Scolaris Content Display Scolaris Content Display

Tratamientos orales a base de hierbas para la artrosis

Collapse all Expand all

References

Referencias de los estudios incluidos en esta revisión

Adegbehingbe 2008 {published data only}

Adegbehingbe OO, Adesanya SA, Idowu TO, Okimi OC, Oyelami OA, Iwalewa EO, et al. Clinical effects of Garcinia kola in knee osteoarthritis. Journal of Orthopedic Surgery 2008;3:34.

Altman 2001 {published data only}

Altman RD, Marcussen KC. Effects of a ginger extract on knee pain in patients with osteoarthritis. Arthritis & Rheumatism 2001;44(11):2531‐8.

Appelboom 2001 {published data only}

Appelboom T, Schuermans J, Verbruggen G, Henrotin Y, Reginster JY. Symptoms modifying effect of avocado/soyabean unsaponifiables (ASU) in knee osteoarthritis: A double‐blind, prospective, placebo‐controlled study. Scandinavian Journal of Rheumatology 2001;30(4):242‐7.

Badria 2002 {published data only}

Badria FA, El‐Farahaty T, Shabana AA, Hawas SA, El‐Batoty MF. Boswellia‐curcumin preparation for treating knee osteoarthritis: A clinical evaluation. Alternative & Complementary Therapies 2002;8(6):341‐8.

Belcaro 2008 {published data only}

Belcaro G, Cesarone MR, Errichi S, Zulli C, Errichi BM, Vinciguerra G, et al. Treatment of osteoarthritis with Pycnogenol®. The SVOS (San Valentino Osteoarthritis Study). Evaluation of signs, symptoms, physical performance and vascular aspects. Phytotherapy Research 2008;22:518‐23. [DOI: 10.1002/ptr.2376]

Bernhardt 1991 {unpublished data only}

Bernhardt M, Keimel A, Belucci G, et al. Double‐blind, randomised, comparative study of Phytodolor N and placebo as well as an open comparison with Felden 20 Tabs involving convalescent inpatients with arthrotic joint alterations. Unpublished Pharma Report1991. [Steigrwald Pharmaceuticals, Germany]

Biegert 2004 {published data only}

Biegert C, Wagner I, Ludtke R, Kotter I, Lohmuller C, Gunaydin I, et al. Efficacy and safety of willow bark extract in the treatment of osteoarthritis and rheumatoid arthritis: Results of 2 randomized double‐blind controlled trials. The Journal of Rheumatology 2004;31(11):2121‐30.

Biller 2002 {published data only}

Biller A. Results of two randomized controlled studies and of a post‐marketing surveillance study investigating a Devil's claw extract [Ergebnisse zweier randomisierter kontrollierter Studien und einer Anwendungsbeobachtung mit Teufelskrallenextrakt]. In: Schulz V, Rietbrock N, Roots I, Loew D editor(s). In: Phylopharmaka VII. Darmstadt: Steinkopf‐Verlag, 2002:81‐92.

Bliddal 2000 {published data only}

Bliddal H, Rosetzsky A, Schlichting P, Weidner MS, Andersen LA, Ibfelt HH, et al. A randomized, placebo‐controlled, cross‐over study of ginger extracts and ibuprofen in osteoarthritis. Osteoarthritis and Cartilage 2000;8:9‐12.

Blotman 1997 {published data only}

Blotman F, Maheu E, Wulwik A, Caspard H, Lopez A. Efficacy and safety of avocado/soybean unsaponifiables in the treatment of symptomatic osteoarthritis of the knee and hip [Efficacite et tolerance des insaponifiables d'avocat/soja dans le traitement de la gonarthrose et de la coxarthrose symptomatiques]. Revue du Rhumatisme [Éd Fr Joint Bone Spine] 1997;64(12):825‐34.

Cao 2005 {published data only}

Cao Y, Shi Y, Zheng Y, Shi M, Lo SK. Blood‐nourishing and hard‐softening capsule costs less in the management of osteoarthritis knee pain: A randomized controlled trial. eCAM 2005;2(3):363‐8.

Cheras 2010 {published data only}

Cheras PA, Myers SP, Paul‐Brent P, Outerbridge KH, Nielsen GVL. Randomised double‐blind placebo‐controlled trial on the potential modes of action of SheaFlex70 in osteoarthritis. Phytotherapy Research 2010;24:1126‐31. [DOI: 10.1002/ptr.3075]

Chopra 2004 {published data only}

Chopra A, Lavin P, Patwardhan B, Chitre D. A 32‐week randomized, placebo‐controlled clinical evaluation of RA‐11, an Ayurvedic drug, on osteoarthritis of the knees. Journal of Clinical Rheumatology 2004;10(5):236‐45.

Chopra 2011 {published data only}

Chopra A, Saluja M, Tillu G, Venugopalan A, Sarmukaddam S, Raut AK, et al. A randomized controlled exploratory evaluation of standardized Ayurvedic formulations in symptomatic osteoarthritis knees: A Government of India NMITLI project. Evidence‐based Complementary and Alternative Medicine 2011;8:1‐12. [DOI: 10.1155/2011/724291]

Chopra 2013 {published data only}

Chopra A, Saluja M, Tillu G, Sarmukkaddam S, Venugopalan A, Narsimulu G, Handa R, et al. Ayurvedic medicine offers a good alternative to glucosamine and celecoxib in the treatment of symptomatic knee osteoarthritis: a randomized, double‐blind, controlled equivalence trial. Rheumatology 2013;52(8):1408‐17. [DOI: 10.1093/rheumatology/kes414]

Cisar 2008 {published data only}

Cisár P, Jány R, Waczulíková I, Sumegová K, Muchová J, VojtaSSák J, et al. Effect of pine bark extract (Pycnogenol®) on symptoms of knee osteoarthritis. Phytotherapy Research 2008;22:1087‐92. [DOI: 10.1002/ptr.2461]

Farid 2007 {published data only}

Farid R, Mirfeizi Z, Mirheidari M, Rezaieyazdi Z, Mansouri H, Esmaelli H, et al. Pycnogenol supplementation reduces pain and stiffness and improved physical function in adults with knee osteoarthritis. Nutrition Research 2007;27:692‐7. [DOI: 10.1016/j.nutres.2007.09.007]

Ferraz 1991 {published data only}

Ferraz MB, Pereira RB, Iwata NM, Atra E. Tipi. A popular analgesic tea: A double‐blind cross‐over trial in osteoarthritis (letter). Clinical and Experimental Rheumatology 1991;9:205‐12.

Frerick 2001 {published data only}

Frerick H, Biller A, Schmidt U. A treatment schedule for coxarthrosis: A double‐blind study with Devil's claw [Stufenschema bei coxarthrose: Doppelblindstudie mit Teufelskralle]. Der Kassenarzt 2001;5:34‐41.

Gupta 2011 {published data only}

Gupta AK, Acharya K, Sancheti PS, Joshi RS. A double‐blind, randomized, multicentric, placebo‐controlled clinical trial of Antarth, a phytomedicine, in the treatment of osteoarthritis. Indian Journal of Pharmacology 2011;43:69‐72. [DOI: 10.4103/0253‐7613.75674]

Huber 1991 {published data only (unpublished sought but not used)}

Huber B. Therapy of degenerative rheumatic diseases: Use of analgesic medication with Phytodolor N [Therapie degenerativer rheumatischer erkrankungen: Bedarf an zusatzlicher analgetischer medikation unter Phytodolor N]. Fortschritte der Medizin 1991;109:248‐50.

Jung 2001 {published data only}

Jung YB, Roh KJ, Jung JA, Jung K, Yoo H, Cho YB, et al. Effect of SKI306X, a new herbal anti‐arthritic agent, in patients with osteoarthritis if the knee: A double‐blind placebo controlled study. American Journal of Chinese Medicine 2001;29(3‐4):485‐91.

Jung 2004 {published data only}

Jung YB, Seong SC, Lee MC, Shin YU, Kim DH, Kim JM, et al. A four‐week randomized, double‐blind trial of the efficacy and safety of SKI306X: A herbal anti‐arthritic agent versus diclofenac in osteoarthritis of the knee. American Journal of Chinese Medicine 2004;32(2):291‐301.

Kimmatkar 2003 {published data only}

Kimmatkar N, Thawani V, Hingorani L, Khiyani R. Efficacy and tolerability of Boswellia serrata extract in treatment of osteoarthritis of knee ‐ A randomized double blind placebo controlled trial. Phytomedicine 2003;10:3‐7.

Kuptniratsaikul 2009 {published data only}

Kuptniratsaikul V, Thanakhumtorn S, et al. Efficacy and safety of Curcumica domestica extracts in patients with knee osteoarthritis. Journal of Alternative and Complementary Medicine 2009;15:891‐7. [DOI: 10.1089/acm.2008.0186]

Kuptniratsaikul 2011 {published data only}

Kuptniratsaikul V, Pinthong T, Bunjob M, Thanakhumtorn S, Chinswangwatanakul P, Thamlikitkul V. Efficacy and safety of Derris scandens benth extracts in patients with knee osteoarthritis. Journal of Alternative and Complementary Medicine 2011;17:147‐53. [DOI: 10.1089/acm.2010.0213]

Leblan 2000 {published data only}

Leblan D, Chantre P, Fournié B. Harpogophytum procumbens in the treatment of knee and hip osteoarthritis: Four‐month results of a prospective, multicenter, double‐blind trial versus diacerhein [L'harpagophyton dans le traitement de la gonarthrose et de la coxarthrose. Résultats à quatre mois d'une étude prospective multicentrique, contrôlée en double aveugle, versus diacerhéine]. Revue du Rhumatisme [Éd Fr Joint Bone Spine] 2000;67(5):462‐7.

Lequesne 2002 {published data only}

Lequesne M, Maheu E, Cadet C, Dreiser RL. Structural effect of avocado/soyabean unsaponifiables on joint space loss in osteoarthritis of the hip. Arthritis Care & Research 2002;47:50‐8.

Maheu 1998 {published data only}

Maheu E, Mazieres B, Valat J‐P, Loyau G, Le Loet X, Bourgeois P, et al. Symptomatic efficacy of avocado/soybean unsaponifiables in the treatment of osteoarthritis of the knee and hip. Arthritis & Rheumatism 1998;41(1):81‐91.

Maheu 2013 {published data only}

Maheu E, Cadet C, Marty M, Moyse D, Kerloch I, Coste P, Dougados M, Mazières B, Spector TD, Halhol H, Grouin JM, Lequesne M. Randomised, controlled trial of avocado‐soybean unsaponifiable (Piascledine) effect on structure modification in hip osteoarthritis: the ERADIAS study. Annals of the Rheumatic Diseases 2013;Jan 23:[Epub ahead of print]. [DOI: 10.1136/annrheumdis‐2012‐202485]

Majima 2012 {published data only}

Majima T, Inoue M, Kasahara Y, Onodera T, Takahashi D, Minami A. Effect of the Japanese herbal medicine, Boiogito,on the osteoarthritis of the knee with jointeffusion. Sports Medicine, Arthroscopy, Rehabilitation, Therapy & Technology 2012;4:3. [DOI: 10.1186/1758‐2555‐4‐3]

Medhi 2009 {published data only}

Medhi B, Kishore K, Singh U, Seth SD. Comparative clinical trial of castor oil and diclofenac sodium in patients with osteoarthritis. Phytotherapy Research 2009;23:1469‐73. [DOI: 10.1002/ptr.2804]

Mehta 2007 {published data only}

Mehta K, Gala J, Bhasale S, Naik S, Modak M, Thakur H, et al. Comparison of glucosamine sulfate and a polyherbal supplement for the relief of osteoarthritis of the knee: a randomized controlled trial [ISRCTN25438351]. BMC Complementary and Alternative Medicine 2007;7:34. [DOI: 10.1186/1472‐6882‐7‐34]

Mills 1996 {published data only}

Mills SY, Jacoby RK, Chacksfield M, Willoughby M. Effect of a proprietary herbal medicine on the relief of chronic arthritic pain: A double‐blind study. British Journal of Rheumatology 1996;35:874‐8.

Oben 2009 {published data only}

Oben J, Enonchong E, Kothari S, Chambliss W, Garrison R, Dolnick D. Phellodendron and citrus extracts benefit joint health in osteoarthritis patients: a pilot, double‐blind, placebo‐controlled study. Nutrition Journal 2009;8:38. [DOI: 10.1186/1475‐2891‐8‐38]

Pavelka 2010 {published data only}

Pavelka K, Coste P, Géher P, Krejci G. Efficacy and safety of piascledine 300 versus chondroitin sulfate in a 6 months treatment plus 2 months observation in patients with osteoarthritis of the knee. Clinical Rheumatology 2010;29:659‐70. [DOI: 10.1007/s10067‐010‐1384‐8]

Piscoya 2001 {published data only}

Piscoya J, Rodriguez Z, Bustamante SA, Okuhama NN, Miller MJS, Sandoval M. Efficacy and safety of freeze‐dried cat's claw in osteoarthritis of the knee: Mechanism of action of the species Uncaria guianensis . Inflammation Research 2001;50:442‐8.

Rein 2004a {published data only}

Rein E, Kharazami A, Winther K. A herbal remedy, Hyben Vital (stand. powder of a subspecies of Rosa canina fruits), reduces pain and improves general wellbeing in patients with osteoarthritis ‐ a double‐blind, placebo‐controlled, randomised trial. Phytomedicine 2004;11:383‐91.

Schadler 1988 {published data only (unpublished sought but not used)}

Schadler W. Phytodolor for the treatment of activated arthrosis. Rheuma 1988;8:288‐90.

Schmelz 1997 {published data only}

Schmelz H, Hammerle HD, Springorum HW. Analgesic effect of a Devil's claw extract in various chronic degenerative joint diseases [Analgetische Wirkung eines Teufelskrallenwurzel‐Extraktes bei verschiedenen chronisch‐degenerativen Gelenkerkrankungen]. In: Chrubasik S, Wink M editor(s). Rheumatherapie mit Phytopharmaka. Stuttgart: Hippokrates Verlag, 1997:86‐9.

Schmid 2000 {published data only}

Schmid B, Ludtke R, Selbmann HK, Kotter I, Tschirdewahn B, Schaffner W, et al. Efficacy and tolerability of a standardized willow bark extract in patients with osteoarthritis: Randomized placebo‐controlled double blind clinical trial [Wirksamkeit und vertraglichkeit eines standardisierten weidenrindenextraktes bei arthose‐patienten: Randomisierte, placebo‐kontrollierte doppelblindstudie]. Zeitschrift für Rheumatologie 2000;59:1‐7.

Sengupta 2008 {published data only}

Sengupta K, Alluri KV, Satish AR, Mishra S, Golakoti T, Sarma KVS, et al. A double blind, randomized, placebo controlled study of the efficacy and safety of 5‐Loxin® for treatment of osteoarthritis of the knee. Arthritis Research & Therapy 2008;10:R85. [DOI: 10.1186/ar2461]

Sengupta 2010 {published data only}

Sengupta K, Krishnaraju AV, Vishal AA, Mishra A, Trimurtulu G, Sarma KVS, et al. Comparative efficacy and tolerability of 5‐Loxin® and Aflapin® against osteoarthritis of the knee: A double blind, randomized, placebo controlled clinical study. International Journal of Medical Sciences 2010;7(6):366‐77.

Sontakke 2007 {published data only}

Sontakke S, Thawani V, Pimpalkhute S, Kambra P, Babhulkar S, Hingorani L. Open, randomized, controlled trial of Boswellia serrata extract as compared to valdecoxib in osteoarthritis of knee. Indian Journal of Pharmacology 2007;39:27‐9.

Teekachunhatean 2004 {published data only}

Teekachunhatean S, Kunanusom P, Rojanasthien N, Sananpanich K, Pojchamarnwitputh S, Lheiochaiphunt S, Pruksakorn S. Chinese herbal recipe versus diclofenac in symptomatic treatment of osteoarthritis of the knee: A randomized controlled trial. BMC Comlementary and Alternative Medicine 2004;4:19. [DOI: 10.1186/1472‐6882‐4‐19]

Vishal 2011 {published data only}

Vishal AA, Mishra A, Raychaudhuri SP. A double blind, randomized, placebo controlled clinical study evaluates the early efficacy of Aflapin® in subjects with osteoarthritis of the knee. International Journal of Medical Sciences 2011;8(7):615‐22.

Warholm 2003 {published data only}

Warholm O, Skaar S, Hedman E, Molmen HM, Eik L. The effects of a standardized herbal remedy made from a subtype of Rosa canina in patients with osteoarthritis: A double‐blind, randomized, placebo‐controlled clinical trial. Current Therapeutic Research 2003;64(1):21‐31.

Wigler 2003 {published data only}

Wigler I, Grotto I, Caspi D, Yaron M. The effects of Zintona EC (a ginger extract) on symptomatic gonarthritis. Osteoarthritis and Cartilage 2003;11:783‐9.

Winther 2005 {published data only}

Winther K, Apel K, Thamsborg G. A powder made from seeds and shells of a rose‐hip subspecies (Rosa canina) reduces symptoms of knee and hip osteoarthritis: A randomized, double‐blind, placebo‐controlled clinical trial. Scandinavian Journal of Rheumatology 2005;34:302‐8.

Referencias de los estudios excluidos de esta revisión

Anonymous 1993 {published data only}

Anonymous. The garlic treatment: detoxifying with garlic. [Die knoblauchkur: entschlackung durch knoblauch]. Natur & Heilen 1993;70(10):520‐1.

Belcaro 2010 {published data only}

Belcaro G, Cesarone MR, Dugall M, Pellegrini L, Ledda A, Grossi MG, et al. Efficacy and safety of Meriva®, a curcumin‐phosphatidylcholine complex, during extended administration in osteoarthritis patients. Alternative Medicine Review 2010;15(4):337‐44.

Biswas 1997 {published data only}

Biswas NR, Biswas A, Pandey RM. EASE ‐ a herbal preparation for rheumatoid arthritis, non specific arthritis and osteoarthritis. FACT: Focus on Alternative and Complementary Therapies. Proceedings of 4th Annual Symposium on Complementary Health Care. Exeter (UK), 1997; Vol. 2, issue 4:186‐7.

Biswas 1998 {published data only}

Biswas NR, Biswas K, Pandey M, Pandy RM. Treatment of osteoarthritis, rheumatoid arthritis and non‐specific arthritis with a herbal drug: a double‐blind, active drug controlled parallel study. JK Practitioner 1998;5(2):129‐32.

Brien 2006 {published data only}

Brien S, Lewith GT, McGregor G. Devil's Claw (Harpagophytum procumbens) as a treatment for osteoarthritis: A review of safety and efficacy. The Journal of Alternative and Complementary Medicine 2006;12:981‐93.

Chantre 2000 {published data only}

Chantre P, Cappelaere A, Leblan D, Guedon D, Vandermander J, Fournie B. Efficacy and tolerance of Harpagophytum procumbens versus diacerhein in treatment of osteoarthritis. Phytomedicine 2000;7(3):177‐83.

Chrubasik 1998 {published data only}

Chrubasik S, Wink M. Traditional herbal therapy for the treatment of rheumatic pain: Preparations from Devil's claw and stinging nettle. Pain Digest 1998;8:94‐101.

Dharmananda 1985 {published data only}

Dharmananda S. Chinese herbal therapies for chronic joint pain. The American Chiropractor 1985;January:38‐42.

Du 2006 {published data only}

Du T‐X, Han X‐F, Gao S‐T. Effect of hanshibi granule on rheumatism due to blockage of cold and damp. Chinese Journal of Clinical Rehabilitation 2006;10:148‐50.

Falch 1997 {published data only}

Falch VB. Ginger ‐ not only a spice. Review of its effect and efficacy profile [Ingwer ‐ nicht nur ein Gewurz. Untersuchungen zu Wirkungen und Wirksamkeit]. Deutsche Apotheker Zeitung 1997;137:47‐52, 55‐60.

Fang 2008 {published data only}

Fang R, Meng Q‐C, Deng Y‐J, Song G, Bai Y‐P. Effects of Bu Shen Tong Luo decoction on matrix metalloproteinase‐1, matrix metalloproteinase‐3, tissue inhibitor of matrix metalloprotease‐1 and interleukin‐1beta content in the synovial fluid and serum of patients with knee osteoarthritis. Journal of Clinical Rehabilitative Tissue Engineering Research 2008;12:5581‐5.

Gendo 1997 {published data only}

Gendo U. Chronische Polyarthritis in view of traditional Chinese medicine (TCM) [Die chronische Polyarthritis aus der Sicht der traditionellen Chinesischen Medizin (TCM)]. Naturamed 1997;12(10):37‐40.

Grahame 1981 {published data only}

Grahame R, Robinson BV. Devil's claw (Harpagophytum procumbens): Pharmacological and clinical studies. Annals of the Rheumatic Diseases 1981;40:632.

Guyader 1984 {published data only}

Guyader M. Herbal anti‐rheumatic therapies: A longitudinal, pharmacological and clinical study of the treatment of 50 osteoarthritic patients with Harpagophytum procumbens (Devil's Claw) following hospital intervention [Les plantes anti‐rhumatismales. Etude historique et pharmacologique, et etude clinique du nebulisat d'harpagophytum procumbens D.C. chez 50 patients arthrosiques suivis en service hospitalier]. Thesis for the award of the degree Doctor of Medicine.1984.

Hamblin 2008 {published data only}

Hamblin L, Laird A, Parkes E, Walker AF. Improved arthritic knee health in a pilot RCT of phytotherapy. The Journal of the Royal Society for the Promotion of Health 2008;128(5):255‐62. [CENTRAL: 00651201]

Jacquet 2009 {published data only}

Jacquet A, Girodet P, Pariente A, Forest K, Mallet, Moore N. Phytalgic, a food supplement, vs placebo in patients with osteoarthritis of the knee or hip: a randomised double‐blind placebo‐controlled clinical trial. Arthritis Research & Therapy 2009;11:R192. [DOI: 10.1186/ar2891]

Kagore 2011 {published data only}

Kogure T, Tatsumi T, Shigeta T, Fujinaga H, Sato T, Niizawa A. Effect of Kampo medicine on pain and range of motion of osteoarthritis of the hip accompanied by acetabular dysplasia: Case report and literature review. Integrative Medicine Highlights 2011;6:13‐7. [DOI: 10.4137/IMI.S7884]

Kielczynski 1997 {published data only}

Kielczynski W. Osteoarthritis ‐ clinical outcomes after uniform, long‐term herbal treatment. The European Journal of Herbal Medicine 1997;3(2):29‐35.

Kulkarni 1991 {published data only}

Kulkarni RR, Patki PS, Jog VP, Gandage SG, Patwardhan B. Treatment of osteoarthritis with a herbomineral formulation: A double‐blind, placebo‐controlled, cross‐over study. Journal of Ethnopharmacology 1991;33:91‐5.

Lechner 2011 {published data only}

Lechner M, Steirer I, Brinkhaus B, Chen Y, Krist‐Dungl C, Koschier A, et al. Efficacy of individualized Chinese herbal medication in osteoarthrosis of hip and knee: a double‐blind, randomized‐controlled clinical study. Journal of Alternative and Complementary Medicine 2011;17(6):539‐47.

Levy 2009 {published data only}

Levy RM, Saikovsky R, Shmidt E, Khokhlov A, Burnett BP. Flavocoxid is as effective as naproxen for managing the signs and symptoms of osteoarthritis of the knee in humans: a short‐term randomized, double‐blind pilot study. Nutrition Research 2009;29:298‐304.

Linsheng 1997 {published data only}

Linsheng W. Treatment of bony arthritis with herbal medicine and by massotherapy ‐ analysis of 121 cases. Journal of Traditional Chinese Medicine 1997;17(1):32‐6.

Loew 1996 {published data only}

Loew D, Schuster O, Mollerfeld J. Stability and biopharmaceutical quality. A postulate for bioavailability and effectiveness of Harpagophytum procumbens [Stabilitat und biopharmazeutische Qualitat. Voraussetzung fur Bioverfugbarkeit und Wirksamkeit von Harpagophytum procumbens]. In: Loew D, Rietcrock N editor(s). Phytopharmaka II. Forschung und klinische Anwendung. Darmstadt: Steinkopf‐Verlag, 1996:83‐93.

Long 2001 {published data only}

Long L, Soeken K, Ernst E. Herbal medicines for the treatment of osteoarthritis: A systematic review. Rheumatology 2001;40:779‐93.

Lung 2004 {published data only}

Lung YB, Seong SC, Lee MC, Shin YU, Kim DH, Kim JM, et al. A four‐week, randomized, double‐blind trial of the efficacy and safety of SKI306X: a herbal anti‐arthritic agent versus diclofenac in osteoarthritis of the knee. The American Journal of Chinese Medicine 2004;32:291‐301.

Mishra and Singh 2003 {published data only}

Mishra LC, Singh BB, Vinjamury SP. The effectiveness of Commiphora mukul for osteoarthritis of the knee: an outcomes study. Althernative Therapies in Health and Medicine 2003;9:74‐9.

Myers 2010 {published data only}

Myers S, O'Connor J, Fitton JH, Brooks L, Rolfe M, Connellan P, et al. A combined phase I and phase II open label study on the effects of a seaweed extract nutrient complex on osteoarthritis. Biologics: Targets & Therapy 2010;4:33‐44.

Park 2009 {published data only}

Park SH, Kim SK, et al. Effects of AIF on knee osteoarthritis patients: Double‐blind, randomized, placebo‐controlled study. Korean Journal of Physiology and Pharmacology 2009;13:33‐7.

Rein 2004b {published data only}

Rein E, Kharazmi A, Thamsborg G, Winther K. A herbal remedy, made from a subspecies of rose‐hip Rosa canina, reduces symptoms of knee and hip osteoarthritis. Osteoarthritis and Cartilage 2004;12 Suppl 2:S80.

Reuss 1981 {published data only}

Ruess D. Echinacin in the treatment of primary chronic polyarthritis [Echinacin in der Therapie der primar‐chronischen Polyarthritis]. ZFA ‐ Stuttgart 1981;57(11):865.

Rosen 2013 {published data only}

Rosen PD, Liakeas GP, Sadigim E, Bied AM. A pilot study assessing the short term use of Tanacetum parthenium for treatment of osteoarthritis. Integrative Medicine 2013;12(3):31‐6.

Sagar 1988 {published data only}

Sagar VMV. A clinical study of Amavata with special reference to some indigenous drugs. Rheumatism 1988‐89;24(3):3‐7.

Saley 1987 {published data only}

Saley SR, Tilak MN, Deshmukh SS. Amavata ‐ a clinical study of 41 cases. Rheumatism 1987;22(2):46‐50.

Schaffner 1997 {published data only}

Schaffner W. Willow bark ‐ an antirheumatic in modern phytotherapy? [Weidenrinde ‐ Ein Antirheumatikum der modernen Phytotherapie?]. In: Chrubasik S, Wink M editor(s). Rheumatherapie mit Phytopharmaka. Stuttgart: Hippokrates Verglag, 1997:125‐7.

Schmid 1998a {published data only}

Schmid B, Tschirdewahn B, Kotter I, Gunaydin I, Ludtke R, Selbmann HK, et al. Analgesic effects of willow bark extract in osteoarthritis: results of a clinical double‐blind trial. FACT:Focus on Alternative and Complementary Therapies 1998;3(4):186.

Schmid 2001 {published data only}

Schmid B, Lüdtke R, Selbmann H‐K, Kötter I, Tschirdewahn B, Schaffner W, Heide L. Efficacy and tolerability of a standardized willow bark extract in patients with osteoarthritis: Randomized placebo‐controlled double blind clinical trial. Phytotherapy Research 2001;15:344‐50. [DOI: 10.1002/ptr.981]

Srivastava 1989 {published data only}

Srivastava KC, Mustafa T. Ginger (Zingiber officinale) and rheumatic disorders. Medical Hypotheses 1989;29:25‐8.

Wang 1985 {published data only}

Wang ZM, Chang JY, et al. A report on 310 cases of articular rheumatism treated with Feng Shi Han Tong tablet. Chung Hsi I Chieh Ho Tsa Chih 1985;5(5):284‐5, 260.

Wegener 2003 {published data only}

Wegener T, Lupke NP. Treatment of patients with arthrosis of hip or knee with an aqueous extract of Devil's claw (Harpagophytum procumbens DC). Phytotherapy Research 2003;17(10):1165‐72.

Winther 2004 {published data only}

Winther K, Kharazmi A, Rein E. A powder made from a subspecies of rosehip (Rosa canina) reduces WOMAC symptoms scores as well as cholesterol levels in patients suffering from osteoarthritis. Osteoarthritis and Cartilage 2004;13 Suppl A:S93.

Xu 2005 {published data only}

Xu J‐W, Ding J‐X. An analysis on the clinical therapeutic effect of the manipulation matched with Chinese medical herb washing for treatment of osteoarthritis of the knee. Journal of Beijing Teachers College of Physical Education 2005;1:9.

Yuelong 2011 {published data only}

Yuelong C, Hongsheng Z, Jian P, Feiyue L, Shaojian X, Jinghua G, et al. Individually integrated traditional Chinese medicine approach in the management of knee osteoarthritis: study protocol for a randomized controlled trial. Trials 2011;12:160. [DOI: 10.1186/1745‐6215‐12‐160]

Zell 1993 {published data only}

Zell J, Ecker R, Batz H, Vestweber AM. Mistletoe for hip and knee arthrosis: Segment ‐ or immunotherapy? [Misteltherapie bei Gon‐ und Coxarthrose: Segment ‐ oder Immuntherapie? Eine Pilotstudie]. Heilkunst 1993;106(9):23‐6.

Zeng 2008 {published data only}

Zeng Y‐R, Fan Y‐G, Wu F, Li X‐P, Fan H‐J. Effects of compound herb of invigorating kidney and promoting blood flow on serum matrix metalloproteinases‐3, tumor necrosis factor alpha, interleukin‐1, hyaluronic acid, lipid peroxidation levels and superoxide dismutase activity in patients with knee osteoarthritis. Journal of Clinical Rehabilitative Tissue Engineering Research 2008;12:5436‐9.

Referencias de los estudios en espera de evaluación

Gao 2012 {published data only}

Gao G, Wu H, Tian J, Du J, Xie X, Gao J. Clinical efficacy of bushen huoxue qubi decoction on treatment of knee‐osteoarthritis and its effect on hemarheology, anti‐inflammation and antioxidation. Zhongguo Zhong Yao Za Zhi 2012;37(3):390‐6.

Hochberg 2012 {published data only}

Hochberg MC, Lao LX, Langenberg P, Fong HHS, Lee DYW, Berman B. Efficacy and Safety of the Chinese Herbal Compound Hou‐Lou‐Xiao‐Ling Dan in Patients with Osteoarthritis of the Knee: Results of a Phase II International Study [Abstract]. Arthritis and Rheumatism 2012;64(10):S112. [DOI: 10.1002/art.37993]

Kang 2011 {published data only}

Kang XZ, Wu QF, Jie HY. Clinical study on the treatment of knee osteoarthritis by wangbi tablet. Zhongguo Zhong Xi Yi Jie He Za Zhi [Chinese Journal of integrated Traditional and Western Medicine] 2011;31(9):1205‐8.

Liu 2006 {published data only}

Liu H‐B, Zhang J‐F, Zhong L‐W, Yang H. Manipulation plus pyrola compound traditional Chinese medicine in treatment of senile osteoarthritis of knee. Chinese Journal of Clinical Rehabilitation 2006;10(23):30‐2. [CENTRAL: 00613036]

Pinsornsak 2012 {published data only}

Pinsornsak P, Niempoog S. The efficacy of Curcuma longa L. extract as an adjuvant therapy in primary knee osteoarthritis: A randomized controlled trial. Journal of the Medical Association of Thailand 2012;95 Suppl 1:S51‐8.

Tao 2009 {published data only}

Tao QW, Xu Y, Jin DE, Yan XP. Clinical efficacy and safety of Gubitong Recipe () in treating osteoarthritis of knee joint. Chinese Journal of Integrative Medicine 2009;15(6):458‐61.

Zhong 2006 {published data only}

Zhong Q‐S, Ye G‐H, Wang H‐Z, Lin S‐H. Treatment of knee osteoarthritis by invigorating the kidney, dispelling the cold and activating the collaterals: A randomized controlled study. Chinese Journal of Clinical Rehabilitation 2006;10:177‐9.

Altman 1986

Altman R, Asch E, Bloch D, Bole G, Borenstein D, Brandt K, et al. The American College of Rheumatology crtieria for the classification and reporting of osteoarthritis of the knee. Arthritis & Rheumatism 1986;29:1039‐49.

Altman 1990

Altman R, Alarcon G, Appelrouth D, Bloch D, Borenstein D, Brandt K. The American College of Rheumatology criteria for the classification and reporting of osteoarthritis of the hand. Arthritis & Rheumatism 1990;33:1601‐10.

Altman 1991

Altman R, Alarcon G, Appelrouth D, Bloch D, Borenstein D, Brandt K, et al. The American College of Rheumatology criteria for the classification and reporting of osteoarthritis of the hip. Arthritis & Rheumatism 1991;34:505‐14.

Basch 2004

Basch E, Boon H, Davies‐Heerema T, Foppo I, Hashmi S, Hasskarl J, et al. Boswellia: an evidence‐based systematic review by the Natural Standard Research Collaboration. Journal of Herbal Pharmacotherapy 2004;4(3):63‐83.

Begg 1996

Begg C, Cho M, Eastwood S, Horeton R, Moher D, Olkin I, et al. Improving the quality of reporting of randomized controlled trials. The CONSORT Statement. JAMA 1996;276(8):637‐9.

Blumenthal 1998

Blumenthal M. The complete German Commission E Monographs. Austin, TX: American Botanical Council, 1998.

Boullata 2003

Boullata JI, McDonnell PJ, Oliva CD. Anaphylactic reaction to a dietary supplement containing willow bark. Annals of Pharmacotherapy 2003;37:832‐5.

Cameron 2011

Cameron M, Gagnier JJ, Chrubasik S. Herbal therapy for treating rheumatoid arthritis. Cochrane Database of Systematic Reviews 2011, Issue 2. [DOI: 10.1002/14651858.CD002948]

Cameron 2013

Cameron M, Chrubasik S. Topical herbal therapies for treating osteoarthritis. Cochrane Database of Systematic Reviews 2013, Issue 5. [DOI: 10.1002/14651858.CD010538]

Cates 2008

Cates CD. Visual Rx NNT Calculator. Dr Chris Cates EBM website. Available from http://www.nntonline.net/, 2008.

Choi 2002

Choi JH, Choi JH, Kim DY, Yoon JH, Youn HY, Yi JB, et al. Effects of SKI 306X, a new herbal agent, on proteoglycan degradation in cartilage explant culture and collagenase‐induced rabbit osteoarthritis model. Osteoarthritis and Cartilage 2002;10:471‐8.

Christensen 2008a

Christensen R, Bartels EM, Astrup A, Bliddal H. Symptomatic efficacy of avocado‐soybean unsaponifiables (ASU) in osteoarthritis (OA) patients: a meta‐analysis of randomized controlled trials.. Osteoarthritis and Cartilage 2008;16:399‐408.

Christensen 2008b

Christensen R, Bartels EM, Altman RD, Astrup A, Bliddal H. Does the hip powder of Rosa canina (rosehip) reduce pain in osteoarthritis patients? A meta‐analysis of randomized controlled trials. Osteoarthritis and Cartilage 2008;16:965‐72.

Chrubasik 1996

Chrubasik S,  Sporer F,  Wink M. Active ingredients in medicines from Harpagophytum procumbens [Zum Wirkstoffgehalt in Arzneimitteln aus Harpagophytum procumbens]. Forschende Komplementarmedizin 1996;3:57‐63.

Chrubasik 1999

Chrubasik S, Junck H, Breitschwerdt H, Conradt C, Zappe H. Effectiveness of Harpagophytum extract WS 1531 in the treatment of exacerbation of low back pain: A randomized, placebo‐controlled, double‐blind study. European Journal of Anaesthesiology 1999;16:118‐29.

Chrubasik 2000

Chrubasik S, Eisenberg E, Balan E, Weinberger T, Luzzati R, Conradt C. Treatment of low back pain exacerbations with willow bark extract: A randomized double‐blind study. American Journal of Medicine 2000;109:9‐14.

Chrubasik 2001

Chrubasik S, Künzel O, Model A, Conradt C, Black A. Treatment of low back pain with a herbal or syntheticantirheumatic: a randomized controlled study. Rheumatology 2001;40:1388‐93.

Chrubasik 2002

Chrubasik S, Fiebich B, Black A, Pollak S. Treating low back pain with an extract of Harpagophytum that inhibits cytokine release. European Journal of Anaesthesiology 2002;19:209.

Chrubasik 2003

Chrubasik S, Roufogalis B. Bioequivalence of herbal medicines. New Zealand Journal of Pharmacy 2003;53:39‐44.

Chrubasik 2005

Chrubasik S, Pittler MH, Roufogalis BD. Zingiberis rhizoma: a comprehensive review on the ginger effect and efficacy profiles. Phytomedicine 2005;12(9):684‐701.

Chrubasik 2006

Chrubasik JE, Lindhorst E, Neumann E, Gerlach U, Faller‐Marquardt M, Torda T, Muller‐Ladner U, Chrubasik S. Potential molecular basis of the chondroprotective effect of Harpagophytum procumbens . Phytomedicine 2006;13:598‐600.

Chrubasik 2007

Chrubasik JE, Roufogalis BD, Chrubasik S. Evidence of effectiveness of herbal anti‐inflammatory drugs in the treatment of painful osteoarthritis including chronic low back pain. Phytotherapy Research 2007;21:675‐83.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors).Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011)The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

EMA 2009

European Medicines Agency (EMA). Assessment report on salicis cortex (willow bark) and herbal preparation(s) thereof with well‐established use and traditional use. EMA, London, 20092009.

ESCOP 2003

European Scientific Cooperative on Phytotherapy (Eds). ESCOP Monographs. New York: Thieme‐Verlag Stuttgart, 2003.

ESCOP 2009

European Scientific Cooperative on Phytotherapy (Eds). ESCOP Monographs. Vol. Supplement, Stuttgart, New York: Thieme Press, 2009.

Gabriel 1991

Gabriel SE, Jaakkimainen L, Bombardier C. Risk for serious gastrointestinal complications related to use of nonsteroidal anti‐inflammatory drugs. A meta‐analysis. Annals of Internal Medicine 1991;115:787‐96.

Glinko 1998

Glinko A. Pharmacological properties of a standardized extract from Willow Bark (Cortex salicis) [treatise]. Chair of Pharmacology and Toxicology. Pomeranian Academy of Medicine. Szczecin, 1998.

Gundermann 2001

Gundermann KJ. Phytodolor solution. Clinical expert report. Darmstadt, Germany: Steigerwald, 2001.

Hadhyiski 2006

Hadzhiyski H, Lindhorst E, Raif W, Torda T, Chrubasik S. Impact of Harpagophytum procumbens on the urinary pyridinoline deoxypyridinoline ratio in experimental osteoarthritis [Abstract]. FACT: Focus on Alternative and Complementary Therapies 2006;11:13.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies.. Higgins JPT, Green S (editors). The Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration. Available from www.cochrane‐handbook.org, 2011.

ICH 2004

ICH. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. Synopsis of ICH guidelines and topics. Geneva, Switzerland: Author, 2004.

Jäger 2007

Jäger AK, Eldeen IM, van Staden J. COX‐1 and ‐2 activity of rose hip. Phytotherapy Research 2007;21(12):1251‐2.

Jäger 2008

Jäger AK, Petersen KN, Thomasen G, Christensen SB. Isolation of linoleic and alpha‐linolenic acids as COX‐1 and ‐2 inhibitors in rose hip. Phytotherapy Research 2008;22(7):982‐4.

Krishnaraju 2010

Krishnaraju AV, Sundararaju D, Vamsikrishna U, Suryachandra R, Machiraju G, Sengupta K, Trimurtulu G. Safety and toxicological evaluation of Aflapin: a novel Boswellia‐derived anti‐inflammatory product. Toxicology Mechanisms and Methods 2010;20(9):556‐63.

Krivoy 2001

Krivoy N, Pavlotzky E, Chrubasik S, Eisenberg E, Brooks G. Effect of Salicis cortex extract on human platelet aggregation. Planta Medica 2001;67:209‐13.

Lawrence 2008

Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al. National Arthritis Data Workgroup. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part II. Arthritis & Rheumatism 2008;58:26‐35.

Lee 2010

Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Effect of glucosamine or chondroitin sulfate on the osteoarthritis progression: a meta‐analysis. Rheumatology International 2010;30(3):357‐63.

Malemud 2010

Malemud CJ. Anticytokine therapy for osteoarthritis: Evidence to date. Drugs & Aging 2010;27:95‐115. [DOI: 10.2165/11319950‐000000000‐00000]

Mazieres 1993

Mazieres B, Tempesta C, Tiechard M, Vaguier G. Pathologic and biochemical effects of a lipidic avocado and soya extract on an experimental post‐contusive model of OA [Abstract]. Osteoarthritis and Cartilage 1993;1:46.

Moher 2001

Moher D, Schulz KF, Altman DG. The CONSORT statement: Revised recommendations for improving the quality of reports of parallel‐group randomized trials. Annals of Internal Medicine 2001;134:657‐62.

Ofman 2002

Ofman JJ, MacLean CH, Straus WL, Morton SC, Berger ML, Roth EA, Shekelle P. A metaanalysis of severe upper gastrointestinal complications of nonsteroidal antiinflammatory drugs. Journal of Rheumatology 2002;29:804‐12.

Ofman 2003

Ofman JJ, Maclean CH, Straus WL, Morton SC, Berger ML, Roth EA, Shekelle PG. Meta‐analysis of dyspepsia and nonsteroidal antiinflammatory drugs. Arthritis & Rheumatism 2003;49:508‐18.

Olivier 2010

Olivier P, Montastruc JL. Réseau français des centres régionaux de pharmacovigilance [Post‐marketing safety profile of avocado‐soybean unsaponifiables]. Presse Medicale 2010;39(10):e211‐6.

Pham 2003

Pham T, Van Der Heijde D, Lassere M, Altman RD, Anderson JJ, Bellamy N, et al. Outcome variables for osteoarthritis clinical trials: The OMERACT‐OARSI set of responder criteria. Journal of Rheumatology 2003;30:1645‐54.

Pham 2004

Pham T, van der Heijde D, Altman RD, Anderson JJ, Bellamy N, Hochberg M, et al. OMERACT‐OARSI initiative: Osteoarthritis Research Society International set of responder criteria for osteoarthritis clinical trials revisited. Osteoarthritis and Cartilage 2004;12:389‐99.

Reginister 2007

Reginster JY, Malmstrom K, Mehta A, Bergman G, Ko AT, Curtis SP, Reicin AS. Evaluation of the efficacy and safety of etoricoxib compared with naproxen in two, 138‐week randomised studies of patients with osteoarthritis. Annals of the Rheumatic Diseases 2007;66:945‐51.

Reichenbach 2007

Reichenbach S, Sterchi R, Scherer M, Trelle S, Bürgi E, Bürgi U, et al. Meta‐analysis: chondroitin for osteoarthritis of the knee or hip. Annals of Internal Medicine 2007;146(8):580‐90.

Sangha 2000

Sangha O. Epidemiology of rheumatic diseases. Rheumatology 2000;39 Suppl 2:3‐12.

Schmid 1998b

Schmid B. Treatment of hip and knee osteoarthritis with willow bark extract [Behandlung von Cox‐ und Gonarthrosen mit einem Weidenrindenextrakt]. Dissertation Universität Tübingen1998.

Schmitz 2010

Schmitz N, Kraus VB, Aigner T. Targets to tackle‐‐the pathophysiology of the disease. Current Drug Targets 2010;11:521‐7.

Schoonees 2012

Schoonees A, Visser J, Musekiwa A, Volmink J. Pycnogenol® (extract of French maritime pine bark) for the treatment of chronic disorders. Cochrane Database of Systematic Reviews 2012, Issue 4. [DOI: 10.1002/14651858.CD008294.pub4]

Schunemann 2011a

Schunemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and 'Summary of findings' tables. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration, updated March 2011.

Schunemann 2011b

Schunemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, Guyatt GH. Chapter 12: Interpreting results and drawing conclusions. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration, updated March 2011.

Smalley 1996

Smalley WE, Griffin MR, Fought RL, Ray WA. Excess costs from gastrointestinal disease associated with nonsteroidal anti‐inflammatory drugs. Journal of General Internal Medicine 1996;11:461‐9.

Sporer 1999

Sporer F, Chrubasik S. Preparations from Devil's claw (Harpagophytum procumbens) [Präparate aus der Teufelskralle (Harpagophytum procumbens)]. Zeitschrift für Phytotherapie 1999;20:235‐6.

Stohs 2011

Stohs SJ, Preuss HG, Shara M. The safety of Citrus aurantium (bitter orange) and its primary protoalkaloid p‐synephrine. Phytotherapy Research 2011;25(10):1421‐8. [DOI: 10.1002/ptr.3490]

Stone 1763

Stone E. An account of the success of the bark of the willow in the cure of agues. Philosophy. Trans: Royal Society London 1763;53:195‐200.

Valerio 2005

Valerio LG, Gonzales GF. Toxicological aspects of the South American herbs cat's claw (Uncaria tomentosa) and Maca (Lepidium meyenii) : a critical synopsis. Toxicology Review 2005;24(1):11‐35.

van den Berg 2011

van den Berg WB. Osteoarthritis year 2010 in review: Pathomechanisms. Osteoarthritis and Cartilage 2011;19:338‐41.

Vlachojannis 2008

Vlachojannis JE, Roufogalis BD, Chrubasik S. Systematic review on the safety of Harpagophytum preparations for osteoarthritic and low back pain. Phytotherapy Research 2008;22(2):149‐52. [DOI: 10.1002/ptr.2314]

Vlachojannis 2009

Vlachojannis JE, Duke RK, Tran VH, Duke CC, Chrubasik S. Anti‐inflammatory herbal medicines for the control of pain. In: Carpinella M C, Rai M editor(s). Novel Therapeutic Agents from Plants. Enfield, NH, USA: Science Publisher, 2009:339‐67.

Vlachojannis 2013

Vlachojannis C, Magora F, Chrubasik S. Pro and contra duration restriction of willow bark treatment. Phytotherapy Research 2014;28(1):148‐9.

Wandel 2010

Wandel S, Jüni P, Tendal B, Nüesch E, Villiger PM, Welton NJ, Reichenbach S, Trelle S. Effects of glucosamine, chondroitin, or placebo in patients with osteoarthritis of hip or knee: network meta‐analysis. BMJ 2010;341:c4675. [DOI: 10.1136/bmj.c4675]

Wenzig 2008

Wenzig EM, Widowitz U, Kunert O, Chrubasik S, Bucar F, Knauder E, Bauer R. Phytochemical composition and in vitro pharmacological activity of two rose hip (Rosa canina L.) preparations. Phytomedicine 2008;15(10):826‐35.

Zhang 2009

Zhang W, Doherty M, Leeb BF, Alekseeva L, Arden NK, Bijlsma JW, et al. EULAR evidence based recommendations for the diagnosis of hand osteoarthritis ‐ report of a task force of the EULAR Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT). Annals of Rheumatic Diseases 2009;68:8‐17.

Zhang 2010a

Zhang Y, Jordan JM. Epidemiology of osteoarthritis. Clinical Geriatric Medicine 2010;26:355‐69.

Zhang 2010b

Zhang W, Doherty M, Peat G, Bierma‐Zeinstra MA, Arden NK, Bresnihan B, et al. EULAR evidence‐based recommendations for the diagnosis of knee osteoarthritis. Annals of Rheumatic Diseases 2010;69(3):483‐9.

Referencias de otras versiones publicadas de esta revisión

Cameron 2007

Cameron M, Chrubasik S, Parsons T, Gagnier J, Blümle A, Little C. Herbal therapy for treating osteoarthritis: Update of a Cochrane review. Annals of the Rheumatic Diseases 2007;66 Supp II:494.

Cameron 2009

Cameron M, Gagnier J, Little C, Parsons T, Blümle A, Chrubasik S. Evidence of the effectiveness of herbal medicinal products in the treatment of arthritis. Part A: Osteoathritis. Phytotherapy Research 2009;23:1497‐515. [DOI: 10.1002/ptr.3007]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Adegbehingbe 2008

Methods

Randomised, double‐blind, placebo and active controls, 4 parallel groups, single centre study. Duration 6 weeks

Participants

Randomised n=143, Completed n=84. Mean age: placebo control 53.2 yrs, active control 1 (naproxen 1000mg) 51.0 yrs, active control 2 (Celebrex 400mg) 52.5 yrs, intervention 54.1 yrs. M:F placebo control 7:14, active control A 6:15, active control B 6:15, intervention 5:16. Inclusion: primary or secondary OA knee (ACR criteria), pain at rest VAS 0‐100 >45mm at baseline

Interventions

Tradename not provided: Garcinia kola 400mg (2 x 200mg), tablets

Active control A: naproxen 1000mg (2 x 500mg), tablets

Active control B: celecoxib (Celebrex) 400mg (2 x 200mg), tablets

Placebo control: ascorbic acid 200mg (2 x 100g), tablets

Outcomes

WOMAC‐VAS (Pain), WOMAC 0‐4 (Function), walking distance, time to pain relief

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results favour intervention and active controls over placebo. Reductions in pain were not significantly different between Garcinia kola and the active controls, although onset of pain relief was most rapid and most persistent in the celebrex group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four within each stratum, using computer generated random number sequences

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment: Medications prepared by nursing staff, administered by blinded senior orthopaedic registrar

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention, placebo, and active controls not distinguished by look, taste, smell, packaging, or medication regimen. Baseline and outcome assessor blinded to allocations

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Outcome data reported as change scores, percentages, confidence intervals, and P values only, insufficient for extraction (unclear risk).

WOMAC subscales for pain and physical function used independently, and in different forms (VAS and 0‐4).

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Altman 2001

Methods

Randomised, double‐blind, placebo control, 2 parallel groups, 10 centre study. Duration 12 weeks

Participants

Randomised n=261, Completed n=247. Mean age 65 yrs. M:F 37:63. Inclusion: OA knee stage II‐IV (ACR criteria), knee pain on standing 40‐90mm on VAS 100mm

Interventions

EV.EXT 77: mixture of Zingiber officinale (ginger) and Alpina galanga (galangal) extracts, 510mg (2 x 255mg), capsules

Placebo control: coconut oil, capsules

Rescue medication permitted: acetominophen, up to 4000mg (4 x 2 x 500mg) daily PRN

Concurrent medication permitted: aspirin, up to 325mg daily for anticoagulation

Outcomes

Pain on standing, pain walking 50ft, WOMAC‐VAS (normalised units), SF‐12, patient global 1‐5

Notes

Confirmatory study design; statistical power not reported, but post‐hoc calculation of power based on sample size and design indicates adequate power to detect medium to large effects (if d=0.5, then P=0.97). Reported compliance with ICH GCP guidelines and ethics committee approval. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of two groups using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "both the investigators and the patients were blinded to treatment assignment"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Appelboom 2001

Methods

Randomised, double‐blind, placebo control, 3 parallel groups, multicentre study. Duration 90 days (˜12 weeks)

Participants

Randomised n=260, Completed n=206. Age range 45‐80 yrs. M:F 55:205. Inclusion: OA knee (ACR criteria), VAS 0‐100 pain on standing 40‐90mm, baseline analgesia 90‐110mg diclofenac equivalents

Interventions

Piascledine 300*: Persa gratissma and Glycine max, avocado / soyabean unsaponifiables, 300mg / 600mg, OD, tablets

Placebo control: ingredients not reported

Outcomes

NSAID use (diclofenac equivalents), days without NSAIDs, pain VAS 0‐100, Lesquesne index, patient efficacy assessment, clinician efficacy assessment, adverse events

Notes

Confirmatory study; statistical power not reported, but post‐hoc calculation of power based on sample size and design indicates adequate power to detect medium to large effects (if Cohen's f=0.25, then P=0.90). Reported ethics committee approval. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method not reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included per protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Badria 2002

Methods

Randomised, double‐blind, placebo control, 3 parallel groups (intervention, placebo control, non‐intervention control). Erroneously described as "cross‐over trial". Duration 3 months (˜12 weeks)

Participants

Randomised n=60; intervention n=30, placebo n=15, non‐intervention control n=15. Age and gender data not reported. Inclusion: OA knee (criteria not specified)

Interventions

Tradename not provided. Boswellia‐curcuma extract mixture, 1500mg (3 x 500mg), capsules

Placebo control: ingredients not reported

Outcomes

Nocturnal pain, pain with active movement 0‐3, pain with passive movement 0‐3, tenderness 0‐3, knee effusion 0‐3, pain‐free walking time minutes, antioxidant enzyme SOD, free radical damage markers NO, nitrate, nitrite, and CD, CD4

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported. Group sizes dissimilar and likely to have been determined a priori

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method not reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawals not reported. Age and gender data not reported

Selective reporting (reporting bias)

High risk

Described as a crossover trial, but method of crossover and data from second arm not reported. Considered as a parallel trial for this review (high risk)

Adverse events not reported (high risk)

Conclusion not supported by data: Reported efficacy and tolerability of boswellia‐curcumin as superior to diclofenac, but this trial did not include diclofenac as an active control (high risk)

Other bias

High risk

Diagnosis of OA not established at baseline (high risk)

Unvalidated outcome measures (unclear risk)

Belcaro 2008

Methods

Randomised, double‐blind, placebo control, 2 parallel group study. Duration 3 months (˜12 weeks)

Participants

Randomised n=156; intervention n=77, control n=79. Completed n=143. Mean age: control 47.8 yrs, intervention 48.6 yrs. M:F: control 39:40, intervention 39:38. Inclusion: OA knee (radiographic criteria)

Interventions

Pycnogenol®: Pinus pinaster, pine bark extract, 100mg (2 x 50mg), tablets

Placebo control: ingredients not reported, tablets

Outcomes

WOMAC 0‐4, mobility (treadmill walking)

Notes

Confirmatory study design; statistical power 80%, alpha set at 0.05. Reported ethics committee approval and compliance with Declaration of Helsinki. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants allocated to treatment groups using randomisation by block allocation sequences created from a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Unclear whether analysis is per protocol or intention‐to‐treat (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Adverse events reported generally, but not individually (unclear risk)

Reported WOMAC subscale scores but no standard deviations: standard deviations computed from item scores for extraction and re‐analysis (unclear risk)

Other bias

Unclear risk

Diagnosis and assessment based on radiographic criteria only (unclear risk)

Bernhardt 1991

Methods

Randomised, double‐blind, placebo control, active control (unblinded), 3 parallel groups. Duration 4 weeks

Participants

Randomised n=108; intervention n=36, placebo n=36, piroxicam n=36. Completed n=108. Mean age 52 yrs. M:F 22:50. Inclusion: OA (criteria not specified), acute or recurrent degenerative arthritic complaints

Interventions

PhytodolorRN: standardised extract mixture of ash bark, aspen leaf, aspen bark, golden rod herb, 3 x 30 drops, tincture

Active control: piroxicam (Feldene 20), 20mg, OD

Placebo control: ingredients not reported

Concurrent treatment permitted: balneology (thermal baths), and physiotherapy

Outcomes

Pain with movement 0‐3, enduring pain 0‐3, mobility impairment 0‐3, finger‐ground distance, grip strength, PGA 0‐6, patient perception efficacy 0‐3

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention. Some outcome measures (eg: finger‐ground distance) are non‐specific and may be of limited use in rheumatological assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of three groups using a table of random numbers

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind. In PhytodolorRN and placebo groups, active intervention and placebo not distinguished by look, taste, smell or packaging (low risk)

Piroxicam group not blinded (unclear risk)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported no withdrawals (low risk)

Intention‐to‐treat analysis can be assumed

Selective reporting (reporting bias)

Unclear risk

Most outcome data reported as change scores, percentages, graphs and p values only, insufficient for extraction (unclear risk)

Reported adverse events (low risk)

Other bias

High risk

Diagnosis not based on ACR criteria. Non‐homogenous sample (any degenerative arthropathy, any site) (high risk)

Unvalidated outcome measures (unclear risk)

Biegert 2004

Methods

Randomised, double‐blind, placebo control, active control, 3 parallel groups. Duration 6 weeks

Participants

Randomised n=127, Completed n=106. Mean age 62 yrs. M:F 53:74. Inclusion: OA knee or hip (ACR criteria), WOMAC >30mm, aspirin 100mg/d

Interventions

Assalix*: Salix daphnoides cortex (willow bark), ethanolic extract, 1572.96mg (2 x 2 x 393.24mg, equivalent to 240mg salicin), tablets

Active control: diclofenac, 100mg (2 x 2 x 25mg), tablets

Placebo control: ingredients not reported, tablets

Outcomes

WOMAC‐VAS (normalised units), SF‐36, patient efficacy assessment VAS 0‐100, physician efficacy assessment VAS 0‐100, HAQ‐DI (German)

Notes

Confirmatory study; statistical power 80%, alpha set at 0.05 (2 tailed). Reported compliance with ICH GCP guidelines and the Declaration of Helsinki. Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of three groups using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1. Authors contacted for confirmation, but details of allocation concealment not provided

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active interventions and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat and per protocol analyses

Selective reporting (reporting bias)

Unclear risk

Mid‐point data reported as mean scores only (no standard deviations), therefore not adequate for extraction and re‐analysis (unclear risk)

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Biller 2002

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 20 weeks

Participants

Randomised n=78, Completed n=77. Age and gender data not reported. Inclusion: OA knee (ACR criteria)

Interventions

LoHar 45 flexi‐loges®*: Harpagophytum procumbens (devil's claw), ethanolic extract, 960mg, tablets

Placebo control: ingredients not reported

Outcomes

WOMAC‐VAS (German version). Post hoc "responders” to treatment were defined as participants whose WOMAC pain scores increased by not more than 20% without additional rescue medication in weeks 17‐20

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported compliance with ICH GCP guidelines. Results equivocal: no improvement on primary outcome measure (WOMAC).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised, method not reported1. Authors contacted: provided full details of computer generated randomisation sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1. Authors contacted for confirmation, but details of allocation concealment not provided

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

High risk

Brief report. Full report not available (high risk)

Reported withdrawals (low risk)

Unclear whether analysis is per protocol or intention‐to‐treat analysis (unclear risk)

Selective reporting (reporting bias)

High risk

Age and gender data not reported (high risk)

Reasons for withdrawal (ie: adverse events) not reported (unclear risk)

Outcome data reported as percentages only, insufficient for extraction (unclear risk)

Results showed no improvement on planned primary outcome measure (WOMAC). Alternate outcome measure and definition of improvement constructed post hoc

Other bias

Unclear risk

Diagnosis consistent with ACR criteria (low risk)

Post‐hoc created outcome measure is unvalidated (unclear risk)

Bliddal 2000

Methods

Randomised, double‐blind, placebo control, active control, 3 group crossover. Duration 12 weeks (1 week washout followed by 3 weeks intervention)

Participants

Randomised n=67, Completed n=56. Mean age 66 yrs, range 24‐87 yrs. M:F 15:41. Inclusion: OA hip or knee, radiologically verified Kellgren grade I‐IV, VAS 0‐100 pain on mvt >30mm

Interventions

Eurovita.EXT 33: Zingiber officinale (Chinese ginger) extract, 510mg (3 x 170mg), capsules

Active control: ibuprofen, 400mg, tablets

Placebo control: ingredients not reported, capsules and tablets

Rescue medicine permitted: paracetamol (acetominophen), 3000mg daily, PRN

Outcomes

Pain VAS 0‐100, Lequesne index, range of motion (hip or knee), acetominophen use, investigator treatment preference, daily pain diary (4 point Likert scale)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval and compliance with ICH GCP guidelines. Results favour ibuprofen over ginger, ginger over placebo.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised, method of randomisation incompletely reported1. Reported as randomised in blocks of six to one of three groups, with further randomisation of treatment sequence within blocks. Authors contacted for confirmation, but further details not provided

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1. Authors contacted for confirmation, but details of allocation concealment not provided

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Double‐dummy method, placebo controls for both intervention and active controls. Active interventions and placebos not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Included per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety (low risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Blotman 1997

Methods

Randomised, double‐blind, placebo control, 2 parallel groups, multicentre (n not specified). Duration 90 days (˜12 weeks)

Participants

Completed n=163. Mean age 63 yrs. M:F 55:108. Inclusion: OA knee (n=101) or hip (n=62) (ACR criteria), Kellgren grade IB‐III, pain requiring NSAIDs for 3 months

Interventions

Piascledine 300*: Persa gratissma and Glycine max, avocado / soyabean unsaponifiables, 300mg / 600mg, OD, tablets

Placebo control: ingredients not reported

Rescue medication permitted: one of 7 predefined NSAIDs taken by all participants for first 45 days. Resumption of same NSAID allowed during second 45 days

Outcomes

Resumption of NSAIDS, time off NSAIDS, NSAID use (diclofenac equivalents), Lequesne index, pain VAS 0‐100, patient global 0‐4, physician global 0‐4

Notes

Confirmatory study design, power 80%, alpha 0.05. Reported compliance with Helsinki Declaration and ethics committee approval. Results favour intervention for reduced use of NSAIDs, but pain scores are similar in the two groups.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised, in blocks of four, stratified according to site of arthritis (hip or knee), to one of two groups, using a table of random numbers

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Included per protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Cao 2005

Methods

Randomised, unblinded, active control, 2 parallel groups, two centres. Duration 4 weeks

Participants

Randomised n=120, intervention (n=60; n=30 at x centres), Chinese control n=30, Western control n=30. Completed n=116. Inclusion: OA knee (ACR criteria), at least 5 days on NSAIDs, adverse events with NSAIDs, pain with walking at least 20mm (VAS 0‐100) in the previous 48h

Interventions

Tradename not provided. Chinese herbal mixture (blood‐nourishing, hard‐softening; BNHS), 3150mg

Active control (Chinese): Chinese mixture to counter osteophytes, 5250mg, capsules

Active control (Western): Viatril‐s 2250mg (crystalline glucosamine sulphate 1884mg equivalent to glucosamine sulphate 1500mg, sodium chloride 384mg)

Rescue medication permitted: paracetamol (acetominophen), up to 4000mg daily, PRN; and aspirin, up to 100mg daily in a stable dose

Outcomes

WOMAC‐VAS (normalised scores), pain during walking 0‐100 VAS, patient global 0‐4, physician global 0‐4

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval and compliance with ICH GCP guidelines. Improvements occured in all groups over time. Results indicate that BNHS is not inferior to counter osteophyte Chinese mixture or Viatril‐s.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method inadequately reported: "sealed envelope method"

Allocation concealment (selection bias)

Low risk

Allocation concealment can be inferred: "sealed envelope method"

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as blinded, method inadequately reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Cheras 2010

Methods

Ramdonised, double‐blind, placebo control, 2 parallel groups, single centre. Duration 18 weeks (3 weeks washout, 15 weeks trial)

Participants

Completed n=89, intervention n=39, control n=50. OA hip or OA knee (ACR criteria), overall WOMAC score=30 at baseline

Interventions

SheaFlex70: Vitellaria paradoxia, 100% sheabutter extract with 75% triterpene esters, 2250mg (3 x 750mg), capsules

Placebo control: 100% canola oil, 2250mg (3 x 750mg), capsules

Outcomes

WOMAC, Comprehensive Osteoarthritis Test (COAT)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval and clinical trials registration (ACTRN12606000162516). Results equivocal on clinical outcomes. Changes in WOMAC scores were not significant either within or between groups. Significant decrease in COAT pain subscale score within the shea group over time was not significantly different from this outcome in the control group at the end of the trial. Significant differences in some inflammatory markers were reported, but are not of relevance to this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of two groups using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Allocation "held by a third party to the investigator and trial sponsor"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention, placebo, and active controls not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

High risk

Withdrawals not reported (high risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

High risk

Clinical outcome data reported as percentages and P values (non‐significant) only, insufficient for extraction (unclear risk)

Adverse events not reported (high risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Chopra 2004

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 32 weeks

Participants

Randomised n=90, intervention n=45, control n=45. Midpoint (16 weeks) n=78. Completed n=62, intervention n=31, control n=31. Age 35+ years. OA knee (ACR criteria). Stable NSAIDs for 1 month at baseline. Not pregnant

Interventions

RA‐11: Ayurvedic medication, 2 capsules

Placebo control: ingredients not reported

Rescue medication not permitted

Concurrent medication permitted: stable medication for concomitant diseases

Outcomes

WOMAC 0‐4 (Asian ‐ Indian modification), VAS 0‐100, 50 feet walk time (seconds), physician global assessment 0‐4, patient global assessment 0‐4, early morning stiffness (minutes), knee swelling 0‐3

Notes

Confirmatory study design, power 80%, alpha 0.05 (2 tailed). Did not report ethical oversight or compliance with guidelines. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised: "...assigned to the active or placebo groups as per a predetermined computer generated randomisation schedule..."

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "A sealed copy of the randomization code was kept with the sponsor and the chief investigator but was not revealed to the subjects or the clinical staff until completion of the study."

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals. Included intention‐to‐treat analysis. Last observation carried forward to replace missing data (low risk)

Three participants were withdrawn by the investigators due to "efficacy failure", which may confound results (unclear risk)

Selective reporting (reporting bias)

Low risk

Pre‐determined levels of improvement (MCID) (low risk)

Reported adverse events. Two participants in intervention group died, but these deaths were attributed to concomitant cardiovascular disease

Other bias

Low risk

Diagnosis consistent with ACR criteria (low risk)

Chopra 2011

Methods

Randomised, double‐blind, placebo and active control, 7 parallel groups, multicentre (n not specified). Duration 16 weeks

Participants

Randomised n=245, all groups n=35. Completed n=202. Data available for analysis n=236. Mean age: placebo control 54 yrs, active control 54.2 yrs, intervention A 57.5 yrs, intervention B 56.6 yrs, intervention C 56.8 yrs, intervention D 56.2 yrs, intervention E 56.2 yrs. M:F not reported. Inclusion: OA knee (ACR criteria with lower age limit reduced to 40 years)

Interventions

Tradenames not provided. five Ayurvedic formulations containing Zingiber officinale and Tinospora cordifolia and combinations ofEmblica officinale, Withania somnifera, or Tribulus terrestris, variable doses (4 x approx 500mg), capsules

Active control: glucosamine sulphate, 1000mg (4 x 250mg). Capsule mass approx 500mg

Placebo control: charcoal and synthetic ginger essence, 2000mg (4 x 500mg), capsules

Rescue medication permitted: paracetamol (acetominophen), 2000mg (4 x 500mg) PRN

Outcomes

Pain on weightbearing VAS 0‐10, WOMAC 0‐4 (Indian version), paracetamol use

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results equivocal for primary outcomes. Trend to favour intervention C for pain relief, paracetamol use, and knee function

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, but participants allocated directly to groups on order of enrolment into the trial (quasi‐randomised)

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active interventions, active control, and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Unclear risk

Outcome data reported as change scores and percentages only, insufficient for extraction (unclear risk)

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Synthetic ginger extract in placebo may not be inert

Chopra 2013

Methods

Randomised, double‐blind, active control, multicentre (n=3), 4 parallel groups. Duration 24 weeks (+2 to 5 days washout for participants using NSAIDs)

Participants

Randomised n=440, intervention SGC n=110, intervention SGCG n=110, active control celecoxib n=110, active control glucosamine n=110. Completed n=314, SGC n=75, SGCG n=75, celecoxib n=78, glucosamine n=86. Age range 40‐70 years. Inclusion: OA knee (ACR criteria with modified age range), unilateral or bilateral knee OA, baseline VAS 0‐100 (pain on weightbearing) >54mm. Not pregnant or lactating, not taking medications likely to influence pain / functional outcomes, no known GIT bleeding

Interventions

Tradename not provided. Standardised Ayurvedic formulation (shunthi‐guduchi, SGCG), 2400mg (2 x 400mg, TID), capsules

Tradename not provided. Standardised Ayurvedic formulation (shunthi‐guduchi with guggal, SGCG), 2400mg (2 x 400mg, TID), capsules

Active control A: celecoxib, 200mg (2 x 33.3mg, TID), capsules

Active control B: glucosamine sulphate, 2000mg (2 x 333mg, TID), capsules

Rescue medication permitted: 500mg acetominophen (paracetamol), PRN

Outcomes

Pain on weightbearing VAS 0‐10, WOMAC 0‐4 (Indian version for hip and knee; pain and function subscales only), patient global 1‐5, physical global 1‐5, HAQ

Notes

Confirmatory study; statistical power 80%, alpha set at 0.05 (2 tailed). Reported compliance with ICH GCP guidelines, and Declaration of Helsinki. Reported clinical trial registration (CTRI/2008/091/000063). Results for Ayurvedic interventions show equivalent outcomes in pain and function to glucosamine sulphate and celecoxib, but the more participants in the Ayurvedic group reported (unexpected) adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in order of enrolment in the trial. "The study biostatistician (S.S.) used a standard software program to generate a randomized schedule of permuted block randomization with block size 4 for blinded (coded) drug allotment."

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active interventions, active control, and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included both intention‐to‐treat and per protocol analyses

Selective reporting (reporting bias)

Low risk

Outcome data reported as means and confidence intervals only, standard deviations computed for extraction and re‐analysis

Pain VAS 0‐10 converted to 100mm scale for data extraction and re‐analysis

Reported adverse events

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Active control B, glucosamine sulphate, is not an analgesic, and may be a poor choice of control in a trial using pain as a primary outcome measure

Cisar 2008

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 12 weeks intervention, plus 2 weeks washout/follow‐up

Participants

Randomised n=100, Completed intervention n=90, Completed washout n=81. Mean age 54 yrs. M:F control 18:32, intervention 14:36. Inclusion: OA knee (ACR criteria), Kellgren grade I or II, mild‐moderate pain in target knee for at least 3 months, morning stiffness, knee crepitus, age > 25 years. Female participants not pregnant, nor planning pregnancy for > 12 months post study

Interventions

Pycnogenol®: Pinus pinaster, pine bark extract with 90% proanthocyanines, 150mg (3 x 50mg), in 50mg doses TID with meals, tablets

Placebo control: ingredients not reported, tablets

Concurrent medication permitted: stable NSAIDs and analgesics

Outcomes

WOMAC 0‐4 (Slovak version), Pain VAS 0‐100

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis and assessment consistent with ACR criteria

Farid 2007

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 3 months (˜12 weeks)

Participants

Randomised n=37; intervention n=19, control n=18. Completed n=35. Mean age: control 48.9 yrs, intervention 47.5yrs. M:F: control 1:17, intervention 2:18. Inclusion: OA knee (ACR criteria)

Interventions

Pycnogenol®: Pinus pinaster, pine bark extract with 70% proanthocyanines, 150mg (3 x 50mg), tablets

Placebo control: "inactive ingredient", ingredients not report, tablets

Outcomes

WOMAC‐VAS (aggregated scores), NSAID/COX‐2 inhibitor use

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Adequate concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Reported identical per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Unclear risk

Reported no adverse events (low risk)

Other bias

Low risk

Diagnosis / assessment based on ACR criteria

Ferraz 1991

Methods

Randomised, double‐blind, placebo control, 2 group crossover. Duration 3 weeks (2 x 1 week crossover, 1 week washout)

Participants

Randomised n=22, Completed n=20. Mean age 62 yrs, range 47‐78 yrs. Inclusion: OA hip or knee, clinical and radiographic verification (criteria not specified)

Interventions

Tipi tea: Petiveria alliacea, aqueous extract, 3 x 200ml tea (equivalent to 9gm tipi)

Placebo control: Sape, Imperata exaltata (dose not specified)

Outcomes

Pain (scale not reported) at rest, pain with mvt, pain at night, 15 metre walking time, MACTAR patient preference questionnaire

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method not reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

Brief report. Full report not available (high risk)

Reported withdrawals (low risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

High risk

Criteria for diagnosis of OA not specified (high risk)

Financial and in kind support not reported

Frerick 2001

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 20 weeks

Participants

Randomised n=46; intervention n=24, control n=22. Completed n=41, intevention n=21, control n=20. Mean age: intervention 58 yrs, control 61 yrs. Gender data not reported. Inclusion: OA hip (ACR criteria)

Interventions

LoHar‐45 flexi‐loges®: Harpagophytum procumbens (devil's claw), 960mg, ethanolic extract, tablets

Placebo control: ingredients not reported

Outcomes

WOMAC‐VAS (German version). Post hoc "responders” to treatment were defined as participants whose WOMAC pain scores did not increase by more than 20% in weeks 17 to 20 of the study

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported compliance with ICH GCP guidelines. Results equivocal: no improvement on primary outcome measure (WOMAC).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised, method not reported1. Authors contacted: provided full details of computer generated randomisation sequence

Allocation concealment (selection bias)

Low risk

Allocation concealment not reported1. Authors contacted for confirmation, but details of allocation concealment not provided

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Reported intention‐to‐treat analysis

Selective reporting (reporting bias)

High risk

Reasons for withdrawals and adverse events not reported (high risk)

Outcome data reported as change scores, percentages, and bar charts only, insufficient for extraction (unclear risk)

Results show no improvement on planned primary outcome measure (WOMAC). Alternate outcome measure and definition of improvement constructed post hoc (high risk)

Other bias

Unclear risk

Diagnosis consistent with ACR criteria (low risk)

Post‐hoc created outcome measure not validated (unclear risk)

Gupta 2011

Methods

Randomised, double‐blind, placebo control, 2 parallel groups, multicentre (n=3). Duration 3 months (˜12 weeks)

Participants

Randomised n=90. Completed n=88; intervention n=44, control n=44. Inclusion: OA knee (knee pain, swelling, stiffness, tenderness, age 45+ years, one or more radiological signs)

Interventions

Antarth, Ayurvedic phytomedicine (mixture), dose not stated, 2 x BID, capsules

Placebo control: ingredients not reports, capsules

Rescue medication permitted: diclofenac sodium, up to 50mg BID; ranitidine up to 150mg OD

Outcomes

Pain VAS 0‐10, pain walking 0‐4, pain squatting 0‐4, pain crossing legs 0‐4, pain climbing stairs 0‐4, physician global (descriptive), patient global (descriptive), rescue medication use

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results slightly favour intervention. Participants receiving Antarth used less rescue medication and may be more satisfied than participants receiving placebo.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo control not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Outcome data reported as VAS 0‐10, converted to 100mm scale for data extraction (low risk)

Reported no adverse events (low risk)

Other bias

Unclear risk

Diagnosis not consistent with ACR criteria, but likely to be OA (unclear risk)

Huber 1991

Methods

Double‐blind, placebo control, 2 parallel groups. Duration 3 weeks

Participants

Recruited n=40, Completed n=38. Age range 50‐80 yrs. M:F 4:24. Inclusion: OA (criteria not specified), at least one indication for treatment with antirheumatics

Interventions

PhytodolorRN: standardised extract mixture of ash bark, aspen leaf, aspen bark, golden rod herb, 3 x 30 drops, tincture

Placebo control: ingredients not reported

Outcomes

Rescue medication use, joint size, maximum ROM, pain at rest, pain with mvt, pressure pain, finger‐ground distance (spine only), Schober index, percussion pain, serum biochemistry

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention. Some outcome measures (eg: Schober index) are non‐specific and may be of limited use in rheumatological assessment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Not randomised

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method not reported. In other studies of PhytodolorRN, active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

High risk

Outcome data variances reported as standard error of measurement (SEM). When converted to standard deviation (SD), data are skewed, violating an assumption of the inferential analyses (unclear risk)

Adverse events not reported (high risk)

Other bias

Unclear risk

Criteria for diagnosis of OA not specified (unclear risk)

Unvalidated outcome measures (unclear risk)

Jung 2001

Methods

Randomised, double‐blind, placebo control, 4 parallel groups, multicentre (n=2). Duration 4 weeks

Participants

Randomised n=96, Completed n=93. Mean age 58 yrs. M:F 9:84. Inclusion: OA knee, clinical and radiographic verification (criteria not specified), pain VAS 0‐100 >35mm

Interventions

SKI306X: standardised extract mixture of Clematis mandshurica, Prunella vulgaris, Trichosanthes kirilowii, 600mg (3 x 200mg), 1200mg (3 x 400mg), 1800mg (3 x 600mg), tablets

Placebo control: ingredients not reported, tablets

Outcomes

Pain VAS 0‐100, Lequesne index, patient opinion of efficacy 1‐5, investigator opinion of efficacy 1‐5, tolerability, serum biochemistry, heamatology, urinanalysis

Notes

Confirmatory study design, but statistical power not reported. Reported compliance with Helsinki Declaration and ethics committee approval. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method incompletely reported. Assume active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Unclear risk

Criteria for diagnosis of OA not specified, clinical and radiographic verification (unclear risk)

Jung 2004

Methods

Randomised, double‐blind, active control, 2 parallel groups. Duration 4 weeks

Participants

Randomised n=249, Completed n=214. Mean age 60 yrs. M:F 18:231. Inclusion: OA knee (ACR criteria), pain VAS 0‐100 >35mm

Interventions

SKI306X: standardised extract mixture of Clematis mandshurica, Prunella vulgaris, Trichosanthes kirilowii, 600mg (3 x 200mg), tablets

Active control: diclofenac SR, 100mg OD, tablets

Placebo controls: ingredients not reported, tablets (double dummy)

Concurrent medication permitted: medications for conditions unrelated to OA, if known not to interact with either study medications

Outcomes

Pain VAS 0‐100, Lequesne, patient global 1‐5, physician global 1‐5, tolerability, serum biochemistry, haematology, urinanalysis

Notes

Confirmatory study design, statistical power 80%, alpha 0.05.Reported compliance with the Declaration of Helsinki and institutional review board oversight. Results equivocal: SKI306X equally effective as diclofenac on pain, Lequesne index, patient and physician global scores. Participants using SKI306X reported fewer adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four or six to one of two groups using a computer generated random number sequence

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Double‐dummy method, placebo controls for both intervention and active controls. Active interventions and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria (low)

Kimmatkar 2003

Methods

Randomised, double‐blind, placebo control, crossover. Duration 19 weeks (2 x 8 weeks intervention + 3 week washout)

Participants

Randomised n=30, Completed n=30. No withdrawals. Mean age 59 yrs, range 45‐72 yrs. M:F 12:18. Inclusion: OA knee, clinical and radiographic verification (criteria not specified), currently using physiotherapy and NSAIDs

Interventions

Cap Wokvel™: Boswellia serrata (Gajabhakshya) extract with 40% boswellic acid, 1000mg (3 x 333mg), capsules

Placebo control: starch powder, capsules

Outcomes

Joint pain 0‐3, loss of function 0‐3, swelling 0‐3

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Reported that study formed part of an academic coursework requirement. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of two groups using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "The clinical orthopedic investigator and the patients were blind for the interventions"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported 100% compliance, no withdrawals

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

Unclear risk

Criteria for diagnosis of OA not specified: "clinoradiographic verification" (unclear risk)

Kuptniratsaikul 2009

Methods

Randomised, single blind, active control, 2 parallel groups. Duration 6 weeks

Participants

Randomised n=107, Completed n=91. Mean age intervention 61.4 yrs, active control 60.0 yrs. Primary OA knee (ACR criteria)

Interventions

Tradename not provided.Curcuma domestica extract, 2000mg (4 x 500mg) with 1000mg curcuminoids, capsules

Active control: ibuprofen, 800mg (2 x 400mg), method of administration not reported

Outcomes

Pain on level walking NRS 0‐10, pain on stair climbing NRS 0‐10, 100m walk (seconds), stair climb and descent (seconds)

Notes

Confirmatory study design, power 80%, alpha 0.05. Reported ethics committee approval. Efficacy of Curcuma domestica is not significantly different from active control (ibuprofen).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method of randomisation incompletely reported. Reported as "a computer randomisation code kept by a research assistant"

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

High risk

Blinding incomplete: research assistant not blind to allocation, and medication regimens differ between active control and intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

Unclear risk

Diagnosis consistent with ACR criteria (low risk)

Outcome assessments not validated measures (unclear risk)

Kuptniratsaikul 2011

Methods

Randomised, single blind, active control, 2 parallel groups. Duration 4 weeks

Participants

Randomised n=125; intervention n=63, control n=62. Completed n=107; intervention n=55, control n=52. Inclusion: primary OA knee (ACR criteria)

Interventions

Tradename not provided. Derris scandens extract, 800mg (400mg BID)

Active control: naproxen, 500mg (250mg BID)

Outcomes

WOMAC‐VAS (10cm normalised scores), 6 minute walk, patient global (categorical 1‐6), patient satisfaction (categorical 1‐6)

Notes

Confirmatory study design; power 80%, alpha 0.05. Reported ethics committee approval. Efficacy of Derris scandens is not significantly different from active control (naproxen).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation according to computer generated randomisation code

Allocation concealment (selection bias)

High risk

Allocation not concealed from participants or research assistant

Blinding (performance bias and detection bias)
All outcomes

High risk

Blinding incomplete: research assistant not blind to allocation, and interventions may be distinguishable between active control and intervention. interventions distinguishable

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Outcome data WOMAC‐VAS converted to 100mm scale for data extraction. Error identified during data extraction (unclear risk).

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Reported financial and in kind support, declared no competing financial interests

Leblan 2000

Methods

Randomised, double blind, active control, 2 parallel groups, multicentre (n=30 rheumatology practices). Duration 4 months (˜20 weeks)

Participants

Randomised n=122, Completed n=92. Mean age 61 yrs. M:F 45:77. Inclusion: primary OA knee or hip (ACR criteria), Kellgren stage I‐III

Interventions

Harpadol®: Harpagophytum procumbens (devil's claw), freeze‐ground powder, 2610 mg (6 x 435mg), equivalent to 60mg harpagoside, capsules.

Active control: diacerhein, 100mg (2 x 50mg), capsules

Placebo controls: ingredients not reports, capsules (double dummy)

Rescue medication permitted: acetaminophen‐caffeine OD PRN, followed by diclofenac 150mg (3 x 50mg) PRN

Outcomes

Pain VAS 0‐100, disability VAS 0‐100, Lequesne index, rescue medication use, patient global, investigator treatment preference

Notes

Confirmatory study; statistical power 90%, alpha 0.05 (1 tailed). Reported compliance with Declaration of Helsinki and ethics committee approval. Results indicate Harpagophytum equally effective as diacerhein on pain, function, and Lequesne index. Participants using Harpagophytum used less rescue medication (acetaminophen‐caffeine or diclofenac) and reported significantly fewer adverse effects.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method of randomisation incompletely reported. Reported as randomised in blocks of four patients at each study centre

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Double‐dummy method, placebo controls for both intervention and active controls. Active interventions and placebos not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included per‐protocol and intention‐to‐treat analyses. Missing data replaced using the last observation carried forward method

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Lequesne 2002

Methods

Randomised, placebo control, 2 parallel groups, multicentre (50 rheumatology practices). Duration 2 years (˜104 weeks)

Participants

Randomised n=163, Completed n=96, Returned 2 radiographs n=108. Mean age 63 years. M:F 102:61. Inclusion: OA hip (ACR criteria), Kellgren stage I‐III, joint space narrowing >1mm, Lequesne index >4

Interventions

Piascledine 300: Persa gratissma and Glycine max (avocado‐soyabean unsaponifiables), 300mg, capsules

Placebo control: ingredients not reported, capsules

Rescue medication permitted: NSAIDs measured in diclofenac equivalents, and analgesics (not specified), PRN

Concurrent medication permitted: all concomitant medications for medical diseases

Outcomes

Joint space width, Lequesne index, global pain VAS 0‐100, NSAID use (diclofenac equivalents), patient global (verbal 7 point scale), investigator global (verbal 4 point scale), days of sick leave, n participants requiring hip replacements

Notes

Confirmatory study; statistical power 80%, alpha 0.05. Reported ethics committee approval. Results equivocal; results favour intervention in subgroup of participants with advanced joint space narrowing.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four, to one of two groups, by an independent statistical unit

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria (ie: EULAR criteria)

Maheu 1998

Methods

Randomised, double‐blind, placebo control, 2 parallel groups, multicentre (n not specified). Duration 8.5 months (˜34 weeks); 15 day washout, 6 month intervention (˜24 weeks), 2 month follow‐up

Participants

Randomised n=164, Completed n=144. Mean age 64 yrs. M:F 46:118. Inclusion: OA knee or hip (ACR criteria), Kellgren stage IB‐III, active OA for 6 months, regular pain for 3 months

Interventions

Piascledine 300: Persa gratissma and Glycine max (avocado‐soyabean unsaponifiables), 300mg, capsules

Placebo control: ingredients not reported, capsules

Rescue medication permitted: analgesics PRN, up to 1 intra‐articular injection of corticosteroid "if absolutely necessary"

Outcomes

Lequesne index, pain VAS 0‐100, disability VAS 0‐100, number of participants using NSAIDs

Notes

Confirmatory study design; statistical power 90%, alpha 0.05. Reported review board approval, but unclear whether a formally constituted HREC approved the study protocol. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four, to one of two groups, using a table of random numbers

Allocation concealment (selection bias)

Low risk

Adequate. Allocation completed by an independent statistician

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria (low risk)

Maheu 2013

Methods

Randomised, double‐blind, placebo control, 2 parallel groups, multicentre (n=122; 52 rheumatology clinics, 70 general practices). Duration 3 years

Participants

Randomised n=399, Completed n=345. Mean age 62 yrs. M:F 46:54. Inclusion: OA hip (ACR criteria), joint space width (JSW) 1‐4mm, Lequesne index 3‐10 (scale 0‐24), pain for at least 1 year. Most symptomatic hip selected as target joint

Interventions

Piascledine 300: Persa gratissma and Glycine max (avocado/soyabean unsaponifiables), 300mg, capsules

Placebo control: ingredients not reported, capsules

Rescue medication permitted: analgesics or NSAIDs PRN recorded in self‐report diary

Outcomes

Change in joint space width (JSW; narrowest point on pelvis / hip AP view), WOMAC‐VAS, Lequesne index (normalised 0‐100)

Notes

Confirmatory study design; planned recruitment n=380 for statistical power 90%, alpha 0.05; actual power exceeded 75%. Reported ethics committee approval. Results equivocal for clinical outcomes. Fewer participants (20%) in the intervention group showed progression of joint space narrowing.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised: "Randomisation...by blocks of two for each stratum defined by baseline JSW"

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "Randomisation list established by an independent company"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Unclear risk

Variances reported as standard error of measurement (SEM). When converted to standard deviation (SD), data are not normally distributed, violating an assumption of the inferential analyses (unclear risk)

Change in JSW reported as joint loss in mm. Negative scores converted to positive for reanalysis so that higher scores mean worse (low risk).

Reported adverse events. Discussed intervention safety (low risk)

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Majima 2012

Methods

Randomised, unblinded, adjunct to active treatment, 2 parallel groups. Duration 12 weeks

Participants

Randomised n=50; intervention n=25, control n=25. Inclusion: primary knee OA (criteria not specified) with joint effusion

Interventions

Boiogito: Japanese herbal mixture containing extract of Sinomenium acutum, Astragalus, Atractylodes Lancea, Jujube, Glycyrrhiza, ginger, 7.5g (2.5g TID); and loxoprofen 60mg (20mg TID)

Control: loxoprofen 60mg (20mg TID)

Boiogito provided as adjunct therapy to loxoprofen

Outcomes

Knee Society Rating System, SF‐36, joint effusion (joint puncture)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported institutional review board oversight, but unclear whether a formally consistuted Human Research Ethics Committee approved the research design. Improvement in pain and function occured in both groups over time. Interventiong roup showed reduction in joint effusion as well as other measures.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

High risk

Open trial. Medication regimens differ between active control and intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Unclear risk

Criteria for diagnosis of OA not specified (unclear risk)

Medhi 2009

Methods

Randomised, double‐blind, active control, 2 parallel groups. Duration 4 weeks

Participants

Randomised n=110; intervention n=55, control n=55. Completed n=100; intervention n=50, control n=50. Age 40+ years. OA knee (not ACR criteria), knee pain, knee swelling

Interventions

Tradename not provided. Ricinus officinalis. Castor oil, 2.7ml (3 x 0.9ml), capsule

Active control: diclofenac sodium, 150mg (3 x 50mg), capsule

Concurrent intervention permitted: all participants encouraged to have physiotherapy

Outcomes

Pain VAS 0‐100

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results favour diclofenac over castor oil for improvement in osteoarthritic knee pain. Pain improved in both intervention and active control groups, but improvement was greater in the diclofenac group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported. Participants "selected from outpatients" may imply that allocation was unconcealed, or that participation in the study was not voluntary (unclear risk)

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double‐blind, method incompletely reported. Assume active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Five participants withdrew due to "efficacy failure", which may confound results

Selective reporting (reporting bias)

Unclear risk

Clinical outcome data reported as percentages and P values only, insufficient for extraction (unclear risk)

Reported adverse events (low risk)

Other bias

Unclear risk

Diagnosis / assessment not consistent with ACR criteria (unclear risk)

Mehta 2007

Methods

Randomised, double blind, active control (glucosamine sulfate), 2 parallel groups. Duration 8 weeks

Participants

Randomised n=95; intervention n=48, control n=47. Completed n=79; intervention n=41, control n=38. Mean age: control 55.1 yrs, intevention 51.9 yrs. OA knee (ACR criteria), Kellgren II or III, function VAS 0‐100 between 40mm and 80mm at baseline

Interventions

Reparagen®: combination of Uncaria guianensis (cat's claw; 300mg) and Lepidium meyenii (1500mg), 1800mg (2 x 2 x 450mg)

Active control: glucosamine sulfate, 1500mg (2 x 2 x 375mg), capsules

Rescue medication permitted: paracetamol (acetaminophen), up to 1500mg (3 x 500mg) per day in weeks 1‐4, 1000mg (2 x 500mg) per day in weeks 5‐8

Outcomes

WOMAC 0‐4, pain VAS 0‐100, rescue medication use

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported compliance with Helsinki Declaration and ethics committee approval. Reported clinical trials registration (ISRCTN25438351). Efficacy of Reparagen® is not significantly different from glucosamine sulfate.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Used a fixed allocation randomisation procedure

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Intervention and active control not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Unclear risk

Outcome data reported as change scores, percentages, graphs, and p values only, insufficient for extraction (unclear risk)

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Mills 1996

Methods

Randomised, double‐blind, placebo control, 2 parallel groups. Duration 2 months (˜8 weeks)

Participants

Randomised n=82 (all participants, OA and RA), Completed n=52 (plus RA n=20). Mean age (all participants, OA and RA) 62 yrs. Gender data not reported. Inclusion: Self‐identified arthritis pain, subsequently assessed by rheumatologist (ACR criteria), AIMS2 pain score of at least 3, not using prescribed salicylates, NSAIDs, or analgesics

Interventions

Reumalex: polyherbal mixture including extracts of willow bark, guaiacum resin, black cohosh, sarsparilla, and poplar bark, 2 "at a time", tablets.

Placebo control: calcium phosphate, tablets

Concurrent medication permitted: stable self‐prescribed analgesics

Outcomes

Pain AIMS 2, modified Ritchie index, analgesic use (diary)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results equivocal; medium effect size improvements in pain, but no reduction in analgesic use.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, but participants allocated directly to groups on order of enrolment into the trial (quasi‐randomised): "Assigned by accession to pre‐set lists of allocations randomised for equalisation in every ten, and after stratification by clinical condition, to one of two groups"

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active interventions, active control, and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Oben 2009

Methods

Randomised, double blind, placebo control, 4 parallel groups (2 x normal weight patients, 2 x overweight patients). Duration 8 weeks

Participants

Randomised n=80, Completed n=45. Age range 25‐60 yrs (mean age not reported). Gender data not reported. OA knee (ACR criteria) in adults of normal weight and overweight

Interventions

NP 06‐1: mixture of Phellodendron amurense tree bark extract with 50% berberine and Citrus sinensis peel extract a minimum of 30% polymethoxylated flavones, 1480mg (2 x 2 x 370mg), capsules

Placebo control: ingredients not reported, capsules

Outcomes

Lequesne index, BMI, CRP, ESR

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported university oversight, but unclear whether a formally consistuted human research ethics committee approved the research design. Results favour intervention. In the overweight intervention group, weight loss during the intervention period may have contributed to improvement.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number sequence

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Incompletely reported adverse events

Other bias

Unclear risk

Diagnosis / assessment consistent with ACR criteria (low risk)

Possible confounder: the berberine component of the intervention may have contributed to weight loss

Pavelka 2010

Methods

Randomised, double blind, active control, 2 parallel groups, multicentre (26 centres in 5 countries). Duration 8 months (6 months intervention, 2 months follow up)

Participants

Randomised n=361; intervention n=183, control n=178. Completed n=263; intervention n=142, control n=121. Included in ITT analyses n=357, intervention n=181, control n=176. Age 45+ years (range not reported). M:F 62:299. OA knee (ACR criteria)

Interventions

Piascledine 300: Persa gratissma and Glycine max (avocado‐soyabean unsaponifiables), 300mg, capsules

Active control: chondroitin sulphate, 1200mg (3 x 400mg), capsules

Placebo control: ingredients not reported, capsules

Rescue medication permitted: paracetamol (acetominophen), dose not reported

Outcomes

WOMAC‐VAS (aggregated scores), pain with active movement VAS 0‐100, pain at rest VAS 0‐100, Lequesne index (0‐24), use of rescue medication

Notes

Confirmatory study; statistical power 80%, alpha 0.05. Reported ethics committee application (approval not specified), and compliance with ICH GCP guidelines. WOMAC aggregated scores converted to normalised scores for data extraction and re‐analysis. Results show that ASU is not inferior to chondroitin sulphate on any outcome.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Described as randomised, method of randomisation incompletely reported1. "Randomisation to the two treatment groups was performed using the computer program Rancode 1.0". Author contacted: provided full details of computer generated randomisation

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1. Author contacted: confirmed allocation concealment using single sealed opaque envelopes

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Double‐dummy method, placebo controls for both intervention and active controls. Active interventions and placebos not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Piscoya 2001

Methods

Randomised, placebo control, 2 parallel groups, multicentre study. Duration 4 weeks

Participants

Randomised n=45, Completed n=45. Reported no withdrawals. Assume that group sizes were allocated a priori; intervention n=30, control n=15. Mean age 60 yrs, range 45‐75 yrs. All male. Inclusion: OA knee (ACR criteria), Kellgren stage II‐III, pain most days of the month, NSAIDs for 3 months

Interventions

Tradename not provided. Uncaria guianensis (cat's claw), aqueous extract, freeze‐dried, 100mg, tablets

Placebo control: ingredient not reported, "same excipient but without cat's claw", tablets

Outcomes

Pain at rest VAS 0‐10, pain at night VAS 0‐10, tenderness 0‐3, global tolerance 0‐4, blood variables

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported compliance with Declaration of Helsinki. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported. Assume that group sizes were allocated a priori

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported 100% compliance, no withdrawals (intervention over 4 weeks) (low risk)

Per‐protocol analysis (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Outcome measure VAS 0‐10 converted to 100mm scale for data extraction (low risk)

Variances reported as standard error of measurement (SEM). When converted to standard deviation (SD), data are skewed, violating an assumption of the inferential analyses (unclear risk)

Reported adverse events (low risk)

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Rein 2004a

Methods

Randomised, placebo control, 2 group crossover. Duration 6.5 months; 14 days run‐in, 2 x 3 months (˜12 weeks) intervention, no washout period)

Participants

Randomised n=112, Completed stage 1 n=97, Completed stage 2 n=85. Mean age 67 yrs. M:F 41:71. Inclusion: primary OA hip, knee, hand, shoulder, or neck, radiographic verification (criteria not specified), mild to moderate pain

Interventions

Hyben Vital: Rosa canina lito (rosehip and seed), 5000mg (2 x 5 x 500mg) equivalent to 1.5mg galactolipid, capsules

Placebo control: ingredients not reported, capsules

Rescue medication permitted: self‐prescribed analgesics, measured in paracetamol equivalents

Concurrent medication permitted: stable prescribed NSAIDs

Outcomes

Pain change 0‐4, rescue medication use (paracetamol equivalents), joint stiffness change 0‐4, point in time severity of pain, joint stiffness, wellbeing diary (mood, energy, sleep), patient treatment preference

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results moderately favour intervention. Evidence of carry‐over effect in the group receiving the intervention prior to the placebo.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four, to one of two groups, using a computer generated allocation schedule

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals

Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Unclear risk

Reported adverse events (low risk)

Subgroup analyses published separately (unclear risk)

Other bias

High risk

Criteria for diagnosis of OA not specified (high risk)

Schadler 1988

Methods

Randomised, double blind, placebo control, 2 group crossover. Duration 14 days (˜2 weeks); 2 x 7 days (˜1 week) intervention, no washout period

Participants

Randomised n=30, Completed n=30. Mean age 66 yrs, range 45‐81 yrs. M:F 7:23. Inclusion: OA knee, hip, thumb, shoulder (criteria not specified)

Interventions

PhytodolorRN: standardised extract mixture of ash bark, aspen leaf, aspen bark, golden rod herb, 3 x 40 drops, tincture

Placebo control: ingredients not reported

Outcomes

Diclofenac use, pain at rest 0‐3, pressure pain 0‐3, pain with mvt 0‐3, mobility impairment (scale not reported)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention (reduced used of diclofenac only, results equivocal for other outcomes).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double blind, method not reported. In other studies of PhytodolorRN, active intervention and placebo not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported 100% compliance, no withdrawals: likely in intervention over 7 days

Intention‐to‐treat analysis can be assumed

Selective reporting (reporting bias)

Unclear risk

Reported adverse events (low risk)

Other bias

Unclear risk

Criteria for diagnosis of OA not specified (unclear risk)

Schmelz 1997

Methods

Randomised, placebo control, 2 parallel groups. Duration 30 days (˜4 weeks)

Participants

Randomised n=56, Completed n=56. Age and gender data not reported. Inclusion: OA spine (criteria not specified), acute exacerbations

Interventions

Arthrotabs®: Harpagophytum procumbens (devil's claw), aqueous extract, 4290mg (2 x 3 x 715mg), tablets

Placebo control: ingredients not reported

Outcomes

Pain 0‐4

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Described as double blind, method not reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported 100% compliance, no withdrawals

Selective reporting (reporting bias)

Unclear risk

Outcome data reported as percentages and bar charts only, insufficient for extraction (unclear risk)

Reported adverse events (low risk)

Other bias

High risk

Criteria for diagnosis of OA not specified (high risk)

Unvalidated outcome measure (unclear risk)

Schmid 2000

Methods

Randomised, placebo control, 2 parallel, stratified groups. Duration 2 weeks

Participants

Randomised n=78, Completed n=68. Mean age 53 yrs. M:F 59:19. Inclusion: OA hip or knee (ACR criteria), clinical, radiographic and laboratory verification

Interventions

Tradename not provided. Salix purpurea x daphnoides cortex (willow bark) extract, 1360mg (2 x 2 x 340mg, equivalent to 240mg salicin), tablets

Placebo control: cellulose and lactose, tablets

Outcomes

WOMAC, patient global, physician global, haematology, urinanalysis

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval and compliance with ICH GCP guidelines. Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four, using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1. Authors contacted for confirmation, but details of allocation concealment not provided

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included per‐protocol and intention‐to‐treat analyses

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety (low risk)

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria

Sengupta 2008

Methods

Randomised, double blind, placebo control, 3 parallel groups (2 x intervention). Duration 90 days (˜12 weeks)

Participants

Randomised n=75, Completed n=70, intervention low dose (100mg/day 5‐Loxin) n=24, intervention high dose (250mg/day 5‐Loxin) n=23, control n=23 Mean age: control 52.4 yrs, intervention low dose 52.3 yrs, intervention high dose 53.2 yrs. M:F intervention low dose 7:17, intervention high dose 8:15, control 5:18. OA knee (ACR criteria), must report pain with movement VAS 0‐100 between 40mm and 70mm and Lequesne index > 7 points at baseline

Interventions

5‐Loxin®: extract of Boswellia serrata with 30% 3‐O‐acetyl‐11‐keto‐beta‐boswellic acid, low dose 100mg (2 x 50mg), high dose 250mg (5 x 50mg), capsules

Placebo control: rice bran, capsules

Rescue medication permitted: ibuprofen, up to 1200mg (3 x 400mg) PRN

Outcomes

VAS, Lequesne index, WOMAC

Notes

Confirmatory study design: statistical power 80%, alpha set at 0.05 ( 2 tailed). Reported institutional review board oversight, but unclear whether a formally constituted HREC approved the research design. Reported clinical trails registration (ISRCTN05212803). Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated randomisation table

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Unclear risk

Reported adverse events

Standard errors of measure erroneously labelled as standard deviations for some outcome data (WOMAC function subscale). Errors corrected during data extraction for re‐analysis (Analysis 2.2, Analysis 3.2)

Other bias

Unclear risk

Diagnosis/assessment consistent with ACR crtieria (low risk)

Non‐comparable groups: Intervention low‐dose group reported markedly greater pain (WOMAC subscale) than the other groups at baseline (unclear risk)

Sengupta 2010

Methods

Randomised, placebo control, 3 parallel groups (2 active products derived from the same herb), single centre trial. Duration 90 days (˜12 weeks)

Participants

Randomised n=60, Completed n=57, intervention A (100mg/day 5‐Loxin) n=19, intervention B (100mg/day Aflapin) n=19, control n=19. Mean age: intervention A 51.6 yrs, intervention B 53.2 yrs, control 52.4 yrs. M:F intervention A 3:16, intervention B 7:12, control 9:10. OA knee (ACR criteria), regular NSAID or acetominophen use for pain, must report pain VAS 0‐100 between 40mm and 70mm and Lequesne index > 7 points at baseline after regular medication washout

Interventions

5‐Loxin®: extract of Boswellia serrata with 30% 3‐O‐acetyl‐11‐keto‐beta‐boswellic acid 100mg (2 x 50mg), capsules

Aflapin®: extract ofBoswellia serrata, enriched with 30% 3‐O‐acetyl‐11‐keto‐beta‐boswellic acid, and non‐volatile Boswellia serrata oil, 100mg (2 x 50mg), capsules

Placebo control: ingredients not reported, "filled with a suitable excipient", capsules

Rescue medication permitted: ibuprofen, up to 1200mg (3 x 400mg) PRN

Outcomes

Pain VAS 0‐100, Lequesne index, WOMAC‐VAS (normalised units) pain, stiffness, physical function subscales

Notes

Confirmatory study design; statistical power 80%, alpha set at 0.05 (2 tailed). Reported institutional review board oversight, but unclear whether a formally constituted human research ethics committee approved the research design. Reported clinical trials registration (ISRCTN80793440). Results favour Aflapin over 5‐Loxin, and both products over placebo.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated randomisation table

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "The clinical trial pharmacist and statistician ensured that treatment codes remained confidential"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Low risk

Diagnosis/assessment consistent with ACR criteria (low risk)

Sontakke 2007

Methods

Randomised, unblinded, active control (valdecoxib), 2 parallel groups. Duration 7 months; 6 months (˜24 weeks) intervention, plus 1 month follow‐up

Participants

Randomised n=66; intervention n=33, control n=33. Completed n=57; intervention n=31, control n=27. Age 40‐70 years. OA knee (ACR criteria). Not pregnant or lactating

Interventions

Cap Wovkel™: containing Boswellia serrata extract with 65% organic acids, 999mg (3 x 333mg), capsules

Active control: valdecoxib, 10mg OD, tablets

Rescue medication permitted: ibuprofen, up to 1200mg (400mg TID), PRN

Outcomes

WOMAC‐VAS (aggregated scores)

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported university oversight, but unclear whether a formally constituted HREC approved the research design. WOMAC aggregated scores converted to normalised VAS 0‐100 scale during data extraction for re‐analysis. Results reported to favour intervention, but re‐analysis of data indicates that results slightly favour intervention for WOMAC pain subscale scores only. Results favour control on all other outcomes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised using an independent computerised system: "The patients were randomly allocated by SAS for Windows..."

Allocation concealment (selection bias)

High risk

Open trial. Allocation not concealed

Blinding (performance bias and detection bias)
All outcomes

High risk

Open trial. Medication regimens differ between active control and intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events and rescue medication use

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Teekachunhatean 2004

Methods

Randomised, double blind, active control, 2 parallel groups. Duration 5 weeks; 1 week run‐in, 4 weeks intervention

Participants

Randomised n=200, Completed n=188. Mean age 62 yrs. M:F 41:159. Inclusion: OA knee unilateral or bilateral (ACR criteria), Kellgren stage II‐IV

Interventions

Duhuo Jisheng Wan (DJW): herbal mixture containing angelica root and mulberry mistletoe, 9000mg (6 x 3 x 500mg), capsules

Active control: diclofenac sodium (Voltaren), 75mg (3 x 25mg), tablets packed in capsules

Placebo controls: cane sugar, tablets packed in capsules, and capsules

Outcomes

Battery of pain VAS 0‐100 (night pain, pain with standing, pain with movement, pain with stair climbing, resting pain, total pain), battery of stiffness VAS 0‐100 (morning stiffness, stiffness at rest, total stiffness), Lequesne index, time to climb 10 stairs, patient opinion of improvement VAS 0‐100, investigator opinion of improvement VAS 0‐100

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval and compliance with Declaration of Helsinki. Reported clinical trials registration (ISRCTN70292892). DJW equally effective as diclofenac on pain, stiffness, and Lequesne index. Participants using DJW reported improvements after a longer period of intervention that participants using diclofenac. Toxicity profiles of interventions are approximately equal. DJW is a large dose (9g, administered as 18 capsules per day) which may be a barrier to long‐term clinical compliance.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as randomised, method not reported. Baseline parameters compared for significant differences

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Double‐dummy method, placebo controls for both intervention and active controls. Active interventions and placebos not distinguished by look, taste, smell or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Reported adverse events. Discussed intervention safety

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Vishal 2011

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 30 days (˜5 weeks)

Participants

Randomised n=60; intervention n=30, control n=30. Completed n=59; intervention n=30, control n=29. Age 40‐80 years. OA knee (ACR criteria), most painful knee VAS >40mm, Lequesne index >7

Interventions

Aflapin®: extract ofBoswellia serrata with 30% 3‐O‐acetyl‐11‐keto‐beta‐boswellic acid and non‐volatile Boswellia serrata oil, 100mg (2 x 50mg), capsules.

Placebo control: ingredients not reported, "similar organoleptic properties", capsules

Rescue medication permitted: ibuprofen, up to 1200mg (3 x 400mg) PRN

Outcomes

VAS, Lequesne index, WOMAC‐VAS (normalised units), pain, stiffness, physical function subscales

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported institutional review board oversight, but unclear whether a formally constituted HREC approved the research design. Reported clinical trials registration (ISRCTN69643551). Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated randomisation schedule

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "Randomization codes were secured confidentially by the clinical trial pharmacist and statistician"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Reported withdrawals (low risk)

Per‐protocol analysis only (unclear risk)

Selective reporting (reporting bias)

Low risk

Reported adverse events

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Warholm 2003

Methods

Randomised, placebo control, 2 parallel groups. Duration 4 months (˜20 weeks)

Participants

Randomised n=100; intervention n=50, control n=50. Completed n=96; intervention n= 48, control n=48. Mean age 65 yrs. M:F 35:65. Inclusion: OA hip or knee, radiographic verification (criteria not specified)

Interventions

Hyben Vital: Rosa canina lito (rosehip and seed), 5000mg (2 x 5 x 500mg), equivalent to 1.5mg galactolipid, capsules

Placebo control: ingredients not reported, capsules

Outcomes

Active and passive range of motion (goniometer), activities of daily living VAS 0‐10, pain relief 0‐4, NSAID use

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Did not report ethical oversight or compliance with guidelines. Results favour intervention for some ranges of motion and pain, but are less convincing for activities of daily living.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised using an independent computerised system

Allocation concealment (selection bias)

Unclear risk

Allocation concealment not reported

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat and per protocol analyses

Selective reporting (reporting bias)

Unclear risk

Pain data reported as percentages and graphs only, insufficient for data extraction (unclear risk)

Reported adverse events (low risk)

Other bias

Unclear risk

Criteria for diagnosis of OA not specified, radiographic verification only (unclear risk)

Wigler 2003

Methods

Randomised, double blind, placebo control, 2 group crossover. Duration 48.5 weeks; 4 days run‐in, 2 x 12 weeks crossover, 24 weeks open follow‐up

Participants

Randomised n=29, Completed stage 1 n=24, Completed stage 2 n=20, Completed stage 3 (open trial) n=17. Mean age 62 yrs, range 42‐85 yrs Inclusion: OA knee (ACR criteria), Kellgren II‐IV, pain VAS 0‐100 >35mm. Not pregnant or lactating

Interventions

Zintona EC: Zingiber officinale (ginger) extract, 1000mg (4 x 250mg), equivalent to 40mg gingerol, capsules. Capsule contains 10mg gingerol absorbed on maltodextrin

Placebo control: maltodextrin only

Outcomes

WOMAC (Hebrew), knee circumference

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results equivocal in stage 1. Results in stage 2 and stage 3 favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised to one of two groups using a computer generated random number sequence

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred: "Both patients and investigators were blinded to treatment assignment"

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, packaging, or medication regimen

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis (success versus failure)

Selective reporting (reporting bias)

Unclear risk

Outcome data reported as means and confidence intervals. Standard deviations calculated for data extraction (unclear risk)

Reported adverse events

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Winther 2005

Methods

Randomised, placebo control, 2 group crossover. Duration 6.5 months; 14 days run‐in, 2 x 3 months (˜12 weeks) intervention, no washout period

Participants

Randomised n=94, Completed stage 1 n=94, Completed stage 2 n=80. Mean age 66 yrs. M:F 40:54. Inclusion: aged 35+ years, primary OA hip or knee, radiographic verification (ACR criteria), mild to moderate pain

Interventions

LitoZin: Rosa canina lito (rosehip and seed), 5000mg (2 x 5 x 500mg), equivalent to 1.5mg galactolipid, capsules

Placebo control: ingredients not reported, "inactive powder of similar taste, smell, and colour", capsules

Outcomes

WOMAC‐VAS

Notes

Exploratory study design; power, effect, and sample size not determined a priori. Reported ethics committee approval. Results moderately favour intervention. Evidence of carryover effect in the group receiving the intervention prior to the placebo.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in blocks of four, to one of two groups, using a computer generated allocation schedule

Allocation concealment (selection bias)

Low risk

Adequate allocation concealment can be inferred1

Blinding (performance bias and detection bias)
All outcomes

Low risk

Active intervention and placebo not distinguished by look, taste, smell, or packaging

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analysis

Selective reporting (reporting bias)

Unclear risk

Reported adverse events (low risk)

Subgroup analyses published separately (unclear risk)

Other bias

Low risk

Diagnosis / assessment consistent with ACR criteria

Unless otherwise stated, all oral medications are reported as total daily doses, which may have been administered in single or divided doses.

Unless subscales are named, outcome measures (eg: WOMAC, HAQ, COAT) were used in entirety. Unless specified, all outcome measures were administered, scored, and scaled according to Osteoarthritis Research Society International (OARSI) standards.

1. Reported compliance with ICH GCP guidelines (ICH 2004) anchored in European law, or ethics committee oversight that would require the same, so adequate randomisation, allocation concealment, and blinding can be assumed.

2. Indicates that the tradename was not provided in the manuscript, but has been determined through communication with the manufacturing company noted in the acknowledgements.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Anonymous 1993

Discussion paper

Belcaro 2010

Intervention included extracted or synthetic curcumin and another synthetic compound from soy lecithin (isolated compounds), therefore not herbal as per WHO definition

Biswas 1997

Abstract only. Unable to identify individual herbal interventions. Ingredients not listed in sufficient detail to allow replication of the study

Biswas 1998

Mixed sample, including people with rheumatoid arthritis and non‐specific arthritis. Unable to extract data on OA only

Brien 2006

Review paper

Chantre 2000

Repeat publication of Leblan 2000

Chrubasik 1998

Review paper

Dharmananda 1985

Discussion paper

Du 2006

Mixed sample, including people with "rheumatism due to blockage of cold and damp". Unable to extract data on OA only

Falch 1997

Discussion paper

Fang 2008

Individualised treatments, not a standardised dose, therefore, considered as a case series rather than RCT. Metabolic outcomes only. No functional or clinical outcomes

Gendo 1997

Discussion paper

Grahame 1981

Not RCT

Guyader 1984

Case series, not RCT. Used inappropriate statistical analyses

Hamblin 2008

Individualised treatments, not a standardised plant product or dose, therefore, a case series rather than RCT. Abstract only available from CENTRAL. Cannot locate full manuscript

Jacquet 2009

Intervention not purely herbal. Unable to identify effects of herbal intervention alone

Kagore 2011

Case study

Kielczynski 1997

Discussion paper

Kulkarni 1991

Intervention not purely herbal. Unable to identify effects of herbal interventions alone. Ingredients not listed in sufficient detail to allow replication of the study

Lechner 2011

Individualised treatments, not a standardised plant product or dose, therefore, considered as a case series rather than RCT

Levy 2009

Intervention (Limbrel) included extracted biacalin and catechin (flavanoids, isolated compounds), therefore not herbal as per WHO definition. According to the manufacturer, "each capsule of Limbrel also contains 50 mg of citrated zinc bisglycinate, which provides 10 mg of elemental zinc", a potentially active substance (http://www.limbrel.com/limbrel.php). Study is only repeatable using the proprietary product

Linsheng 1997

Not RCT

Loew 1996

Not RCT. Primary measures not consistent with the topic of this review

Long 2001

Review paper

Lung 2004

Repeat analysis (safety data only) of Jung 2004

Mishra and Singh 2003

Not RCT (single group trial). Author list in indexed citation differs from actual publication

Myers 2010

Not RCT (open label, uncontrolled study)

Park 2009

Intervention included magnesium (mineral), therefore not herbal as per WHO definition. Magnesium is a potentially active substance that may alter the active principle of the herbal extracts, unless it has been demonstrated that they are essentially similar

Rein 2004b

Abstract only. Subgroup analysis of Rein 2004a

Reuss 1981

Discussion paper

Rosen 2013

Not RCT. Open‐label, uncontrolled, cohort (single group) study

Sagar 1988

Not RCT

Saley 1987

Not RCT

Schaffner 1997

Mixed sample, including people with back pain not due to osteoarthritis. Unable to extract data on OA only

Schmid 1998a

Abstract only. Abstract to Schmid 2000 and Schmid 2001

Schmid 2001

Repeat publication of Schmid 2000

Srivastava 1989

Not RCT

Wang 1985

Not RCT

Wegener 2003

Not RCT

Winther 2004

Abstract only. Subgroup analysis of Rein 2004a

Xu 2005

Not RCT (case series)

Yuelong 2011

Not RCT (protocol for RCT)

Zell 1993

Not RCT

Zeng 2008

Metabolic outcomes only. No functional or clinical outcomes

Characteristics of studies awaiting assessment [ordered by study ID]

Gao 2012

Methods

RCT, 2 parallel groups. Duration: 4 weeks

Participants

n=96 (intervention n=48, active control n=48)

Interventions

Intervention: Bushen Huoxue Qubi decoction (ingredients unknown), one bag/day

Active control: diacerein (50 mg Bid, Po) and celecoxib (0.2 Qd, Po)

Outcomes

VAS (outcome and scale unknown), WOMAC, relative viscosity, aggregation index and IL‐1beta, NO, iNOS, LPO, SOD in serum, adverse events

Notes

Abstract only available. China Journal of Chinese materia medica indexed, but not held in known collections. Full manuscript sought in hard copy via inter‐library loan.

Hochberg 2012

Methods

RCT, multicentre (n=2; USA and Hong Kong), 2 parallel groups. Duration: 8 weeks

Participants

n=92 (USA n=53, Hong Kong n=39)

Interventions

Intervention: Hou‐Lou‐Xiao‐Ling Dan 5,180 mg/day (15 capsules in 3 divided doses)

Control: placebo (ingredients unknown), dose matched to intervention

Outcomes

WOMAC, patient global, patient global assessment of response to therapy

Notes

Abstract only available. Abstracts from the annual ACR meeting are published in Arthritis and Rheumatism. This study will be classified if a full manuscript published.

Kang 2011

Methods

RCT, 3 parallel groups

Participants

n=160 (A: intervention n=76, B: active control n=42, C: intervention plus active control n=46)

Interventions

Intervention: wangbi (ingredients and dose unknown)

Active control: voltaren (dose unknown)

Outcomes

Morning stiffness, joint tenderness, swelling, pain, functional activities, adverse reactions (outcome measures unknown). Results favour combination therapy

Notes

Abstract only available. Chinese Journal of Intergrated Traditional and Western medicine indexed, but not held in known collections. Full manuscript sought in hard copy via inter‐library loan.

Liu 2006

Methods

RCT

Participants

n=86. 4 groups: manipulation plus pyrola (n=22), manipulation (n=22), pyrola compound traditional Chinese medicine (n=22), and self‐exercise (n=20)

Interventions

The manipulation group: Prone position, rolling manipulation to the affected thigh for 5 minutes, mainly the Weizhong and Weiyang of the fossa poplitea and the posterolateral part of the leg; Supine position, rolling manipulation for 5 minutes to affected side of quadriceps femoris and superior part of the whirbone; Alternately manipulation of pressing‐kneading, flicking‐poking and digital‐pressing to Dubi, xiyan, Yanglingquan, Heding, Xiyangguan and Liangqiu; Rotating of the knee joint cooperated by the passive stretch, flexion, inward and lateral rotation; At last, spreading some ointment of Chinese holly leaf on the affected knee joint and scrubbing until give the patient a warm sensation. The treatment was done three times weekly for 4 weeks.

Pyrola group: The patients were treated with Chinese herbs orally (modified pyrola compound traditional Chinese medicine) twice a day with water for weeks.

Manipulation plus pyrola group: Patients were treated by Chinese herb orally besides the manipulation with the same processes as the manipulation group for 4 weeks.

Self‐exercise group: Patients were told to do exercise themselves such as stretching exercises, active and passive range‐of‐motion exercises, strengthening exercises and so on for 4 weeks. 

Outcomes

WOMAC 0‐100, 20m walk time

Notes

Abstract only available. Chinese Journal of Clinical Rehabilitation not indexed. Full manuscript sought in hard copy via inter‐library loan.

Pinsornsak 2012

Methods

RCT

Participants

n=88 (intervention n=44, control n=44)

Interventions

Intervention: diclofenac 75mg/day with 1000mg/day curcumin (Curcuma longa extract)

Active control: diclofenac 75mg/day with placebo (unknown)

Outcomes

Pain VAS, KOOS. Results equivocal

Notes

Abstract only available. Journal of the Medical Association of Thailand indexed, but not held in known collections. Full manuscript sought in hard copy via inter‐library loan.

Tao 2009

Methods

RCT

Participants

n=90 (intervention n=45, control n=45)

Interventions

Intervention: gubitong decoction (dose and ingredients unknown)

Control: glucosamine sulfate 1500mg/day (3 x 500mg)

Outcomes

WOMAC, symptom VAS. Results equivocal: statistically significant improvement in treatment and control groups

Notes

Abstract only available. Chinese Journal of Integrative Medicine not indexed. Full manuscript sought in hard copy via inter‐library loan.

Zhong 2006

Methods

RCT

Participants

n=88 (intervention n=44, control n=44)

Interventions

Intervention: Bushen Quhan Tongluo herbs by orally or externally washing

Bushen Quhan. Tongluo: Hutaorou (12 g), Buguzhi (12 g), Chaoduzhong (12 g), Shudi (15 g), Dahuixiang (9 g), Luoshiteng (15 g), Zhichuanwu (9 g), Sanqi (6 g), Wugong (3 g), Jixieteng (15 g). The prescription for external washing: Tuogucao (40 g), Danggui (15 g), Sumu (15 g), Shengdahuang (15 g), Shengnanxing (10 g), Ruxiang (10 g), Meyao (10 g), Bingpian (3 g). Oral administration: The medicine shall be taken with water of 37 degrees C one dose a day.

Patients in the control group were given sulfated glucosamine (Weiguli Capsule. Each capsule contains 314 mg of sulfated glucosamine crystal, which is equal to 250 mg of sulfated glucosamine) two capsules a time and 3 times a day as well as piroxicam (Yantong Xikang Pill) once a day and 20 mg each time. Patients in both groups were administrated for 12 weeks

Outcomes

WOMAC

Notes

Abstract only available. Chinese Journal of Clinical Rehabilitation not indexed. Full manuscript sought in hard copy via inter‐library loan.

Unable to distinguish oral administration internvention group from topical administration intervention group results from abstract alone.

Data and analyses

Open in table viewer
Comparison 1. Boswellia serrata 999 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 1 Pain (0 to 3).

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 1 Pain (0 to 3).

2 Function: loss of function (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 2 Function: loss of function (0 to 3).

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 2 Function: loss of function (0 to 3).

3 Participants (n) reported adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 3 Participants (n) reported adverse effects.

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 3 Participants (n) reported adverse effects.

Open in table viewer
Comparison 2. Boswellia serrata (enriched) 100 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 90 days Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐16.57 [‐24.67, ‐8.47]

Analysis 2.1

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

2 WOMAC‐VAS (Function) Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐8.21 [‐14.21, ‐2.22]

Analysis 2.2

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

3 Adverse event episodes (n) reported Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Open in table viewer
Comparison 3. Boswellia serrata (enriched) 250 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 90 days Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 3.1

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

3 Adverse event episodes (n) reported Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Open in table viewer
Comparison 4. Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

2

97

Mean Difference (IV, Random, 95% CI)

‐16.09 [‐20.37, ‐11.81]

Analysis 4.1

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 1 Pain VAS 0‐100.

1.1 At 90 days

1

38

Mean Difference (IV, Random, 95% CI)

‐18.10 [‐24.95, ‐11.25]

1.2 At 30 days

1

59

Mean Difference (IV, Random, 95% CI)

‐14.80 [‐20.29, ‐9.31]

2 WOMAC‐VAS (Function) Show forest plot

2

97

Mean Difference (IV, Random, 95% CI)

‐15.01 [‐19.21, ‐10.81]

Analysis 4.2

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 2 WOMAC‐VAS (Function).

2.1 At 30 days

1

59

Mean Difference (IV, Random, 95% CI)

‐14.30 [‐20.07, ‐8.53]

2.2 At 90 days

1

38

Mean Difference (IV, Random, 95% CI)

‐15.8 [‐21.92, ‐9.68]

3 Participants (n) reported adverse events Show forest plot

2

97

Odds Ratio (M‐H, Random, 95% CI)

0.98 [0.13, 7.29]

Analysis 4.3

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 3 Participants (n) reported adverse events.

3.1 At 30 days

1

59

Odds Ratio (M‐H, Random, 95% CI)

0.97 [0.06, 16.20]

3.2 At 90 days

1

38

Odds Ratio (M‐H, Random, 95% CI)

1.0 [0.06, 17.25]

Open in table viewer
Comparison 5. Boswellia serrata 999 mg versus valdecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

58

Mean Difference (IV, Random, 95% CI)

‐0.51 [‐7.26, 6.24]

Analysis 5.1

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 1 WOMAC‐VAS (Pain).

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 1 WOMAC‐VAS (Pain).

2 WOMAC‐VAS (Function) Show forest plot

1

58

Mean Difference (IV, Random, 95% CI)

2.49 [‐4.07, 9.05]

Analysis 5.2

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 2 WOMAC‐VAS (Function).

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

1

66

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.39, 10.18]

Analysis 5.3

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 3 Participants (n) reported adverse events.

4 Participants (n) withdrew due to adverse events Show forest plot

1

66

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 71.07]

Analysis 5.4

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 4 Participants (n) withdrew due to adverse events.

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 4 Participants (n) withdrew due to adverse events.

Open in table viewer
Comparison 6. Curcuma domestica versus ibuprofen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain on walking NRS 0‐10 Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 1 Pain on walking NRS 0‐10.

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 1 Pain on walking NRS 0‐10.

2 Function: 100m walk time (seconds) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 2 Function: 100m walk time (seconds).

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 2 Function: 100m walk time (seconds).

3 Participants (n) reported adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.46, 1.25]

Analysis 6.3

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 3 Participants (n) reported adverse events.

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 7. Derris scandens versus naproxen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) change from baseline Show forest plot

1

107

Mean Difference (IV, Random, 95% CI)

5.0 [‐1.84, 11.84]

Analysis 7.1

Comparison 7 Derris scandens versus naproxen, Outcome 1 WOMAC‐VAS (Pain) change from baseline.

Comparison 7 Derris scandens versus naproxen, Outcome 1 WOMAC‐VAS (Pain) change from baseline.

2 WOMAC‐VAS (Function) change from baseline Show forest plot

1

107

Mean Difference (IV, Random, 95% CI)

5.10 [‐0.13, 10.33]

Analysis 7.2

Comparison 7 Derris scandens versus naproxen, Outcome 2 WOMAC‐VAS (Function) change from baseline.

Comparison 7 Derris scandens versus naproxen, Outcome 2 WOMAC‐VAS (Function) change from baseline.

3 Participants (n) reported adverse events. Show forest plot

1

125

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.49, 1.15]

Analysis 7.3

Comparison 7 Derris scandens versus naproxen, Outcome 3 Participants (n) reported adverse events..

Comparison 7 Derris scandens versus naproxen, Outcome 3 Participants (n) reported adverse events..

Open in table viewer
Comparison 8. Harpagophytum procumbens versus diacerhein

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline at 120 days Show forest plot

1

92

Mean Difference (IV, Random, 95% CI)

‐5.10 [‐6.52, ‐3.68]

Analysis 8.1

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 1 Pain VAS 0‐100 change from baseline at 120 days.

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 1 Pain VAS 0‐100 change from baseline at 120 days.

2 Participants (n) reported adverse events Show forest plot

1

92

Risk Ratio (M‐H, Random, 95% CI)

0.4 [0.21, 0.75]

Analysis 8.2

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 2 Participants (n) reported adverse events.

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 2 Participants (n) reported adverse events.

Open in table viewer
Comparison 9. Petiveria alliacea versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (scale unknown) with mvt change from baseline Show forest plot

1

40

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐1.31, 1.11]

Analysis 9.1

Comparison 9 Petiveria alliacea versus placebo, Outcome 1 Pain (scale unknown) with mvt change from baseline.

Comparison 9 Petiveria alliacea versus placebo, Outcome 1 Pain (scale unknown) with mvt change from baseline.

2 Participants (n) reported adverse events Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.5 [0.28, 8.04]

Analysis 9.2

Comparison 9 Petiveria alliacea versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 9 Petiveria alliacea versus placebo, Outcome 2 Participants (n) reported adverse events.

Open in table viewer
Comparison 10. Pinus pinaster (Pycnogenol® 150 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

37

Mean Difference (IV, Random, 95% CI)

‐142.0 [‐199.55, ‐84.45]

Analysis 10.1

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 1 WOMAC‐VAS (Pain).

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 1 WOMAC‐VAS (Pain).

2 WOMAC‐VAS (Function) Show forest plot

1

37

Mean Difference (IV, Random, 95% CI)

‐529.0 [‐741.59, ‐316.41]

Analysis 10.2

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

2

137

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.08, 1.97]

Analysis 10.3

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 11. Pinus pinaster (Pycnogenol® 100 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC 0‐4 (Pain) Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐7.50 [‐8.43, ‐6.57]

Analysis 11.1

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 1 WOMAC 0‐4 (Pain).

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 1 WOMAC 0‐4 (Pain).

2 WOMAC 0‐4 (Function) Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐29.3 [‐30.99, ‐27.61]

Analysis 11.2

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 2 WOMAC 0‐4 (Function).

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 2 WOMAC 0‐4 (Function).

Open in table viewer
Comparison 12. Ricinus officinale versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants (n) reported adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.04 [0.00, 0.66]

Analysis 12.1

Comparison 12 Ricinus officinale versus placebo, Outcome 1 Participants (n) reported adverse events.

Comparison 12 Ricinus officinale versus placebo, Outcome 1 Participants (n) reported adverse events.

Open in table viewer
Comparison 13. Rosa canina versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relief of pain (0 to 4) at 3 months Show forest plot

1

97

Mean Difference (IV, Random, 95% CI)

0.43 [‐0.12, 0.98]

Analysis 13.1

Comparison 13 Rosa canina versus placebo, Outcome 1 Relief of pain (0 to 4) at 3 months.

Comparison 13 Rosa canina versus placebo, Outcome 1 Relief of pain (0 to 4) at 3 months.

2 WOMAC‐VAS (Pain) Show forest plot

1

94

Mean Difference (IV, Random, 95% CI)

‐2.5 [‐10.20, 5.20]

Analysis 13.2

Comparison 13 Rosa canina versus placebo, Outcome 2 WOMAC‐VAS (Pain).

Comparison 13 Rosa canina versus placebo, Outcome 2 WOMAC‐VAS (Pain).

3 WOMAC‐VAS (Function) Show forest plot

1

94

Mean Difference (IV, Random, 95% CI)

‐1.20 [‐8.98, 6.58]

Analysis 13.3

Comparison 13 Rosa canina versus placebo, Outcome 3 WOMAC‐VAS (Function).

Comparison 13 Rosa canina versus placebo, Outcome 3 WOMAC‐VAS (Function).

4 Participants (n) reported adverse events Show forest plot

2

194

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.63, 4.43]

Analysis 13.4

Comparison 13 Rosa canina versus placebo, Outcome 4 Participants (n) reported adverse events.

Comparison 13 Rosa canina versus placebo, Outcome 4 Participants (n) reported adverse events.

Open in table viewer
Comparison 14. Salix purpurea x daphnoides versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 14 days Show forest plot

1

68

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 14.1

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 1 Pain VAS 0‐100 at 14 days.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 1 Pain VAS 0‐100 at 14 days.

2 Function VAS 0‐100 at 14 days Show forest plot

1

68

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 14.2

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 2 Function VAS 0‐100 at 14 days.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 2 Function VAS 0‐100 at 14 days.

3 Participants (n) reported adverse events Show forest plot

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.57, 1.43]

Analysis 14.3

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 15. Salix purpurea x daphnoides versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 15.1

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 1 WOMAC‐VAS (Pain).

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 1 WOMAC‐VAS (Pain).

1.1 At 14 days

1

86

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 42 days

1

86

Mean Difference (IV, Random, 95% CI)

15.0 [5.91, 24.09]

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 15.2

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 2 WOMAC‐VAS (Function).

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 2 WOMAC‐VAS (Function).

2.1 At 14 days

1

86

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 At 42 days

1

86

Mean Difference (IV, Random, 95% CI)

12.0 [2.70, 21.30]

3 Participants (n) reported adverse events Show forest plot

1

86

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.43, 0.93]

Analysis 15.3

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 16. Uncaria guianensis versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (night) Show forest plot

1

45

Mean Difference (IV, Random, 95% CI)

‐11.10 [‐26.44, 4.24]

Analysis 16.1

Comparison 16 Uncaria guianensis versus placebo, Outcome 1 Pain VAS 0‐100 (night).

Comparison 16 Uncaria guianensis versus placebo, Outcome 1 Pain VAS 0‐100 (night).

2 Participants (n) reported adverse events Show forest plot

1

45

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.54, 5.17]

Analysis 16.2

Comparison 16 Uncaria guianensis versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 16 Uncaria guianensis versus placebo, Outcome 2 Participants (n) reported adverse events.

Open in table viewer
Comparison 17. Zingiber officinale (Zintona EC) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (movement) Show forest plot

1

24

Mean Difference (IV, Random, 95% CI)

‐9.0 [‐31.12, 13.12]

Analysis 17.1

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 1 Pain VAS 0‐100 (movement).

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 1 Pain VAS 0‐100 (movement).

2 Function (handicap) VAS 0‐100 Show forest plot

1

24

Mean Difference (IV, Random, 95% CI)

‐6.0 [‐27.25, 15.25]

Analysis 17.2

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 2 Function (handicap) VAS 0‐100.

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 2 Function (handicap) VAS 0‐100.

3 Participants (n) reported adverse events Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

3.5 [0.16, 78.19]

Analysis 17.3

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 18. Boswellia carteri + Curcuma longa versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Function: pain free walking time (minutes) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 18.1

Comparison 18 Boswellia carteri + Curcuma longa versus placebo, Outcome 1 Function: pain free walking time (minutes).

Comparison 18 Boswellia carteri + Curcuma longa versus placebo, Outcome 1 Function: pain free walking time (minutes).

1.1 At 1 month

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 2 months

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.3 At 3 months

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 19. Persea gratissma + Glycine max (ASU 300 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

4

651

Mean Difference (IV, Random, 95% CI)

‐8.47 [‐15.90, ‐1.04]

Analysis 19.1

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

1.1 At 3 months

2

326

Mean Difference (IV, Random, 95% CI)

‐11.90 [‐23.95, 0.15]

1.2 At 6 months

1

162

Mean Difference (IV, Random, 95% CI)

‐10.40 [‐17.20, ‐3.60]

1.3 At 12 months

1

163

Mean Difference (IV, Random, 95% CI)

1.0 [‐6.58, 8.58]

2 Pain VAS 0‐100 change from baseline at 36 months Show forest plot

1

345

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐7.39, 6.07]

Analysis 19.2

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 2 Pain VAS 0‐100 change from baseline at 36 months.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 2 Pain VAS 0‐100 change from baseline at 36 months.

3 Pain VAS 0‐100 grouped by joint Show forest plot

1

324

Mean Difference (IV, Random, 95% CI)

‐9.06 [‐15.24, ‐2.88]

Analysis 19.3

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 3 Pain VAS 0‐100 grouped by joint.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 3 Pain VAS 0‐100 grouped by joint.

3.1 VAS (hip OA)

1

162

Mean Difference (IV, Random, 95% CI)

‐13.80 [‐25.22, ‐2.38]

3.2 VAS (knee OA)

1

162

Mean Difference (IV, Random, 95% CI)

‐7.10 [‐14.45, 0.25]

4 Function: disability VAS 0‐100 Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 19.4

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 4 Function: disability VAS 0‐100.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 4 Function: disability VAS 0‐100.

5 WOMAC‐VAS (Function) change from baseline at 36 months Show forest plot

1

345

Mean Difference (IV, Random, 95% CI)

‐1.0 [‐7.14, 5.14]

Analysis 19.5

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 5 WOMAC‐VAS (Function) change from baseline at 36 months.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 5 WOMAC‐VAS (Function) change from baseline at 36 months.

6 Lequesne algofunctional index Show forest plot

3

480

Mean Difference (IV, Random, 95% CI)

‐1.17 [‐2.54, 0.20]

Analysis 19.6

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 6 Lequesne algofunctional index.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 6 Lequesne algofunctional index.

6.1 At 3 months

2

317

Mean Difference (IV, Random, 95% CI)

‐1.80 [‐2.68, ‐0.92]

6.2 At 12 months

1

163

Mean Difference (IV, Random, 95% CI)

0.10 [‐0.78, 0.98]

7 Function (various tools) Show forest plot

4

642

Std. Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.73, ‐0.11]

Analysis 19.7

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 7 Function (various tools).

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 7 Function (various tools).

8 Participants (n) reported adverse events Show forest plot

5

1050

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.97, 1.12]

Analysis 19.8

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 8 Participants (n) reported adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 8 Participants (n) reported adverse events.

9 Participants (n) withdrew due to adverse events Show forest plot

1

398

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.73, 1.80]

Analysis 19.9

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 9 Participants (n) withdrew due to adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 9 Participants (n) withdrew due to adverse events.

10 Particpants (n) reported serious adverse events Show forest plot

1

398

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.94, 1.59]

Analysis 19.10

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 10 Particpants (n) reported serious adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 10 Particpants (n) reported serious adverse events.

11 JSW change from baseline Show forest plot

2

453

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.43, 0.19]

Analysis 19.11

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 11 JSW change from baseline.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 11 JSW change from baseline.

11.1 < median group, at 24 months

1

55

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.73, ‐0.13]

11.2 > median group, at 24 months

1

53

Mean Difference (IV, Random, 95% CI)

0.16 [‐0.31, 0.63]

11.3 At 36 months

1

345

Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.22, 0.16]

Open in table viewer
Comparison 20. Persea gratissma + Glycine max (ASU 600 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐14.2 [‐20.82, ‐7.58]

Analysis 20.1

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

2 Lequesne algofunctional index Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐1.30 [‐2.38, ‐0.22]

Analysis 20.2

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 2 Lequesne algofunctional index.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 2 Lequesne algofunctional index.

3 Participants (n) reported adverse events Show forest plot

1

174

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.66, 1.74]

Analysis 20.3

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 21. Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 21.1

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 1 WOMAC‐VAS (Pain).

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 1 WOMAC‐VAS (Pain).

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 21.2

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 2 WOMAC‐VAS (Function).

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

1

357

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.59, 1.26]

Analysis 21.3

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 3 Participants (n) reported adverse events.

4 Paricipants (n) reported serious adverse events Show forest plot

1

357

Risk Ratio (M‐H, Random, 95% CI)

2.92 [0.31, 27.78]

Analysis 21.4

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 4 Paricipants (n) reported serious adverse events.

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 4 Paricipants (n) reported serious adverse events.

Open in table viewer
Comparison 22. Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lequesne algofunctional index Show forest plot

1

45

Mean Difference (IV, Random, 95% CI)

‐3.82 [‐7.05, ‐0.59]

Analysis 22.1

Comparison 22 Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo, Outcome 1 Lequesne algofunctional index.

Comparison 22 Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo, Outcome 1 Lequesne algofunctional index.

1.1 Normal BMI participants

1

18

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐3.37, ‐1.03]

1.2 Overweight BMI participants

1

27

Mean Difference (IV, Random, 95% CI)

‐5.50 [‐6.95, ‐4.05]

Open in table viewer
Comparison 23. Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants (n) reported adverse events Show forest plot

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.18, 3.24]

Analysis 23.1

Comparison 23 Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate, Outcome 1 Participants (n) reported adverse events.

Comparison 23 Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate, Outcome 1 Participants (n) reported adverse events.

Open in table viewer
Comparison 24. Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain immediately after walking 50 feet VAS 0‐100 Show forest plot

1

247

Mean Difference (IV, Random, 95% CI)

‐9.60 [‐16.81, ‐2.39]

Analysis 24.1

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 1 Pain immediately after walking 50 feet VAS 0‐100.

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 1 Pain immediately after walking 50 feet VAS 0‐100.

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 24.2

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 24.3

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 25. SKI306X versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 25.1

Comparison 25 SKI306X versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 25 SKI306X versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

1.1 Low dose (600mg) SKI306X

1

47

Mean Difference (IV, Random, 95% CI)

‐16.1 [‐25.19, ‐7.01]

1.2 Medium dose (1200mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐14.5 [‐23.04, ‐5.96]

1.3 High dose (1800mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐22.3 [‐31.82, ‐12.78]

2 Lequesne algofunctional index change from baseline Show forest plot

1

139

Mean Difference (IV, Random, 95% CI)

‐2.73 [‐3.71, ‐1.74]

Analysis 25.2

Comparison 25 SKI306X versus placebo, Outcome 2 Lequesne algofunctional index change from baseline.

Comparison 25 SKI306X versus placebo, Outcome 2 Lequesne algofunctional index change from baseline.

2.1 Low dose (600mg) SKI306X

1

47

Mean Difference (IV, Random, 95% CI)

‐2.40 [‐4.05, ‐0.75]

2.2 Medium dose (1200mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐2.8 [‐4.62, ‐0.98]

2.3 High dose (1800mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐3.0 [‐4.68, ‐1.32]

3 Participants (n) reported adverse events Show forest plot

2

139

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.49, 1.79]

Analysis 25.3

Comparison 25 SKI306X versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 25 SKI306X versus placebo, Outcome 3 Participants (n) reported adverse events.

3.1 Low dose (600mg) SKI306X

1

47

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.32, 2.88]

3.2 Medium dose (1200mg) SKI306X

1

46

Risk Ratio (M‐H, Random, 95% CI)

1.2 [0.43, 3.38]

3.3 High dose (1800mmg) SKI306X

1

46

Risk Ratio (M‐H, Random, 95% CI)

0.6 [0.16, 2.22]

Open in table viewer
Comparison 26. SKI306X (600 mg) versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

249

Mean Difference (IV, Random, 95% CI)

1.31 [‐2.78, 5.40]

Analysis 26.1

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 1 Pain VAS 0‐100 change from baseline.

2 Lequesne algofunctional index change from baseline Show forest plot

1

249

Mean Difference (IV, Random, 95% CI)

0.77 [0.10, 1.44]

Analysis 26.2

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 2 Lequesne algofunctional index change from baseline.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 2 Lequesne algofunctional index change from baseline.

3 Participants (n) reported adverse events Show forest plot

1

249

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.38, 0.97]

Analysis 26.3

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 27. Phytodolor N versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Enduring pain (0 to 3) Show forest plot

1

72

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 27.1

Comparison 27 Phytodolor N versus placebo, Outcome 1 Enduring pain (0 to 3).

Comparison 27 Phytodolor N versus placebo, Outcome 1 Enduring pain (0 to 3).

2 Function: mobility limitations (0 to 3) Show forest plot

1

72

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 27.2

Comparison 27 Phytodolor N versus placebo, Outcome 2 Function: mobility limitations (0 to 3).

Comparison 27 Phytodolor N versus placebo, Outcome 2 Function: mobility limitations (0 to 3).

3 Participants (n) reported adverse events Show forest plot

3

140

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.92]

Analysis 27.3

Comparison 27 Phytodolor N versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 27 Phytodolor N versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 28. Reumalex versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 AIMS2 arthritis pain score change from baseline Show forest plot

1

52

Mean Difference (IV, Random, 95% CI)

‐0.89 [‐1.73, ‐0.05]

Analysis 28.1

Comparison 28 Reumalex versus placebo, Outcome 1 AIMS2 arthritis pain score change from baseline.

Comparison 28 Reumalex versus placebo, Outcome 1 AIMS2 arthritis pain score change from baseline.

2 Participants (n) reported adverse events Show forest plot

1

52

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.40, 2.91]

Analysis 28.2

Comparison 28 Reumalex versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 28 Reumalex versus placebo, Outcome 2 Participants (n) reported adverse events.

Open in table viewer
Comparison 29. Chinese DJW versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (total) Show forest plot

1

200

Mean Difference (IV, Random, 95% CI)

11.81 [‐9.67, 33.29]

Analysis 29.1

Comparison 29 Chinese DJW versus diclofenac, Outcome 1 Pain VAS 0‐100 (total).

Comparison 29 Chinese DJW versus diclofenac, Outcome 1 Pain VAS 0‐100 (total).

2 Lequesne algofunctional index Show forest plot

1

200

Mean Difference (IV, Random, 95% CI)

0.28 [‐0.89, 1.45]

Analysis 29.2

Comparison 29 Chinese DJW versus diclofenac, Outcome 2 Lequesne algofunctional index.

Comparison 29 Chinese DJW versus diclofenac, Outcome 2 Lequesne algofunctional index.

3 Participants (n) reported adverse events Show forest plot

1

200

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.66, 1.63]

Analysis 29.3

Comparison 29 Chinese DJW versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 29 Chinese DJW versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 30. Chinese BNHS versus Chinese active control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (walking) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

2.0 [‐7.12, 11.12]

Analysis 30.1

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 1 Pain VAS 0‐100 (walking).

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 1 Pain VAS 0‐100 (walking).

2 WOMAC‐VAS (Function) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐7.57, 3.57]

Analysis 30.2

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 2 WOMAC‐VAS (Function).

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

1

60

Risk Ratio (M‐H, Random, 95% CI)

9.00 [0.51, 160.17]

Analysis 30.3

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 3 Participants (n) reported adverse events.

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 31. Chinese BNHS versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (walking) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐6.81, 2.81]

Analysis 31.1

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 (walking).

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 (walking).

2 WOMAC‐VAS (Function) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.0 [‐2.53, 2.53]

Analysis 31.2

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 2 WOMAC‐VAS (Function).

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 2 WOMAC‐VAS (Function).

3 Participants (n) reported adverse events Show forest plot

1

60

Risk Ratio (M‐H, Random, 95% CI)

9.00 [0.51, 160.17]

Analysis 31.3

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 32. Ayurvedic A to E versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse event episodes (n) reported Show forest plot

1

454

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.71, 1.28]

Analysis 32.1

Comparison 32 Ayurvedic A to E versus placebo, Outcome 1 Adverse event episodes (n) reported.

Comparison 32 Ayurvedic A to E versus placebo, Outcome 1 Adverse event episodes (n) reported.

1.1 Formula A versus placebo

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.63, 1.45]

1.2 Formula B versus placebo

1

108

Risk Ratio (M‐H, Random, 95% CI)

1.35 [0.92, 1.98]

1.3 Formula C versus placebo

1

82

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.50, 1.21]

1.4 Formula D versus placebo

1

72

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.34, 0.93]

1.5 Formula E versus placebo

1

102

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.82, 1.80]

Open in table viewer
Comparison 33. Ayurvedic Antarth versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

88

Mean Difference (IV, Random, 95% CI)

‐1.0 [‐9.79, 7.79]

Analysis 33.1

Comparison 33 Ayurvedic Antarth versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 33 Ayurvedic Antarth versus placebo, Outcome 1 Pain VAS 0‐100.

Open in table viewer
Comparison 34. Ayurvedic RA‐II versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

90

Mean Difference (IV, Random, 95% CI)

‐1.03 [‐1.18, ‐0.88]

Analysis 34.1

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 1 Pain VAS 0‐100.

2 WOMAC 0‐4 (Function) Show forest plot

1

90

Mean Difference (IV, Random, 95% CI)

‐5.80 [‐6.72, ‐4.88]

Analysis 34.2

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 2 WOMAC 0‐4 (Function).

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 2 WOMAC 0‐4 (Function).

3 Participants (n) reported adverse events Show forest plot

1

90

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.69, 1.58]

Analysis 34.3

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 35. Ayurvedic SGC versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

3.0 [‐3.28, 9.28]

Analysis 35.1

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

2.0 [‐0.72, 4.72]

Analysis 35.2

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

3 Participants (n) reported adverse events Show forest plot

1

210

Odds Ratio (M‐H, Random, 95% CI)

1.04 [0.58, 1.86]

Analysis 35.3

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 36. Ayurvedic SGC versus celecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

‐3.0 [‐8.98, 2.98]

Analysis 36.1

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

1.0 [‐1.60, 3.60]

Analysis 36.2

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

3 Participants (n) reported adverse events Show forest plot

1

207

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.56, 1.79]

Analysis 36.3

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 37. Ayurvedic SGCG versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

4.0 [‐1.42, 9.42]

Analysis 37.1

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

1.38 [‐1.40, 4.16]

Analysis 37.2

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

3 Participants (n) reported adverse events Show forest plot

1

211

Odds Ratio (M‐H, Random, 95% CI)

0.85 [0.47, 1.54]

Analysis 37.3

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 38. Ayurvedic SGCG versus celecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐7.42, 3.42]

Analysis 38.1

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

0.19 [‐2.59, 2.97]

Analysis 38.2

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

3 Participants (n) reported adverse events Show forest plot

1

208

Odds Ratio (M‐H, Random, 95% CI)

0.82 [0.45, 1.48]

Analysis 38.3

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 39. Japanese Boiogito + loxoprofen versus loxoprofen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain: Knee Society Rating System 0‐100 (knee) Show forest plot

1

47

Mean Difference (IV, Random, 95% CI)

‐1.30 [‐8.90, 6.30]

Analysis 39.1

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 1 Pain: Knee Society Rating System 0‐100 (knee).

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 1 Pain: Knee Society Rating System 0‐100 (knee).

2 Function: Knee Society Rating System 0‐50 (stairs) Show forest plot

1

47

Mean Difference (IV, Random, 95% CI)

3.60 [0.51, 6.69]

Analysis 39.2

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 2 Function: Knee Society Rating System 0‐50 (stairs).

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 2 Function: Knee Society Rating System 0‐50 (stairs).

3 Participants (n) reported adverse events Show forest plot

1

47

Risk Ratio (M‐H, Random, 95% CI)

2.88 [0.12, 67.29]

Analysis 39.3

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 3 Participants (n) reported adverse events.

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 3 Participants (n) reported adverse events.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 1 Pain (0 to 3).
Figures and Tables -
Analysis 1.1

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 1 Pain (0 to 3).

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 2 Function: loss of function (0 to 3).
Figures and Tables -
Analysis 1.2

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 2 Function: loss of function (0 to 3).

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 3 Participants (n) reported adverse effects.
Figures and Tables -
Analysis 1.3

Comparison 1 Boswellia serrata 999 mg versus placebo, Outcome 3 Participants (n) reported adverse effects.

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.
Figures and Tables -
Analysis 2.1

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 2.2

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.
Figures and Tables -
Analysis 2.3

Comparison 2 Boswellia serrata (enriched) 100 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.
Figures and Tables -
Analysis 3.1

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 1 Pain VAS 0‐100 at 90 days.

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 3.2

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.
Figures and Tables -
Analysis 3.3

Comparison 3 Boswellia serrata (enriched) 250 mg versus placebo, Outcome 3 Adverse event episodes (n) reported.

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 1 Pain VAS 0‐100.
Figures and Tables -
Analysis 4.1

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 4.2

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 4.3

Comparison 4 Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 1 WOMAC‐VAS (Pain).
Figures and Tables -
Analysis 5.1

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 1 WOMAC‐VAS (Pain).

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 5.2

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 2 WOMAC‐VAS (Function).

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 5.3

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 4 Participants (n) withdrew due to adverse events.
Figures and Tables -
Analysis 5.4

Comparison 5 Boswellia serrata 999 mg versus valdecoxib, Outcome 4 Participants (n) withdrew due to adverse events.

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 1 Pain on walking NRS 0‐10.
Figures and Tables -
Analysis 6.1

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 1 Pain on walking NRS 0‐10.

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 2 Function: 100m walk time (seconds).
Figures and Tables -
Analysis 6.2

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 2 Function: 100m walk time (seconds).

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 6.3

Comparison 6 Curcuma domestica versus ibuprofen, Outcome 3 Participants (n) reported adverse events.

Comparison 7 Derris scandens versus naproxen, Outcome 1 WOMAC‐VAS (Pain) change from baseline.
Figures and Tables -
Analysis 7.1

Comparison 7 Derris scandens versus naproxen, Outcome 1 WOMAC‐VAS (Pain) change from baseline.

Comparison 7 Derris scandens versus naproxen, Outcome 2 WOMAC‐VAS (Function) change from baseline.
Figures and Tables -
Analysis 7.2

Comparison 7 Derris scandens versus naproxen, Outcome 2 WOMAC‐VAS (Function) change from baseline.

Comparison 7 Derris scandens versus naproxen, Outcome 3 Participants (n) reported adverse events..
Figures and Tables -
Analysis 7.3

Comparison 7 Derris scandens versus naproxen, Outcome 3 Participants (n) reported adverse events..

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 1 Pain VAS 0‐100 change from baseline at 120 days.
Figures and Tables -
Analysis 8.1

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 1 Pain VAS 0‐100 change from baseline at 120 days.

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 2 Participants (n) reported adverse events.
Figures and Tables -
Analysis 8.2

Comparison 8 Harpagophytum procumbens versus diacerhein, Outcome 2 Participants (n) reported adverse events.

Comparison 9 Petiveria alliacea versus placebo, Outcome 1 Pain (scale unknown) with mvt change from baseline.
Figures and Tables -
Analysis 9.1

Comparison 9 Petiveria alliacea versus placebo, Outcome 1 Pain (scale unknown) with mvt change from baseline.

Comparison 9 Petiveria alliacea versus placebo, Outcome 2 Participants (n) reported adverse events.
Figures and Tables -
Analysis 9.2

Comparison 9 Petiveria alliacea versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 1 WOMAC‐VAS (Pain).
Figures and Tables -
Analysis 10.1

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 1 WOMAC‐VAS (Pain).

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 10.2

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 10.3

Comparison 10 Pinus pinaster (Pycnogenol® 150 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 1 WOMAC 0‐4 (Pain).
Figures and Tables -
Analysis 11.1

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 1 WOMAC 0‐4 (Pain).

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 2 WOMAC 0‐4 (Function).
Figures and Tables -
Analysis 11.2

Comparison 11 Pinus pinaster (Pycnogenol® 100 mg) versus placebo, Outcome 2 WOMAC 0‐4 (Function).

Comparison 12 Ricinus officinale versus placebo, Outcome 1 Participants (n) reported adverse events.
Figures and Tables -
Analysis 12.1

Comparison 12 Ricinus officinale versus placebo, Outcome 1 Participants (n) reported adverse events.

Comparison 13 Rosa canina versus placebo, Outcome 1 Relief of pain (0 to 4) at 3 months.
Figures and Tables -
Analysis 13.1

Comparison 13 Rosa canina versus placebo, Outcome 1 Relief of pain (0 to 4) at 3 months.

Comparison 13 Rosa canina versus placebo, Outcome 2 WOMAC‐VAS (Pain).
Figures and Tables -
Analysis 13.2

Comparison 13 Rosa canina versus placebo, Outcome 2 WOMAC‐VAS (Pain).

Comparison 13 Rosa canina versus placebo, Outcome 3 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 13.3

Comparison 13 Rosa canina versus placebo, Outcome 3 WOMAC‐VAS (Function).

Comparison 13 Rosa canina versus placebo, Outcome 4 Participants (n) reported adverse events.
Figures and Tables -
Analysis 13.4

Comparison 13 Rosa canina versus placebo, Outcome 4 Participants (n) reported adverse events.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 1 Pain VAS 0‐100 at 14 days.
Figures and Tables -
Analysis 14.1

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 1 Pain VAS 0‐100 at 14 days.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 2 Function VAS 0‐100 at 14 days.
Figures and Tables -
Analysis 14.2

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 2 Function VAS 0‐100 at 14 days.

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 14.3

Comparison 14 Salix purpurea x daphnoides versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 1 WOMAC‐VAS (Pain).
Figures and Tables -
Analysis 15.1

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 1 WOMAC‐VAS (Pain).

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 15.2

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 2 WOMAC‐VAS (Function).

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 15.3

Comparison 15 Salix purpurea x daphnoides versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 16 Uncaria guianensis versus placebo, Outcome 1 Pain VAS 0‐100 (night).
Figures and Tables -
Analysis 16.1

Comparison 16 Uncaria guianensis versus placebo, Outcome 1 Pain VAS 0‐100 (night).

Comparison 16 Uncaria guianensis versus placebo, Outcome 2 Participants (n) reported adverse events.
Figures and Tables -
Analysis 16.2

Comparison 16 Uncaria guianensis versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 1 Pain VAS 0‐100 (movement).
Figures and Tables -
Analysis 17.1

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 1 Pain VAS 0‐100 (movement).

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 2 Function (handicap) VAS 0‐100.
Figures and Tables -
Analysis 17.2

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 2 Function (handicap) VAS 0‐100.

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 17.3

Comparison 17 Zingiber officinale (Zintona EC) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 18 Boswellia carteri + Curcuma longa versus placebo, Outcome 1 Function: pain free walking time (minutes).
Figures and Tables -
Analysis 18.1

Comparison 18 Boswellia carteri + Curcuma longa versus placebo, Outcome 1 Function: pain free walking time (minutes).

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 1 Pain VAS 0‐100.
Figures and Tables -
Analysis 19.1

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 2 Pain VAS 0‐100 change from baseline at 36 months.
Figures and Tables -
Analysis 19.2

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 2 Pain VAS 0‐100 change from baseline at 36 months.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 3 Pain VAS 0‐100 grouped by joint.
Figures and Tables -
Analysis 19.3

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 3 Pain VAS 0‐100 grouped by joint.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 4 Function: disability VAS 0‐100.
Figures and Tables -
Analysis 19.4

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 4 Function: disability VAS 0‐100.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 5 WOMAC‐VAS (Function) change from baseline at 36 months.
Figures and Tables -
Analysis 19.5

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 5 WOMAC‐VAS (Function) change from baseline at 36 months.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 6 Lequesne algofunctional index.
Figures and Tables -
Analysis 19.6

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 6 Lequesne algofunctional index.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 7 Function (various tools).
Figures and Tables -
Analysis 19.7

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 7 Function (various tools).

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 8 Participants (n) reported adverse events.
Figures and Tables -
Analysis 19.8

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 8 Participants (n) reported adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 9 Participants (n) withdrew due to adverse events.
Figures and Tables -
Analysis 19.9

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 9 Participants (n) withdrew due to adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 10 Particpants (n) reported serious adverse events.
Figures and Tables -
Analysis 19.10

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 10 Particpants (n) reported serious adverse events.

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 11 JSW change from baseline.
Figures and Tables -
Analysis 19.11

Comparison 19 Persea gratissma + Glycine max (ASU 300 mg) versus placebo, Outcome 11 JSW change from baseline.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 1 Pain VAS 0‐100.
Figures and Tables -
Analysis 20.1

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 2 Lequesne algofunctional index.
Figures and Tables -
Analysis 20.2

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 2 Lequesne algofunctional index.

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 20.3

Comparison 20 Persea gratissma + Glycine max (ASU 600 mg) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 1 WOMAC‐VAS (Pain).
Figures and Tables -
Analysis 21.1

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 1 WOMAC‐VAS (Pain).

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 21.2

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 2 WOMAC‐VAS (Function).

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 21.3

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 4 Paricipants (n) reported serious adverse events.
Figures and Tables -
Analysis 21.4

Comparison 21 Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate, Outcome 4 Paricipants (n) reported serious adverse events.

Comparison 22 Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo, Outcome 1 Lequesne algofunctional index.
Figures and Tables -
Analysis 22.1

Comparison 22 Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo, Outcome 1 Lequesne algofunctional index.

Comparison 23 Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate, Outcome 1 Participants (n) reported adverse events.
Figures and Tables -
Analysis 23.1

Comparison 23 Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate, Outcome 1 Participants (n) reported adverse events.

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 1 Pain immediately after walking 50 feet VAS 0‐100.
Figures and Tables -
Analysis 24.1

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 1 Pain immediately after walking 50 feet VAS 0‐100.

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 24.2

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 2 WOMAC‐VAS (Function).

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 24.3

Comparison 24 Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 25 SKI306X versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 25.1

Comparison 25 SKI306X versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 25 SKI306X versus placebo, Outcome 2 Lequesne algofunctional index change from baseline.
Figures and Tables -
Analysis 25.2

Comparison 25 SKI306X versus placebo, Outcome 2 Lequesne algofunctional index change from baseline.

Comparison 25 SKI306X versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 25.3

Comparison 25 SKI306X versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 26.1

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 2 Lequesne algofunctional index change from baseline.
Figures and Tables -
Analysis 26.2

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 2 Lequesne algofunctional index change from baseline.

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 26.3

Comparison 26 SKI306X (600 mg) versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 27 Phytodolor N versus placebo, Outcome 1 Enduring pain (0 to 3).
Figures and Tables -
Analysis 27.1

Comparison 27 Phytodolor N versus placebo, Outcome 1 Enduring pain (0 to 3).

Comparison 27 Phytodolor N versus placebo, Outcome 2 Function: mobility limitations (0 to 3).
Figures and Tables -
Analysis 27.2

Comparison 27 Phytodolor N versus placebo, Outcome 2 Function: mobility limitations (0 to 3).

Comparison 27 Phytodolor N versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 27.3

Comparison 27 Phytodolor N versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 28 Reumalex versus placebo, Outcome 1 AIMS2 arthritis pain score change from baseline.
Figures and Tables -
Analysis 28.1

Comparison 28 Reumalex versus placebo, Outcome 1 AIMS2 arthritis pain score change from baseline.

Comparison 28 Reumalex versus placebo, Outcome 2 Participants (n) reported adverse events.
Figures and Tables -
Analysis 28.2

Comparison 28 Reumalex versus placebo, Outcome 2 Participants (n) reported adverse events.

Comparison 29 Chinese DJW versus diclofenac, Outcome 1 Pain VAS 0‐100 (total).
Figures and Tables -
Analysis 29.1

Comparison 29 Chinese DJW versus diclofenac, Outcome 1 Pain VAS 0‐100 (total).

Comparison 29 Chinese DJW versus diclofenac, Outcome 2 Lequesne algofunctional index.
Figures and Tables -
Analysis 29.2

Comparison 29 Chinese DJW versus diclofenac, Outcome 2 Lequesne algofunctional index.

Comparison 29 Chinese DJW versus diclofenac, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 29.3

Comparison 29 Chinese DJW versus diclofenac, Outcome 3 Participants (n) reported adverse events.

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 1 Pain VAS 0‐100 (walking).
Figures and Tables -
Analysis 30.1

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 1 Pain VAS 0‐100 (walking).

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 30.2

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 2 WOMAC‐VAS (Function).

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 30.3

Comparison 30 Chinese BNHS versus Chinese active control, Outcome 3 Participants (n) reported adverse events.

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 (walking).
Figures and Tables -
Analysis 31.1

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 (walking).

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 2 WOMAC‐VAS (Function).
Figures and Tables -
Analysis 31.2

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 2 WOMAC‐VAS (Function).

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 31.3

Comparison 31 Chinese BNHS versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 32 Ayurvedic A to E versus placebo, Outcome 1 Adverse event episodes (n) reported.
Figures and Tables -
Analysis 32.1

Comparison 32 Ayurvedic A to E versus placebo, Outcome 1 Adverse event episodes (n) reported.

Comparison 33 Ayurvedic Antarth versus placebo, Outcome 1 Pain VAS 0‐100.
Figures and Tables -
Analysis 33.1

Comparison 33 Ayurvedic Antarth versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 1 Pain VAS 0‐100.
Figures and Tables -
Analysis 34.1

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 2 WOMAC 0‐4 (Function).
Figures and Tables -
Analysis 34.2

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 2 WOMAC 0‐4 (Function).

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 34.3

Comparison 34 Ayurvedic RA‐II versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 35.1

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.
Figures and Tables -
Analysis 35.2

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 35.3

Comparison 35 Ayurvedic SGC versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 36.1

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.
Figures and Tables -
Analysis 36.2

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 36.3

Comparison 36 Ayurvedic SGC versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 37.1

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.
Figures and Tables -
Analysis 37.2

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 37.3

Comparison 37 Ayurvedic SGCG versus glucosamine sulphate, Outcome 3 Participants (n) reported adverse events.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.
Figures and Tables -
Analysis 38.1

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.
Figures and Tables -
Analysis 38.2

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 2 WOMAC 0‐4 (Function) change from baseline.

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 38.3

Comparison 38 Ayurvedic SGCG versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 1 Pain: Knee Society Rating System 0‐100 (knee).
Figures and Tables -
Analysis 39.1

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 1 Pain: Knee Society Rating System 0‐100 (knee).

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 2 Function: Knee Society Rating System 0‐50 (stairs).
Figures and Tables -
Analysis 39.2

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 2 Function: Knee Society Rating System 0‐50 (stairs).

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 3 Participants (n) reported adverse events.
Figures and Tables -
Analysis 39.3

Comparison 39 Japanese Boiogito + loxoprofen versus loxoprofen, Outcome 3 Participants (n) reported adverse events.

Summary of findings for the main comparison. Boswellia serrata for treating osteoarthritis

Boswellia serrata for treating osteoarthritis

Patient or population: patients with treating osteoarthritis
Settings: Community: India
Intervention:Boswellia serrata 999 mg

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Boswellia serrata

Pain
Global pain 0‐3 (higher scores mean worse)
Follow‐up: mean 8 weeks

Mean pain in the control group at the end of treatment was 2.50 (0 to 3 scale).

Mean pain in the intervention groups was
2.24 lower
(2.64 to 1.84 lower).

30
(1 study)

⊕⊕⊝⊝
low1,2,3,4

Absolute improvement in pain was 56% (46% to 66%); Relative improvement in pain was 80% (66% to 94%)5; NNTB = 1 (95% CI 1 to 2).

Function
Loss of function 0‐3 (higher scores mean worse)
Follow‐up: mean 8 weeks

Mean disability in the control group at the end of treatment was 2.46 (0 to 3 scale).

Mean disability in the intervention groups was
2.16 lower
(2.56 to 1.76 lower).

30
(1 study)

⊕⊕⊝⊝
low1,2,3,4

Absolute improvement in function was 54% (44% to 64%); Relative improvement was 76% (62% to 90%)5; NNTB = 1 (95% CI 1 to 3).

Adverse events
Participants (n) reported adverse effects
Follow‐up: mean 8 weeks

No (n=0) participants in the control group reported adverse events.

0 per 1000

Two (n=2) participants in the intervention group reported adverse events.

0 per 1000

RR 5.00
(0.26 to 96.13)

30
(1 study)

⊕⊕⊝⊝
low1,2,3,4

Absolute risk of adverse events was 13% higher in the Boswellia serrata group (6% lower to 33% higher); Relative percentage change 400% worsening (74% to 9513% worsening); NNT n/a.6

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

30
(1 study)

See comment

Reported NIL withdrawals due to adverse events.

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

See comment

Serious adverse events not reported as discrete outcome.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Criteria for diagnosis of OA not specified.
2 Exploratory study design; power, effect, and sample size not determined a priori.
3 Ethical oversight not reported.

4 Downgrade estimate due to single study.

5 Control group baseline pain (SD) 2.80 (0.41), baseline disability 2.86 (0.35), from Kimmatkar 2003.

6 Number needed to treat (NNT) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/). Assumed a minimal clinically important difference of 1 point of a 0 to 3 point scale (pain, function).

Figures and Tables -
Summary of findings for the main comparison. Boswellia serrata for treating osteoarthritis
Summary of findings 2. Boswellia serrata (enriched) 100 mg for treating osteoarthritis

Boswellia serrata (enriched) 100 mg for treating osteoarthritis

Patient or population: patients with treating osteoarthritis
Settings: Community: India
Intervention:Boswellia serrata (enriched) 100 mg

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Boswellia serrata (enriched) 100mg

Pain

Global pain VAS 0‐100 (higher scores mean worse)
Follow‐up: mean 90 days

Weighted mean pain in the control groups at the end of treatment was 40.02 (0 to 100 scale).

The weighted mean pain in the intervention groups was
16.57 lower
(26.47 to 8.47 lower)

85
(2 studies)

⊕⊕⊕⊝
moderate2

Absolute improvement in pain was 17% (8% to 26%); Relative improvement in pain was 29% (15% to 43%)3; NNTB 2 (95% CI 1 to 6).

Function
WOMAC‐VAS (Function)1 0‐100 (higher scores mean worse)
Follow‐up: mean 90 days

Weighted mean disability in the control groups at the end of treatment was 33.13 (0 to 100 scale).

The weighted mean disability in the intervention groups was
8.21 lower
(14.21 to 2.22 lower)

85
(2 studies)

⊕⊕⊕⊝
moderate2

Absolute improvement was 8% (14% to 2%); Relative improvement was 20% (5% to 34%)3; NNTB 4 (95% CI 2 to 18).

Adverse events
Adverse event episodes (n) reported
Follow‐up: mean 90 days

625 per 1000

375 per 1000
(211 to 577)

RR 0.60
(0.39 to 0.92)

96
(1 study)

⊕⊕⊕⊝
moderate4

Absolute risk of adverse events was 25% lower in the Boswellia serrata group (6% to 44% lower); Relative percentage change 40% improvement (61% improvement to 9% worsening); NNT = 4 (95% CI 3 to 22).

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

96
(1 study)

See comment

Reported NIL withdrawals due to adverse events.

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

96
(1 study)

See comment

Reported NIL serious adverse events.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Sengupta 2008, Sengupta 2010, Vishal 2011: WOMAC scores presented as subscale scores only. Overall WOMAC not reported.

2 Confirmatory study design: statistical power 80%, alpha set at 0.05, but downgraded due to potential imprecision due to small number of participants; and lower limit of 95% CI does not preclude clincially insignificant change

3 Control group baseline measures taken from Sengupta 2008, the study most heavily weighted in the meta‐analyses. Control group baseline pain (SD) 56.88 (12.04), baseline disability 41.3 (9.6).

4 Downgrade estimate due to potential imprecision, eg, small number of events and participants from a single study.

5 Number needed to treat (NNT) is not applicable (n/a) when result is not statistically significant. NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/); NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). Assumed a minimal clinically important difference of 15 points on 0 to 100 mm pain scale, and 10 points on 0 to 100 mm function scale.

Figures and Tables -
Summary of findings 2. Boswellia serrata (enriched) 100 mg for treating osteoarthritis
Summary of findings 3. Boswellia serrata (enriched) 250 mg for treating osteoarthritis

Boswellia serrata (enriched) 250mg for treating osteoarthritis

Patient or population: patients with treating osteoarthritis
Settings: Community: India
Intervention:Boswellia serrata (enriched) 250mg

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Boswellia serrata (enriched) 250mg

Pain
Global pain VAS 0‐100 (higher scores mean worse)
Follow‐up: mean 90 days

Mean pain in the control group at the end of treatment was 41.76 (0 to 100 scale).

Mean pain in the intervention group was
27.54 lower
(34.64 to 20.44 lower).

47
(1 study)

⊕⊕⊕⊝
moderate2

Absolute improvement in pain was 28% (20% to 35%); Relative improvement in pain was 48% (36% to 61%)3 ; NNT = 1 (95% CI 1 to 2).

Function
WOMAC‐VAS (Function)1

(higher scores mean worse)
Follow‐up: mean 90 days

Mean disability in the control group at the end of treatment was 34.07 (0 to 100 scale).

Mean disability in the intervention group was
16.8 lower
(21.23 to 12.37 lower).

47
(1 study)

⊕⊕⊕⊝
moderate2

Absolute improvement in disability was 17% (12% to 21%); Relative improvement in disability was 41% (30% to 51%)3; NNT = 1 (95% CI 1 to 2).

Adverse events
Adverse event episodes (n) reported
Follow‐up: mean 90 days

526 per 1000

474 per 1000
(302 to 653)

RR 0.90
(0.62 to 1.30)

114
(1 study)

⊕⊕⊕⊝
moderate2

Absolute risk of adverse events was 5% lower in the Boswellia serrata group (24% lower to 13% higher); Relative percentage change 10% improvement (38% improvement to 30% worsening); NNT n/a.4

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

114
(1 study)

See comment

Reported NIL withdrawals due to adverse events.

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

114
(1 study)

See comment

Reported NIL serious adverse events.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Sengupta 2008: WOMAC scores presented as subscale scores only. Overall WOMAC not reported.

2 Downgrade estimate due to single study.

3 Control group baseline pain (SD) 56.88 (12.04), baseline disability 41.3 (9.6), from Sengupta 2008.

4 Number needed to treat (NNT) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/).

Figures and Tables -
Summary of findings 3. Boswellia serrata (enriched) 250 mg for treating osteoarthritis
Summary of findings 4. Boswellia serrata (enriched) plus non‐volatile oil for treating osteoarthritis

Boswellia serrata (enriched) plus non‐volatile oil for treating osteoarthritis

Patient or population: patients with treating osteoarthritis
Settings: Community: India
Intervention:Boswellia serrata (enriched) 100mg plus non‐volatile oil

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Boswellia serrata (enriched) plus non‐volatile oil

Pain
Global pain VAS 0‐100 (higher scores mean worse)
Follow‐up: 30‐90 days1

Weighted mean pain in the control groups at the end of treatment was 38.90 (0 to 100 scale).

Weighted mean pain in the intervention groups was
16.09 lower
(20.37 to 11.81 lower).

97
(2 studies)

⊕⊕⊕⊝
moderate2

Absolute improvement in pain was 16% (12% to 20%); Relative improvement in pain was 34%(25% to 42%)3; NNTB 2 (1 to 4)4

Function
WOMAC‐VAS (Function)5 normalised units

(higher scores mean worse)
Follow‐up: 30‐90 days

Weighted mean disability in the control groups at the end of treatment was 34.90 (0 to 100 scale).

Weighted mean disability in the intervention groups was
15.01 lower
(19.21 to 10.81 lower).

97
(2 studies)

⊕⊕⊕⊝
moderate2

Absolute improvement in disability was 15% (11% to 19%); Relative improvement in disability was 37% (27% to 47%)3; NNTB 2 (1 to 3).

Adverse events
Participants (n) reported adverse events
Follow‐up: 30‐90 days

42 per 1000

41 per 1000
(6 to 241)

RR 0.98
(0.14 to 6.69)

97
(2 studies)

⊕⊕⊕⊝
moderate2

Absolute risk of adverse events was 0% lower in the Boswellia serrata group (8% lower to 8% higher); Relative percentage change 2% improvement (86% improvement to 569% worsening); NNT n/a.5

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

See comment

Reported NIL withdrawals due to adverse events.

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

See comment

Reported NIL serious adverse events.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Vishal 2011: 30 day intervention. Sengupta 2010: 90 day intervention.
2Vishal 2011: Exploratory study design; power, effect, and sample size not determined a priori.
3 Control group baseline measures taken from Vishal 2011, the study most heavily weighted in the meta‐analyses. Control group baseline pain 47.6 (9.7), baseline disability 40.6 (9.5).

4 Number needed to treat to benefit (NNTB), and harm (NNTH) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/).

5Sengupta 2010, Vishal 2011: WOMAC scores presented as subscale scores only. Overall WOMAC not reported.

Figures and Tables -
Summary of findings 4. Boswellia serrata (enriched) plus non‐volatile oil for treating osteoarthritis
Summary of findings 5. Boswellia serrata compared to valdecoxib for treating osteoarthritis

Boswellia serrata compared to valdecoxib for treating osteoarthritis

Patient or population: patients with treating osteoarthritis
Settings: Community: India
Intervention:Boswellia serrata 999 mg
Comparison: valdecoxib

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Valdecoxib

Boswellia serrata

Pain
WOMAC‐VAS (Pain)

(higher scores mean worse)
Follow‐up: mean 6 months

Mean pain in the valdecoxib group at the end of treatment was 17.08 (0 to 100 scale).

Mean pain in the intervention groups was
0.51 lower
(7.26 lower to 6.24 higher).

58
(1 study)

⊕⊝⊝⊝
very low1,2,3

Absolute improvement in pain was 1% (7% improvement to 6% worsening); Relative improvement in pain was 1%4; NNT n/a.5

Function
WOMAC‐VAS (Function)5
(higher scores mean worse)

Follow‐up: mean 6 months

Mean disability in the valdecoxib group at the end of treatment was 16.64 (0 to 100 scale).

Mean disability in the intervention groups was
2.49 higher
(4.07 lower to 9.05 higher).

58
(1 study)

⊕⊝⊝⊝
very low1,2,3

Absolute worsening in disability was 3% (4% improvement to 9% worsening); Relative improvement in disability was 4%4; NNT n/a.5

Adverse events
Participants (n) reported adverse events
Follow‐up: mean 6 months

61 per 1000

121 per 1000
(23 to 448)

RR 2.0
(0.39 to 10.18)

66
(1 study)

⊕⊝⊝⊝
very low1,2,3

Absolute risk of adverse events was 6% higher in the Boswellia serrata group (8% lower to 20% higher); Relative percentage change 100% worsening (61% improvement to 918% worsening); NNT n/a.5

Adverse events

Participants (n) withdrew due to adverse effects

RR 3.0

(0.13 to 71.07)

66
(1 study)

⊕⊝⊝⊝
very low1,2,3

Reported one (1) withdrawal possibly due to adverse events.

Absolute risk of withdrawal due to adverse events was 3% higher in the Boswellia serrata group (5% lower to 11% higher); Relative percentage change 200% worsening (87% improvement to 7007% worsening); NNT n/a.5

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

66
(1 study)

See comment

Reported NIL serious adverse events.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Open trial. Medication regimens differ between active control and intervention.

2 Downgrade estimate due to single study. Treatment effect crosses midline (no effect).
3 Exploratory study design; power, effect, and sample size not determined a priori.

4 Baseline pain in valdecoxib group 49.2, baseline disability 51.6. Aggregate WOMAC scores converted to normalised scores for re‐analysis.

5 Number needed to treat (NNT) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/).

Figures and Tables -
Summary of findings 5. Boswellia serrata compared to valdecoxib for treating osteoarthritis
Summary of findings 6. Persea gratissma + Glycine max (ASU 300 mg) for treating osteoarthritis

Persea gratissma + Glycine max (ASU 300 mg) for treating osteoarthritis

Patient or population: patients with osteoarthritis
Settings: Community: France (3), Belgium (1).
Intervention:Persea gratissma + Glycine max (ASU 300 mg)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Persea gratissma + Glycine max (ASU 300mg)

Pain
Global pain VAS 0‐100 (higher scores mean worse)
Follow‐up: 3 to 12 months

Weighted mean pain in the control groups at end of treatment was 40.53 (0 to 100 scale).

Weighted mean pain in the intervention groups was
8.47 lower
(15.90 to 1.04 lower)

651
(4 studies)

⊕⊕⊕⊝
moderate1

Absolute improvement in pain was 8% (1% to 16%); Relative improvement in pain was 15% (2% to 29%)2; NNTB 8 (4 to 77)3

Function
Multiple tools4
Follow‐up: 3 to 12 months

Mean disability in the control group at end of treatment was 47.10 mm, on VAS 0 to 100 mm scale (higher scores mean worse)5.

Mean disability in the intervention groups was
7 mm lower
(12 mm to 2 mm lower6)

642
(4 studies)

⊕⊕⊕⊝
moderate1

SMD ‐0.42 (95% CI ‐0.73 to ‐0.11), in favour of ASU 300mg

Absolute improvement in disability was 7% (2% to 12%); Relative improvement in disability was 13% (4% to 23%)7; NNTB 5 (3 to 19)3

Adverse events
Participants (n) reported adverse events
Follow‐up: 3 to 36 months

510 per 1000

531 per 1000
(495 to 572)

RR 1.04
(0.97 to 1.12)

1050
(5 studies)

⊕⊕⊕⊝
moderate1

Absolute risk of adverse events is 2% higher in the ASU group (2% lower to 7% higher); Relative percentage change 4% worsening (9% improvement to 12% worsening); NNT n/a3

Adverse events

Participants (n) withdrew due to adverse effects

148 per 1000

169 per 100

(108 to 267)

RR 1.14

(0.73 to 1.80)

398

(1 study)

⊕⊕⊕⊝
moderate8

Absolute risk of participants withdrawing due to adverse events in 2% higher in ASU group (5% lower to 9% higher); Relative percentage change 14% worsening (27% improvement to 90% worsening); NNT n/a.3,9

Adverse events

Participants (n) reported serious adverse events

325 per 1000

397 per 1000

(306 to 517)

RR 1.22

(0.94 to 1.59)

398

(1 study)

⊕⊕⊕⊝
moderate8

Absolute risk of serious adverse events is 7% higher in the ASU group (2% lower to 17% higher); Relative percentage change 22% worsening (6% improvement to 59% worsening); NNT n/a.3,9

Radiographic joint changes

Change in Joint Space Width (JSW) from baseline

(higher scores mean worse).

Follow up: 24 to 36 months.

Weighted mean JSW change from baseline in the control groups at end of treatment was 0.65.

Mean JSW change from baseline in the intervention groups was 0.12 lower (0.43 lower to 0.19 higher)

453

(2 studies)

⊕⊕⊕⊝
moderate8

Absolute change

NNT n/a.3,9

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Downgrade due to heterogeneity, inconsistency

2 Calculations based on control group baseline pain measure taken from Blotman 1997, the most heavily weighted study in the meta‐analysis. Control group baseline mean (SD) pain 54.3 (11.9).

3 Number needed to treat to benefit (NNTB), or to harm (NNTH) = not applicable (n/a) when result is not statistically significant. NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/)NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office), assuming a minimal clinically important difference of 15 mm on a 0 to 100 mm pain scale, and 10 mm on a 0 to 100 mm function scale.

4 Multiple tools: Disability VAS reported in one study only (Maheu 1998); WOMAC change score reported in one study (Maheu 2013); Lequesne algofunctional index reported in four studies, but to avoid over‐reporting, data were extracted on this outcome from three studies only (Appelboom 2001, Blotman 1997, Lequesne 2002)

5 From Maheu 1998: follow‐up disability score in the control group 47.10 mm (VAS 0 to 100 mm scale)

6 Four trials pooled (Appelboom 2001, Blotman 1997, Lequesne 2002, Maheu 1998) using SMD, and re‐expressed as MD by multiplying the SMD (95% CI) by the baseline SD in the control group of Maheu 1998 (16.78).

7 Calculations based on data from Maheu 1998: control group baseline mean (SD) disability 52.5 (16.78), 0 to 100 mm VAS scale.

8 Downgrade estimate due to imprecision: few participants.

9 Treatment effect crosses midline (no effect).

Figures and Tables -
Summary of findings 6. Persea gratissma + Glycine max (ASU 300 mg) for treating osteoarthritis
Summary of findings 7. Persea gratissma + Glycine max (ASU 600 mg) for treating osteoarthritis

Persea gratissma + Glycine max (ASU 600 mg) for treating osteoarthritis

Patient or population: patients with osteoarthritis
Settings: Community: Belgium
Intervention:Persea gratissma + Glycine max (ASU 600 mg)

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Persea gratissma + Glycine max (ASU 600mg)

Pain
Global pain VAS 0‐100

(higher scores mean worse)

Follow up: 3 months

Mean pain in the control group at the end of treatment was 42.4 (0 to 100 scale).

Mean pain in the intervention group was
14.2 lower
(20.82 to 7.58 lower)

156
(1 study)

⊕⊕⊕⊝
moderate1

Absolute improvement in pain was 14% (21% to 8%); Relative improvement in pain was 26.5%2; NNT =

Function
Lequesne algofunctional index 0‐24

(higher scores mean worse)
Follow‐up: 3 months

Mean disability in the control group at the end of treatment was 7.8 (0 to 24 scale).

Mean disability in the intervention group was
1.3 lower
(2.38 to 0.22 lower)

156
(1 study)

⊕⊕⊕⊝
moderate1

Absolute improvement in disability was 1% (1% to 0%); Relative improvement in disability was 13.7%2; NNT =

Adverse events
Participants (n) reported adverse events
Follow‐up: 3 months

261 per 1000

278 per 1000
(165 to 431)

RR 1.07
(0.66 to 1.74)

174
(1 study)

⊕⊕⊕⊝
moderate1

Absolute risk of adverse events is 2% higher in the ASU group (11% lower to 15% higher); Relative percentage change 7% worsening (34% improvement to 74% worsening); NNT n/a.3

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

174
(1 study)

See comment

Withdrawals due to adverse events not reported as a discrete outcome in ASU 600mg subgroup.

Adverse events

Participants (n) reported serious adverse events

See comment

See comment

Not estimable

174
(1 study)

See comment

Serious adverse events not reported as a discrete outcome in ASU 600mg subgroup.

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Single study.

2 Control group baseline mean (SD) pain 53.5 (13.9), baseline mean (SD) disability 9.5 (2.2), from Appelboom 2001.

3 Number needed to treat (NNT) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/).

Figures and Tables -
Summary of findings 7. Persea gratissma + Glycine max (ASU 600 mg) for treating osteoarthritis
Summary of findings 8. Persea gratissma + Glycine max (ASU 300 mg) compared to chondroitin sulphate for treating osteoarthritis

Persea gratissma + Glycine max (ASU 300 mg) compared to chondroitin sulphate for treating osteoarthritis

Patient or population: patients with osteoarthritis
Settings: Community: Czech Republic, Slovak Republic, Hungary, Poland, Romania
Intervention:Persea gratissma + Glycine max (ASU 300mg)
Comparison: chondroitin sulphate

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Chondroitin sulphate

Persea gratissma + Glycine max (ASU 300mg)

Pain
WOMAC‐VAS (Pain)

(higher scores mean worse)
Follow‐up: mean 6 months

Mean pain in the chondroitin sulphate group at the end of treatment was 22.88 (0 to 100 scale).

The mean pain in the intervention group was
1.41 higher
(2.68 lower to 5.50 higher)

357
(1 study)

⊕⊕⊝⊝
low1,2

Absolute worsening of pain was 10% (10% improvement to 31% worsening); Relative worsening of pain was 3%3; NNT n/a.4

Function
WOMAC‐VAS (Function)

(higher scores mean worse)
Follow‐up: mean 6 months

Mean function in the chondroitin sulphate group at the end of treatment was 25.14 (0 to 100 scale).

The mean disability in the intervention group was
1.63 higher
(2.51 lower to 5.77 higher)

357
(1 study)

⊕⊕⊝⊝
low1,2

Absolute worsening of disability was 28% (43% improvement to 98% worsening); Relative worsening of disability was 3%3; NNT n/a.4

Adverse events
Participants (n) reported adverse events

244 per 1000

210 per 1000
(139 to 304)

RR 0.86
(0.59 to 1.26)

357
(1 study)

⊕⊕⊝⊝
low1,2

Absolute risk of adverse events was 3% lower in the ASU group (12% lower to 5% higher); Relative percentage change 14% improvement (41% improvement to 26% worsening); NNT n/a.4

Adverse events

Participants (n) withdrew due to adverse effects

See comment

See comment

Not estimable

357
(1 study)

Withdrawals due to adverse events not reported as a discrete outcome.

Adverse events

Participants (n) reported serious adverse events

6 per 1000

17 per 1000

(2 to 158)

RR 2.92

(0.31 to 27.78)

357
(1 study)

⊕⊕⊝⊝
low1,2

Absolute risk of serious adverse events was 1% higher in the ASU group (1% lower to 3% higher); Relative percentage change 192% worsening (69% improvement to 2678% worsening); NNT n/a.4

Radiographic joint changes

See comment

See comment

Not estimable

See comment

Radiographic joint changes not measured.

Quality of life

See comment

See comment

Not estimable

See comment

Quality of life not measured.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 SIngle study. Treatment effect crosses midline (no effect).

2 Chondroitin sulfate might not be active control. Non‐inferiority hypothesis may be flawed.

3 Chrondroitin sulfate group baseline pain 49.08, baseline disability 49.07. Aggregate WOMAC scores converted to normalised scores for re‐analysis.

4 Number needed to treat (NNT) = not applicable (n/a) when result is not statistically significant. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office). NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/).

Figures and Tables -
Summary of findings 8. Persea gratissma + Glycine max (ASU 300 mg) compared to chondroitin sulphate for treating osteoarthritis
Table 1. Herbal medicinal products used for the treatment of OA

PLANT

MEDICINAL PRODUCT

DOSE

MARKER

Botanical name

Part/s

Tradename

Preparation

Drug:Extract

mg/day

Constituent marker

Quantity of marker

References

Medicinal products from single plants

Boswellia serrata

gum resin

CapWokvelTM

extraction solvent not stated

not stated

999

boswellic acid

(total organic acids 65%)

40%

Kimmatkar 2003, Sontakke 2007

5‐Loxin

100 or 250

AKBA

30%

Sengupta 2008

Sengupta 2010

Aflapin

100

AKBA + non‐volatile oil

20%

Sengupta 2010

Vishal 2011

Curcuma domestica

root

study medication

ethanolic extract

not stated

curcumoids

500mg

Kuptniratsaikul 2009

Derris scandens

stem

study medication

ethanolic (50%) extract

not stated

800

genistein derivatve

not stated

Kuptniratsaikul 2011

Garcinia kola

seed

study medication

freeze‐dried aqueous extract

not stated

400

not stated

Adegbehingbe 2008

Harpagophytum procumbens

root

Arthrotabs

aqueous extract

1.5‐2.5:1

2400

harpagoside1

30 mg

Schmelz 1997.

Flexiloges

ethanolic (60%) extract

4.5‐5.5:1

960

<30 mg

Frerick 2001, Biller 2002.

Harpadol

cryoground powder

2610

60 mg

Leblan 2000.

Petiveria alliacea

herb

Tipi tea

aqueous extract

9g / 600 ml

600 ml

not stated

Ferraz 1991

Pinus pinaster (synonym Pinus maritima)

bark

Pycnogenol®

polyphenol concentrate

150

proanthocyanidins

45 (90%)

Cisar 2008

100

not stated

Belcaro 2008

150

70%

Farid 2007

Ricinus officinalis

seed

study medication

oil

not stated

2,7 ml

ricinoleic acid

not stated

Medhi 2009

Rosa canina lito

rose hip and seed

Hyben Vital or Litozin

powder

5000

galactolipid

1.5mg

Rein 2004aWarholm 2003

Winther 2005

Salix daphnoides

bark

study medication

ethanolic (70%) extract

8‐14:1

1573

salicin

240 mg

Biegert 2004.

Salix pupurea x daphnoides

bark

study medication

ethanolic (70%) extract2

10‐20:1

1360

salicin

240 mg

Schmid 2000.

Uncaria guianensis

bark

study medication

freeze‐dried aqueous extract

not stated

100

not stated

Piscoya 2001.

Vitellaria paradoxa 

seed

study medication

patented extract

not stated

2250

triterpenes

75%

Cheras 2010

Zingiber officinale3

root

EV.EXT 33

acetone extract3

20:1

510

not stated

Bliddal 2000.

Zingiber officinale

root

Zintona EC

CO2 extract

not stated

1000

gingerol

40 mg

Wigler 2003

Medicinal products from two plants

Boswellia carteri + Curcuma longa

gum + root

study medication

extract, solvent not stated

not stated

not stated

boswellic acid

37.5%

Badria 2002

Persea gratissma (P) + Glycine max (G)

oils

Piascledine 300

unsaponifiable fraction 1/3 P;2/3 G

300 or 600

not stated

Appelboom 2001, Blotman 1997, Lequesne 2002, Maheu 1998, Maheu 2013.

Phellondenron amurense + Citrus sinensis

bark

peel

NP 06‐1

extract, solvent not stated

not stated

370 mixture

berberine

polymethoxylated flavones

50%

30%

Oben 2009

Uncaria guianensis + Lepidium meyenii

bark

Reparagen®

freeze‐dried aqueous extract

not stated

1500

300

not stated

Mehta 2007

Zingiber officinale + Alpinia galanga

root

EV.EXT 77

acetone extract3

20:1

not stated

not stated

Altman 2001

Medicinal products from three or more plants

Clematis mandshurica + Prunella vulgaris + Trichosanthes kirilowii

root, flower, root; 1:1:2

SKI306X

ethanol 30% extracts, thereafter butanol extraction

7:1

600‐1800

oleanolic acid 4%, rosmarinic acids 0.2%, ursolic acids 0.5%, hydroxybenzoic acid 0.03%,
hydroxymethoxybenzoic acid 0.03%, trans‐cinnamic
acid 0.05%

Jung 2001, Jung 2004.

Fraxinus excelsior

bark

Phytodolor

fresh plant ethanolic (45,6%) extract

3:1:1

5‐8 ml

total flavonoids

0.34 ‐ 0.56 mg

Bernhardt 1991, Huber 1991, Schadler 1988.

salicyl alcohol

0.48 ‐ 0.8 mg

Solidago virgaurea

herb

isofraxidin

0.67 ‐ 1.1 mg

Populus tremula

bark and leaf

salicin

4.8 ‐ 8 mg

Salix alba

bark

Reumalex

powder

200

salicin

40‐80mg

Mills 1996

Guaiacun officinale

resin

powder

80

Cimicifuga racemosa

root

powder

70

Smilax (species not stated)

root

extract, solvent not stated

4:1

50

Populus (species not stated)

bark

extract, solvent not stated

7:1

34

Chinese mixture4

herb

Duhuo Jisheng Wan

powder

3 x 3 g

not stated

Teekachunhatean 2004.

Paeoniae alba

root

Chinese mixture:

Blood nourishing, hard softening (BNHS)

extract, solvent not stated

not stated

3150

paconiflorin

not stated

Cao 2005

Gentiana macrophylla

gentianine

Glycyrrhiza (species not stated)

not stated

Auryvedic formaulae5

powder

not stated

1000

total gingerols

not stated

Chopra 2011

Zingiber officinale 

rhizome

component of formulae A, B, C, D, and E

Tinospora cordifolia

stem

component of formulae A, B, C, D, and E

aqueous extract

220

tinosporosides

not stated

Withania somnifera

root

component of formulae B and E

aqueous extract

600

total withanolides

not stated

Emblica officinale

fruit

component of formulae C

aqueous extract

500

tannins

galic acid

not stated

Tribulus terrestris

fruit

component of formulae A and B

aqueous extract

216

total saponins

not stated

Ayuvedic formula6

Antarth3 (for sandhigata vata)

not stated

not stated

not stated

not stated

Gupta 2011

Ayuvedic formula

RA‐11

not stated

not stated

not stated

not stated

Chopra 2004

Ayuvedic formula

SGC

Chopra 2013

Ayuvedic

SGCG

Chopra 2013

Japanese mixture7

Boiogito

not stated

not stated

7.5g

not stated

not stated

Majima 2012

1. Harpagoside content estimated indirectly and approximately from iridoid glycoside content in daily dose of raw material (Sporer 1999).

2. Ethanolic extract stated in unpublished thesis but not in published paper (Schmid 1998b).

3. Information provided by manufacturer but not reported in paper.

4. Chinese herbal medicine contains 7.75% each of: radix angelicae pubescentiis, radix gentianae macrophyllae, cortex eucommiae, radix achyranthis bidentatae, radix angelicae sinensis, herba taxilli, radix rehmanniae preparata, rhizoma chuanxiong, cortex cinnamomi, radix ledebouriellae. 5% each of: radix paeoniae alba, radix codonopis, radix glycyrrhizae, poria. 2.5% herba asari.

5. All Ayurvedic formulae A‐E contain Zingiber officinale (dried rhizome powder, total gingerols as marker), and Tinospora cordifolia (dried stem aqueous extract, marker tinosporosides). Some formulae also included Emblica officinale, Withania somnifera, or Tribulus terrestris. Drug:extract ratio and marker content not stated.

6. Ayurvedic phytomedicine Antarth contains Boswellia serrata, Commiphora mukul, Curcuma longa and Vitex negundo, Alpinia galangal, Withania somnifera, Tribulus terrestris, and Tinospora cordifolia.

7. Japanese herbal medicine Boiogito contains Sinomenium acutum, Astragalus (species not stated) root, Atractylodes lancea rhizome, Jujube (probably Ziziphus zizyphus), Glycyrrhiza (species not stated), and ginger (species not stated, probably Zingiber officinale).

Figures and Tables -
Table 1. Herbal medicinal products used for the treatment of OA
Comparison 1. Boswellia serrata 999 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 Function: loss of function (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Participants (n) reported adverse effects Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figures and Tables -
Comparison 1. Boswellia serrata 999 mg versus placebo
Comparison 2. Boswellia serrata (enriched) 100 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 90 days Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐16.57 [‐24.67, ‐8.47]

2 WOMAC‐VAS (Function) Show forest plot

2

85

Mean Difference (IV, Random, 95% CI)

‐8.21 [‐14.21, ‐2.22]

3 Adverse event episodes (n) reported Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figures and Tables -
Comparison 2. Boswellia serrata (enriched) 100 mg versus placebo
Comparison 3. Boswellia serrata (enriched) 250 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 90 days Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Adverse event episodes (n) reported Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figures and Tables -
Comparison 3. Boswellia serrata (enriched) 250 mg versus placebo
Comparison 4. Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

2

97

Mean Difference (IV, Random, 95% CI)

‐16.09 [‐20.37, ‐11.81]

1.1 At 90 days

1

38

Mean Difference (IV, Random, 95% CI)

‐18.10 [‐24.95, ‐11.25]

1.2 At 30 days

1

59

Mean Difference (IV, Random, 95% CI)

‐14.80 [‐20.29, ‐9.31]

2 WOMAC‐VAS (Function) Show forest plot

2

97

Mean Difference (IV, Random, 95% CI)

‐15.01 [‐19.21, ‐10.81]

2.1 At 30 days

1

59

Mean Difference (IV, Random, 95% CI)

‐14.30 [‐20.07, ‐8.53]

2.2 At 90 days

1

38

Mean Difference (IV, Random, 95% CI)

‐15.8 [‐21.92, ‐9.68]

3 Participants (n) reported adverse events Show forest plot

2

97

Odds Ratio (M‐H, Random, 95% CI)

0.98 [0.13, 7.29]

3.1 At 30 days

1

59

Odds Ratio (M‐H, Random, 95% CI)

0.97 [0.06, 16.20]

3.2 At 90 days

1

38

Odds Ratio (M‐H, Random, 95% CI)

1.0 [0.06, 17.25]

Figures and Tables -
Comparison 4. Boswellia serrata (enriched) 100 mg plus non‐volatile oil versus placebo
Comparison 5. Boswellia serrata 999 mg versus valdecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

58

Mean Difference (IV, Random, 95% CI)

‐0.51 [‐7.26, 6.24]

2 WOMAC‐VAS (Function) Show forest plot

1

58

Mean Difference (IV, Random, 95% CI)

2.49 [‐4.07, 9.05]

3 Participants (n) reported adverse events Show forest plot

1

66

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.39, 10.18]

4 Participants (n) withdrew due to adverse events Show forest plot

1

66

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 71.07]

Figures and Tables -
Comparison 5. Boswellia serrata 999 mg versus valdecoxib
Comparison 6. Curcuma domestica versus ibuprofen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain on walking NRS 0‐10 Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2 Function: 100m walk time (seconds) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3 Participants (n) reported adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.46, 1.25]

Figures and Tables -
Comparison 6. Curcuma domestica versus ibuprofen
Comparison 7. Derris scandens versus naproxen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) change from baseline Show forest plot

1

107

Mean Difference (IV, Random, 95% CI)

5.0 [‐1.84, 11.84]

2 WOMAC‐VAS (Function) change from baseline Show forest plot

1

107

Mean Difference (IV, Random, 95% CI)

5.10 [‐0.13, 10.33]

3 Participants (n) reported adverse events. Show forest plot

1

125

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.49, 1.15]

Figures and Tables -
Comparison 7. Derris scandens versus naproxen
Comparison 8. Harpagophytum procumbens versus diacerhein

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline at 120 days Show forest plot

1

92

Mean Difference (IV, Random, 95% CI)

‐5.10 [‐6.52, ‐3.68]

2 Participants (n) reported adverse events Show forest plot

1

92

Risk Ratio (M‐H, Random, 95% CI)

0.4 [0.21, 0.75]

Figures and Tables -
Comparison 8. Harpagophytum procumbens versus diacerhein
Comparison 9. Petiveria alliacea versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (scale unknown) with mvt change from baseline Show forest plot

1

40

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐1.31, 1.11]

2 Participants (n) reported adverse events Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.5 [0.28, 8.04]

Figures and Tables -
Comparison 9. Petiveria alliacea versus placebo
Comparison 10. Pinus pinaster (Pycnogenol® 150 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

37

Mean Difference (IV, Random, 95% CI)

‐142.0 [‐199.55, ‐84.45]

2 WOMAC‐VAS (Function) Show forest plot

1

37

Mean Difference (IV, Random, 95% CI)

‐529.0 [‐741.59, ‐316.41]

3 Participants (n) reported adverse events Show forest plot

2

137

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.08, 1.97]

Figures and Tables -
Comparison 10. Pinus pinaster (Pycnogenol® 150 mg) versus placebo
Comparison 11. Pinus pinaster (Pycnogenol® 100 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC 0‐4 (Pain) Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐7.50 [‐8.43, ‐6.57]

2 WOMAC 0‐4 (Function) Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐29.3 [‐30.99, ‐27.61]

Figures and Tables -
Comparison 11. Pinus pinaster (Pycnogenol® 100 mg) versus placebo
Comparison 12. Ricinus officinale versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants (n) reported adverse events Show forest plot

1

100

Risk Ratio (M‐H, Random, 95% CI)

0.04 [0.00, 0.66]

Figures and Tables -
Comparison 12. Ricinus officinale versus placebo
Comparison 13. Rosa canina versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relief of pain (0 to 4) at 3 months Show forest plot

1

97

Mean Difference (IV, Random, 95% CI)

0.43 [‐0.12, 0.98]

2 WOMAC‐VAS (Pain) Show forest plot

1

94

Mean Difference (IV, Random, 95% CI)

‐2.5 [‐10.20, 5.20]

3 WOMAC‐VAS (Function) Show forest plot

1

94

Mean Difference (IV, Random, 95% CI)

‐1.20 [‐8.98, 6.58]

4 Participants (n) reported adverse events Show forest plot

2

194

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.63, 4.43]

Figures and Tables -
Comparison 13. Rosa canina versus placebo
Comparison 14. Salix purpurea x daphnoides versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 at 14 days Show forest plot

1

68

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 Function VAS 0‐100 at 14 days Show forest plot

1

68

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

3 Participants (n) reported adverse events Show forest plot

1

84

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.57, 1.43]

Figures and Tables -
Comparison 14. Salix purpurea x daphnoides versus placebo
Comparison 15. Salix purpurea x daphnoides versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 At 14 days

1

86

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 42 days

1

86

Mean Difference (IV, Random, 95% CI)

15.0 [5.91, 24.09]

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 At 14 days

1

86

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 At 42 days

1

86

Mean Difference (IV, Random, 95% CI)

12.0 [2.70, 21.30]

3 Participants (n) reported adverse events Show forest plot

1

86

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.43, 0.93]

Figures and Tables -
Comparison 15. Salix purpurea x daphnoides versus diclofenac
Comparison 16. Uncaria guianensis versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (night) Show forest plot

1

45

Mean Difference (IV, Random, 95% CI)

‐11.10 [‐26.44, 4.24]

2 Participants (n) reported adverse events Show forest plot

1

45

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.54, 5.17]

Figures and Tables -
Comparison 16. Uncaria guianensis versus placebo
Comparison 17. Zingiber officinale (Zintona EC) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (movement) Show forest plot

1

24

Mean Difference (IV, Random, 95% CI)

‐9.0 [‐31.12, 13.12]

2 Function (handicap) VAS 0‐100 Show forest plot

1

24

Mean Difference (IV, Random, 95% CI)

‐6.0 [‐27.25, 15.25]

3 Participants (n) reported adverse events Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

3.5 [0.16, 78.19]

Figures and Tables -
Comparison 17. Zingiber officinale (Zintona EC) versus placebo
Comparison 18. Boswellia carteri + Curcuma longa versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Function: pain free walking time (minutes) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.1 At 1 month

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 2 months

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.3 At 3 months

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Figures and Tables -
Comparison 18. Boswellia carteri + Curcuma longa versus placebo
Comparison 19. Persea gratissma + Glycine max (ASU 300 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

4

651

Mean Difference (IV, Random, 95% CI)

‐8.47 [‐15.90, ‐1.04]

1.1 At 3 months

2

326

Mean Difference (IV, Random, 95% CI)

‐11.90 [‐23.95, 0.15]

1.2 At 6 months

1

162

Mean Difference (IV, Random, 95% CI)

‐10.40 [‐17.20, ‐3.60]

1.3 At 12 months

1

163

Mean Difference (IV, Random, 95% CI)

1.0 [‐6.58, 8.58]

2 Pain VAS 0‐100 change from baseline at 36 months Show forest plot

1

345

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐7.39, 6.07]

3 Pain VAS 0‐100 grouped by joint Show forest plot

1

324

Mean Difference (IV, Random, 95% CI)

‐9.06 [‐15.24, ‐2.88]

3.1 VAS (hip OA)

1

162

Mean Difference (IV, Random, 95% CI)

‐13.80 [‐25.22, ‐2.38]

3.2 VAS (knee OA)

1

162

Mean Difference (IV, Random, 95% CI)

‐7.10 [‐14.45, 0.25]

4 Function: disability VAS 0‐100 Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5 WOMAC‐VAS (Function) change from baseline at 36 months Show forest plot

1

345

Mean Difference (IV, Random, 95% CI)

‐1.0 [‐7.14, 5.14]

6 Lequesne algofunctional index Show forest plot

3

480

Mean Difference (IV, Random, 95% CI)

‐1.17 [‐2.54, 0.20]

6.1 At 3 months

2

317

Mean Difference (IV, Random, 95% CI)

‐1.80 [‐2.68, ‐0.92]

6.2 At 12 months

1

163

Mean Difference (IV, Random, 95% CI)

0.10 [‐0.78, 0.98]

7 Function (various tools) Show forest plot

4

642

Std. Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.73, ‐0.11]

8 Participants (n) reported adverse events Show forest plot

5

1050

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.97, 1.12]

9 Participants (n) withdrew due to adverse events Show forest plot

1

398

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.73, 1.80]

10 Particpants (n) reported serious adverse events Show forest plot

1

398

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.94, 1.59]

11 JSW change from baseline Show forest plot

2

453

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.43, 0.19]

11.1 < median group, at 24 months

1

55

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.73, ‐0.13]

11.2 > median group, at 24 months

1

53

Mean Difference (IV, Random, 95% CI)

0.16 [‐0.31, 0.63]

11.3 At 36 months

1

345

Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.22, 0.16]

Figures and Tables -
Comparison 19. Persea gratissma + Glycine max (ASU 300 mg) versus placebo
Comparison 20. Persea gratissma + Glycine max (ASU 600 mg) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐14.2 [‐20.82, ‐7.58]

2 Lequesne algofunctional index Show forest plot

1

156

Mean Difference (IV, Random, 95% CI)

‐1.30 [‐2.38, ‐0.22]

3 Participants (n) reported adverse events Show forest plot

1

174

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.66, 1.74]

Figures and Tables -
Comparison 20. Persea gratissma + Glycine max (ASU 600 mg) versus placebo
Comparison 21. Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 WOMAC‐VAS (Pain) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Participants (n) reported adverse events Show forest plot

1

357

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.59, 1.26]

4 Paricipants (n) reported serious adverse events Show forest plot

1

357

Risk Ratio (M‐H, Random, 95% CI)

2.92 [0.31, 27.78]

Figures and Tables -
Comparison 21. Persea gratissma + Glycine max (ASU 300 mg) versus chondroitin sulphate
Comparison 22. Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Lequesne algofunctional index Show forest plot

1

45

Mean Difference (IV, Random, 95% CI)

‐3.82 [‐7.05, ‐0.59]

1.1 Normal BMI participants

1

18

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐3.37, ‐1.03]

1.2 Overweight BMI participants

1

27

Mean Difference (IV, Random, 95% CI)

‐5.50 [‐6.95, ‐4.05]

Figures and Tables -
Comparison 22. Phellondendron amurense + Citrus sinensis (NP 06‐1) versus placebo
Comparison 23. Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Participants (n) reported adverse events Show forest plot

1

95

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.18, 3.24]

Figures and Tables -
Comparison 23. Uncaria guianensis + Lepidium meyenii versus glucosamine sulphate
Comparison 24. Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain immediately after walking 50 feet VAS 0‐100 Show forest plot

1

247

Mean Difference (IV, Random, 95% CI)

‐9.60 [‐16.81, ‐2.39]

2 WOMAC‐VAS (Function) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figures and Tables -
Comparison 24. Zingiber officinale + Alpinia galanga (EV.EXT77) versus placebo
Comparison 25. SKI306X versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 Low dose (600mg) SKI306X

1

47

Mean Difference (IV, Random, 95% CI)

‐16.1 [‐25.19, ‐7.01]

1.2 Medium dose (1200mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐14.5 [‐23.04, ‐5.96]

1.3 High dose (1800mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐22.3 [‐31.82, ‐12.78]

2 Lequesne algofunctional index change from baseline Show forest plot

1

139

Mean Difference (IV, Random, 95% CI)

‐2.73 [‐3.71, ‐1.74]

2.1 Low dose (600mg) SKI306X

1

47

Mean Difference (IV, Random, 95% CI)

‐2.40 [‐4.05, ‐0.75]

2.2 Medium dose (1200mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐2.8 [‐4.62, ‐0.98]

2.3 High dose (1800mg) SKI306X

1

46

Mean Difference (IV, Random, 95% CI)

‐3.0 [‐4.68, ‐1.32]

3 Participants (n) reported adverse events Show forest plot

2

139

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.49, 1.79]

3.1 Low dose (600mg) SKI306X

1

47

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.32, 2.88]

3.2 Medium dose (1200mg) SKI306X

1

46

Risk Ratio (M‐H, Random, 95% CI)

1.2 [0.43, 3.38]

3.3 High dose (1800mmg) SKI306X

1

46

Risk Ratio (M‐H, Random, 95% CI)

0.6 [0.16, 2.22]

Figures and Tables -
Comparison 25. SKI306X versus placebo
Comparison 26. SKI306X (600 mg) versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

249

Mean Difference (IV, Random, 95% CI)

1.31 [‐2.78, 5.40]

2 Lequesne algofunctional index change from baseline Show forest plot

1

249

Mean Difference (IV, Random, 95% CI)

0.77 [0.10, 1.44]

3 Participants (n) reported adverse events Show forest plot

1

249

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.38, 0.97]

Figures and Tables -
Comparison 26. SKI306X (600 mg) versus diclofenac
Comparison 27. Phytodolor N versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Enduring pain (0 to 3) Show forest plot

1

72

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 Function: mobility limitations (0 to 3) Show forest plot

1

72

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

3 Participants (n) reported adverse events Show forest plot

3

140

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.92]

Figures and Tables -
Comparison 27. Phytodolor N versus placebo
Comparison 28. Reumalex versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 AIMS2 arthritis pain score change from baseline Show forest plot

1

52

Mean Difference (IV, Random, 95% CI)

‐0.89 [‐1.73, ‐0.05]

2 Participants (n) reported adverse events Show forest plot

1

52

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.40, 2.91]

Figures and Tables -
Comparison 28. Reumalex versus placebo
Comparison 29. Chinese DJW versus diclofenac

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (total) Show forest plot

1

200

Mean Difference (IV, Random, 95% CI)

11.81 [‐9.67, 33.29]

2 Lequesne algofunctional index Show forest plot

1

200

Mean Difference (IV, Random, 95% CI)

0.28 [‐0.89, 1.45]

3 Participants (n) reported adverse events Show forest plot

1

200

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.66, 1.63]

Figures and Tables -
Comparison 29. Chinese DJW versus diclofenac
Comparison 30. Chinese BNHS versus Chinese active control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (walking) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

2.0 [‐7.12, 11.12]

2 WOMAC‐VAS (Function) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐7.57, 3.57]

3 Participants (n) reported adverse events Show forest plot

1

60

Risk Ratio (M‐H, Random, 95% CI)

9.00 [0.51, 160.17]

Figures and Tables -
Comparison 30. Chinese BNHS versus Chinese active control
Comparison 31. Chinese BNHS versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 (walking) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐6.81, 2.81]

2 WOMAC‐VAS (Function) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.0 [‐2.53, 2.53]

3 Participants (n) reported adverse events Show forest plot

1

60

Risk Ratio (M‐H, Random, 95% CI)

9.00 [0.51, 160.17]

Figures and Tables -
Comparison 31. Chinese BNHS versus glucosamine sulphate
Comparison 32. Ayurvedic A to E versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adverse event episodes (n) reported Show forest plot

1

454

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.71, 1.28]

1.1 Formula A versus placebo

1

90

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.63, 1.45]

1.2 Formula B versus placebo

1

108

Risk Ratio (M‐H, Random, 95% CI)

1.35 [0.92, 1.98]

1.3 Formula C versus placebo

1

82

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.50, 1.21]

1.4 Formula D versus placebo

1

72

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.34, 0.93]

1.5 Formula E versus placebo

1

102

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.82, 1.80]

Figures and Tables -
Comparison 32. Ayurvedic A to E versus placebo
Comparison 33. Ayurvedic Antarth versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

88

Mean Difference (IV, Random, 95% CI)

‐1.0 [‐9.79, 7.79]

Figures and Tables -
Comparison 33. Ayurvedic Antarth versus placebo
Comparison 34. Ayurvedic RA‐II versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 Show forest plot

1

90

Mean Difference (IV, Random, 95% CI)

‐1.03 [‐1.18, ‐0.88]

2 WOMAC 0‐4 (Function) Show forest plot

1

90

Mean Difference (IV, Random, 95% CI)

‐5.80 [‐6.72, ‐4.88]

3 Participants (n) reported adverse events Show forest plot

1

90

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.69, 1.58]

Figures and Tables -
Comparison 34. Ayurvedic RA‐II versus placebo
Comparison 35. Ayurvedic SGC versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

3.0 [‐3.28, 9.28]

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

2.0 [‐0.72, 4.72]

3 Participants (n) reported adverse events Show forest plot

1

210

Odds Ratio (M‐H, Random, 95% CI)

1.04 [0.58, 1.86]

Figures and Tables -
Comparison 35. Ayurvedic SGC versus glucosamine sulphate
Comparison 36. Ayurvedic SGC versus celecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

‐3.0 [‐8.98, 2.98]

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

1.0 [‐1.60, 3.60]

3 Participants (n) reported adverse events Show forest plot

1

207

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.56, 1.79]

Figures and Tables -
Comparison 36. Ayurvedic SGC versus celecoxib
Comparison 37. Ayurvedic SGCG versus glucosamine sulphate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

4.0 [‐1.42, 9.42]

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

1.38 [‐1.40, 4.16]

3 Participants (n) reported adverse events Show forest plot

1

211

Odds Ratio (M‐H, Random, 95% CI)

0.85 [0.47, 1.54]

Figures and Tables -
Comparison 37. Ayurvedic SGCG versus glucosamine sulphate
Comparison 38. Ayurvedic SGCG versus celecoxib

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

‐2.0 [‐7.42, 3.42]

2 WOMAC 0‐4 (Function) change from baseline Show forest plot

1

220

Mean Difference (IV, Random, 95% CI)

0.19 [‐2.59, 2.97]

3 Participants (n) reported adverse events Show forest plot

1

208

Odds Ratio (M‐H, Random, 95% CI)

0.82 [0.45, 1.48]

Figures and Tables -
Comparison 38. Ayurvedic SGCG versus celecoxib
Comparison 39. Japanese Boiogito + loxoprofen versus loxoprofen

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain: Knee Society Rating System 0‐100 (knee) Show forest plot

1

47

Mean Difference (IV, Random, 95% CI)

‐1.30 [‐8.90, 6.30]

2 Function: Knee Society Rating System 0‐50 (stairs) Show forest plot

1

47

Mean Difference (IV, Random, 95% CI)

3.60 [0.51, 6.69]

3 Participants (n) reported adverse events Show forest plot

1

47

Risk Ratio (M‐H, Random, 95% CI)

2.88 [0.12, 67.29]

Figures and Tables -
Comparison 39. Japanese Boiogito + loxoprofen versus loxoprofen