Next Article in Journal
Analysis of Osmotic Pump-Administered Xylitol in a Syngeneic Mouse Melanoma Model
Previous Article in Journal
A Novel Liposomal Palmitoylethanolamide (PEA) with Enhanced Gastrointestinal Permeating Properties
Previous Article in Special Issue
Anti-Inflammatory and Antinociceptive Potential of Hydroethanolic Extract of Bauhinia guianensis Aubl. in Zebrafish (Danio rerio)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effect of Ageratum Conyzoides on Osteoarthritis in an Ageing Adult Population: A Double-Blind Randomized Placebo-Controlled Parallel Study

1
RDC Clinical, Brisbane 4006, Australia
2
School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane 4072, Australia
3
Gencor Pacific Ltd., Discovery Bay, Hong Kong
*
Author to whom correspondence should be addressed.
Nutraceuticals 2025, 5(4), 35; https://doi.org/10.3390/nutraceuticals5040035
Submission received: 1 September 2025 / Revised: 14 October 2025 / Accepted: 22 October 2025 / Published: 3 November 2025
(This article belongs to the Special Issue Nutraceuticals and Their Anti-inflammatory Effects)

Abstract

This randomized, double-blind, placebo-controlled study investigated Ageratum conyzoides (A. conyzoides) for alleviating osteoarthritis symptoms and improving quality of life. Conducted in Australia between 2021 and 2024, the study included 70 adults aged ≥45 years with clinically diagnosed osteoarthritis. Participants consumed 250 mg of pyrrolizidine alkaloid-free A. conyzoides extract or a placebo daily for 12 weeks. Pain and function were assessed using the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) every three weeks. Secondary measures included pain assessed by the Visual Analogue Scale (VAS), the SF-36 quality-of-life questionnaire, inflammatory markers, and safety parameters. A. conyzoides supplementation resulted in significant reductions in total WOMAC scores at weeks 9 and 12 (p < 0.05) compared to placebo. VAS pain scores were significantly lower at weeks 9 and 12 (p < 0.05). SF-36 scores improved significantly in the pain and role limitations due to physical health domains (p < 0.05). Plasma inflammatory markers IL-6 and IL-8 showed significant reductions compared with placebo (p < 0.05). No between-group differences were observed for adverse events. These findings demonstrate that A. conyzoides supplementation is a safe and effective option for reducing osteoarthritis symptoms, with significant improvements observed in pain, function, and inflammatory markers.

1. Introduction

Osteoarthritis (OA) is the most prevalent joint disorder globally, affecting approximately 9.3% of Australians according to the Australian Bureau of Statistics (2017–2018), with prevalence rapidly increasing among individuals over 45 years of age [1]. The prevalence of OA is estimated to range from 45 to 95 percent among affected populations, often resulting in restricted daily activity, work and school absenteeism, and significant personal distress [2,3]. Pain typically begins with joint use and arises from inflammatory processes within the joint structure, persisting throughout daily activities and significantly impacting quality of life [4,5]. Despite its high prevalence and substantial impact on daily life, OA remains inadequately treated, largely due to a limited understanding of disease mechanisms and lack of disease-modifying therapies [6].
The primary mechanisms underlying OA are not yet fully understood. However, painful symptoms are primarily attributed to the excessive production of inflammatory biomarkers, particularly interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8), which peak during disease progression and contribute to cartilage degradation [2,7]. Elevated inflammatory marker levels are directly linked to increased joint inflammation, irregular cartilage matrix degradation through matrix metalloproteinases (MMPs), and synovial tissue changes, all of which contribute to pain perception and heightened sensitivity of nerve endings [8,9].
Nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin, naproxen, and ibuprofen remain the first-line treatment, inhibiting cyclooxygenase enzymes responsible for inflammatory mediator production [10]. While effective for most patients, frequent NSAID use can cause gastrointestinal complications (e.g., ulcers, bleeding) and systemic side effects such as nausea, dizziness, and organ damage [11,12]. Additionally, a large portion of OA patients do not achieve satisfactory symptom control or are unable to tolerate NSAID therapy. In a cross-sectional study, 50% of patients or their physicians expressed dissatisfaction with NSAID treatment, citing poor efficacy, side effects, or tolerance issues [13]. Moreover, real-world data indicate that approximately 10% of participants with moderate-to-severe OA exhibit NSAID intolerance, highlighting the need for safer, more tolerable alternatives [14,15].
Ageratum conyzoides L. (Asteraceae), commonly known as billygoat weed, is an annual herb native to Africa, Asia, and South America [16]. Ageratum has a long tradition of use in ethnomedicine for a wide range of conditions, including fever, rheumatism, headaches, toothaches, skin disorders, leprosy, inflammatory pain, spasm-related ailments, and as a topical dressing for wounds (e.g., as a febrifuge, purgative, anti-inflammatory agent, and wound healer) [17,18]. This therapeutic versatility is attributed to its diverse pharmacological activities, including antioxidant, antimicrobial, analgesic, and anti-inflammatory properties [16,19].
Phytochemical analysis has identified numerous bioactive compounds in A. conyzoides, including benzofurans, phenols, phenolic esters, alkaloids, coumarins, chromenes, terpenoids, steroids, and flavonoids [16,20]. The plant’s anti-inflammatory activity is particularly associated with its flavonoid fraction, which has been extensively documented for its anti-inflammatory properties [21,22].
Several preclinical studies have demonstrated the anti-arthritic properties of pyrrolizidine alkaloid-free A. conyzoides (A. conyzoides). Bahtiar et al. (2017) found that ethanolic extracts of A. conyzoides at doses of 80 mg and 160 mg per 200 g body weight decreased edema volume, increased articular cartilage area and thickness, and elevated proteoglycan levels in monosodium iodoacetate (MIA)-induced OA rats through anti-inflammatory actions, including inhibition of TNF-α and MMP expression [23]. Similarly, Permawati et al. (2019) demonstrated that a nanoemulgel containing A. conyzoides (in combination with Oldenlandia corymbosa) reduced inflammatory biomarkers in MIA-induced OA rats [24].
Given A. conyzoides’ well-documented analgesic and anti-inflammatory properties, particularly those derived from polymethoxyflavone-rich extracts (e.g., SEPAc), and its demonstrated safety during prolonged therapeutic use, it represents a promising alternative for OA management [21,22]. Although preclinical studies have consistently demonstrated the analgesic and anti-inflammatory effects of A. conyzoides, to date no published double-blind, placebo-controlled clinical trials have evaluated its efficacy in OA management. The present study therefore represents the first such clinical trial, addressing this important gap in the literature. This study aimed to evaluate the effectiveness of A. conyzoides supplementation compared with a placebo in alleviating OA symptoms in ageing adults, hypothesizing that A. conyzoides supplementation would significantly reduce pain severity and improve joint function compared with a placebo.

2. Materials and Methods

2.1. Study Design

This was a 12-week, randomized, double-blind, placebo-controlled clinical trial utilizing an active group (A. conyzoides extract) and a placebo group (microcrystalline cellulose). The study was conducted in Australia between November 2021 and December 2024.

2.2. Study Population

Participants were recruited from an existing participant database and via social media outlets from across Australia. Interested individuals were provided with a participant information sheet and screened against the inclusion and exclusion criteria (detailed below) via telehealth consultation.
Seventy (70) adults aged 45 years and older with clinically diagnosed osteoarthritis were enrolled in this study. Participants were included if they provided informed consent, had a clinical diagnosis of osteoarthritis by a qualified health professional with written evidence, reported joint pain ≥ 10 mm on the Visual Analogue Scale (VAS) not associated with acute injury or prescription medication use, had a BMI ≤ 35 kg/m2, and agreed not to change their current diet or exercise programs during the trial. Participants were also required to agree not to undertake another clinical trial while enrolled. Participants with clinically diagnosed mild-to-moderate osteoarthritis were included; those with severe OA were excluded.
Exclusion criteria included unstable or serious illness (e.g., serious mood disorders, neurological disorders, kidney disease, liver disease, heart conditions, diabetes, or thyroid dysfunction), current malignancy (excluding basal cell carcinoma) or chemotherapy/radiotherapy treatment within the previous two years, clinically significant acute or chronic inflammation or connective tissue diseases (rheumatoid arthritis, tendinitis, bursitis, fibromyalgia, gout), history of knee or hip surgery or trauma within the previous 12 months, use of anticoagulation therapy, or treatment for joint pain including corticosteroids or supplements (without a required four-week washout period). Other exclusion criteria included active smoking or substance abuse, chronic alcohol use (>14 drinks per week), allergy to study ingredients, pregnancy or breastfeeding, participation in related studies within the previous month, or infection within the month prior to study commencement.

2.3. Chemical Analysis

Both the active and placebo products were manufactured according to Good Manufacturing Practice standards (Pharmaceutical Inspection Co-operation Scheme (PIC/S). Guide to Good Manufacturing Practice for Medicinal Products (PE009-17). Geneva: PIC/S, 2023) in a Therapeutic Goods Administration–approved facility. The active product, supplied by Gencor Pacific Ltd. (Lantau Island, Hong Kong), contained 250 mg of pyrrolizidine alkaloid–free Ageratum conyzoides extract. The final composition of the extract was 6% water, 0.75% palmitic acid, 10% saponin glycosides, 5% tannins, 2% bitters, 10% fatty acids, 15% flavonoids, 10% terpenes, 5% maltodextrin, and 36.25% other compounds co-extracted from the aerial parts of A. conyzoides. The placebo consisted solely of microcrystalline cellulose.

2.4. Study Procedure

The study was conducted remotely, with no face-to-face interaction with participants. All communication between investigators and participants was conducted via phone (primary contact method), email, or text message (for reminders). Eligible participants completed baseline measures [self-reported demographics and questionnaires—International Prostate Symptom Score (IPSS), Aging Males Symptoms (AMS) for males, and Women’s Health Questionnaire (WHQ) for females], after which the trial product was mailed to them.
Compliance was monitored using participant self-reported diaries returned at weeks 6 and 12, as well as by capsule counts of unused product at study completion. Participants received a 6-week supply of capsules at baseline and again at week 6. Weekly phone calls and electronic reminders (email or SMS) were used to reinforce adherence and monitor adverse events.
Participants were randomly allocated (1:1 ratio) to receive either 250 mg of A. conyzoides (active) or a placebo in identical opaque capsules. Randomization was performed online from: https://www.sealedenvelope.com/simple-randomiser/v1/lists (accessed on 13 October 2021) by an individual not involved in the trial to ensure both participants and investigators remained blinded to allocation.
Participants consumed one capsule, once daily in the evening with food throughout the 12-week study period. All participants, investigators, and laboratory personnel remained blinded to treatment allocation throughout the study and analysis phases.
Adverse events (AEs) were monitored throughout the study through spontaneous participant reporting and at scheduled study visits. Investigators recorded the type, duration, severity, and possible relationship of each AE to the investigational product. Severity was graded according to standard criteria (mild, moderate, severe), and any serious adverse events were reported immediately to the ethics committee and trial sponsor in accordance with Good Clinical Practice requirements.

2.5. Outcome Measures

The primary outcome measure for this study was osteoarthritis symptom severity assessed using the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC, version 3.1). The WOMAC is a validated self-administered health status measure consisting of 24 questions assessing pain, stiffness, and function in people with osteoarthritis of the hip or knee, completed every three weeks throughout the trial.
Secondary outcome measures included joint pain severity assessed using the VAS where participants scored their morning and evening pain daily for seven days at weeks 3, 6, 9, and 12 on a 100 mm scale (0 = “no pain” to 100 = “worst pain”); the SF-36 Quality of Life Questionnaire completed at baseline, week 6, and week 12; male-specific questionnaires (IPSS and AMS); female-specific questionnaire (WHQ); plasma inflammatory biomarkers (TNF-α, IL-6, IL-8, IL-10, CRP) measured at baseline, week 6, and week 12; safety biomarkers (liver function tests, complete metabolic panel, PSA for males); anthropometric measurements; rescue medication use; and adverse event reporting. Rescue medication use was defined as any self-reported use of analgesics (e.g., ibuprofen, paracetamol) or physiotherapy for OA-related pain, recorded in participants’ study diaries.

2.6. Statistical Analysis

Sample size was calculated to detect a 15-point difference in WOMAC scores between groups with 95% power and α = 0.05. Twenty-seven participants per group were required, with 40 participants per group (80 total) enrolled to account for an expected 30% dropout.
Statistical analyses were conducted using GraphPad Prism 8.0 and SPSS 25. Continuous data were tested for normality using the Shapiro–Wilk test. Changes in outcome measures over time were analyzed using repeated-measures analysis of variance, with time and treatment condition as factors. For post hoc comparisons, paired t-tests evaluated differences between time points within each treatment condition. Between-group differences at each time point were analyzed using unpaired t-tests. Missing data were handled using mixed-effects models. All tests were two-tailed, with statistical significance set at p < 0.05.

2.7. Ethical Considerations

All participants provided written informed consent prior to commencing the study. This study was conducted in compliance with the International Conference on Harmonization Guideline for Good Clinical Practice and the Therapeutic Goods Administration. Ethical approval was obtained from the Bellberry Limited Human Research Ethics Committee (Adelaide, Australia). This clinical trial was registered with the Australian New Zealand Clinical Trials Registry (ACTRN12621000694819). The trial protocol and statistical plan were not publicly archived.

3. Results

Seventy participants (25 male, 45 female) with clinically diagnosed osteoarthritis were randomized (Figure 1). Twelve participants withdrew during the study, leaving 58 who completed the trial and were included in the final analysis (A. conyzoides n = 28 and placebo n = 30). Of the withdrawals, three participants (A. conyzoides n = 2 and placebo n = 1) reported adverse events, none of which were related to the study product. Baseline demographic and anthropometric characteristics were comparable between groups, with no statistical differences observed (Table 1).
Participants with both knee and hip osteoarthritis recorded data for both joints, resulting in data from 84 joints total. Progressive reductions in WOMAC scores occurred over the 12-week intervention period, with the A. conyzoides group showing greater improvements than placebo (Table 2, Figure 2).
Total WOMAC scores were lower in the A. conyzoides group compared to placebo at week 9 (p = 0.031) and week 12 (p = 0.021). Compared to the placebo group, joint pain in the A. conyzoides group decreased significantly at week 12 (Table 2, p < 0.05). Between-group differences were also observed for joint function at weeks 9 and 12, and for pain scores at week 12. No differences were found for joint stiffness at any timepoint.
VAS results were consistent with WOMAC findings, showing significantly lower pain scores in the A. conyzoides group at weeks 9 and 12 compared to placebo (p < 0.05) (Figure 3, Table 3).
The A. conyzoides group had significant improvements in SF-36 scores for “pain” and “role limitations due to physical health” domains compared to placebo group (Table 4). No significant changes were observed in male-specific questionnaires (IPSS and AMS) or female-specific questionnaires (WHQ).
Significant reductions from baseline were observed in the A. conyzoides group for plasma IL-6 (−2.40 ± 5.47 pg/mL, p < 0.05) and IL-8 (−6.35 ± 16.37 pg/mL, p < 0.05) compared to placebo (Table 5). No significant changes were observed for other inflammatory markers.
A significant reduction in rescue medication use over the 12 weeks was seen in the A. conyzoides group (25 to 16 participants), while the placebo group showed no change (21 to 19 participants) (χ2 = 4.97, p = 0.026).
Five adverse events were recorded during the study. Two events in the A. conyzoides group were deemed unrelated to the study product. Three events occurred in the placebo group: one unrelated to the product and two gastrointestinal events (flatulence and nausea) possibly related to treatment.

4. Discussion

This randomized, double-blind, placebo-controlled trial is the first to demonstrate the clinical efficacy of A. conyzoides extract in managing OA symptoms among ageing adults. The daily dose of 250 mg administered over 12 weeks resulted in statistically significant and clinically meaningful improvements in pain, functional capacity, quality of life, and reductions in inflammatory biomarkers.
Progressive reductions in WOMAC scores, particularly within the pain and function domains, suggest the ability of A. conyzoides to target central features of OA symptoms. Improvements increased over the intervention period, reflecting a potential cumulative therapeutic benefit. While the observed effect did not reach the postulated 15-point difference, consistency across secondary measures, including VAS pain scores and SF-36 quality of life assessments, provides strong support for the clinical relevance of these findings [10,25].
Marked decreases in circulating plasma IL-6 and IL-8 concentrations are noteworthy, given their established associations with OA pathology. IL-6 plays a central role in synovial inflammation and the astimulation of matrix metalloproteinases (MMPs) leading to cartilage degradation, while IL-8 contributes to leukocyte infiltration and sustained joint inflammation [7,8]. Attenuation of these cytokines suggests that A. conyzoides may act through anti-inflammatory pathways to reduce structural and symptomatic disease burden. The results align with preclinical studies in animal models showing inhibition of TNF-α and MMP expression following treatment with A. conyzoides extracts [23,24]. In the present study, no between-group differences were observed in TNF-α, CRP or IL-10. This may indicate a mechanism of action targeting a specific anti-inflammatory pathway or a discrete stage within one. Differences from preclinical studies may reflect dose or exposure variation, model design or species context. Further investigations focusing on biomarkers isolated from synovial fluid may provide a clearer delineation of the mechanism.
Osteoarthritis is a multifactorial disease involving multiple cell types, including chondrocytes, synoviocytes, and infiltrating immune cells, with NF-κB signaling, IL-6/IL-8 release, and leukocyte infiltration identified as central drivers of inflammation and joint damage [26,27]. Preclinical studies of A. conyzoides provide mechanistic support for our findings. Flavonoid-rich extracts of A. conyzoides have been shown to suppress NF-κB activation, reduce pro-inflammatory cytokine release (including IL-6 and IL-8), and attenuate leukocyte infiltration in animal models [17,21,22]. These actions align with our observed reductions in circulating IL-6 and IL-8, suggesting that A. conyzoides may exert targeted anti-inflammatory effects relevant to OA pathophysiology.
Reduced use of rescue medication in the A. conyzoides group provides an important indicator of meaningful symptom relief in daily life. Findings also confirmed a favourable safety profile, with no significant alterations in liver function tests or clinical biochemistry, and minimal adverse events, comparable with placebo. As such, A. conyzoides may offer a safer alternative or help reduce reliance on NSAIDs—a clinically significant outcome given the cardiovascular, renal, and gastrointestinal risks associated with long-term NSAID consumption in older adults [3].
The symptomatic benefits observed may derive from the plant’s diverse phytochemical composition. Flavonoids and other bioactive compounds present in A. conyzoides have been reported to modulate inflammatory signalling cascades, inhibit prostaglandin synthesis, and decrease the release of pro-inflammatory mediators [17,21,22].
However, several potential limitations must be acknowledged. The modest sample size may limit statistical power for secondary outcomes and subgroup comparisons. The 12-week intervention period, while sufficient to establish efficacy, does not inform long-term effects, durability of benefit, or optimal dosing schedules. Additionally, participants presented with mild-to-moderate OA; thus, extrapolation to individuals with severe disease may not be appropriate [1,2]. Another limitation is the absence of imaging (X-ray or MRI) to assess structural joint changes such as cartilage thickness or synovial inflammation. Future studies should incorporate imaging analyses to complement symptomatic and biomarker outcomes.
Further investigation is warranted to determine the long-term impact of A. conyzoides on disease progression and potential adverse effects. Trials of extended duration with larger cohorts could evaluate sustained symptom relief, radiographic or MRI-based structural changes, and possible disease-modifying effects. Exploration of synergistic effects with conventional therapies and optimal dosing regimens would also enhance translational value.

5. Conclusions

Daily supplementation with 250 mg of A. conyzoides extract over 12 weeks was associated with significant improvements in pain, physical function, and quality of life in individuals with osteoarthritis, accompanied by reductions in key inflammatory cytokines. Consistent improvements across patient-reported outcomes, functional measures, and biomarker assessments underscores the therapeutic potential of A. conyzoides in OA management. Importantly, the favourable safety profile and reduced reliance on rescue medication further strengthens its suitability in the long-term management of osteoarthritis.
Collectively, the results of this study position A. conyzoides as a promising, safe, and well-tolerated adjunct or alternative to current pharmacological approaches. Further investigations of longer duration and larger sample sizes, incorporating imaging measures, are warranted to confirm efficacy.

Author Contributions

Conceptualization, A.R. and D.B.; Methodology, A.R. and D.B.; Software, A.R. and D.B.; Validation, A.R. and D.B.; Formal Analysis, A.R.; Investigation, A.R. and A.G.; Resources, A.R. and D.B.; Data Curation, A.R., A.G. and D.B.; Writing—Original Draft Preparation, S.S., S.M., N.B. and D.B.; Writing—Review & Editing, A.R., A.G. and D.B.; Visualization, A.R. and D.B.; Supervision, A.R. and D.B.; Project Administration, A.R.; Funding Acquisition, A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research and APC was funded by Gencor Pacific Ltd.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by Bellberry Limited Human Ethics Committee, approval number 2021-03-223, approved date: 14 September 2021.

Informed Consent Statement

All participants provided written informed consent prior to participating in the study.

Data Availability Statement

The datasets generated and/or analysed during the current study are not publicly available due to intellectual property rights but are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors A.R., A.G. and D.B. declare that they have no competing interests. Authors S.S., S.M. and N.B. are employees of the trial sponsor, Gencor Pacific Ltd. These authors had no role in the study design, data collection, analysis, or interpretation, and thus had no influence on the study outcome. The authors declare that this study received funding from Gencor Pacific Ltd. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article or the decision to submit it for publication.

Abbreviations

AEAdverse Events
AMSAgeing Male Symptom
E/LFTEnzyme And Liver Function Test
ILInterleukin
IPSSInternational Prostate Symptom Score
MMPsMatrix Metalloproteinases
MIAMonosodium Iodoacetate
NSAIDSNonsteroidal Anti-Inflammatory Drugs
OAOsteoarthritis
TNFTumor Necrosis Factor
VASVisual Analogue Scale
WOMACWestern Ontario and McMaster Universities Osteoarthritis Index
WHQWomen’s Health Questionnaire

References

  1. Australian Institute of Health and Welfare. Osteoarthritis, What Is Osteoarthritis? Available online: https://www.aihw.gov.au/reports/chronic-musculoskeletal-conditions/osteoarthritis?utm_source=chatgpt.com#description (accessed on 21 August 2025).
  2. Chen, D.; Shen, J.; Zhao, W.; Wang, T.; Han, L.; Hamilton, J.L.; Im, H.-J. Osteoarthritis: Toward a comprehensive understanding of pathological mechanism. Bone Res. 2017, 5, 16044. [Google Scholar] [CrossRef] [PubMed]
  3. Grassel, S.; Muschter, D. Recent advances in the treatment of osteoarthritis. F1000Research 2020, 9, F1000 Faculty Rev-325. [Google Scholar] [CrossRef] [PubMed]
  4. Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A disease of the joint as an organ. Arthritis Rheumatol. 2012, 64, 1697–1707. [Google Scholar] [CrossRef] [PubMed]
  5. Tong, L.; Yu, H.; Huang, X.; Shen, J.; Xiao, G.; Chen, L.; Wang, H.; Xing, L.; Chen, D. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022, 10, 60. [Google Scholar] [CrossRef]
  6. Salman, L.A.; Ahmed, G.; Dakin, S.G.; Kendrick, B.; Price, A. Osteoarthritis: A narrative review of molecular approaches to disease management. Arthritis Res. Ther. 2023, 25, 27. [Google Scholar] [CrossRef]
  7. Molnar, V.; Matišić, V.; Kodvanj, I.; Bjelica, R.; Jeleč, Ž.; Hudetz, D.; Rod, E.; Čukelj, F.; Vrdoljak, T.; Vidović, D.; et al. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int. J. Mol. Sci. 2021, 22, 9208. [Google Scholar] [CrossRef]
  8. Akeson, G.; Malemud, C.J. A Role for Soluble IL-6 Receptor in Osteoarthritis. J. Funct. Morphol. Kinesiol. 2017, 2, 27. [Google Scholar] [CrossRef]
  9. Vuolteenaho, K.; Koskinen, A.; Kukkonen, M.; Nieminen, R.; Paivarinta, U.; Moilanen, T.; Moilanen, E. Leptin enhances synthesis of proinflammatory mediators in human osteoarthritic cartilage--mediator role of NO in leptin-induced PGE2, IL-6, and IL-8 production. Mediat. Inflamm. 2009, 2009, 345838. [Google Scholar] [CrossRef]
  10. Hunter, D.J.; Felson, D.T. Osteoarthritis. BMJ 2006, 332, 639–642. [Google Scholar] [CrossRef]
  11. Fendrick, A.M.; Greenberg, B.P. A review of the benefits and risks of nonsteroidal anti-inflammatory drugs in the management of mild-to-moderate osteoarthritis. Osteopath. Med. Prim. Care 2009, 3, 1. [Google Scholar] [CrossRef]
  12. Wongrakpanich, S.; Wongrakpanich, A.; Melhado, K.; Rangaswami, J. A Comprehensive Review of Non-Steroidal Anti-Inflammatory Drug Use in The Elderly. Aging Dis. 2018, 9, 143–150. [Google Scholar] [CrossRef] [PubMed]
  13. Taylor, S.D.; Everett, S.V.; Taylor, T.N.; Watson, D.J.; Taylor-Stokes, G. A measure of treatment response: Patient and physician satisfaction with traditional NSAIDs for osteoarthritis control. Open Access Rheumatol. 2013, 5, 69–76. [Google Scholar] [CrossRef] [PubMed]
  14. Magni, A.; Agostoni, P.; Bonezzi, C.; Massazza, G.; Menè, P.; Savarino, V.; Fornasari, D. Management of Osteoarthritis: Expert Opinion on NSAIDs. Pain Ther. 2021, 10, 783–808. [Google Scholar] [CrossRef] [PubMed]
  15. Castro-Dominguez, F.; Vargas-Negrin, F.; Perez, C.; Gutierrez-Prieto, H.; Rebollo, P. Unmet Needs in the Osteoarthritis Chronic Moderate to Severe Pain Management in Spain: A Real Word Data Study. Rheumatol. Ther. 2021, 8, 1113–1127. [Google Scholar] [CrossRef]
  16. Okunade, A.L. Ageratum conyzoides L. (Asteraceae). Fitoterapia 2002, 73, 1–16. [Google Scholar] [CrossRef]
  17. Galati, E.M.; Miceli, N.; Taviano, M.F.; Sanogo, R.; Raneri, E. Anti-inflammatory and Antioxidant Activity of Ageratum conyzoides. Pharm. Biol. 2001, 39, 336–339. [Google Scholar] [CrossRef]
  18. Yadav, N.; Ganie, S.A.; Singh, B.; Chhillar, A.K.; Yadav, S.S. Phytochemical constituents and ethnopharmacological properties of Ageratum conyzoides L. Phytother. Res. 2019, 33, 2163–2178. [Google Scholar]
  19. Adebayo, H.A.; Zheng, G.Z.; Fan, J.T.; Ji, C.; He, W.J.; Xu, J.J.; Zhang, Y.M.; Akindahunsi, A.A.; Kela, R.; Tan, N.H. Biochemical, haematological and histopathological studies of extract of Ageratum conyzoides L. in Sprague Dawley rats. J. Med. Plants Res. 2010, 4, 2264–2272. [Google Scholar]
  20. Chahal, R.; Nanda, A.; Akkol, E.K.; Sobarzo-Sánchez, E.; Arya, A.; Kaushik, D.; Dutt, R.; Bhardwaj, R.; Rahman, M.H.; Mittal, V. Ageratum conyzoides L. and Its Secondary Metabolites in the Management of Different Fungal Pathogens. Molecules 2021, 26, 2933. [Google Scholar] [CrossRef]
  21. Moura, A.C.; Silva, E.L.; Fraga, M.C.; Wanderley, A.G.; Afiatpour, P.; Maia, M.B. Antiinflammatory and chronic toxicity study of the leaves of Ageratum conyzoides L. in rats. Phytomedicine 2005, 12, 138–142. [Google Scholar] [CrossRef]
  22. Faqueti, L.G.; Brieudes, V.; Halabalaki, M.; Skaltsounis, A.L.; Nascimento, L.F.; Barros, W.M.; Santos, A.R.; Biavatti, M.W. Antinociceptive and anti-inflammatory activities of standardized extract of polymethoxyflavones from Ageratum conyzoides. J. Ethnopharmacol. 2016, 194, 369–377. [Google Scholar] [CrossRef]
  23. Bahtiar, A.; Nurazizah, M.; Roselina, T.; Tambunan, A.P.; Arsianti, A. Ethanolic extracts of babandotan leaves (Ageratum conyzoides L.) prevents inflammation and proteoglycan degradation by inhibiting TNF-alpha and MMP-9 on osteoarthritis rats induced by monosodium iodoacetate. Asian Pac. J. Trop. Med. 2017, 10, 270–277. [Google Scholar] [CrossRef]
  24. Permawati, M.; Anwar, E.; Arsianti, A.; Bahtiar, A. Anti-inflammatory activity of nanoemulgel formulated from Ageratum conyzoides (L.) L. and Oldenlandia corymbosa L. extracts in rats. J. Nat. Remedies 2019, 19, 124–134. [Google Scholar] [CrossRef]
  25. Hunter, D.J.; Lo, G.H. The management of osteoarthritis: An overview and call to appropriate conservative treatment. Rheum. Dis. Clin. N. Am. 2008, 34, 689–712. [Google Scholar] [CrossRef]
  26. Zhang, W.; Ouyang, H.; Dass, C.R.; Xu, J. Current research on pharmacologic and regenerative therapies for osteoarthritis. Bone Res. 2016, 4, 15040. [Google Scholar] [CrossRef]
  27. Usher, K.M.; Zhu, S.; Mavropalias, G.; Carrino, J.A.; Zhao, J.; Xu, J. Pathological mechanisms and therapeutic outlooks for arthrofibrosis. Bone Res. 2019, 7, 9. [Google Scholar] [CrossRef]
Figure 1. Consort flow diagram. Flow diagram of the progress through the two arms of a randomised, parallel trial with two interventions.
Figure 1. Consort flow diagram. Flow diagram of the progress through the two arms of a randomised, parallel trial with two interventions.
Nutraceuticals 05 00035 g001
Figure 2. Change in Womac pain, stiffness, function and total scores over 12 weeks. * significant difference between groups; p < 0.05.
Figure 2. Change in Womac pain, stiffness, function and total scores over 12 weeks. * significant difference between groups; p < 0.05.
Nutraceuticals 05 00035 g002
Figure 3. VAS pain over time. * Statistically significant difference between groups (p < 0.05).
Figure 3. VAS pain over time. * Statistically significant difference between groups (p < 0.05).
Nutraceuticals 05 00035 g003
Table 1. Demographics of participants completing the study.
Table 1. Demographics of participants completing the study.
CharacteristicA. conyzoides (n = 28)Placebo (n = 30)
Age (years)63.9 (8.4)64.8 (9.6)
Weight (kg)75.1 (14.2)78.7 (15.1)
Height (m)1.7 (0.1)1.7 (0.1)
BMI (kg/m2)26.7 (4.1)27.8 (4.7)
Waist (cm)90.4 (11.2)92.5 (12.2)
Hip (cm)104.8 (10.8)106.5 (10.8)
All values are mean (SD).
Table 2. WOMAC total and sub-scores.
Table 2. WOMAC total and sub-scores.
A. conyzoides (n = 28)Placebo (n = 30)
BaselineWeek 3Week 6Week 9Week 12BaselineWeek 3Week 6Week 9Week 12
Joint Pain6.2 (2.9)4.9 (2.9)4.2 (3.0)4.1 (2.4)3.6 (2.6) *6.4 (3.1)6.4 (3.3)5.5 (3.0)5.2 (3.7)5.4 (3.4)
Joint Stiffness3.2 (1.6)2.5 (1.5)2.1 (1.5)2.2 (1.3)2.1 (1.3)3.1 (1.7)3.0 (1.4)2.4 (1.4)2.6 (1.7)2.5 (1.4)
Joint Function21.9 (10.2)17.0 (8.5)15.3 (8.7)14.1 (8.6) *13.9 (7.6) *21.0 (12.1)21.1 (10.9)17.8 (10.2)18.5 (12.6)18.0 (11.5)
WOMAC Total31.4 (13.9)24.5 (12.1)21.6 (12.4)20.4 (11.5) *19.6 (10.8) *30.4 (16.1)30.5 (14.7)25.7 (14.1)26.2 (17.5)25.9 (15.6)
* Statistically significant difference between active and placebo groups (p < 0.05).
Table 3. VAS Pain scores.
Table 3. VAS Pain scores.
TimeA. conyzoides (n = 28)Placebo (n = 30)
Baseline38.4 (17.5)41.9 (18.9)
Week 325.8 (14.8)31.4 (20.0)
Week 621.2 (13.7)27.4 (18.0)
Week 919.0 (13.3) *27.3 (19.3)
Week 1216.9 (11.9) *25.5 (20.1)
* Statistically significant difference between groups (p < 0.05).
Table 4. SF-36 Quality of Life Questionnaire.
Table 4. SF-36 Quality of Life Questionnaire.
DomainA. conyzoides (n = 28)Placebo (n = 30)
BaselineWeek 6Week 12BaselineWeek 6Week 12
Physical Functioning53.3 (20.8)62.2 (22.5)65.2 (18.7)56.3 (24.4)62.2 (22.0)60.3 (22.2)
Role limitations (physical)54.2 (43.1)61.7 (40.3)84.2 (29.0) *64.7 (37.5)72.1 (36.3)67.6 (42.4)
Role limitations (emotional)73.3 (39.5)88.9 (26.7)88.9 (25.3)75.5 (37.9)82.4 (32.0)81.4 (36.0)
Energy/fatigue59.0 (17.1)63.7 (14.9)65.8 (17.1)58.8 (16.4)61.2 (15.4)60.1 (17.1)
Emotional well-being80.3 (12.4)82.7 (11.0)81.9 (14.2)77.5 (10.6)80.9 (10.9)79.3 (14.3)
Social functioning84.2 (18.0)87.5 (13.9)90.4 (13.0)84.9 (20.1)90.4 (11.5)87.5 (16.6)
Pain58.1 (17.9)70.3 (12.5)73.5 (13.9) *56.1 (16.1)67.1 (18.3)66.6 (15.1)
General Health75.7 (12.7)76.7 (12.0)76.0 (11.7)74.7 (14.4)76.2 (14.1)73.7 (17.7)
* Statistically significant difference between groups (p < 0.05).
Table 5. Plasma biomarker changes from baseline.
Table 5. Plasma biomarker changes from baseline.
MarkerA. conyzoides (n = 28)Placebo (n = 30)
BaselineChange at Week 12BaselineChange at Week 12
CRP (mg/L)3.72 (4.59)−0.26 (6.06)3.84 (3.41)0.77 (4.13)
IL-10 (pg/mL)33.13 (56.73)−5.39 (18.27)19.33 (32.64)0.71 (9.20)
IL-6 (pg/mL)17.84 (26.78)−2.40 (5.47) *10.79 (20.14)0.45 (4.56)
IL-8 (pg/mL)29.78 (41.93)−6.35 (16.37) *22.62 (38.83)1.80 (9.71)
TNF-α (pg/mL)14.29 (13.57)0.65 (4.93)11.94 (11.07)−0.10 (3.93)
PSA (ng/mL) a2.25 (2.38)0.04 (0.22)1.22 (1.20)0.04 (0.36)
a measured in males only; * Significant difference between groups (p < 0.05).
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

Rao, A.; Gorman, A.; Subah, S.; Muntha, S.; Bogoda, N.; Briskey, D. Effect of Ageratum Conyzoides on Osteoarthritis in an Ageing Adult Population: A Double-Blind Randomized Placebo-Controlled Parallel Study. Nutraceuticals 2025, 5, 35. https://doi.org/10.3390/nutraceuticals5040035

AMA Style

Rao A, Gorman A, Subah S, Muntha S, Bogoda N, Briskey D. Effect of Ageratum Conyzoides on Osteoarthritis in an Ageing Adult Population: A Double-Blind Randomized Placebo-Controlled Parallel Study. Nutraceuticals. 2025; 5(4):35. https://doi.org/10.3390/nutraceuticals5040035

Chicago/Turabian Style

Rao, Amanda, Alanna Gorman, Silma Subah, Sedratul Muntha, Nathasha Bogoda, and David Briskey. 2025. "Effect of Ageratum Conyzoides on Osteoarthritis in an Ageing Adult Population: A Double-Blind Randomized Placebo-Controlled Parallel Study" Nutraceuticals 5, no. 4: 35. https://doi.org/10.3390/nutraceuticals5040035

APA Style

Rao, A., Gorman, A., Subah, S., Muntha, S., Bogoda, N., & Briskey, D. (2025). Effect of Ageratum Conyzoides on Osteoarthritis in an Ageing Adult Population: A Double-Blind Randomized Placebo-Controlled Parallel Study. Nutraceuticals, 5(4), 35. https://doi.org/10.3390/nutraceuticals5040035

Article Metrics

Back to TopTop