicate detection, and management. During the de-duplication process, 9 duplicate records were detected. 4 duplicates were resolved and 5 was removed, leaving 78 articles for title and abstract screening. The search strategy combined controlled vocabulary terms and free-text keywords related to network pharmacology, molecular docking, and herbal medicine. The PubMed search string was: (“Network Pharmacology” [Title/Abstract] OR “Systems Pharmacology” [Title/Abstract]) AND (“Molecular Docking” [Title/Abstract] OR “In Silico Docking” [Title/Abstract]) AND (“Herbal Medicine” [Title/Abstract] OR “Medicinal Plants” [Title/Abstract] OR “Phytochemicals” [Title/Abstract]) AND (“2010/01/01” [Date-Publication]: “2025/12/31” [Date-Publication]). Equivalent search terms were adapted for Scopus and Web of Science to ensure consistency while accommodating database-specific syntax.

2.4 Study selection and screening

The study selection process followed a structured and transparent approach consistent with PRISMA-ScR guidelines. After the initial search and de-duplication, 78 articles were screened for title and abstract. Two reviewers independently assessed each article against the defined eligibility criteria. Articles that met the inclusion criteria were selected for full-text screen. During the screening process, any disagreements between the two reviewers were addressed through discussion. When consensus could not be reached, a third reviewer was consulted to resolve the discrepancy. Full-text articles were then examined to confirm compliance with the predefined inclusion criteria. Decisions at each stage of the screening process, including reasons for exclusion, were documented systematically. Out of the screened records, 36 studies met all criteria and were included in the final synthesis as depicted by Figure 1.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure1.gif

Figure 1. PRISMA flow chart.

2.5 Data extraction

Data extraction was carried out using a structured and predefined data collection framework drafted in excel (version 2019) to ensure consistency and completeness. Key study characteristics and methodological details were systematically extracted from each included article. Extracted information included bibliographic data (author, year, journal, and country), herbal species or formulations investigated, plant part used, extraction or preparation method (where applicable), and the class of bioactive compounds assessed. Technical information relevant to methodologies and computational workflows was also collected. This included the databases used for compound identification and target prediction, software and algorithms applied in network construction, molecular docking tools and scoring functions, validation strategies, and major biological pathways or molecular targets identified. In addition, details related to experimental design, such as in vitro or in vivo validation, were captured when reported. The extraction process was conducted independently by two reviewers, and extracted data were cross-checked for accuracy. Any discrepancies were resolved through joint review and consensus discussion. Where information was incomplete or ambiguous, efforts were made to confirm details directly from study text and supplementary materials to ensure accurate interpretation.

2.6 Quality assessment

Quality assessment of included studies focused on methodological transparency, reliability of computational procedures, and the validity of reported outcomes. Reviewers evaluated the completeness of reporting regarding source databases, compound screening criteria, target identification procedures, network construction parameters, docking protocols, scoring metrics, and validation methods. Emphasis was placed on whether computational pipelines adhered to accepted standards in network pharmacology and in silico molecular docking, including appropriate justification for bioinformatics tools and docking algorithms. The review process also assessed the presence of experimental verification when applicable, consistency between predicted mechanisms and biological evidence, and clarity in reporting key outputs such as compound–target associations, pathway enrichment results, and docking affinity scores. The quality appraisal was performed in parallel by two reviewers. Disagreements were resolved through discussion, and when necessary, a third reviewer provided adjudication. This iterative and collaborative assessment ensured that only methodologically sound and scientifically credible studies contributed to the final synthesis.

2.7 Data synthesis and analysis

A descriptive synthesis was undertaken to map study characteristics, herbal taxa, phytochemicals, disease indications, target families, and pathway/enrichment outputs. Quantitative tabulations summarized frequencies and percentages for commonly used compound/target databases, target-prediction platforms, network tools, docking suites, validation modalities, and pathway resources (e.g., KEGG, GO, DAVID), ensuring category-level totals were normalized within 100% when analyses were mutually exclusive. Subgroup analyses were conducted by database category (e.g., herbal/phytochemical repositories, disease-gene resources, protein annotation/interaction platforms), by target-prediction tools, and by network software/version (e.g., Cytoscape sub-versions), reflecting the methodological heterogeneity observed. Temporal trends (2010–2025), country distribution, and therapeutic domains were summarized to contextualize the evolution of methods and application areas. Visual maps of herb–compound–target–pathway networks were synthesized from the extracted data to illustrate mechanistic linkages where reporting permitted.

3. Results

3.1 Study characteristics

A total of 36 studies met the inclusion criteria and were incorporated into the final synthesis. The studies were published between 2013 and 2025, with a significant increase in publications from 2020 onward. Majority of the studies were conducted in Asian countries, particularly China (n = 20, 55.6%)4,10,11 and India (n = 9,12 25.0%), followed by Pakistan, Saudi Arabia, Egypt,13,14 Ethiopia,8 Indonesia, and Taiwan.15 Significant number of studies employed fully in silico approaches integrating network pharmacology and molecular docking (n = 22, 61.1%).12 The remaining studies combined computational pipelines with experimental laboratory validation, including in vitro assays (n = 8, 22.2%) and in vivo animal models (n = 6, 16.7%). Sample sizes for experimental arms varied, with most in vivo studies utilizing murine models, while in vitro analyses frequently employed human cancer cell lines and macrophage systems as summarized in Table 1. Plant materials analyzed varied widely and included whole plants, roots, rhizomes, barks, fruits,10,12 seeds, leaves, and formulated polyherbal preparations. Solvents used for extraction, where specified, included methanol,16 ethanol, chloroform, and n-hexane; however, solvent reporting was inconsistent across studies, with nearly half not specifying extraction media. Both single-herb studies (e.g., Centella asiatica, Piper longum, 10 Saussurea lappa) and polyherbal formulations (e.g., Huangqin Tang, Dashamoola, HSXFF, Krishnadi Churna, Qi-Enriching and Blood-Tonifying herbs) were reported. Biological validation where present included evaluation in disease-specific cell lines such as MCF-7 and A549, immune cells such as RAW264.7 macrophages and human peripheral blood cells, and rodent models of neurodegeneration, inflammation, ulcerative colitis, and metabolic dysfunction. Most purely computational studies validated predicted targets and pathways using publicly available protein structures, genomic databases, and docking or molecular dynamics outputs.

Table 1. Systematic review study characteristics table.

IDAuthor(s)YearCountrySample SizeStudy Type Plant Part Solvent Medicinal Plant/Formula Biological Source/Tissue
1172024ChinaIn vivo & in vitro (n not specified)Experimental + NP + in vivo + in vitroNot reportedNot reportedCentella asiatica/AsiaticosideMPTP mouse model, BV2 cells
2182022India194 NCs screenedIn silico NP & DockingNot reportedNot reportedClerodendrum sp.Protein models
3192024India145 compoundsIn silico NP & DockingFruit & rootNot reportedPiper longum Fruit & root tissues
4102024ChinaNot specifiedExperimental + NPFruit spikesEthanolPiper longum L.Mouse inflammation model
5122025IndiaNot specifiedExperimental + Bioinformatics + NPFruitsNot specifiedCaryota urens MCF7 cells
6202025Chinan=3 per groupExperimental + NP + DockingFruitNot specifiedFicus hispida HASMCs, mouse aorta
7162025Chinan=6 per groupExperimental + NP + Proteomics + DockingRootsMethanolJiegeng Gancao DecoctionColon tissue (UC mice)
8212025ChinaIn silicoNP + Docking + Meta-analysis + DFT + MDRoots & barksNot specifiedDashamoolaHuman intestinal GEO datasets
9222024ChinaIn silicoNP + Docking + MD + DFT + ADMENot specifiedNot specifiedMagnolia spp.In-silico molecular models
10232022ChinaIn silicoBioinformatics + DockingRhizomeNot specifiedPicrorhizae Rhizoma Human genes
11242024ChinaIn vitroNP + Docking + ExperimentsRoots/stems/rhizomesNot specifiedHuangqin TangMCF-7 & MDA-MB-231 cells
12252024PakistanIn silicoNP + Docking + MDWhole plantNot specifiedCassia spp.Target proteins
13262024IndiaIn silicoNP + Docking + MD + MM-GBSAWhole plantNot specifiedDrymaria cordata Protein structures
14272024India40 phytochemicalsNP + Docking + MDS + MMGBSARoot & StemHexane/MethanolPotentilla nepalensis Cancer proteins
15282024ChinaA549 cellsExperimental + in silicoRootChloroform + EthanolSaussurea lappa A549 lung cells
16292023Egypt32 plants, 2,154 compoundsNP + Docking + MD + in vitroWhole plants70% EthanolMulti-herb panelHuman WBCs
17302023Saudi ArabiaNot specifiedNP + Docking + MDNot specifiedNot statedDodonaea angustifolia Not reported
18312023ChinaIn silicoNP + DockingWhole plantNot specifiedSolanum nigrum Breast cancer genes
19112023ChinaIn silico + in vitroNP + Docking + CETSAWhole plantNot specifiedSolanum nigrum L.CT26 cells
20322025IndiaIn silico + in vitroNP + Docking + MDWhole plantNot statedAlchornea laxiflora Not stated
21332024ChinaIn silico + in vitroNP + DockingWhole plantNot specifiedPolygonum cuspidatum HEK-293T cells
22142022Saudi ArabiaIn silicoComputationalStemMethanolArgyreia capitiformis Stem extracts
23342022ChinaIn silico + experimentalNP + DockingSeedsNot specifiedRheum tanguticum Seed tissue
24132021Saudi ArabiaIn silico + in vivoExperimental + NPLeavesMethanolCnesmone javanica Leaf extracts
25352021Egypt100 compoundsIn silico + in vitroPropolisEthanolEgyptian propolisPropolis extract
26362022ChinaIn silicoNP studyWhole plantNot specifiedHSXFF Formula
27152013ChinaIn silicoNP modelingRoots/rhizomes/tubersNot specifiedQi-Enriching & Blood-Tonifying herbsMolecular models
28372024IndiaIn silico + in vivoNP + DockingNot reportedNot reportedKrishnadi ChurnaNot stated
29382016ChinaIn silicoComputationalWhole plantNot specifiedSH FormulaNot stated
3082022Ethiopia10 aloe speciesIn silico profilingLeavesNot statedAloe speciesLeaves
31392022IndonesiaIn silicoVirtual screeningNot specifiedNot statedMoringa & Psidium Not specified
3272016Taiwan/ChinaNot specifiedVirtual ScreeningNot statedNot statedMultiple TCM herbsNot reported
33402020PakistanIn silico + in vitroPhytochemical + DockingRootEthyl acetateZiziphus oxyphylla Not specified
34412021ChinaNot specifiedNP + transcriptomics + in vitroXYPI compoundNot reportedXYPIRAW264.7 macrophages
35422025ChinaNot specifiedIn silicoNot statedNot statedBK002 formulaPatient genomic data
36432023IndiaNot specifiedIn silicoTea leavesNot statedCamellia sinensis Extracts

3.2 Temporal distribution of included studies

The temporal distribution of studies indicates a steady increase in the use of network pharmacology and molecular docking in herbal medicine research presented in Figure 2. Early activity was limited, with only 2.78% of studies published in 201315 and 5.56% in 2016, reflecting the initial stage of adoption. A small increased appeared in 2020 (2.78%), followed by more noticeable growth in 2021 (8.33%), marking the beginning of wider implementation of computational methods in the field. A significant expansion occurred from 2022 onward, where publication frequencies reached 19.44% in 2022 and 13.89% in 2023.11 The highest proportion of studies was observed in 2024 (30.56%),19,11 and a substantial number continued to be published in 2025 (16.67%).4

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure2.gif

Figure 2. Temporal distribution of studies applying network pharmacology and molecular docking in herbal medicine research.

The results show limited research activity during the early years (2.78% in 2013; 5.56% in 2016), gradual adoption in 2020–2021 (2.78% and 8.33%), and a marked increase from 2022 onward, including 19.44% in 2022, 13.89% in 2023, and a peak in 2024 (30.56%). A sustained level of publications continued in 2025 (16.67%).

3.3 Distribution of database categories reported

The included studies utilized a range of computational resources for compound identification and target screening. Databases clustered into four primary categories: herbal/phytochemical repositories (e.g., TCMSP, IMPPAT, HERB), chemical-structure sources (PubChem, ChEMBL, Dictionary of Natural Products), target-prediction platforms (SwissTargetPrediction,16 SEA, PharmMapper), and disease-gene resources (GeneCards, DisGeNET, OMIM, GEO, NCBI).12,20 Additional tools, including SwissADME,19 admetSAR, PreADMET, and BindingDB, supported ADME-toxicity screening and protein-interaction mapping.43 Together, these database categories formed the core evidence pipeline for phytochemical retrieval, drug-likeness evaluation, and mechanistic network construction.

3.3.1 Herbal/phytochemical repositories

Across the included studies, TCMSP was the most frequently used herbal database, accounting for 52.9% of all phytochemical database citations as presented in Figure 3. This was followed by IMPPAT (17.6%) and TCM Database@Taiwan (11.8%),15 whereas HERB, ETCM, and NPASS were each used in 5.9% of studies. These findings suggest that most researchers rely on widely established Chinese herbal databases, with relatively lower utilization of newer or region-specific phytochemical repositories.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure3.gif

Figure 3. Distribution of herbal and phytochemical databases used for compound retrieval in included studies.

The figure illustrates the frequency of herbal database utilization across the reviewed literature (n = 17 mentions). TCMSP was the most commonly employed platform (52.9%), followed by IMPPAT (17.6%) and TCM Database@Taiwan (11.8%). HERB, ETCM, and NPASS were each used in 5.9% of studies.

3.3.2 Target-prediction platforms used in included studies

Analysis of target-prediction tools demonstrated heterogeneous platform use across the included studies as presented in Table 2. SwissTargetPrediction was the most frequently applied tool, appearing in 36.36% of studies,12 highlighting its central role in ligand-based target inference. DIGEP-Pred, SEA (Similarity Ensemble Approach), and non-specified computational approaches each accounted for 9.09% of studies, indicating moderate use of transcriptomic-based and chemical-similarity prediction methods. PharmMapper, Way2Drug, SuperPred,27 ImaGEO,16 STITCH, STRING,21 TTD, and PharmGKB were each reported in 4.55% of studies, demonstrating selective and context-specific utilization for structure-guided screening, protein-interaction mapping, and disease-gene associations.

Table 2. Target-prediction platforms used in included studies.

Target prediction platformFrequency (n = 22)Percentage (%) Reference
SwissTargetPrediction836.36%16
DIGEP-Pred 29.09%27
SEA29.09%26
Not specified/model-based 29.09%35
PharmMapper14.55%18
Way2Drug14.55%19
SuperPred14.55%26
ImaGEO14.55%12,16
STITCH (target assignment)14.55%8
STRING (target assignment)14.55%35
TTD14.55%29
PharmGKB14.55%17

The results show that SwissTargetPrediction was the most commonly employed platform (36.36%), followed by DIGEP-Pred, SEA, and non-specified computational approaches (each 9.09%). PharmMapper, Way2Drug, SuperPred, ImaGEO, STITCH, STRING, TTD, and PharmGKB were each used in 4.55% of studies, indicating selective adoption of specialized tools for structure-based prediction, transcriptomic inference, and protein-interaction mapping.

3.3.3 Chemical structure databases

Chemical structure databases were used to support compound identification and structural verification across the included studies as depicted in Figure 4. PubChem was the most frequently used resource (31.25%), followed by SwissADME (18.75%). Other tools including ChEMBL,22 admetSAR, ChemDraw,23 NIST, Dictionary of Natural Products, and CDRUG were each reported in 6.25% of cases. This distribution reflects a primary reliance on established public chemical repositories such as PubChem, with supplementary use of cheminformatics and property-prediction tools for compound validation.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure4.gif

Figure 4. Chemical structure databases used for compound characterization across included studies.

The result show that pubChem was most commonly employed (31.25%), followed by SwissADME (18.75%), with ChEMBL, admetSAR, ChemDraw, NIST, Dictionary of Natural Products, and CDRUG each used in 6.25% of studies.

3.3.4 Disease-gene annotation databases used across included studies

Disease-gene retrieval in the included studies that primary relied on GeneCards (44.44%), followed by OMIM and DisGeNET (22.22% each) as depicted by Figure 5. GEO, NCBI, and HPO were each utilized in 5.56% of studies. This pattern indicates a primary dependence on comprehensive disease-gene compendiums, particularly GeneCards,16 supplemented by clinically annotated resources (OMIM) and disease-association platforms (DisGeNET). Use of curated transcriptomic repositories (GEO)21 was less common, reflecting fewer studies integrating omics-driven disease gene mining.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure5.gif

Figure 5. Disease-gene annotation databases used across included studies.

The figure results of the Disease-gene retrieval revealed GeneCards as the most frequently used disease-gene source (44.44%), followed by OMIM and DisGeNET (22.22% each). GEO, NCBI, and HPO accounted for 5.56% each, indicating selective incorporation of transcriptomic and phenotype-ontology sources.

3.3.5 Protein annotation and interaction databases used in the included studies

Across the included studies, protein annotation and interaction resources were used to support target validation and functional characterization as presented in Figure 6. UniProt/UniProtKB was the most frequently applied database (62.5%), indicating its central role in annotating protein targets and linking phytochemicals to biological functions. BindingDB appeared in 25.0% of studies, primarily for retrieving experimental protein ligand interaction data, while STITCH was used in 12.5% of studies for identifying chemical protein interaction networks. Together, these tools provided essential support for confirming target relevance and mapping molecular interaction profiles in network-pharmacology workflows.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure6.gif

Figure 6. Protein annotation and interaction databases used in the included studies.

The results show that UniProt/UniProtKB was the most widely used platform, accounting for 62.5% of studies, followed by BindingDB (25.0%) and STITCH (12.5%). These resources were primarily applied for protein functional annotation, target confirmation, and mapping chemical–protein interaction networks within network-pharmacology pipelines.

3.3.6 Computational multi-database integrators

Among the included studies, a subset employed computational multi-database integrators, reflecting advanced informatics approaches that combine multiple chemical, genomic, proteomic, and pathway resources within a unified analytical pipeline. These systems were generally used to enhance compound annotation, strengthen target prediction confidence, and support holistic network construction. Examples identified in this review included HERB-linked compound and serum-metabolite integration frameworks, the TCMIP22 v2.0 Traditional Chinese Medicine Integrated Platform, and multi-omics heterogeneous network pipelines combining drug–target, protein–protein interaction, and pathway data. All were reported once accounting for 12.5%. Additional approaches incorporated Metascape-based enrichment, cross-database compound validation pipelines (e.g., PubChem + ChEMBL + Dictionary of Natural Products),22 and hybrid LC-MS/GC-MS experimental data coupled with public disease-gene repositories such as DrugBank, OMIM, DisGeNET, and GeneCards.16 One study also applied a combined Indonesian Herbal Database and viral-target literature platform to support antiviral repositioning.

3.3.7 Literature-derived compound sources

Literature-derived compound sources were also employed across the included studies. These approaches relied on extracting phytochemical information directly from published research and authoritative herbal references, either as standalone lists or to supplement cheminformatics platforms. Reported sources included literature-curated compound datasets combined with PreADMET and Osiris Property Explorer filters, manual literature searches integrated with IMPPAT and SwissADME,26 and published phytochemical reports cross-validated using SwissTargetPrediction,16 GeneCards,21 and DisGeNET. Some studies generated in-house compound libraries based entirely on literature mining, while others used literature-supported compound lists alongside established herbal databases such as TCM Database@Taiwan. Additional examples included literature-based repurposing datasets for antiviral compounds (e.g., SARS/MERS/SARS-CoV-2 studies) and disease-specific compound mining from published evidence (e.g., prostate cancer patient data). Overall, ten studies (12.5%) incorporated literature-derived phytochemical sources as part of their compound identification strategy.

3.4 Therapeutic domains in the included studies

Across the included studies, different therapeutic domains were used as summarized in Table 3. Inflammatory and immune-mediated conditions represented the largest focus area (14.04%), followed by oncology-related investigations. Specifically, breast cancer (8.77%),22 lung cancer (5.26%), colon/colorectal cancer (3.51%),21 prostate cancer (3.51%),23 hepatocellular carcinoma (1.75%), cervical cancer (1.75%) and cancer of unspecified type (10.53%)18 collectively accounted for a substantial proportion of studies, emphasizing the dominant interest in cancer-targeting phototherapeutics. COVID-19-related studies comprised 7.02% of the dataset, underscoring sustained research interest in antiviral phytocompounds for emerging infectious diseases. Neurological and psychiatric conditions including Alzheimer’s disease (1.75%), Parkinson’s disease (1.75%), and anxiety/depression (3.51%) were also represented, along with metabolic and cardiovascular disorders such as diabetes (1.75%), hypertension (1.75%),25 vascular calcification (1.75%), and oxidative stress-associated pathology (1.75%). Gastrointestinal immune disorders, including ulcerative colitis (3.51%) and Crohn’s disease (1.75%), were additionally identified.

Table 3. Therapeutic domains in the included studies.

ConditionFrequency (n)Percentage (%) Reference
Inflammation/Anti-inflammatory 814.0414
Cancer (unspecified)610.5327
Breast cancer58.7737
COVID-1947.0239
Lung cancer35.2619
Colorectal/Colon cancer23.5121
Anxiety/Depression23.5144
CNS diseases (unspecified)23.5115
Hematologic diseases (unspecified)23.5115
Immune system diseases (unspecified)23.5129
Prostate cancer23.5123
Ulcerative colitis23.5116
Alzheimer’s disease11.7540
Cervical cancer11.7526
Crohn’s disease11.7521
Diabetes11.7540
Digestive diseases (unspecified)11.7515
Gout11.7510
Hepatocellular carcinoma11.7532
HIV-111.758,38
Hypertension11.7525
Oxidative stress/Antioxidant11.7534
Parkinson’s disease11.7533
Vascular calcification11.7520
Infectious diseases (unspecified)11.758
Lung cancer (SCLC)11.7528

The results show that inflammatory and immune-mediated disorders were the most frequent focus (14.04%), followed by cancer-related investigations (breast cancer 8.77%, cancer-unspecified 10.53%, lung cancer 5.26%, colon and prostate cancer each 3.51%, and hepatocellular and cervical cancer each 1.75%). COVID-19 accounted for 7.02% of studies, while neurological disorders (Parkinson’s and Alzheimer’s disease), metabolic and cardiovascular conditions (diabetes, hypertension, vascular calcification), and gastrointestinal immune diseases (ulcerative colitis, Crohn’s disease) each represented 1.75–3.51% of the literature.

3.5 Network-pharmacology software used in included studies

Across the included studies, Cytoscape emerged as the primary network-pharmacology platform, appearing in 63.89% of cases as presented in Table 4. Among these, versions v3.7.2 and v3.8.223 were most frequently reported (8.33% each), while other versions including v3.7.0,24 v3.10, v3.9.0, v3.9.1, v3.7.1, and v3.2 were each identified in 2.78% of studies. Nevertheless, version information was omitted in 30.56% of Cytoscape-using studies, indicating incomplete reporting of software details. Beyond Cytoscape, STRING23 was the next most commonly used platform (22.22%), followed by AutoDock-based pipelines (11.11%) and STITCH (5.56%). Less frequently adopted tools included GEPIA2, KEGG/GO16,20 enrichment platforms, Python NetworkX, and machine-learning-driven approaches (each 2.78%). Additionally, 19.44% of studies applied network-pharmacology workflows without specifying the software used, highlighting variability and reporting gaps in computational methodology.

Table 4. Network-pharmacology software used in included studies.

Software/PlatformFrequency (n)Percentage (%)
Cytoscape v3.7.238.33%18
Cytoscape v3.8.238.33%23
Cytoscape v3.7.012.78%24
Cytoscape v3.1012.78%27
Cytoscape v3.9.012.78%32
Cytoscape v3.9.112.78%30
Cytoscape v3.7.112.78%33
Cytoscape v3.212.78%7
Cytoscape (version not specified)1130.56%17,21
STRING822.22%23
AutoDock/PyRx docking workflow411.11%17
STITCH25.56%18
GEPIA212.78%19
KEGG/GO enrichment platforms12.78%16
Python NetworkX12.78%8
Machine-learning pipeline (SVM/MLP/RF)12.78%40
Unspecified network-pharmacology tools719.44%42

The results indicate different computational workflows, with multiple versions of Cytoscape widely used (8.33% each for v3.7.2 and v3.8.2; 30.56% unspecified). STRING was applied in 22.22% of studies, and AutoDock-based pipelines in 11.11%. STITCH and other tools including GEPIA2, KEGG/GO enrichment tools, Python NetworkX,8 and machine-learning models each appeared in 2.78% of studies. Additionally, 19.44% of studies referenced network-pharmacology pipelines without specifying software, highlighting reporting variability.

3.6 Distribution of pathway and enrichment analysis tools among included studies

Across the included studies, pathway and enrichment analysis with majority of the studies relying on KEGG20 and Gene Ontology resources as presented in Table 5. KEGG accounted for the most common tool reported with (37.88%). Gene Ontology methods constituted 28.79% of all studies, reflecting frequent application for biological process, molecular function, and cellular component annotation. DAVID contributed 16.67% of tool reported, functioning primarily as an analytical interface for KEGG and GO integration.25 Application of alternative enrichment platforms was comparatively limited. Reactome and BIOCARTA each contributed 3.03% of tool usage, while individual tools such as ShinyGO, KOBAS, BBID, WikiPathways, GOBP, C–T–D network enrichment, and enrichment-factor analysis were reported in 1.52%.

Table 5. Distribution of Pathway/Enrichment tools.

Tool/PlatformFrequencies (n)Percentages (%) Reference
KEGG Pathway Analysis2537.8817,20
Gene Ontology (GO)1928.7917
DAVID1116.6718
Reactome23.0340
BIOCARTA23.0329
ShinyGO11.5230
KOBAS11.5233
BBID11.5235
WikiPathways11.5240
GOBP (GO Biological Process)11.5241
C–T–D network enrichment11.528
Enrichment factor (EF) analysis11.528

Distribution of pathway and enrichment analysis tools used across included studies. The results show that KEGG (37.88%) and Gene Ontology (28.79%) were the most frequently applied enrichment resources, followed by DAVID (16.67%). Other tools, including Reactome, BIOCARTA, ShinyGO, KOBAS, and WikiPathways, accounted for smaller shares (≤3.03%), demonstrating limited adoption beyond core KEGG- and GO-based platforms.

3.7 Distribution of core protein targets identified across included studies

The analysis reveals a concentration of research efforts on key molecular regulators implicated in cancer progression, inflammation, cell survival, apoptosis, and angiogenesis as presented in Table 6. The most frequently investigated targets included ESR1 (27.78%), EGFR (25.00%),20 and AKT1 (22.22%), reflecting strong emphasis on hormone-dependent and growth-factor-mediated signaling pathways.23 Inflammation-related proteins such as TNF (19.44%), IL6 (11.11%), and IL1B (8.33%),17 alongside apoptosis-linked mediators (CASP3 and PTGS2, each 16.67%) were also prominently represented. Similarly, tumor suppressors and stress-response proteins (TP53 and HSP90AA1, each 13.89%) and angiogenesis and matrix-remodeling factors (VEGFA and MMP9, each 13.89%) appeared consistently across studies.

Table 6. Frequency distribution of key protein targets identified across studies.

Protein TargetBiological Role CategoryFrequency (n)Percentage (%) Reference
ESR1Hormonal/Cancer signaling1027.78%22
EGFRGrowth factor/Cancer signaling925.00%20
AKT1Cell survival/Metabolism822.22%23
TNFInflammation/Immune regulation719.44%21
CASP3Apoptosis execution616.67%23
PTGS2 (COX-2)Inflammation/Pro-tumorigenic 616.67%16
TP53Tumor suppressor/DNA repair513.89%27
STAT3Immune & growth signaling513.89%24
HSP90AA1Heat-shock stress response/Cancer513.89%20
MMP9Extracellular matrix remodeling513.89%30
VEGFAAngiogenesis513.89%27
MAPK1MAPK pathway signaling411.11%25
IL6Cytokine signaling/Inflammation411.11%17
CTNNB1Wnt/β-catenin signaling38.33%37
IL1BCytokine signaling38.33%16

Overall, these patterns highlight a research emphasis on interconnected oncogenic and immuno-inflammatory pathways, demonstrating that herbal-network-pharmacology investigations predominantly target biologically relevant hubs associated with cancer, metabolic disorders, and inflammation-related diseases.

The results show that ESR1 (27.78%), EGFR (25%), and AKT1 (22.22%) were the most frequently investigated proteins, followed by inflammation-related factors such as TNF (19.44%) and CASP3/PTGS2 (16.67% each). Targets predominantly reflected pathways of cancer progression, inflammatory signaling, and metabolic regulation.

3.8 Distribution of docked phytochemicals across included studies

Across the included studies, phytochemical docking was more prevalence with focused on flavonoid compounds, with quercetin (25%),24 kaempferol (13.9%),16 apigenin (11.1%), and luteolin (8.3%) being the most frequently evaluated ligands as presented in Table 7. Phytosterols such as β-sitosterol (8.3%) and polyphenols including gallic acid (5.6%) and EGCG (5.6%) also appeared consistently. The remaining compounds (75%) were evaluated in single studies, indicating ongoing screening of diverse herbal metabolites spanning alkaloids, terpenoids, lignans, catechins, and phenolic acids.

Table 7. Distribution of docked phytochemicals across included studies.

PhytochemicalFrequency (n)Percentage (%) Reference
Quercetin925.0%24
Kaempferol513.9%16
Apigenin411.1%32
Luteolin38.3%34
β-Sitosterol38.3%23
Epigallocatechin gallate (EGCG)25.6%43
Gallic acid25.6%37
Emodin12.8%13
Genistein12.8%13
Naringenin12.8%24
Isovitexin12.8%35
Pipercine/Piperine12.8%37
Resveratrol12.8%37
Allicin/Diallyl trisulfide12.8%34
Silibinin12.8%29
Ecdysterone/20-HE12.8%23
Others (unique single-study phytochemicals)2775.0%16

Across the included studies, flavonoids dominated docking analyses, with quercetin (25%), kaempferol (13.9%), apigenin (11.1%), and luteolin (8.3%) being the most frequently evaluated compounds. Other commonly utilized phytochemicals included β-sitosterol (8.3%), gallic acid (5.6%), and EGCG (5.6%). Alkaloids, phenolic acids, steroidal compounds, and saponins appeared less frequently, typically in disease-specific screening contexts.

3.9 Distribution of reported binding affinities across included studies

The binding affinity ranges reported across the included studies show a consistent trend towards strong ligand–protein interactions as presented in Table 8. Very strong docking scores (≤ −9.0 kcal/mol) were documented in 19.4% of studies, while a further 16.7% reported strong interactions within the −8.0 to −8.9 kcal/mol range. Moderate binding affinity (−7.0 to −7.9 kcal/mol) was observed in 13.9% of studies, with an additional 13.9% reporting moderate binding values between −6.0 and −6.9 kcal/mol.24 Weak-to-moderate affinity scores (−5.0 to −5.9 kcal/mol) accounted for 13.9% of studies, and only 5.6% reported values above −5.0 kcal/mol. A small proportion (8.3%) used broad binding ranges spanning weak to very strong interactions (−3.0 to −11.6 kcal/mol),23 while 8.3% did not provide explicit numerical affinity values. These patterns indicate that most phytochemicals evaluated demonstrated favourable binding strength, supporting their potential as bioactive candidates in computational drug discovery workflows.

Table 8. Distribution of reported binding affinities across included studies.

Binding Affinity Range (kcal/mol)Frequency (n)Percentage (%) Reference
≤ −9.0 (very strong binding)719.4%13
−8.0 to −8.9 (strong binding)616.7%22
−7.0 to −7.9 (moderately strong)513.9%32
−6.0 to −6.9 (moderate affinity)513.9%24
−5.0 to −5.9 (weak-to-moderate)513.9%20
> −5.0/minimal affinity25.6%19
Mixed ranges across all categories (broad −3 to −11.6)38.3%16
Not reported/unclear38.3%35

Across the included studies (n = 36), the majority of phytochemicals demonstrated strong binding affinity, with 19.4% reporting very strong docking scores (≤ −9.0 kcal/mol) and 16.7% showing strong affinities (−8.0 to −8.9 kcal/mol). Moderate binding was observed in ~28% of studies, while weak/variable affinity (> −5 kcal/mol) was uncommon. Approximately 8.3% of studies did not specify binding values.

3.10 Distribution of key molecular pathways identified across included studies

Across the included studies, a total of 142 pathway mentions were identified as sumarised by Figure 7. The analysis showed that PI3K–Akt signaling accounted for the largest share (16.90%),20 followed by MAPK/ERK (14.08%)24 and NF-κB signaling (12.68%), indicating a strong research focus on core cell-survival, proliferation, and inflammation mechanisms.21 Immune and cytokine-related pathways, including TNF, IL-17, and TLR cascades, represented 11.27% of reported pathways, further highlighting the emphasis on inflammation-driven disease processes. Other frequently studied mechanisms included Wnt/β-catenin signaling (7.75%), apoptosis pathways (7.04%), p53-mediated DNA-damage response (6.34%), and VEGF-linked angiogenesis pathways (6.34%). Pathways associated with oxidative stress (AGE–RAGE; 5.63%) and hypoxia response (HIF-1; 4.93%) were also commonly evaluated. In contrast, neurotransmission-related pathways (4.23%) and antiviral signaling mechanisms (2.82%) were less frequently reported.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure7.gif

Figure 7. Distribution of key molecular pathways identified across included studies.

Distribution of molecular signaling pathways investigated across included studies based on total pathway mentions (N = 142). PI3K–Akt (16.90%), MAPK/ERK (14.08%), and NF-κB (12.68%) pathways accounted for the largest proportions, followed by inflammatory cytokine signaling (11.27%), Wnt/β-catenin (7.75%), apoptosis regulation (7.04%), and p53 and VEGF-related pathways (6.34% each). Oxidative stress (5.63%) and hypoxia responses (4.93%) were moderately represented, while neurotransmission (4.23%) and antiviral signaling (2.82%) were least frequently reported.

3.11 ADMET/drug-likeness tools identified in included studies

Across the included studies, ADMET and drug-likeness screening was commonly integrated into computational herbal research workflows as summarized in Table 9. SwissADME19 and TCMSP (OB & DL) were the most frequently used tools, each reported in 17.14% of studies. Lipinski’s rule of five was applied in 14.29% of the studies, highlighting consistent screening for oral bioavailability and small-molecule drug-likeness. Other predictive platforms such as admetSAR (8.57%), QikProp (5.71%), ProTox-II (2.86%), and pkCSM (2.86%) were used less frequently, suggesting selective adoption of advanced pharmacokinetic and toxicity profiling tools. DFT-based stability screening and MMGBSA/MDS simulations appeared in isolated studies (2.86% each), indicating emerging integration of quantum and dynamic computational methods. Notably, 28.57% of studies did not report performing ADMET evaluation, underscoring variability in reporting rigor across the literature.

Table 9. ADMET/Drug-likeness tools identified in included studies.

ADMET/Drug-likeness toolFrequency (n)Percentage (%) Reference
SwissADME617.14%27
TCMSP ADME (OB & DL)617.14%24
Lipinski’s Rule of Five514.29%19
admetSAR38.57%25
QikProp (Schrödinger)25.71%29
ProTox-II 12.86%25
pkCSM12.86%25
PreADMET + Osiris12.86%30
Way2Drug/PASS (toxicity)12.86%26
MMGBSA/MDS (RMSD/RMSF)12.86%27
Density Functional Theory (DFT)12.86%22
Not specified/not performed1028.57%17

Distribution of ADMET and drug-likeness evaluation tools across included studies. SwissADME and TCMSP screening (each 17.14%) were the most commonly used approaches, followed by Lipinski’s criteria (14.29%) and admetSAR (8.57%). Advanced computational pharmacokinetic tools such as QikProp, pkCSM, and ProTox-II were used less frequently (2.86%–5.71%). A considerable proportion of studies (28.57%) did not specify an ADMET assessment tool.

3.12 Molecular dynamics simulation usage

Across the 36 included studies, 17 studies (47.22%) incorporated molecular dynamics (MD) simulations as part of their computational workflow, while 19 studies (52.78%) did not perform MD simulations as depicted by Figure 8. This indicates that although MD simulation has emerged as a valuable technique for validating ligand–protein interactions and assessing structural stability, its adoption remains moderate, with just under half of studies integrating MD into their network-pharmacology or molecular docking analyses. Studies that employed MD simulations typically ran simulations ranging from 10 ns to 300 ns, often accompanied by complementary analyses such as MM-GBSA binding free energy calculation, RMSD, RMSF, PCA, FEL, and DCCM, demonstrating increasing methodological rigor among advanced studies.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure8.gif

Figure 8. Distribution of molecular dynamics simulation usage among included studies.

The results show that 47.22% (n = 17) of the studies conducted molecular dynamics simulations, while 52.78% (n = 19) did not incorporate MD procedures. This reflects a moderate adoption of MD simulation in natural-product-based computational drug-discovery studies.

3.13 Mechanistic evidence across included studies

Across the included studies, phytochemicals and herbal formulations demonstrated broad pharmacological activity mediated through multiple molecular pathways as summarized in Table 10. The most frequently reported mechanisms involved suppression of inflammatory mediators and immune signaling, particularly inhibition of NLRP3 inflammasome activation, NF-κB signalling, and pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6. A substantial proportion of studies also described anticancer effects driven by modulation of key oncogenic pathways including PI3K/Akt, MAPK,10,19 HIF-1α,20 and EGFR, alongside induction of apoptosis, cell-cycle arrest, and caspase activation. Neuroprotective effects were reported through attenuation of oxidative stress, stabilization of neuronal signaling, and inhibition of acetylcholinesterase activity. Additional mechanisms included metabolic and vascular regulation, antiviral activity (particularly SARS-CoV-2 and HIV-related targets), enhancement of intestinal barrier function, and modulation of endocrine signaling pathways. Overall, the results highlight a consistent trend toward multi-target and systems-level regulatory mechanisms, supporting the therapeutic relevance of network-guided phytochemical research.

Table 10. Summary of mechanistic actions across studies.

Mechanistic categoryKey actionsExample targets/PathwaysRepresentative compounds Reference
Anti-inflammatory and Immune ModulationSuppresses inflammatory mediators, blocks inflammasomes, improves intestinal barrierNLRP3, NF-κB, TNF-α, IL-6, IL-1βAsiaticoside, Quercetin, Herbacetin, Costunolide derivatives23
Anti-cancer/Anti-proliferative EffectsInhibits tumor growth, induces apoptosis, blocks cell-cycle progressionEGFR, PI3K/Akt, MAPK, TP53, Cyclins (CDK1, Cyclin B1)Acacetin, Sophoranone, Scutellarein, Episesamin22,23
Cell Cycle Arrest & Apoptosis InductionG2/M arrest, caspase activation, Bcl-2 downregulationCDK1, CDC25A, PLK1, Caspase-3Biochanin A, Myrsininone A12,20
Antioxidant and Oxidative-Stress ReductionEnhances antioxidant enzymes, reduces ROSNrf2-related antioxidant defenseEllagic Acid, EGCG, Kaempferol19
Neuroprotective/Neuro-modulatory Protects neurons, modulates neurotransmittersNLRP3, acetylcholinesteraseAsiaticoside, AChE inhibitors17
Vascular/Metabolic RegulationImproves vascular function, lowers blood pressure, anti-diabetic enzyme inhibitionPI3K/Akt, AMPK, α-amylase, α-glucosidaseAllicin, Skimmin, Umbelliferone21
Antiviral/Host-Immunity SupportViral enzyme inhibition (COVID-19/SARS-CoV-2, HIV)3CLPro, HIV-PR, CXCR4, PD-1/PD-L1Emodin, Genistein40
Gut Barrier & Microbiome SupportEnhances intestinal barrier integrity, regulates gut inflammationRAGE/SLC6A14, STAT3JGD phytochemicals16
Hormone/Endocrine ModulationModulates estrogen/androgen signalingESR1, CYP19A1, 5α-reductaseIsowighteone, Licochalcone A20
Multi-target Network RegulationBroad modulation of interconnected targets and pathwaysPI3K/Akt, MAPK, HIF-1α, VEGFVarious poly-herbal extracts10

Natural compounds and herbal formulations demonstrated multi-target therapeutic actions across inflammation, cancer, metabolic regulation, neuroprotection, antiviral responses, gut barrier function, and hormonal pathways. Core mechanisms included suppression of pro-inflammatory mediators, modulation of PI3K/Akt and MAPK pathways, induction of apoptosis, inhibition of oxidative stress, and regulation of immune and neurotransmitter signaling. These findings support the systems-based therapeutic potential of phytochemicals in diverse chronic disease models.

3.14 Validation modality distribution

Across the 36 studies, in silico validation was the most frequent approach (58.33%), followed by combined in vitro + in silico designs (25.00%)16 as depicted in Figure 9. Fewer studies implemented laboratory animal validation,24 either with in vitro only (2.78%), in vivo + in silico (5.56%),23 or in vitro + in vivo without computational components (8.33%).10,19 No study used in vivo validation alone.

8c6d60e0-a45c-4c2d-a73d-c2827051dbae_figure9.gif

Figure 9. Validation modality distribution.

4. Discussion

This systematic review assessed contemporary applications of network pharmacology combined with molecular docking in herbal medicine research from 2010 to 2025. The findings demonstrate that computational ethnopharmacology has progressed significantly during the last decade, particularly from 2020 onward. This represents global advances in artificial intelligence-driven drug discovery and increased scientific interest in natural products during the COVID-19 era.6 Notably, research activity increases significantly in 2024, highlighting that this field is growing and gaining momentum in mainstream biomedical science.9 Majority of the studies were originated from Asia, particularly China and India, accounting for over 80% of the total output. This geographic trend aligns with global patterns in which Traditional Chinese Medicine (TCM) and Ayurvedic herbal systems continue to drive innovation in plant-based computational pharmacology.4 The World Health Organization has similarly reported that East and South Asian nations lead in integrating computational tools into traditional medical systems.45 In contrast, regions rich in medicinal biodiversity such as Africa and South America remain under-represented. This represents a missed opportunity for global natural product discovery and emphasizing the need for wider international research participation and investment.46

Across the retrieved literature, there was a strong methodological preference for in silico-focused investigations, with over 60% of studies employing purely computational workflows. This confirms broader observations from recent reviews that computational strategies have become central to early-stage natural product screening.47 These strategies have been reported to be associated with improve target prediction, reduce laboratory cost, and minimize animal use.48 However, this trend simultaneously highlights an important translational gap. Few studies combined computational techniques with wet-lab validation, and even fewer extended to animal experimentation. These findings are consistent with prior analyses indicating insufficient experimental verification in network-pharmacology studies, which has raised concerns regarding reproducibility and biological relevance.5 Strengthening bench-to-bedside translation thus remains essential, particularly as network pharmacology continues to expand in clinical and drug-development frameworks.9

Furthermore, molecular dynamics simulation represented an emerging but not universal component of validation, being applied in nearly half of the studies. This adoption rate is higher than earlier reviews reported between 2020–2022, suggesting increasing methodological advancement.3 Nevertheless, variations in simulation duration, reporting formats, and analytic depth indicate that methodological standardization remains incomplete. Likewise, drug-likeness and ADMET filters, were widely acknowledged as necessary, were inconsistently applied, nearly one-third of studies failed to report such screening. This variability is consistent with international observations that computational phytomedicine lacks unified quality standards, reinforcing recent calls for harmonized methodological guidelines.2,49

Therapeutically, the studies addressed different disease systems. The strongest emphasis was observed in cancer, inflammation, immune dysregulation, metabolic disorders, infectious diseases, and neurological conditions. This pattern reflects the global disease burden and growing interest in systems-level natural-product therapeutics.46 Several mechanistic signatures appeared consistently across the literature.50 Phytochemicals frequently modulated PI3K-Akt, MAPK, NF-κB, IL-17, and apoptotic pathways, demonstrating their poly-target properties and supporting the rationale for network-based mechanistic inquiry. This aligns with international findings showing that natural compounds exhibit multi-pathway immunomodulatory and anti-cancer properties and tend to target central biological hubs rather than single pathways.1 In addition, the review identified a consistent focus on flavonoids like quercetin, kaempferol, luteolin, and apigenin molecules well documented in global research for broad anti-inflammatory, antioxidant, and anticancer activity.51,52 However, this reliance on widely studied phytochemicals also highlights a limitation where there is a risk that research pipelines may converge repeatedly on the same metabolites, restricting novel compound discovery. Future research should therefore balance investigation of established phytochemicals with systematic target on the lesser-studied plant metabolites, many of which remain pharmacologically uncharacterized.49 Another notable trend in this review is the frequent use of multi-herbal formulations, especially in studies originating from China and India. This contrasts with Western phytopharmacology, which traditionally prioritizes single-compound drug discovery.53 Multi-herbal computational analysis is particularly relevant for traditional medical systems that rely on poly-herbal synergy. However, it also presents challenges, as validating biological interactions within complex herbal mixtures requires advanced computational and experiment to avoid spurious associations.2

The review also revealed concentrated reliance on specific databases and platforms, including TCMSP, SwissTargetPrediction, STRING, UniProt, KEGG, and GeneCards. These tools indicate modern phytochemical research, dependence on primarily Asian phytochemical databases.14,38 Increased adoption of African and South American plant databases, integration of metabolomics, and broader utilization of global ethnobotanical repositories would improve heterogeneity and enhance natural-compound discovery pipelines.54 The relatively limited use of omics-driven disease-gene databases suggests that full integration of transcriptomic and proteomic signatures into herbal network pharmacology is still developing. As multi-omics approaches become more accessible, future studies should leverage proteogenomic data to improve disease-mechanism fidelity.55

5. Conclusion

Overall, this systematic review demonstrates that network pharmacology and molecular docking have become fundamental components of modern herbal research and are increasingly supported by advancement in computational tools. The convergence of AI, cheminformatics, multi-omics, and natural product pharmacology offers an important potential for drug discovery. However, achieving translational relevance will require stronger validation pipelines, expanded global participation, more consistent methodological standards, and increased discovery of novel plant-based molecules beyond the commonly investigated flavonoids. However, achieving meaningful translational impact requires addressing several gaps. Future studies should prioritize standard validation pipelines integrating in vitro, in vivo, and preferably clinical evidence to substantiate computational predictions. Methodological standardization, transparent reporting of ADMET and molecular dynamics analyses, and greater use of multi-omics data (e.g., proteomics, metabolomics, single-cell sequencing) will strengthen reproducibility and comparability across studies. Importantly, global participation must be broadened to include more contributions from Africa, Latin America, and other biodiversity-rich regions that remain under-represented despite rich traditional medical knowledge.

6. Limitations of the study

Despite providing a comprehensive synthesis of network pharmacology and molecular docking applications in herbal medicine, several limitations should be acknowledged. First, the review was restricted to studies published in English, which may have excluded relevant research, particularly from regions where herbal medicine innovations are frequently reported in local languages. Second, although three leading scientific databases were searched (PubMed, Scopus, and Web of Science), studies indexed in regional repositories or preprint platforms were not included, potentially limiting coverage of emerging work in low-resource research settings. Third, heterogeneity in reporting standards across studies posed challenges for direct comparison. Numerous articles did not clearly document experimental parameters such as solvent systems, extraction conditions, docking software versions, ADMET screening tools, or molecular dynamics protocols, leading to variability in methodological clarity and rigor. Fourth, a substantial proportion of included studies relied solely on in silico approaches without in vitro or in vivo validation, restricting the ability to fully assess biological relevance and translational readiness of computational predictions.

Additionally, the field lacks standardized quality-assessment frameworks for computational herbal pharmacology, and no formal risk-of-bias tool exists for network pharmacology studies, limiting the ability to systematically evaluate study quality. Publication bias is also possible, as studies reporting strong binding affinities or positive mechanistic findings are more likely to be published. Finally, the evolving nature of computational platforms and rapid developments in AI-driven drug-discovery tools mean that some techniques used in earlier studies may now be obsolete, potentially affecting comparability across the review period.

7. Recommendations

Based on the findings of this systematic review, future research and policy efforts should prioritize strengthening methodologies and translational potential in herbal pharmacology. Researchers are encouraged to adopt fully integrated computational pipelines that combine network pharmacology, molecular docking, ADMET profiling, and molecular-dynamics simulation before proceeding to in vitro and in vivo validation. Standardized reporting guidelines for computational herbal studies should also be developed to improve reproducibility and cross-study comparability. In addition, funding bodies and research institutions should support capacity building in computational drug discovery, particularly in low- and middle-income countries, to enhance global research equity. Collaborative frameworks between computational scientists, phytochemists, pharmacologists, and clinicians are essential to accelerate the translation of in silico findings into clinically relevant phytotherapeutics. Finally, fostering open-access databases for phytochemicals, target interactions, and experimental outcomes will facilitate transparency, innovation, and equitable access to scientific resources in natural-product-based drug discovery.

Declarations

Ethics approval and consent to participate

Not applicable. This study is a systematic review based entirely on previously published research and did not involve human participants, animals, or the collection of new biological samples.

Consent for publication

Not applicable. This manuscript does not contain individual person’s data in any form.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary files. Additional materials used during screening and extraction are available from the corresponding author on reasonable request.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 09 Dec 2025
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Terkimbi SD, Mujinya R, Kayanja KL et al. Integrative Network Pharmacology and Molecular Docking Approaches in Herbal Medicine Research. A Systematic Review of Applications, Advances, and Translational Potential [version 1; peer review: awaiting peer review]. F1000Research 2025, 14:1384 (https://doi.org/10.12688/f1000research.173985.1)
NOTE: If applicable, it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status:
AWAITING PEER REVIEW
AWAITING PEER REVIEW
?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 09 Dec 2025
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.