Pulmonary fibrosis (PF) has a complex history, with its understanding evolving significantly over the past century. Initially, the term “pulmonary fibrosis” was used to describe various lung diseases characterized by scarring of lung tissue. The most common form, idiopathic pulmonary fibrosis (IPF), was first recognized in the 1960s, although its symptoms had been documented earlier. The Pulmonary Fibrosis Foundation (PFF) was established in 2000 by the Rosenzweig brothers, motivated by personal loss to the disease. This marked a pivotal moment in advocacy and research, leading to increased awareness and funding for PF research.
Over the years, significant advancements have been made in understanding the disease’s pathophysiology, with a focus on the role of inflammation and fibrosis in lung tissue. The introduction of antifibrotic medications like pirfenidone and nintedanib in the 2010s represented a breakthrough in treatment, providing options to slow disease progression.
As of 2025, pulmonary fibrosis affects over 250,000 individuals in the United States alone, with a growing prevalence worldwide. The disease is characterized by progressive scarring of lung tissue, leading to severe respiratory issues and a median survival rate of 3 to 5 years post-diagnosis. Current treatment strategies focus on managing symptoms and slowing progression, as there is no cure. The landscape of PF treatment is rapidly evolving, with more than 100 therapies currently in development globally, indicating a robust pipeline aimed at addressing this challenging condition.
Key points
- Emerging evidence for nutraceuticals: Research suggests that certain nutraceuticals, such as EGCG, quercetin, and resveratrol, may help mitigate pulmonary fibrosis symptoms by targeting inflammation, oxidative stress, and fibrotic pathways, though human studies are limited and results vary.
- Preclinical promise with human gaps: Animal models indicate potential antifibrotic effects for supplements like melatonin and astragaloside IV, but clinical trials in humans are sparse, and benefits are not yet conclusively proven.
- Safety and dosage considerations: Most of these compounds appear safe at moderate doses based on available data, but interactions and long-term effects remain understudied, especially in patients with pulmonary fibrosis.
- Holistic approach recommended: While evidence leans toward supportive roles in reducing fibrosis progression, nutraceuticals should complement, not replace, standard treatments like antifibrotic drugs, and consultation with healthcare providers is essential due to individual variability.
- Ongoing research needed: The field shows promise but highlights controversies around efficacy, with some studies reporting trends rather than definitive improvements; larger, well-designed trials could clarify their therapeutic potential.
Overview of pulmonary fibrosis and nutraceutical interventions
Pulmonary fibrosis is a progressive lung disease involving scarring that impairs breathing, often idiopathic (IPF) or linked to environmental factors. Standard treatments slow progression but don’t reverse damage. Nutraceuticals—natural compounds with health benefits—offer potential adjunctive support by addressing underlying mechanisms like inflammation and collagen buildup. This review focuses on 10 selected supplements, drawing from preclinical and limited human data.
Potential mechanisms and benefits
These nutraceuticals primarily act as antioxidants, anti-inflammatories, or modulators of fibrotic signaling (e.g., TGF-β pathways). For instance, EGCG from green tea may inhibit collagen cross-linking, while omega-3 fatty acids correlate with better lung function in observational studies. Benefits could include reduced symptom severity and slower disease advancement, but effects are context-dependent and not universal.
Limitations and future directions
Evidence is mostly from animal models, with human trials small-scale or observational. While promising, results are not absolute, and factors like bioavailability vary. Future research should prioritize randomized controlled trials to validate these approaches.
Abstract
Pulmonary fibrosis (PF), including idiopathic pulmonary fibrosis (IPF), is a chronic, progressive interstitial lung disease characterized by excessive extracellular matrix deposition, leading to irreversible lung scarring and impaired respiratory function. Current pharmacological therapies, such as nintedanib and pirfenidone, slow disease progression but offer limited reversal of established fibrosis. Nutraceuticals, derived from natural sources, have garnered attention for their potential antifibrotic, anti-inflammatory, and antioxidant properties. This review synthesizes preclinical and clinical evidence on 10 selected nutraceuticals—EGCG (epigallocatechin gallate from green tea), quercetin, omega-3 fatty acids, resveratrol, melatonin, astragaloside IV (from Astragalus root), neferine (from lotus seed), andrographolide (from Andrographis paniculata), cordycepin (from Cordyceps sinensis mushroom), and fucoidan (from brown seaweed)—in combating PF. Drawing from animal models (e.g., bleomycin-induced PF) and limited human studies, we discuss mechanisms such as TGF-β inhibition, Nrf2 activation, and senescence clearance. While promising, human data remain preliminary, emphasizing the need for larger trials to establish efficacy and safety.
Introduction
Pulmonary fibrosis encompasses a group of disorders marked by fibroblast activation, epithelial-mesenchymal transition (EMT), and collagen accumulation, often triggered by environmental insults like bleomycin (BLM), silica, or radiation. IPF, the most common form, affects over 3 million people globally and carries a poor prognosis, with median survival of 3-5 years post-diagnosis. Pathophysiological hallmarks include dysregulated TGF-β signaling, oxidative stress, inflammasome activation, and cellular senescence. Nutraceuticals offer a complementary strategy, potentially modulating these pathways with fewer side effects than conventional drugs. This paper reviews evidence for the specified compounds, integrating user-provided insights with recent literature, focusing on published studies.
EGCG (Epigallocatechin Gallate from green tea)
EGCG, a polyphenol abundant in green tea, exhibits antifibrotic potential through inhibition of lysyl oxidase-like 2 (LOXL2) and TGF-β1 signaling, reducing collagen cross-linking. A small human study (n=10 IPF patients) demonstrated oral EGCG (400-1200 mg/day) reversed TGF-β1-driven profibrotic markers in lung biopsies after 14 days. Ongoing Phase I trials evaluate safety and dosing, with doses up to 800 mg/day deemed safe. Preclinically, EGCG attenuates BLM-induced PF in mice via Nrf2 activation, promoting epithelial regeneration.
Additional research supports these findings. For example, a 2019 study using next-generation sequencing showed EGCG alters gene expression in IPF fibroblasts, reducing oxidative and fibrotic responses.1 In precision-cut lung slices from IPF patients, EGCG induced collagen turnover by blocking LOXL2 and TGF-β1 receptor kinase.2 A 2024 study highlighted EGCG’s role in reducing secreted frizzled-related protein 2 (sFRP2) near alveolar epithelial cells, mitigating EMT.3 However, interactions with drugs like nintedanib warrant caution, as EGCG may alter bioavailability.4 Derivatives like ABI-171 show enhanced efficacy in BLM mouse models.5
Quercetin
Quercetin, a flavonoid with antioxidant and senolytic properties, targets senescence-associated secretory phenotype (SASP) in fibrotic lungs. Pilot human trials (n=14-20 IPF patients) using dasatinib + quercetin demonstrated safety, reduced frailty, and senescence burden, though pooled analyses showed no major functional gains.6,7 Preclinically, it reduces senescence and fibrosis in BLM/silica models, reversing EMT and collagen deposition (equivalent to 500 mg/day human dose). Bioavailability improves with piperine.
Human studies include a 2023 Phase I trial confirming tolerability in IPF patients.6 A 2019 open-label pilot reported physical function improvements.7 Mechanistically, quercetin clears senescent fibroblasts, as shown in a 2017 study.8 A 2025 review discusses its gene regulatory roles in IPF comorbidities.9 In mice, it inhibits macrophage senescence in silica-induced PF.10
Omega-3 fatty acids
Omega-3 fatty acids (e.g., EPA, DHA) exert anti-inflammatory effects, potentially slowing PF progression. Observational data from over 300 IPF patients link higher plasma levels to improved forced vital capacity (FVC), slower decline, and longer transplant-free survival. No direct IPF trials exist, but preclinical evidence supports lung injury protection.
A 2024 study associated higher omega-3 levels with better PF outcomes.11 In cystic fibrosis (CF), related to fibrotic lung changes, supplementation improved pulmonary function.12 A 2016 pilot in CF patients showed enhanced exercise tolerance after 1 year.13 An 8-month CF trial reported fatty acid profile shifts and clinical benefits.14 Symposium data from 2016 noted PF patient outcomes, indirectly supporting anti-inflammatory roles.15
Resveratrol
Resveratrol activates SIRT1, promoting myofibroblast apoptosis and reducing ECM via NF-κB inhibition. Multiple animal studies (BLM/RA-ILD models) show fibrosis attenuation, with 50% collagen reduction and improved survival. Polydatin enhances bioavailability (3-4x). Human trials (150-500 mg/day) improve endothelial function; synergy with polydatin in peritoneal fibrosis suggests lung potential.
In a 2023 study, resveratrol mitigated diesel exhaust-induced PF in mice via SIRT1/FoxO3.16 BLM rat models showed alleviation via HIF-1α/NF-κB suppression.17 It inhibits miR-21 through MAPK/AP-1.18 TGF-β/Smad/ERK modulation was evident in 2023 research.19 EMT downregulation via TLR4/NF-κB and TGF-β1/Smad3 occurred in rats.20
Melatonin
Melatonin, with antioxidant properties, prevents fibrosis via Hippo/YAP1 and EMT inhibition in BLM/PM2.5 models, improving mitochondrial function. Strong preclinical evidence shows attenuation and partial reversal of established PF.
A 2023 mouse study showed NRF2 activation and galectin-3 inhibition.21 In 2002, it reduced BLM-induced fibrosis in rats.22 ER stress and EMT were inhibited in 2014 mouse models.23 COX-2 modulation alleviated BLM effects in 2015.24 PM2.5-induced PF was prevented in 2024 via EMT targeting.25
Astragaloside IV (from Astragalus Root)
Astragaloside IV regenerates via PTEN/PI3K inhibition and stem cell mobilization. A meta-analysis of 10 animal studies (n=142) showed significant PF score reversal (SMD -2.5) in BLM/silica models (20-80 mg/kg). Safe in TCM for lung conditions, it promotes epithelial repair.
A 2025 meta-analysis confirmed therapeutic effects.26 In COPD-related PF, it targeted RAS/RAF/FoxO.27 Circ_0008898/miR-211-5p/HMGB1 axis was restrained in 2024.28 Synergy with ferulic acid modulated TGF-β1/Smad3.29 LncRNA-ATB/miR-200c/ZEB1 inhibited EMT.30
Neferine (Bisbenzylisoquinoline alkaloid from Lotus Seed)
Neferine reverses EMT via TGF-β suppression, attenuating BLM/amiodarone-induced PF in rats, restoring SOD and reducing MDA (10-40 mg/kg). Human anti-inflammatory effects suggest lung regeneration via apoptosis.
A 2010 study attenuated BLM-induced PF.31 Protective against amiodarone in 2013 mice.32 Inhibited hepatic stellate cells, relevant to PF.33 Pharmacokinetics with amiodarone in 2011.34 Inhibited activation via TGF-β1/Smads/ERK in 2023.35
Andrographolide (from Andrographis paniculata)
Andrographolide inhibits NLRP3 inflammasome and pyroptosis, ameliorating silica/BLM PF (5-20 mg/kg). Radiation models show repair; safe in human respiratory trials (60-180 mg/day).
A 2018 study ameliorated silica PF in mice.36 Protective in BLM mice in 2013.37 Suppressed proliferation via TGF-β1/Smad in 2020.38 Improved MMP-1/TIMP-1 in 2015.39 Reduced cytokines in BLM rats in 2011.40
Cordycepin (from Cordyceps sinensis mushroom)
Cordycepin reverses EMT and mitochondrial stress, alleviating IPF in mice via mitochondrion protection (10-50 mg/kg). A 2024 study reduced collagen; TCM uses for lung tonifying, safe in fatigue trials.
A 2012 study protected against lung fibrosis in rats.41 Ion transport effects in 2008 airway cells.42
Fucoidan (Sulfated Polysaccharide from brown seaweed)
Fucoidan activates Nrf2, degrading ECM in BLM/radiation models (50-100 mg/kg low-MW forms reduce 30-50% fibrosis). Safe in human inflammation trials (1-3 g/day).
A 2023 study isolated fucoidan with anti-IPF activity.43 LMWF inhibited via antioxidants in 2022.44 Attenuated EMT in 2021.45 Inhibited TGF-β1 via ERK in 2019.46 Lung function benefits in viral infections in 2020.47
Summary of evidence levels and mechanisms
Nutraceutical | Primary mechanism | Evidence level | Key models/Studies | Dosage range (Preclinical/Human) |
---|---|---|---|---|
EGCG | LOXL2/TGF-β inhibition, Nrf2 activation | Human (small trials), Preclinical (mice) | BLM-induced PF, IPF biopsies | 400-1200 mg/day |
Quercetin | Senolytic, anti-EMT | Human (pilots n=14-20), Preclinical (mice) | Dasatinib combo, silica models | 500 mg/day equiv. |
Omega-3 Fatty Acids | Anti-inflammatory | Observational (n>300), Preclinical | Plasma levels in IPF, CF trials | Variable (supplements) |
Resveratrol | SIRT1 activation, NF-κB inhibition | Preclinical (rats/mice) | BLM models | 150-500 mg/day |
Melatonin | Antioxidant, EMT inhibition | Preclinical (mice/rats) | BLM/PM2.5 models | Not specified |
Astragaloside IV | PTEN/PI3K inhibition | Meta-analysis (10 studies), Preclinical | BLM/silica | 20-80 mg/kg |
Neferine | TGF-β suppression | Preclinical (mice/rats) | BLM/amiodarone | 10-40 mg/kg |
Andrographolide | NLRP3 inhibition | Preclinical (mice/rats) | Silica/BLM | 5-20 mg/kg; 60-180 mg/day |
Cordycepin | Mitochondrial protection | Preclinical (rats) | Lung fibrosis models | 10-50 mg/kg |
Fucoidan | Nrf2 activation | Preclinical (mice) | BLM/radiation | 50-100 mg/kg; 1-3 g/day |
Discussion
These nutraceuticals target overlapping pathways, suggesting synergistic potential (e.g., EGCG with resveratrol). However, human evidence is limited to small pilots or observational data, with preclinical dominance. Bioavailability challenges (e.g., resveratrol) necessitate formulations like polydatin. Safety profiles are favorable, but drug interactions (e.g., EGCG with nintedanib) require monitoring.
Conclusion
Nutraceuticals offer promising adjunctive strategies for PF, potentially reversing markers via anti-fibrotic mechanisms. Larger RCTs are essential to translate preclinical findings.
Key citations
1Tsai MJ, et al. (2019). The Effects of Epigallocatechin Gallate (EGCG) on Pulmonary Fibroblasts of Idiopathic Pulmonary Fibrosis (IPF)-A Next-Generation Sequencing and Bioinformatic Approach. DOI: 10.3390/ijms20081958
2Wei Y, et al. (2021). Blocking LOXL2 and TGFbeta1 signalling induces collagen I turnover in precision-cut lung slices derived from patients with idiopathic pulmonary fibrosis. DOI: 10.1136/thoraxjnl-2020-215745
3Cohen ML, et al. (2024). A fibroblast-dependent TGF-beta1/sFRP2 noncanonical Wnt signaling axis promotes epithelial metaplasia in idiopathic pulmonary fibrosis. DOI: 10.1172/JCI174598
4Veerman GDM, et al. (2022). The influence of green tea extract on nintedanib’s bioavailability in patients with pulmonary fibrosis. DOI: 10.1016/j.biopha.2022.113101
5Araldi GL, et al. (2024). Development and Evaluation of ABI-171, a New Fluoro-Catechin Derivative, for the Treatment of Idiopathic Pulmonary Fibrosis. DOI: 10.3390/ijms252111827
6Nambiar A, et al. (2023). Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability. DOI: 10.1016/j.ebiom.2023.104481
7Justice JN, et al. (2019). Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. DOI: 10.1016/j.ebiom.2018.12.052
8Schafer MJ, et al. (2017). Cellular senescence mediates fibrotic pulmonary disease. DOI: 10.1038/ncomms14532
9Vásquez-Garzón VR, et al. (2025). Quercetin in Idiopathic Pulmonary Fibrosis and Its Comorbidities: Gene Regulatory Mechanisms and Therapeutic Implications. DOI: 10.3390/genes16080856
10Geng F, et al. (2022). Quercetin Alleviates Pulmonary Fibrosis in Mice Exposed to Silica by Inhibiting Macrophage Senescence. DOI: 10.3389/fphar.2022.912029
11Kim JS, et al. (2024). Associations of Plasma Omega-3 Fatty Acids With Progression and Survival in Pulmonary Fibrosis. DOI: 10.1016/j.chest.2023.09.035
12Bateman RM, et al. (2016). 36th International Symposium on Intensive Care and Emergency Medicine. DOI: 10.1186/s13054-016-1208-6
13Simon MISDS, et al. (2020). Antioxidant Micronutrients and Essential Fatty Acids Supplementation on Cystic Fibrosis Outcomes: A Systematic Review. DOI: 10.1016/j.jand.2020.01.007
14Hanssens L, et al. (2016). The clinical benefits of long-term supplementation with omega-3 fatty acids in cystic fibrosis patients – A pilot study. DOI: 10.1016/j.plefa.2016.03.014
15De Vizia B, et al. (2003). Effect of an 8-month treatment with omega-3 fatty acids (eicosapentaenoic and docosahexaenoic) in patients with cystic fibrosis. DOI: 10.1177/014860710302700152
16Singh N, et al. (2023). Resveratrol mitigates miR-212-3p mediated progression of diesel exhaust-induced pulmonary fibrosis by regulating SIRT1/FoxO3. DOI: 10.1016/j.scitotenv.2023.166063
17Wang Z, et al. (2021). Resveratrol alleviates bleomycin-induced pulmonary fibrosis via suppressing HIF-1alpha and NF-kappaB expression. DOI: 10.18632/aging.202420
18Wang J, et al. (2018). Resveratrol inhibits pulmonary fibrosis by regulating miR-21 through MAPK/AP-1 pathways. DOI: 10.1016/j.biopha.2018.05.104
19Liu L, et al. (2023). Effects of Resveratrol on Pulmonary Fibrosis via TGF-beta/Smad/ERK Signaling Pathway. DOI: 10.1142/S0192415X23500313
20Wang L, et al. (2022). Resveratrol alleviates bleomycin-induced pulmonary fibrosis by inhibiting epithelial-mesenchymal transition and down-regulating TLR4/NF-kappaB and TGF-beta1/smad3 signalling pathways in rats. DOI: 10.1016/j.tice.2022.101953
21Lan YJ, et al. (2023). Melatonin ameliorates bleomycin-induced pulmonary fibrosis via activating NRF2 and inhibiting galectin-3 expression. DOI: 10.1038/s41401-022-01018-x
22Arslan SO, et al. (2002). The effect of melatonin on bleomycin-induced pulmonary fibrosis in rats. DOI: 10.1034/j.1600-079x.2002.10796.x
23Zhao H, et al. (2014). Melatonin inhibits endoplasmic reticulum stress and epithelial-mesenchymal transition during bleomycin-induced pulmonary fibrosis in mice. DOI: 10.1371/journal.pone.0097266
24Karimfar MH, et al. (2015). MELATONIN ALLEVIATES BLEOMYCIN-INDUCED PULMONARY FIBROSIS IN MICE. https://pubmed.ncbi.nlm.nih.gov/26122220/
25Chen PC, et al. (2024). Melatonin prevents pulmonary fibrosis caused by PM(2.5) exposure by targeting epithelial-mesenchymal transition. DOI: 10.1016/j.taap.2024.116949
26Zhang S, et al. (2025). Therapeutic effects and potential mechanisms of astragaloside IV on pulmonary fibrosis: a systematic review and meta-analysis of preclinical studies. DOI: 10.3389/fphar.2025.1564290
27Zhang M, et al. (2023). Astragaloside IV protects against lung injury and pulmonary fibrosis in COPD by targeting GTP-GDP domain of RAS and downregulating the RAS/RAF/FoxO signaling pathway. DOI: 10.1016/j.phymed.2023.155066
28Zhu M, et al. (2024). Astragaloside IV restrains pulmonary fibrosis progression via the circ_0008898/miR-211-5p/HMGB1 axis. DOI: 10.1111/cbdd.14508
29Tong J, et al. (2021). Astragaloside IV Synergizing with Ferulic Acid Ameliorates Pulmonary Fibrosis by TGF-beta1/Smad3 Signaling. DOI: 10.1155/2021/8845798
30Guan Y, et al. (2024). Astragaloside IV inhibits epithelial-mesenchymal transition and pulmonary fibrosis via lncRNA-ATB/miR-200c/ZEB1 signaling pathway. DOI: 10.1016/j.gene.2023.148040
31Zhao L, et al. (2010). Neferine, a bisbenzylisoquinline alkaloid attenuates bleomycin-induced pulmonary fibrosis. DOI: 10.1016/j.ejphar.2009.11.007
32Niu CH, et al. (2013). Protective effects of neferine on amiodarone-induced pulmonary fibrosis in mice. DOI: 10.1016/j.ejphar.2013.06.004
33Ding H, et al. (2011). Neferine inhibits cultured hepatic stellate cell activation and facilitates apoptosis: A possible molecular mechanism. DOI: 10.1016/j.ejphar.2010.10.025
34Wan J, et al. (2011). Effects of neferine on the pharmacokinetics of amiodarone in rats. DOI: 10.1002/bmc.1535
35Ren HL, et al. (2023). Benzylisoquinoline alkaloids inhibit lung fibroblast activation mainly via inhibiting TGF-β1/Smads and ERK1/2 pathway proteins. DOI: 10.1016/j.heliyon.2023.e16849
36Karkale S, et al. (2018). Andrographolide ameliorates silica induced pulmonary fibrosis. DOI: 10.1016/j.intimp.2018.07.012
37Zhu T, et al. (2013). Protective role of andrographolide in bleomycin-induced pulmonary fibrosis in mice. DOI: 10.3390/ijms141223581
38Li J, et al. (2020). Andrographolide ameliorates bleomycin-induced pulmonary fibrosis by suppressing cell proliferation and myofibroblast differentiation of fibroblasts via the TGF-beta1-mediated Smad-dependent and -independent pathways. DOI: 10.1016/j.toxlet.2019.11.003
39Yin JF, et al. (2015). Andrographolide plays an important role in bleomycin-induced pulmonary fibrosis treatment. https://pmc.ncbi.nlm.nih.gov/articles/PMC4612832/
40Zhu HL, et al. (2011). Effects of andrographolide on the concentration of cytokines in BALF and the expressions of type I and III collagen mRNA in lung tissue in bleomycin-induced rat pulmonary fibrosis. https://pubmed.ncbi.nlm.nih.gov/21722520/
41Chen M, et al. (2012). Protective roles of Cordyceps on lung fibrosis in cellular and rat models. DOI: 10.1016/j.jep.2012.06.033
42Yue GG, et al. (2008). Effects of Cordyceps sinensis, Cordyceps militaris and their isolated compounds on ion transport in Calu-3 human airway epithelial cells. DOI: 10.1016/j.jep.2008.01.030
43Wei S, et al. (2023). Isolation, Characterization, and Anti-Idiopathic Pulmonary Fibrosis Activity of a Fucoidan from Costaria costata. DOI: 10.3390/molecules28114343
44Dong H, et al. (2022). Low Molecular Weight Fucoidan Inhibits Pulmonary Fibrosis In Vivo and In Vitro via Antioxidant Activity. DOI: 10.1155/2022/7038834
45Wu N, et al. (2021). Low molecular weight fucoidan attenuating pulmonary fibrosis by relieving inflammatory reaction and progression of epithelial-mesenchymal transition. DOI: 10.1016/j.carbpol.2021.118567
46Wang L, et al. (2019). Low-molecular-weight fucoidan attenuates bleomycin-induced pulmonary fibrosis: possible role in inhibiting TGF-beta1-induced epithelial-mesenchymal transition through ERK pathway. https://pmc.ncbi.nlm.nih.gov/articles/PMC6511757/
47Fitton JH, et al. (2020). Fucoidan and Lung Function: Value in Viral Infection. DOI: 10.3390/md19010004