Scolaris Content Display Scolaris Content Display

Tratamiento a base de hierbas para la artritis reumatoide

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Belch 1988 {published data only}

Belch JJF, Ansell D, Madhok R, O'Dowd A, Sturrock D. Effects of altering dietary essential fatty acids on requirements for non‐steroidal anti‐inflammatory drugs in patients with rheumatoid arthritis: a double blind placebo controlled study. Annals of the Rheumatic Diseases 1988;47:96‐104.

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.

Brzeski 1991 {published data only}

Brzeski M, Madhol R, Capell HA. Evening primrose oil in patients with rheumatoid arthritis and side‐effects of non‐steroidal anti‐inflammatory drugs. British Journal of Rheumatology 1991;30:370‐2.

Chopra 2000 {published data only}

Chopra A, Lavin P, Patwardhan B, Chitre D. Randomized double blind trial of an ayurvedic plant derived formulation for treatment of rheumatoid arthritis. The Journal of Rheumatology 2000;27(6):1365‐72.

Cibere 2003 {published data only}

Cibere J, Deng Z, Lin Y, He Y, Wang Z, Thorne A, et al. A randomized double blind placebo controlled trial of topical Tripterygium wilfordii in rheumatoid arthritis: Reanalysis using logistic regression. The Journal of Rheumatology 2003;30(3):465‐7.

Deal 1991 {published data only}

Deal CL, Schnitzer TJ, Lipstein E, Seibold JR, Stevens RM, Levy MD, et al. Treatment of arthritis with topical capsaicin: A double‐blind trial. Clinical Therapeutics 1991;13(3):383‐95.

Eberl 1988 {unpublished data only}

Eberl R, Dunky A, Leeb B, Wohanka A. Savings of nonsteroidal antirheumatics by Phytodolor: Placebo‐controlled, double blind study over a period of one year per patient. Report for Steigerwald Pharmaceuticals1988.

Goldbach‐Mansky 2009 {published data only}

Goldbach‐Mansky R, Wilson M, Fleischmann R, Olsen N, Silverfield J, Kempf P, et al. Comparison of Tripterygium wilfordii Hook F versus sulfasalazine in the treatment of rheumatoid arthritis. Annals of Internal Medicine 2009;151(4):229‐40.

Jantti 1989 {published data only}

Jantti J, Seppala E, Vapaatalo H, Isomaki H. Evening primrose oil and olive oil in treatment of rheumatoid arthritis. Clinical Rheumatology 1989;8(2):238‐44.

Leventhal 1993 {published data only}

Leventhal LJ, Boyce EG, Zurier RB. Treatment of rheumatoid arthritis with gammalinolenic acid. Annals of Internal Medicine 1993;119(9):867‐73.

Leventhal 1994 {published data only}

Leventhal LJ, Boyce EG, Zurier RB. Treatment of rheumatoid arthritis with blackcurrant seed oil. British Journal of Rheumatology 1994;33:847‐52.

Li 2007 {published data only}

Li Ek, Tam L, Wong CK, Li WC, Lam CWK, Wachtel Galor S, et al. Safety and efficacy of Ganoderma lucidum (Lingzhi) and san miao san supplementation in patients with rheumatoid arthritis: A double‐blind, randomized, placebo‐controlled pilot trial. Arthritis and Rheumatism 2007;57(7):1143‐50.

McCarthy 1992 {published data only}

McCarthy GM, McCarthy DJ. Effect of topical capsaicin in the therapy of painful osteoarthritis of the hands. The Journal of Rheumatology 1992;19(4):604‐7.

Meier 1987 {unpublished data only}

Meier G. Phytodolor N versus placebo in rheumatoid arthritis: Pilot study of the rheumatism clinic. Report for Steigerwald Pharmaceuticals1987.

Mur 2002 {published data only}

Mur E, Hartig F, Eibl G, Schirmer M. Randomised double blind trial of an extract from the pentacyclid alkaloid‐chemotype of Uncaria tomentosa for the treatment of rheumatoid arthritis. The Journal of Rheumatology 2002;29(4):678‐81.

Pattrick 1989 {published data only}

Pattrick M, Heptinstall S, Doherty M. Feverfew in rheumatoid arthritis: a double blind, placebo controlled study. Annals of the Rheumatic Diseases 1989;48:547‐9.

Sandler 1998 {published data only}

Sandler O, Herborn G, Rau R. Is H15 (Extract of Boswelliaserrata, “incense”) an efficient supplementation to established drug therapy in RA? – Results of a double blinded pilot trial [Ist H15 (Harzextrakt von Boswellia serrata,¹Weihrauchª) eine sinnvolle ErgaÈnzungzur etablierten medikamentoÈsen Therapieder chronischen Polyarthritis? Ergebnisse einer doppelblinden Pilotstudie]. Zeitschrift für Rheumatologie 1998;57:11‐6.

Song 2007 {published data only}

Song YW, Lee EY, Koh EM, Cha HS, Yoo B, Lee CK, et al. Assessment of comparative pain relief and tolerability of SKI306X compared with celecoxib in patients with rheumatoid arthritis: a 6‐week, multicenter, randomized, double‐blind, double‐dummy, phase III, noninferiority clinical trial. Clinical Therapeutics 2007;29:862‐73.

Tao 1989 {published data only}

Tao XL, Sun Y, Dong Y, Xiao YL, Hu DW, Shi YP, et al. A prospective, controlled, double‐blind, cross‐over study of Tripterygium wilfordii hook F in treatment of rheumatoid arthritis. Chinese Medical Journal 1989;102(5):327‐2.

Tao 2002 {published data only}

Tao X, Younger J, Fan FZ, Wang B, Lipskey PE. Benefit of an extract of Tripterygium wilfordii Hook F in patients with rheumatoid arthritis. Arthritis and Rheumatism 2002;46(7):1735‐43.

Watson 1993 {published data only}

Watson J, Byars ML, McGill P, Kelman AW. Cytokine and prostaglandin production by monocytes of volunteers and rheumatoid arthritis patients treated with dietary supplements of blackcurrant seed oil. British Journal of Rheumatology 1993;32:1055‐8.

Zurier 1996 {published data only}

Zurier RB, Rossetti RG, Jacobson EW, DeMarco DM, Liu NY, Temming JE, et al. Gamma‐linolenic acid treatment of rheumatoid arthritis. Arthritis and Rheumatism 1996;39(11):1808‐17.

Referencias de los estudios excluidos de esta revisión

Anonymous 1993 {published data only}

Anonymous. The garlic diet: Purification by garlic [Die knoblauchkur: entschlackung durch knoblauch]. Natur und Heilen 1993;70(10):520‐1.

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 Healthcare. Exeter (UK), 1997; Vol. 2, issue 4:186‐7.

Borigini 1996 {published data only}

Borigini MJ, Egger MJ, Williams HJ, Paulus HE, Ward JR. TJ‐114 (Sairei‐To), an herbal medicine in rheumatoid arthritis. A preliminary "go ‐ no go" clinical trial. Journal of Clinical Rheumatology 1996;2(6):309‐16.

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.

Darlington 1987 {published data only}

Darlington LG, Ramsey NW. Olive oil for rheumatoid patients?. British Journal of Rheumatology 1987;26 Suppl:215.

DeLuca 1995 {published data only}

DeLuca P, Rothman D, Zurier RB. Marine and botanical lipids as immunomodulatory and therapeutic agents in the treatment of rheumatoid arthritis. Rheumatic Disease Clinics of North America 1995;21(3):759‐77.

Dharmananda 1985 {published data only}

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

Falch 1997 {published data only}

Falch VB. Ginger ‐ not only one Gewurz investigation of effect and mechanisms. [Ingwer ‐ nicht nur ein Gewurz Untersuchungen zu Wirkungen und Wirksamkeit]. Deutsche Apotheker Zeitung 1997;137(47):47‐52, 55‐60.

Gendo 1997 {published data only}

Gendo U. Traditional Chinese Medicine (TCM) appears to improve chronic polyarthritis. [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.

Guo 1986 {published data only}

Guo J‐I, Gao Z‐G, Zang A‐C, Bai R‐X. Radix Tripterygium wilfordii hook F in rheumatoid arthritis, ankylosing spondylitis and juvenile rheumatoid arthritis. Chinese Medical Journal 1986;99(4):317‐20.

Hansen 1983 {published data only}

Hansen TM, Lerche A, Kassis V, Lorenzen I, Sondergaard J. Treatment of rheumatoid arthritis with prostaglandin E1 precursors CIS‐linolenic acid and y linolenic acid. Scandinavian Journal of Rheumatology 1983;12:85‐8.

Hanyu 1989 {published data only}

Hanyu B. The short‐term effect of childhood rheumatoid arthritis treated with combined Western and Chinese herbal drugs. Journal of Traditional Chinese Medicine 1989;9(2):108‐10.

Huber 1991 {published data only}

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

Jacobs 1991 {published data only}

Jacobs JWG, Rasker JJ, Van riel PLCM, Gribnau FWJ, Van de Putte LBA. Rheumajecta and vasolastine in the treatment of rheumatoid arthritis ‐ the results of a placebo‐controlled, double‐blind trial of a complementary treatment. Scandinavian Journal of Rheumatology 1991;20:434‐40.

Jantti 1989b {published data only}

Jantti J, Nikkari T, Solakivi T, Vapaatalo H, Isomake H. Evening primrose oil in rheumatoid arthritis: changes in serum lipids and fatty acids. Annals of the Rheumatic Diseases 1989;48:124‐7.

Kou 1997 {published data only}

Kou Q, Fang D. Therapeutic effects of the Instant rheumatoid medicinal granules for treatment of acute rheumatoid arthritis ‐ a report of 34 cases. Journal of Traditional Chinese Medicine 1997;17(3):163‐7.

Larsen 1989 {published data only}

Larsen A, Petersson I, Svensson B. Podophyllum derivatives (CPH 82) compared with placebo in the treatment of rheumatoid arthritis. British Journal of Rheumatology 1989;28:124‐7.

Linsheng 1997 {published data only}

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

Lipsky 1997 {published data only}

Lipsky PE, Tao X‐L. A potential new treatment for rheumatoid arthritis: Thunder god vine. Seminars in Arthritis and Rheumatism 1997;26(5):713‐23.

Loew 1996 {published data only}

Loew D, Schuster O, Mollerfeld J. Stability and biopharmaceutical quality: Evidence of biological mechanism and effect 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.

Matsuta 1992 {published data only}

Matsuta K, Kikuna T. Physical therapy, Japanese oriental medicine therapy. Nippon Rinslo 1992;50(3):563‐9.

Mills 1996 {published data only}

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

Ohkaya 1988 {published data only}

Ohkaya M, Shinoriya K, Fujii I, Tomita Y. Effects of Sarei‐to on rheumatoid arthritis (part 1). Journal of Medical Pharmaceutical Society Wakan‐Yaku 1988;5:480‐1.

Ohkaya 1989 {published data only}

Ohkaya M, Fujii I, Tomita Y, Yomotani H, Oike T. Effects of Sarei‐to on rheumatoid arthritis (part 2). Journal of Medical Pharmaceutical Society Wakan‐Yaku 1989;6:264‐5.

Ramakrishanamacharya {published data only}

Ramakrishanamacharya CH, Krishnaswamy MR, Bhima Rao R, Viswanathan S. Anti‐inflammatory efficacy of melothria madraspatana in active rheumatoid arthritis. Clinical Rheumatology 1996;15(2):214‐21.

Ramm 1996 {published data only}

Ramm S, Hansen C. Nettle extract for osteoarthritis and rheumatoid arthritis. [Brennesselblatter‐extrakt bei arthrose und rheumatoider arthritis. Multzentrische anwendungsbeobacktung mit Rheuma‐hek]. Therapiewoche 1996;28:1575‐8.

Reuss 1981 {published data only}

Reuss D. Echinacea as therapy for primary chronic polyarthritis. [Echinacin inder therapite der primar‐chronischen polyarthritis]. ZFA ‐ Stuttgart 1981;57(11):865.

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.

Srivastava 1989 {published data only}

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

Tao 1987 {published data only}

Tao X‐I, Dong Y, Zhang N‐Z, et al. A double‐blind study of T2 in the treatment of rheumatoid arthritis. Chinese Journal of Internal Medicine 1987;26(7):399‐445.

Vyas 1987 {published data only}

Vyas SN, Shukla CP. A clinical study on the effects of Shuddha Guggulu in rheumatoid arthritis. Rheumatism 1987;23(1):15‐26.

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.

Xu 1996 {published data only}

Xu D‐S, Shen Z‐Y, Lu W, et al. Clinical and experimental study of RA mixture in treatment of rheumatoid arthritis. Chung‐Kuo chung Hsi I Chieh Ho Tsa Chih 1996;16(1):14‐7, 23.

Yan 1985 {published data only}

Yan BY. Effects of Tripterygium wilfordii on rheumatoid arthritis ‐ analysis of 165 cases. Chung Hsi I Chieh Ho Tsa Chih 1985;5(5):280‐3.

Zell 1993 {published data only}

Zell J, Ecker R, Batz H, Vestweber AM. Mistletoe for knee and hip arthritis: Segment or immunotherapy? A pilot study. [Misteltherapie bei gon‐und coxarthrose: Segment ‐ oder immuntherapie? Eine pilotstudie]. Heilkunst 1993;106(9):23‐6.

Shamsi 2009 {unpublished data only}

Shamsi Y. A placebo controlled double blind randomized trial of Qurse Mufasil in the treatment of rheumatoid arthritis. http://apps.who.int/trialsearch/Trial.aspx?TrialID=CTRI/2009/091/000746. [CTRI/2009/091/000746]

ACR 2002

American College of Rheumatology Subcommittee on Rheumatoid Arthritis Guidelines. Guidelines for the management of rheumatoid arthritis: 2002 update. Arthritis and Rheumatism 2002;46(2):328‐46.

Arnett 1988

Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis and Rheumatism 1988;31:315‐24.

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.

Boisset 1994

Boisset M, Fitzcharles M. Alternative medicine use by rheumatology patients in a universal health care setting. The Journal of Rheumatology 1994;21(1):148‐52.

Buchbinder 2002

Buchbinder R, Gingold M, Hall S, Cohen M. Non‐prescription complementary treatments used by rheumatoid arthritis patients attending a community‐based rheumatology practice. Internal Medicine Journal 2002;23(5‐6):208‐14.

Buck 1986

Buck SH, Burks TF. The neuropharmacology of capsaicin: Review of some recent observations. Pharmacological Review 1986;38:179‐226.

Canter 2006

Canter PH, Lee HS, Ernst E. A systematic review of randomised clinical trials of Tripterygium wilfordii for rheumatoid arthritis. Phytomedicine 2006;13:371‐7.

Cates 2004

Cates C. Visual Rx 2.0 NNT Calculator. Dr Chris Cates EBM Website www.nntonline.net, 2004.

Chrubasik 2005

Chrubasik S, Pittler M, Roufogalis B. Zingiberis rhizoma: A comprehensive review on the ginger effect and efficacy profiles. Phytomedicine 2005;12:684‐701.

Chrubasik 2007a

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;21:675‐683.

Chrubasik 2007b

Chrubasik JE, Roufogalis BD, Wagner H, Chrubasik S. A comprehensive review on nettle effect and efficacy profiles. Part I: Herba urticae. Phytomedicine 2007;14:423‐435.

Cronan 1989

Cronan TA, Kaplan RM, Posner L, Blumberg E, Franklin K. Prevalence of the use of unconventional remedies for arthritis in a metropolitan community. Arthritis and Rheumatism 1989;32(12):1604‐7.

Dedov 2000

Dedov VN, Roufogalis BD. Mitochondrial calcium accumulation following activation of vanilloid (VR1) receptors by capsaicin in dorsal root ganglion neurons. Neuroscience 2000;95:183‐8.

EFPIA 1996

European Federation of Pharmaceutical Industries and Associations [EFPIA]. Good manufacturing practices for active ingredient manufacturers. EFPIA, 1996.

ESCOP 2009

European Scientific Cooperative on Phytotherapy (ESCOP) (Eds). ESCOP Monographs Supplement 2009. 2nd Edition.. ESCOP Monographs Supplement 2009. 2nd Edition. New York: Thieme Publisher Stuttgart, 2009.

Flynn 1986

Flynn DL, Rafferty MF, Boctor AM. Inhibition of human neutrophil 5‐lipoxygenase activity by gingerdione, shogaol, capsaicin and related pungent compounds. Prostaglandins, Leukotrienes, and Medicine 1986;24:195‐8.

Furlan 2009

Furlan AD, Pennick V, Bombardier C, van Tulder M, editorial board, Cochrane Back Review Group. 2009 updated method guidelines for systematic reviews in the Cochrane Back Review Group. Spine 2009;34(18):1929‐41.

Gagnier 2006a

Gagnier JJ, Rochon PA, Boon H, Barnes J, Moher D, Bombardier C for the CONSORT Group. Improving the reporting of randomized controlled trials of herbal medicine interventions: Explanation and elaboration. Journal of Clinical Epidemiology 2006;59:1134‐49.

Gagnier 2006b

Gagnier JJ, Rochon PA, Boon H, Barnes J, Moher D, Bombardier C for the CONSORT Group. Improving the reporting of randomized controlled trials of herbal medicine interventions: An application of the CONSORT statement. Annals of Internal Medicine 2006;144:364‐7.

Gundermann 2001

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

Higgins 2008

Higgins JPT, Green S (editors). The Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.0 [updated February 2008]. The Cochrane Collaboration, 2008. Available from www.cochrane‐handbook.org, 2008.

ICH 2004

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

Jurgensen 2005

Jurgensen S, Dalbo S, Angers P, Santos AR, Ribeiro‐do‐Valle RM. Involvement of 5‐HT2 receptors in the antinociceptive effect of Uncaria tomentosa. Pharmacology, Biochemistry, and Behavior 2005;81:466‐77.

Katchamart 2010

Katchamart W, Trudeau J, Phumethum V, Bombardier C. Methotrexate monotherapy versus methotrexate combination therapy with non‐biologic disease modifying anti‐rheumatic drugs for rheumatoid arthritis. Cochrane Database of Systematic Reviews 2010, Issue 4. [Art. No.: CD008495. DOI: 10.1002/14651858.CD008495]

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.

MacPherson 1994

MacPherson H, Blackwell R. Rheumatoid arthritis and Chinese medicine. The European Journal of Oriental Medicine 1994;1(3):17‐29.

Mittra 2000

Mittra S, Datta A, Singh SK, Singh A. 5‐Hydroxytryptamine‐inhibiting property of feverfew: Role of parthenolide content. Acta Pharmacologica Sinica 2000;21:1106‐14.

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.

NLM 2009

Evening Primrose Oil. www.nlm.nih.gov/medlineplus/druginfo/natural/patient‐primrose.html Accessed Nov 5th, 2009.

Nolano 1999

Nolano M, Simone DA, Wendelschafer‐Crabb G, Johnson T, Hazen E, Kennedy WR. Topical capsaicin in humans: parallel loss of epidermal nerve fibers and pain sensation. Pain 1999;81:135‐45.

Schmid 2001

Schmid B, Kotter I, Heide L. Pharmacokinetics of salicin after oral administration of a standardized willow bark extract. European Journal of Clinical Pharmacology 2001;57:387‐91.

Schünemann 2008a

Schünemann 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 5.5.0. The Cochrane Collaboration, updated February 2008.

Schünemann 2008b

Schünemann 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 5.0.0. The Cochrane Collaboration, updated February 2008.

Singh 2009

Singh JA, Christensen R, Wells GA, Suarez‐Almazor ME, Buchbinder R, Lopez‐Olivo MA, Tanjong Ghogomu E, Tugwell P. Biologics for rheumatoid arthritis: an overview of Cochrane reviews. Cochrane Database of Systematic Reviews 2009, Issue 4. [Art. No.: CD007848. DOI: 10.1002/14651858.CD007848.pub2]

Tao 1995

Tao X, Cai JJ, Lipsky PE. The identity of immunosuppressive components of the ethyl acetate extract and chloroform methanol extract (T2) of Tripterygium wilfordii Hook. F. Journal of Pharmacoogy Experimental Therapy 1995;272:1305‐1312.

Tarner 2007

Tarner IH, Müller‐Ladner U, Gay S. Emerging targets of biologic therapies for rheumatoid arthritis. Nature Clinical Practice Rheumatology 2007;3(6):336‐45.

Tarner 2008

Tarner IH, Müller‐Ladner U. Drug delivery systems for the treatment of rheumatoid arthritis. Expert Opinion on Drug Delivery 2008;5(9):1027‐37.

Vlachojannis 2009

Vlachojannis JE, Cameron M, Chrubasik S. A systematic review on the effectiveness of willow bark for musculoskeletal pain. Phytotherapy Research 2009;23:897‐900.

WHO 1996

WHO Expert Committee on Specifications for Pharmaceutical Preparations. HO Technical Report Series, No. 863, Annex 11 (Guidelines for the Assessment of Herbal Medicines) ‐ Thirty‐fourth Report1996.

Referencias de otras versiones publicadas de esta revisión

Cameron 2009

Cameron M, Gagnier J, Litle CV, Parsons T, Blumle A, Chrubasik S. Evidence of Effectiveness of Herbal MedicinalProducts in the Treatment of ArthritisPart 2: Rheumatoid Arthritis. Phytotherapy Research 2009;23(12):1647‐62.

Little 2000

Little CV, Parsons T. Herbal therapy for treating rheumatoid arthritis. Cochrane Database of Systematic Reviews 2000, Issue 4. [Art. No.: CD002948. DOI: 10.1002/14651858.CD002948]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Belch 1988

Methods

Randomised, double blind, placebo control, 3 parallel groups. Duration 12 months.

Participants

Randomised n=49, completed n=34. Age range 28‐74 yr. Inclusion: classical or definite RA (ARA criteria), requiring NSAIDs but not DMARDs.

Interventions

Tradename not provided, Oenothera biennis (evening primrose), oil, 6000mg (12x500mg, approx 9% GLA, equivalent to 540mg GLA), capsules, oral. Concurrent intervention: usual NSAIDs for first 3 months only.

Outcomes

Morning stiffness (minutes), grip strength mmHg, Ritchie index, pain VAS 0‐100, patient global.

Notes

Results favour intervention for reduction in pain and NSAID use. No evidence of disease‐modifying effects.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analyses.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Biegert 2004

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 weeks.

Participants

Randomised n=26 (intervention n=13, control n=13), completed n=26 (intervention n=13, control n=13). Age (yr): intervention m=56.5 sd=8.9, control m=60.1 sd=11. Inclusion: ACR criteria RA stage I‐III.

Interventions

Assalix*, Salix daphnoides cortex (willow bark), ethanolic extract, 1572.96mg (2x2x393.24mg, equivalent to 240mg salicin), tablets, oral.

Outcomes

Pain VAS 0‐100, tender joint count, HAQ‐DI, stiffness VAS 0‐100, efficacy VAS 0‐100, SF‐36, ESR, CRP, ACR20.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

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

Allocation concealment?

Low risk

Adequate.

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

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

Free of selective reporting?

Low risk

Confirmatory study, statistical power reported.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Brzeski 1991

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 months.

Participants

Randomised n=40 (intervention n=19, control n=21), completed n=30 (intervention n=13, control n=17). Age range 16‐75 yr. Inclusion: classical or definite RA, all with probable gastro‐intestinal lesions due to NSAIDs.

Interventions

Tradename not provided, Oenothera biennis (evening primrose), oil, 6000mg (12x500mg, approx 9% GLA, equivalent to 540mg GLA), capsules, oral.

Outcomes

Pain VAS 0‐100, well‐being score, morning stiffness (minutes), Ritchie index, HAQ, intake of NSAIDs and analgesics.

Notes

Results favour intervention for morning stiffness, equivocal for all other outcomes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Unclear risk

Reported withdrawals.

Free of other bias?

Unclear risk

Placebo capsules contained olive oil and may not be inert. Reported ethics committee approval.

Chopra 2000

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 16 weeks.

Participants

Randomised n=182 (intervention n=89, control n=93), completed n=165 (intervention n=80, control n=85). Age (yr): intervention m=45, control m=45. Inclusion: ACR criteria RA stage I‐III.

Interventions

RA‐1, Ayurvedic formula, mixture of Withania somnifera, Boswellia serrata, Zingiberis officinale, Ciruma longa, 444mg, (3x2), tablets, oral.

Outcomes

20% or 50% reduction in individual core set variables, patient global assessment, physician global assessment, ARC20.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

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

Allocation concealment?

Low risk

Adequate

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

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

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria. Reported ethics committee approval.

Cibere 2003

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 weeks.

Participants

Randomised n=61 (intervention n=31, control n=30). Dropouts not reported. Age (yr): intervention m=42, control m=39. Inclusion: ACR criteria RA (any stage).

Interventions

Tradename not provided, Tripterygium wilfordii (thunder god vine), tincture, 5‐6 applications/day, topical.

Outcomes

Modified ACR20, 42 tender joint count, 40 swollen joint count, grip strength kPa, morning stiffness (hours), HAQ‐DI, ESR, CRP, patient global, physician global.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

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

Allocation concealment?

Low risk

Adequate

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Unclear risk

Withdrawals not reported. Included intention‐to‐treat analyses.

Free of selective reporting?

Unclear risk

Adverse events not reported. Confirmatory study.

Free of other bias?

Unclear risk

Diagnosis / assessment consistent with ACR criteria. Reanalysis of previous study.

Deal 1991

Methods

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

Participants

Randomised n=31, completed n=29. Age range 20‐79 yr. Inclusion: primary RA one/both knees, moderate to very severe knee pain (scale of 0‐4), at least 3 ACR criteria for classic, definite, or probable RA.

Interventions

Zostrix, capsaicin 0.025% w v cream, topical, QID.

Outcomes

Pain VAS 0‐100, pain 0‐4, physician global ‐1‐3.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging, but placebo validity and blinding may be compromised by burning side effect of topical intervention.

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Unclear 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. Reported adverse events.

Free of other bias?

Unclear risk

Diagnosis / assessment criteria for OA not specified.

Eberl 1988

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 12 months.

Participants

Randomised n=37 (intervention n=20, control n=17), completed n=24 (intervention n=15, control n=9). Age (yr): intervention m=61 sd=12, control m=59 sd=10. M:F=1:36. Inclusion: ACR criteria RA stage II or III.

Interventions

Phytodolor® N, mixture of ash bark, aspen leaf, aspen bark, golden rod herb, tincture, 3x30 drops, oral. Concurrent intervention: diclofenac 25mg/d, oral.

Outcomes

Joint stiffness, grip strength mmHg, Ritchie index.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Unclear risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events. Full data reported.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Goldbach‐Mansky 2009

Methods

Double‐blind, randomised, controlled study. Duration 24 weeks.

Participants

Randomized n=121 (Tripterygium wilfordii n=60; Sulfasalazine n=61), completed n=62 (Tripterygium wilfordii n=37, Sufasalazine n=25). Age (yr): Tripterygium wilfordii m=54 sd=11, Sufasalazine m=51 sd=12. M:F = 1:1.2. Inclusion: ACR criteria RA, > 6 months.

Interventions

Tripterygium wilfordii HF (TwHF) extract, 180 mg/day. Sufasalazine 2g/day.

Outcomes

Primary end point: 20% improvement at 24 weeks, as defined by ACR criteria (ACR 20). Secondary end points: efficacy of TwHF in achieving ACR 50 and ACR 70 responses at 24 weeks, the improvement in the European League Against Rheumatism Disease Activity Score 28 (DAS 28) measure, and a change in the Sharp–van der Heijde score of the hand and foot radiographs

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

Computer‐generated, pseudo‐random code (with random, permuted blocks)

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double blind. Patients are likely blinded, though this is not stated. "A rheumatologist or trained staff member masked to treatment allocation assessed the patients."

Incomplete outcome data addressed?
All outcomes

Unclear risk

There were a large number of drop‐outs, all are accounted for with reasons and they state that they used: "A protocol‐specified, last‐observation‐carried‐forward approach for handling missing data..." It does appear that they did an ITT analysis for several of the outcomes including the primary outcome and report them in the text. All tables appear to be the per‐protocol analyses.

Free of selective reporting?

Low risk

Free of other bias?

Unclear risk

Baseline differences: Less women in the TwHF group (73% vs 87%); CRP appeared to be slightly higher in the TwHF group (255.2 nmol/L vs 236.2 nmol/L); Slightly higher radiographic score in the TwHF group (40.0 vs 34).There is no discussion of differences in medications other than the interventions taking throughout the study. These differences were not tested for significance.

Jantti 1989

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 13 weeks; 1 week washout, 12 weeks intervention.

Participants

Randomised n=20 (intervention n=10, control n=10), completed n=18 (intervention n=9, control n=9). Age: intervention m=50, control m=38. M:F=2:18. Inclusion: definite or classical RA, prepared to abstain from NSAIDs for 13 weeks.

Interventions

Tradename not provided, Oenothera biennis (evening primrose), oil, 20 mls (2x10ml, approx 9% GLA, equivalent to 1800mg of GLA), oral.

Outcomes

Pain VAS 0‐100, joint score (swollen and tender joint counts), duration of morning stiffness, grip strength.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of other bias?

Unclear risk

Placebo capsules contained olive oil and may not be inert. Diagnosis / assessment criteria for OA not specified.

Leventhal 1993

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 24 weeks.

Participants

Randomised n=37 (intervention n=19, control n=18), completed n=27 (intervention n=14, control n=13). Age: intervention m=58, control m=50. Inclusion: 18‐80 yrs, ACR criteria RA stage I‐III, using NSAIDs, not using DMARDs.

Interventions

Boracelle, Borago officinalis (borage seed), oil, 7.2ml (3x4x0.6ml, approx 23% GLA, equivalent to 1400mg GLA), capsules, oral.

Outcomes

Pain VAS 0‐100, pain 0‐4, physician global 0‐4, patient global 0‐4, 68 tender joint count, 66 swollen joint count, joint tenderness score 0‐3, joint swelling score 0‐3, duration of morning stiffness, vocational activity score 0‐3, grip strength mmHg.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

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

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Leventhal 1994

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 24 weeks.

Participants

Randomised n=34 (intervention n=14, control n=20), completed n=14 (intervention n=7, control n=7). Age m=55. Inclusion: 18‐80 yr, ACR criteria RA stage I‐III, using NSAIDs, DMARDs stable for past 3 months.

Interventions

Tradename not provided, Ribes nigrum (blackcurrant seed), oil, 10500mg (15x700mg, approx 19% GLA, equivalent to 2000mg GLA), capsules, oral.

Outcomes

Pain VAS 0‐100, pain 0‐4, physician global 0‐4 and VAS 0‐100, patient global 0‐4 and VAS 0‐100, 68 tender joint count, 66 swollen joint count, joint tenderness score 0‐3, joint swelling score 0‐3, morning stiffness (minutes), vocational activity score 0‐3, grip strength mmHg.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

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

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analyses.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Li 2007

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 24 weeks.

Participants

Randomised n=65 (intervention n=32, control n=33), completed n=58 (intervention n=28, control n=30). Age: intervention m=50, control m=50. M:F=1:1. Inclusion: ACR criteria.

Interventions

Ganoderma lucidum 4g per day together with San Miao San (a combination of Rhizoma atractylodis, Cotex phellodendri, and Radix achyranthes Bidentatae) 2.4 grams per day.

Outcomes

Primary outcome: ACR 20% response; Secondary outcomes: changes in ACR components including tender and swollen joint count, physician’s and patient’s global assessment, HAQ score, and ESR or CRP level, total antioxidant power of plasma, plasma ascorbic acid concentration.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

Computer generated list in blocks of 5

Allocation concealment?

Low risk

The list was generated at the Institute of Chinese Medicine, The Chinese University of Hong Kong. Study medications were dispensed as sealed packages in consecutive numbers. A research nurse was responsible for dispensing study medications. The investigators, research nurse, and participants were not aware of the treatment assignments throughout the study.
Treatment codes were only broken after completion of the study.

Blinding?
All outcomes

Low risk

The investigators, research nurse, and participants were not aware of the treatment assignments throughout the study. Treatment codes were only broken after completion of the
study.

Incomplete outcome data addressed?
All outcomes

High risk

Three participants dropped out of the placebo group (2 due to inefficacy and 1 due to emigration); Four participants dropped out of the treatment group (all due to inefficacy; three at week 8 and one at week 12)

Free of selective reporting?

Low risk

All outcomes were reported

Free of other bias?

High risk

There are a selection of herbal medicines given in the active group. Also, participants in the active group had slightly longer standing RA (9.3 years VS 7.8 years) and a larger number of participants in the active group were taking sulphasalazine (8 VS 4). None of these differences were tested for statistical differences.

McCarthy 1992

Methods

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

Participants

Randomised n=7, completed n=5. Age: m=52, sd=4. Inclusion: ACR criteria RA.

Interventions

Tradename not provided, capsaicin frutescens 0.075% wv cream, topical, QID.

Outcomes

Pain VAS 0‐100, morning stiffness (Landsbury 2 question method), HAQ, grip strength mmHg, swelling (PIP, DIP circumference), tenderness (delorimeter).

Notes

B:1, W:1. Placebo validity and blinding may be compromised by burning side effect of topical intervention. Small sample size, underpowered study. Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method of randomisation incompletely reported. Described as randomised according to a previously established randomisation schedule.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind. Active intervention and placebo not distinguished by look, taste, smell or packaging, but placebo validity and blinding possibly compromised by burning side effect of topical intervention.

Incomplete outcome data addressed?
All outcomes

Unclear risk

Reported no withdrawals. Included per protocol analyses.

Free of selective reporting?

Unclear risk

Variances reported as standard error of measurement (SEM). Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Meier 1987

Methods

Randomised, double blind, placebo control, non‐intervention control, 3 parallel groups. Duration 2 weeks.

Participants

Randomised n=15 (intervention n=5, placebo n=5, non‐intervention n=5), completed n=15 (intervention n=5, placebo n=5, non‐intervention control n=5). Age range 23‐76 yr; intervention m=62 sd=13, control m=63 sd=16. M:F=9:6. Inclusion: ACR crtieria RA stage II or III.

Interventions

PhytodolorRN, mixture of ash bark, aspen leaf, aspen bark, golden rod herb, tincture, 3x30 drops, oral.

Outcomes

Diclofenac use, pain 0‐3, joint swelling 0‐3.

Notes

Results equivocal. Groups dissimilar at baseline. Change (reduction) in diclofenac use and pain was greatest in intervention group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method of randomisation incompletely reported. Described as randomised according to a previously established randomisation schedule.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported. In other studies of Phytodolor® N, active intervention and placebo not distinguished by look, taste, smell or packaging. Non‐intervention control group not blinded.

Incomplete outcome data addressed?
All outcomes

Unclear risk

Reported no withdrawals.

Free of selective reporting?

Low risk

Full data reported.

Free of other bias?

Unclear risk

Diagnosis / assessment consistent with ACR criteria. Groups dissimilar at baseline.

Mur 2002

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 52 weeks; 24 weeks RCT, 28 weeks open trial.

Participants

Randomised n=40 (intervention n=21, control n=19), completed n=38 (intervention n=20, control n=18). Age: intervention m=53.1 sd=13.4, control m=54.9 sd=13.5. M:F intervention=20:1, control=15:4. Inclusion: ACR criteria RA stage II or III, DMARDs (sulfasalazine or hydrochloroquine) for 6 months, dose stable for past 6 weeks.

Interventions

Krallendorn, Uncaria tomentosa (cat's claw), aqueous dry extract of pentacylcic alkaloid chemotype, 60mg (3x20mg), capsules, oral.

Outcomes

66 swollen joint count, 68 tender joint count, Ritchie index, pain VAS 0‐100, disease activity VAS 0‐100, morning stiffness 0‐5, HAQ (baseline and week 24 only).

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Pattrick 1989

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 weeks.

Participants

Randomised n=41 (intervention n=20, control n=21), completed n=40 (intervention n=20, control n=20). Age range 28‐65 yr. Inclusion: female, aged under 65 yr, classical or definite RA, poor symptomatic control.

Interventions

Tanacetum parthenium (feverfew), (70‐86mg), oral.

Outcomes

Morning stiffness (minutes), inactivity stiffness, pain VAS 0‐10, grip strength mmHg, Ritchie index, patient global, physician global.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria.

Sandler 1998

Methods

Randomised, double blind, placebo control, multi‐centre trial, 2 parallel groups. Duration 12 weeks.

Participants

Randomised n=78 (all centres). Data from one centre (Ratingen) available for analysis; randomised n=37 (intervention n=18, control n=19), completed n=36 (intervention n=17, control n=19). Inclusion: Active RA, at least one painful join, stable corticosteroids.

Interventions

Boswellia serrata, 1200‐3600mg, (3x400mg to 3x3x400mg), tablets, oral.

Outcomes

Ritchie index, pain VAS 0‐10, NSAID consumption, patient global VAS 0‐10.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events. Non‐normal data reported as median and range.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria. Reported ethics committee approval.

Song 2007

Methods

Randomised, double blind, active control (celecoxib), 2 parallel group, multicentre trial. Duration 6 weeks.

Participants

Randomised n=183 (intervention n=91, control n=92), completed n=168 (intervention n=84, control n=84). Age (yr): intervention m=52.1 sd=12.6, control m=51.7 sd=10.9. M:F=1:8. Inclusion: ACR criteria RA stage I, II or III, disease duration >3 months, stable medications, pain (VAS 0‐100) increase of 10+mm, and 6+ tender joints, and 3+ swollen joints after NSAID washout.

Interventions

SKI306X, extract mixture of Clematis mandshurica, Prunella vulgaris, Trichosanthes kirilowii, 600mg (3x200mg), tablets, oral.

Outcomes

Pain (VAS 0‐100), rescue medication use (acetaminophen), ACR20.

Notes

Results indicate that intervention is not inferior to active control.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Low risk

Randomised to one of two groups using a computer generated random number sequence. Baseline parameters compared for significant differences.

Allocation concealment?

Low risk

Adequate

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

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

Free of selective reporting?

Low risk

Reported adverse events. Confirmatory study.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria. Reported ethics committee approval.

Tao 1989

Methods

Randomised, double blind, placebo control, 2 groups crossover study. Duration 16 weeks: 12 weeks intervention 1st arm, 4 weeks intervention 2nd arm.

Participants

Randomised n=70, completed first arm n=58. Age: m=47 yr. Inclusion: classic or definite RA of at least 6 months duration with poor response to NSAIDs for at least 2 months.

Interventions

Tripterygium wilfordii hook F (thunder god vine), ethanolic extract, 60mg, capsules, oral.

Outcomes

Joint tenderness and swelling, grip strength, 15 metre walking time, morning stiffness, physician global, patient global.

Notes

Results favour intervention for short‐term use (12 weeks), with cautions regarding adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Reported ethics committee approval.

Tao 2002

Methods

Randomised, double blind, placebo control, 3 parallel groups. Duration 20 weeks.

Participants

Randomised n=35 (low dose n=12, high dose n=11, control n=12), completed 4 weeks n=32, completed n=21. Age: low dose m=54 sd=12, high dose m=57 sd=8, control m=51 sd=12. Inclusion: ACR criteria RA stage II‐IV, for at least 1 year, active disease, 2+ swollen joints, and 2 of 6+ tender joints, morning stiffness >30min, ESR >28mm/h.

Interventions

Tripterygium wilfordii hook F (thunder god vine), ethanolic extract, low dose=180mg; high dose=360mg, capsules, oral.

Outcomes

ACR20, ACR50, ACR70, ESR, CRP, RF.

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

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

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Low risk

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

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals. Included intention‐to‐treat analyses.

Free of selective reporting?

Low risk

Reported adverse events.

Free of other bias?

Low risk

Diagnosis / assessment consistent with ACR criteria. Reported ethics committee approval.

Watson 1993

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 weeks.

Participants

Randomised n=50. Withdrawals not reported. Age: RA group m=40, health controls m=20 yr. Inclusion: definite RA, receiving only NSAIDs.

Interventions

Tradename not provided, Ribes nigrum, (blackcurrant seed), oil, 3000mg (6x500mg, approx 19% GLA, equivalent to 525mg GLA), capsules, oral.

Outcomes

Morning stiffness, grip strength, Ritchie index, pain score, patient global.

Notes

Results equivocal.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

Described as randomised, method not reported.

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double‐blind, method not reported.

Incomplete outcome data addressed?
All outcomes

High risk

Withdrawals not reported.

Free of selective reporting?

High risk

Adverse events not reported.

Free of other bias?

High risk

Data for clinical outcomes not reported.

Zurier 1996

Methods

Randomised, double blind, placebo control, 2 parallel groups. Duration 6 months (followed by 6 month single‐blind phase, followed by 3 month placebo phase).

Participants

Randomised n=56, completed n=41. Age: m=56 yr. Inclusion: ACR criteria RA stage I‐III, 1st line treatment stable for past 1 month, 2nd line treatment stable for past 3 months.

Interventions

GLA‐70, Borago officinalis (borage seed), oil, 4ml (8x0.5ml, approx 70% GLA, equivalent to 2800mg GLA), capsules, oral.

Outcomes

Pain VAS 0‐100, pain 0‐4, physician global, patient global, joint swelling and tenderness, morning stiffness, grip strength, health assessment questionnaire, ACR20 (6 and 12 month follow up).

Notes

Results favour intervention.

Risk of bias

Bias

Authors' judgement

Support for judgement

Adequate sequence generation?

Unclear risk

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

Allocation concealment?

Unclear risk

Unclear

Blinding?
All outcomes

Unclear risk

Described as double blind, method not reported.

Incomplete outcome data addressed?
All outcomes

Low risk

Reported withdrawals.

Free of selective reporting?

Unclear risk

After communication with author, unable to confirm SD for morning stiffness in GLA group (table 2) therefore these data excluded from analysis.

Free of other bias?

Unclear risk

Placebo capsules contained sunflower oil and may not be inert. 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. * Indicates that the trade name was not provided in the manuscript, but has been determined through communication with the manufacturing company noted in the acknowledgements.

ARA: American Rheumatism Association
ACR: American College of Rheumatology

EULAR: European league Against Rheumatism

Allocation concealment may be listed as "unclear" if: (a) the authors reported adherence to the ICH Good Clinical Practice (GCP) guidelines did not describe the method of allocation concealment used, or (b) the reviewers were unable to agree upon the adequacy of allocation concealment as reported.

Unless subscales are named, outcome measures (eg: HAQ, SF‐36, ACR20) were used in entirety. Unless specified, all measures were used, scaled, and scored to ACR/EULAR standards.

ACR core set of disease activity measures comprises tender joint count, swollen joint count, patient's assessment of pain, patient's and physician's global assessment of disease activity, patient's assessment of physical function (global assessment or HAQ‐DI score), and laboratory investigations of one acute‐phase reactant (ESR or C‐reactive protein). ACR20 is defined as 20% improvements in tender joint count, swollen joint count, and three of the other disease activity measures. ACR50 and ACR70 are similarly defined, but at 50% and 70% thresholds.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Anonymous 1993

Discussion paper.

Biswas 1997

Not placebo controlled.

Borigini 1996

Not randomised controlled trial.

Chrubasik 1998

Review paper.

Darlington 1987

Not placebo controlled.

DeLuca 1995

Review paper.

Dharmananda 1985

Discussion paper.

Falch 1997

Discussion paper.

Gendo 1997

Discussion paper.

Grahame 1981

Not randomised controlled trial.

Guo 1986

Not randomised controlled trial.

Hansen 1983

Not randomised controlled trial.

Hanyu 1989

Not randomised controlled trial.

Huber 1991

RA subgroup not distinguishable.

Jacobs 1991

Not a herbal intervention.

Jantti 1989b

No clinical outcomes reported.

Kou 1997

Case series, not a randomised controlled trial.

Larsen 1989

Not truly herbal intervention.

Linsheng 1997

Not randomised controlled trial.

Lipsky 1997

Review paper.

Loew 1996

Not a randomised controlled trial. Primary measures not consistent with the topic of this review.

Matsuta 1992

Discussion paper.

Mills 1996

RA subgroup not distinguishable.

Ohkaya 1988

Abstract only. Full text unavailable.

Ohkaya 1989

Abstract only. Full text unavailable.

Ramakrishanamacharya

Not randomised controlled trial.

Ramm 1996

Not randomised controlled trial.

Reuss 1981

Discussion paper.

Sagar 1988

Not randomised controlled trial.

Saley 1987

Not randomised controlled trial.

Srivastava 1989

Not randomised controlled trial.

Tao 1987

Not randomised controlled trial.

Vyas 1987

Not randomised controlled trial.

Wang 1985

Not randomised controlled trial.

Xu 1996

Not placebo controlled.

Yan 1985

Case series, not a randomised controlled trial.

Zell 1993

Not randomised controlled trial.

Characteristics of ongoing studies [ordered by study ID]

Shamsi 2009

Trial name or title

A clinical trial to study the effects of a herbal drug Qurs Mufasil in patients with joint pain (Arthritis)

Methods

Randomized, parallel group, placebo controlled trial

Participants

Patients of 20‐70 years of age fulfilling the criteria of American College of Rheumatology (ACR) for the diagnosis of Rheumatoid Arthritis, who had never received disease modifying anti/Rheumatoid Drugs (DMARDs). Presence of active disease as defined by the presence of >, 6 tender joints and >; 6 swollen joints.

Interventions

Qurs Mufasil:1000 mg daily for 3 months

Placebo:1000 mg twice daily for 3 months

Outcomes

Reduction in Swollen Joint Count,Tender Joint Count, Intensity of Pain‐VAS (0‐100), Morning Stiffness, ESR and CRP Timepoint:4,8,12 weeks;

Improvement in quality of life as assessed by Health Assessment Questionnaire (HAQ) Time Point: 3 months

Starting date

01‐03‐2003

Contact information

Yasmeen Shamsi

Majeedia Hospital, Jamia Hamdard, 110062 New Delhi, India

Email: [email protected]

Notes

Recruitment complete; http://apps.who.int/trialsearch/Trial.aspx?TrialID=CTRI/2009/091/000746

Data and analyses

Open in table viewer
Comparison 1. Gamma‐linolenic acid 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

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 1 Pain VAS 0‐100.

1.1 At 12 weeks‐ VAS score

1

18

Mean Difference (IV, Random, 95% CI)

6.00 [‐16.36, 28.36]

1.2 At 6 months‐ change from baseline

3

82

Mean Difference (IV, Random, 95% CI)

‐32.83 [‐56.25, ‐9.42]

2 Morning stiffness (minutes) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 2 Morning stiffness (minutes).

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 2 Morning stiffness (minutes).

2.1 At 12 weeks‐ minutes

1

18

Mean Difference (IV, Random, 95% CI)

‐5.0 [‐41.68, 31.68]

2.2 At 6 months‐ change from baseline

3

82

Mean Difference (IV, Random, 95% CI)

‐55.07 [‐76.87, ‐33.27]

3 68 tender joint count percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐53.80 [‐95.61, ‐12.00]

Analysis 1.3

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 3 68 tender joint count percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 3 68 tender joint count percentage change from baseline.

4 66 swollen joint count percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Fixed, 95% CI)

‐14.43 [‐31.43, 2.56]

Analysis 1.4

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 4 66 swollen joint count percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 4 66 swollen joint count percentage change from baseline.

5 Joint tenderness (0 to 3) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐56.64 [‐98.10, ‐15.17]

Analysis 1.5

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 5 Joint tenderness (0 to 3) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 5 Joint tenderness (0 to 3) percentage change from baseline.

6 Joint swelling (0 to 3) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐24.02 [‐70.80, 22.76]

Analysis 1.6

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 6 Joint swelling (0 to 3) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 6 Joint swelling (0 to 3) percentage change from baseline.

7 HAQ disability score percentage change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 1.7

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 7 HAQ disability score percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 7 HAQ disability score percentage change from baseline.

8 Patient global (0 to 4) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐20.87 [‐39.43, ‐2.31]

Analysis 1.8

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 8 Patient global (0 to 4) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 8 Patient global (0 to 4) percentage change from baseline.

9 Physician global (0 to 4) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐21.28 [‐70.52, 27.95]

Analysis 1.9

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 9 Physician global (0 to 4) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 9 Physician global (0 to 4) percentage change from baseline.

10 Participants (n) reported reduced NSAID use Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.10

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 10 Participants (n) reported reduced NSAID use.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 10 Participants (n) reported reduced NSAID use.

10.1 At 6 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 At 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Participants (n) reported adverse events Show forest plot

2

61

Risk Ratio (M‐H, Random, 95% CI)

4.24 [0.78, 22.99]

Analysis 1.11

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 11 Participants (n) reported adverse events.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 11 Participants (n) reported adverse events.

11.1 GLA 540mg

1

34

Risk Ratio (M‐H, Random, 95% CI)

5.59 [0.29, 108.38]

11.2 GLA 1400mg+

1

27

Risk Ratio (M‐H, Random, 95% CI)

3.71 [0.47, 29.06]

12 Participants (n) withdrawn due to worsening disease Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.12

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 12 Participants (n) withdrawn due to worsening disease.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 12 Participants (n) withdrawn due to worsening disease.

Open in table viewer
Comparison 2. Tripterygium wilfordii Hook F 60 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Joint tenderness (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.1

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 1 Joint tenderness (0 to 3).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 1 Joint tenderness (0 to 3).

2 60 swollen joint count Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 2 60 swollen joint count.

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 2 60 swollen joint count.

3 Morning stiffness (hours) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 3 Morning stiffness (hours).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 3 Morning stiffness (hours).

4 Grip strength (mmHg) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.4

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 4 Grip strength (mmHg).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 4 Grip strength (mmHg).

5 15 metre walking time (seconds) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 5 15 metre walking time (seconds).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 5 15 metre walking time (seconds).

Open in table viewer
Comparison 3. Tripterygium wilfordii Hook F 180 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.1

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 1 ACR20 responders.

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 1 ACR20 responders.

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 2 ACR50 responders.

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 2 ACR50 responders.

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 4. Triptrygium wilfordii Hook F 360 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.1

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 1 ACR20 responders.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 1 ACR20 responders.

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.2

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 2 ACR50 responders.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 2 ACR50 responders.

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 4.3

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 5. Tripterygium wilfordii Hook F 180 mg versus sulfasalazine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.1

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 1 ACR20 responders.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 1 ACR20 responders.

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.2

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 2 ACR50 responders.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 2 ACR50 responders.

3 Improvement more than 0.3 units on HAQ Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.3

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 3 Improvement more than 0.3 units on HAQ.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 3 Improvement more than 0.3 units on HAQ.

4 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 5.4

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 4 Participants (n) reported adverse events.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 4 Participants (n) reported adverse events.

5 Participants (n) withdrawn due to adverse events Show forest plot

1

121

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.19, 0.94]

Analysis 5.5

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 5 Participants (n) withdrawn due to adverse events.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 5 Participants (n) withdrawn due to adverse events.

Open in table viewer
Comparison 6. Phytodolor N versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (0 to 3) at 2 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 6.1

Comparison 6 Phytodolor N versus placebo, Outcome 1 Pain (0 to 3) at 2 weeks.

Comparison 6 Phytodolor N versus placebo, Outcome 1 Pain (0 to 3) at 2 weeks.

2 Joint swelling (0 to 3) at 2 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Phytodolor N versus placebo, Outcome 2 Joint swelling (0 to 3) at 2 weeks.

Comparison 6 Phytodolor N versus placebo, Outcome 2 Joint swelling (0 to 3) at 2 weeks.

3 Morning stiffness (minutes) at 12 months Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Phytodolor N versus placebo, Outcome 3 Morning stiffness (minutes) at 12 months.

Comparison 6 Phytodolor N versus placebo, Outcome 3 Morning stiffness (minutes) at 12 months.

4 Ritchie index at 12 months Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.4

Comparison 6 Phytodolor N versus placebo, Outcome 4 Ritchie index at 12 months.

Comparison 6 Phytodolor N versus placebo, Outcome 4 Ritchie index at 12 months.

5 Cumulative NSAID use (diclofenac) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 6.5

Comparison 6 Phytodolor N versus placebo, Outcome 5 Cumulative NSAID use (diclofenac).

Comparison 6 Phytodolor N versus placebo, Outcome 5 Cumulative NSAID use (diclofenac).

5.1 At 1 month (tablets)

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 At 12 months (tablets)

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 7. SKI306X 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

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 7.1

Comparison 7 SKI306X versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 7 SKI306X versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

1.1 At 3 weeks

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 6 weeks

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.2

Comparison 7 SKI306X versus celecoxib, Outcome 2 ACR20 responders.

Comparison 7 SKI306X versus celecoxib, Outcome 2 ACR20 responders.

2.1 At 3 weeks

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 At 6 weeks

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 7.3

Comparison 7 SKI306X versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 7 SKI306X versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Open in table viewer
Comparison 8. Salix purpurea x daphnoides (willow bark) 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)

Totals not selected

Analysis 8.1

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

2 28 tender joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.2

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 2 28 tender joint count change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 2 28 tender joint count change from baseline.

3 28 swollen joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.3

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 3 28 swollen joint count change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 3 28 swollen joint count change from baseline.

4 Patient assessment of efficacy VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.4

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 4 Patient assessment of efficacy VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 4 Patient assessment of efficacy VAS 0‐100 change from baseline.

5 Physician assessment of effiacy VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.5

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 5 Physician assessment of effiacy VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 5 Physician assessment of effiacy VAS 0‐100 change from baseline.

6 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 8.6

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 6 ACR20 responders.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 6 ACR20 responders.

7 HAQ disability index change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.7

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 7 HAQ disability index change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 7 HAQ disability index change from baseline.

8 SF‐36 physical component summary score change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.8

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 8 SF‐36 physical component summary score change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 8 SF‐36 physical component summary score change from baseline.

9 SF‐36 mental component summary score change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 8.9

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 9 SF‐36 mental component summary score change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 9 SF‐36 mental component summary score change from baseline.

Open in table viewer
Comparison 9. Feverfew versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Grip strength (mmHg) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 9.1

Comparison 9 Feverfew versus placebo, Outcome 1 Grip strength (mmHg) at 6 weeks.

Comparison 9 Feverfew versus placebo, Outcome 1 Grip strength (mmHg) at 6 weeks.

2 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 9.2

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

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

Open in table viewer
Comparison 10. RA‐1 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)

Totals not selected

Analysis 10.1

Comparison 10 RA‐1 versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

2 68 tender joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.2

Comparison 10 RA‐1 versus placebo, Outcome 2 68 tender joint count change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 2 68 tender joint count change from baseline.

3 66 swollen joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.3

Comparison 10 RA‐1 versus placebo, Outcome 3 66 swollen joint count change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 3 66 swollen joint count change from baseline.

4 Modified HAQ (Pune) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.4

Comparison 10 RA‐1 versus placebo, Outcome 4 Modified HAQ (Pune) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 4 Modified HAQ (Pune) change from baseline.

5 Patient global (1 to 5) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.5

Comparison 10 RA‐1 versus placebo, Outcome 5 Patient global (1 to 5) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 5 Patient global (1 to 5) change from baseline.

6 Physician global (1 to 5) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 10.6

Comparison 10 RA‐1 versus placebo, Outcome 6 Physician global (1 to 5) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 6 Physician global (1 to 5) change from baseline.

7 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.7

Comparison 10 RA‐1 versus placebo, Outcome 7 ACR20 responders.

Comparison 10 RA‐1 versus placebo, Outcome 7 ACR20 responders.

8 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 10.8

Comparison 10 RA‐1 versus placebo, Outcome 8 ACR50 responders.

Comparison 10 RA‐1 versus placebo, Outcome 8 ACR50 responders.

Open in table viewer
Comparison 11. Boswellia serrata 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

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 11.1

Comparison 11 Boswellia serrata versus placebo, Outcome 1 Participants (n) reported adverse events.

Comparison 11 Boswellia serrata versus placebo, Outcome 1 Participants (n) reported adverse events.

Open in table viewer
Comparison 12. Capsaicin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 percentage change at 4 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 12.1

Comparison 12 Capsaicin versus placebo, Outcome 1 Pain VAS 0‐100 percentage change at 4 weeks.

Comparison 12 Capsaicin versus placebo, Outcome 1 Pain VAS 0‐100 percentage change at 4 weeks.

2 Physician global (‐1 to 3) change from baseline at 4 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 12.2

Comparison 12 Capsaicin versus placebo, Outcome 2 Physician global (‐1 to 3) change from baseline at 4 weeks.

Comparison 12 Capsaicin versus placebo, Outcome 2 Physician global (‐1 to 3) change from baseline at 4 weeks.

Open in table viewer
Comparison 13. Tripterygium wilfordii (topical) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 42 tender joint count at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 13.1

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 1 42 tender joint count at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 1 42 tender joint count at 6 weeks.

2 40 swollen joint count at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 13.2

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 2 40 swollen joint count at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 2 40 swollen joint count at 6 weeks.

3 Grip strength (kPa) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 13.3

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 3 Grip strength (kPa) at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 3 Grip strength (kPa) at 6 weeks.

4 Morning stiffness (hours) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 13.4

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 4 Morning stiffness (hours) at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 4 Morning stiffness (hours) at 6 weeks.

5 ACR20 responders at 6 weeks Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 13.5

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 5 ACR20 responders at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 5 ACR20 responders at 6 weeks.

Open in table viewer
Comparison 14. Ganoderma lucidum and SMS versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.1

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 1 ACR20 responders.

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 1 ACR20 responders.

2 Adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 14.2

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 2 Adverse events.

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 2 Adverse events.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 1 Pain VAS 0‐100.
Figuras y tablas -
Analysis 1.1

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 1 Pain VAS 0‐100.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 2 Morning stiffness (minutes).
Figuras y tablas -
Analysis 1.2

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 2 Morning stiffness (minutes).

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 3 68 tender joint count percentage change from baseline.
Figuras y tablas -
Analysis 1.3

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 3 68 tender joint count percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 4 66 swollen joint count percentage change from baseline.
Figuras y tablas -
Analysis 1.4

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 4 66 swollen joint count percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 5 Joint tenderness (0 to 3) percentage change from baseline.
Figuras y tablas -
Analysis 1.5

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 5 Joint tenderness (0 to 3) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 6 Joint swelling (0 to 3) percentage change from baseline.
Figuras y tablas -
Analysis 1.6

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 6 Joint swelling (0 to 3) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 7 HAQ disability score percentage change from baseline.
Figuras y tablas -
Analysis 1.7

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 7 HAQ disability score percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 8 Patient global (0 to 4) percentage change from baseline.
Figuras y tablas -
Analysis 1.8

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 8 Patient global (0 to 4) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 9 Physician global (0 to 4) percentage change from baseline.
Figuras y tablas -
Analysis 1.9

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 9 Physician global (0 to 4) percentage change from baseline.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 10 Participants (n) reported reduced NSAID use.
Figuras y tablas -
Analysis 1.10

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 10 Participants (n) reported reduced NSAID use.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 11 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 1.11

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 11 Participants (n) reported adverse events.

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 12 Participants (n) withdrawn due to worsening disease.
Figuras y tablas -
Analysis 1.12

Comparison 1 Gamma‐linolenic acid versus placebo, Outcome 12 Participants (n) withdrawn due to worsening disease.

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 1 Joint tenderness (0 to 3).
Figuras y tablas -
Analysis 2.1

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 1 Joint tenderness (0 to 3).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 2 60 swollen joint count.
Figuras y tablas -
Analysis 2.2

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 2 60 swollen joint count.

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 3 Morning stiffness (hours).
Figuras y tablas -
Analysis 2.3

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 3 Morning stiffness (hours).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 4 Grip strength (mmHg).
Figuras y tablas -
Analysis 2.4

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 4 Grip strength (mmHg).

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 5 15 metre walking time (seconds).
Figuras y tablas -
Analysis 2.5

Comparison 2 Tripterygium wilfordii Hook F 60 mg versus placebo, Outcome 5 15 metre walking time (seconds).

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 1 ACR20 responders.
Figuras y tablas -
Analysis 3.1

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 1 ACR20 responders.

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 2 ACR50 responders.
Figuras y tablas -
Analysis 3.2

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 2 ACR50 responders.

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 3 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 3.3

Comparison 3 Tripterygium wilfordii Hook F 180 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 1 ACR20 responders.
Figuras y tablas -
Analysis 4.1

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 1 ACR20 responders.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 2 ACR50 responders.
Figuras y tablas -
Analysis 4.2

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 2 ACR50 responders.

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 3 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 4.3

Comparison 4 Triptrygium wilfordii Hook F 360 mg versus placebo, Outcome 3 Participants (n) reported adverse events.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 1 ACR20 responders.
Figuras y tablas -
Analysis 5.1

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 1 ACR20 responders.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 2 ACR50 responders.
Figuras y tablas -
Analysis 5.2

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 2 ACR50 responders.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 3 Improvement more than 0.3 units on HAQ.
Figuras y tablas -
Analysis 5.3

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 3 Improvement more than 0.3 units on HAQ.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 4 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 5.4

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 4 Participants (n) reported adverse events.

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 5 Participants (n) withdrawn due to adverse events.
Figuras y tablas -
Analysis 5.5

Comparison 5 Tripterygium wilfordii Hook F 180 mg versus sulfasalazine, Outcome 5 Participants (n) withdrawn due to adverse events.

Comparison 6 Phytodolor N versus placebo, Outcome 1 Pain (0 to 3) at 2 weeks.
Figuras y tablas -
Analysis 6.1

Comparison 6 Phytodolor N versus placebo, Outcome 1 Pain (0 to 3) at 2 weeks.

Comparison 6 Phytodolor N versus placebo, Outcome 2 Joint swelling (0 to 3) at 2 weeks.
Figuras y tablas -
Analysis 6.2

Comparison 6 Phytodolor N versus placebo, Outcome 2 Joint swelling (0 to 3) at 2 weeks.

Comparison 6 Phytodolor N versus placebo, Outcome 3 Morning stiffness (minutes) at 12 months.
Figuras y tablas -
Analysis 6.3

Comparison 6 Phytodolor N versus placebo, Outcome 3 Morning stiffness (minutes) at 12 months.

Comparison 6 Phytodolor N versus placebo, Outcome 4 Ritchie index at 12 months.
Figuras y tablas -
Analysis 6.4

Comparison 6 Phytodolor N versus placebo, Outcome 4 Ritchie index at 12 months.

Comparison 6 Phytodolor N versus placebo, Outcome 5 Cumulative NSAID use (diclofenac).
Figuras y tablas -
Analysis 6.5

Comparison 6 Phytodolor N versus placebo, Outcome 5 Cumulative NSAID use (diclofenac).

Comparison 7 SKI306X versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.
Figuras y tablas -
Analysis 7.1

Comparison 7 SKI306X versus celecoxib, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 7 SKI306X versus celecoxib, Outcome 2 ACR20 responders.
Figuras y tablas -
Analysis 7.2

Comparison 7 SKI306X versus celecoxib, Outcome 2 ACR20 responders.

Comparison 7 SKI306X versus celecoxib, Outcome 3 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 7.3

Comparison 7 SKI306X versus celecoxib, Outcome 3 Participants (n) reported adverse events.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.
Figuras y tablas -
Analysis 8.1

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 2 28 tender joint count change from baseline.
Figuras y tablas -
Analysis 8.2

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 2 28 tender joint count change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 3 28 swollen joint count change from baseline.
Figuras y tablas -
Analysis 8.3

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 3 28 swollen joint count change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 4 Patient assessment of efficacy VAS 0‐100 change from baseline.
Figuras y tablas -
Analysis 8.4

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 4 Patient assessment of efficacy VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 5 Physician assessment of effiacy VAS 0‐100 change from baseline.
Figuras y tablas -
Analysis 8.5

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 5 Physician assessment of effiacy VAS 0‐100 change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 6 ACR20 responders.
Figuras y tablas -
Analysis 8.6

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 6 ACR20 responders.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 7 HAQ disability index change from baseline.
Figuras y tablas -
Analysis 8.7

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 7 HAQ disability index change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 8 SF‐36 physical component summary score change from baseline.
Figuras y tablas -
Analysis 8.8

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 8 SF‐36 physical component summary score change from baseline.

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 9 SF‐36 mental component summary score change from baseline.
Figuras y tablas -
Analysis 8.9

Comparison 8 Salix purpurea x daphnoides (willow bark) versus placebo, Outcome 9 SF‐36 mental component summary score change from baseline.

Comparison 9 Feverfew versus placebo, Outcome 1 Grip strength (mmHg) at 6 weeks.
Figuras y tablas -
Analysis 9.1

Comparison 9 Feverfew versus placebo, Outcome 1 Grip strength (mmHg) at 6 weeks.

Comparison 9 Feverfew versus placebo, Outcome 2 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 9.2

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

Comparison 10 RA‐1 versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.
Figuras y tablas -
Analysis 10.1

Comparison 10 RA‐1 versus placebo, Outcome 1 Pain VAS 0‐100 change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 2 68 tender joint count change from baseline.
Figuras y tablas -
Analysis 10.2

Comparison 10 RA‐1 versus placebo, Outcome 2 68 tender joint count change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 3 66 swollen joint count change from baseline.
Figuras y tablas -
Analysis 10.3

Comparison 10 RA‐1 versus placebo, Outcome 3 66 swollen joint count change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 4 Modified HAQ (Pune) change from baseline.
Figuras y tablas -
Analysis 10.4

Comparison 10 RA‐1 versus placebo, Outcome 4 Modified HAQ (Pune) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 5 Patient global (1 to 5) change from baseline.
Figuras y tablas -
Analysis 10.5

Comparison 10 RA‐1 versus placebo, Outcome 5 Patient global (1 to 5) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 6 Physician global (1 to 5) change from baseline.
Figuras y tablas -
Analysis 10.6

Comparison 10 RA‐1 versus placebo, Outcome 6 Physician global (1 to 5) change from baseline.

Comparison 10 RA‐1 versus placebo, Outcome 7 ACR20 responders.
Figuras y tablas -
Analysis 10.7

Comparison 10 RA‐1 versus placebo, Outcome 7 ACR20 responders.

Comparison 10 RA‐1 versus placebo, Outcome 8 ACR50 responders.
Figuras y tablas -
Analysis 10.8

Comparison 10 RA‐1 versus placebo, Outcome 8 ACR50 responders.

Comparison 11 Boswellia serrata versus placebo, Outcome 1 Participants (n) reported adverse events.
Figuras y tablas -
Analysis 11.1

Comparison 11 Boswellia serrata versus placebo, Outcome 1 Participants (n) reported adverse events.

Comparison 12 Capsaicin versus placebo, Outcome 1 Pain VAS 0‐100 percentage change at 4 weeks.
Figuras y tablas -
Analysis 12.1

Comparison 12 Capsaicin versus placebo, Outcome 1 Pain VAS 0‐100 percentage change at 4 weeks.

Comparison 12 Capsaicin versus placebo, Outcome 2 Physician global (‐1 to 3) change from baseline at 4 weeks.
Figuras y tablas -
Analysis 12.2

Comparison 12 Capsaicin versus placebo, Outcome 2 Physician global (‐1 to 3) change from baseline at 4 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 1 42 tender joint count at 6 weeks.
Figuras y tablas -
Analysis 13.1

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 1 42 tender joint count at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 2 40 swollen joint count at 6 weeks.
Figuras y tablas -
Analysis 13.2

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 2 40 swollen joint count at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 3 Grip strength (kPa) at 6 weeks.
Figuras y tablas -
Analysis 13.3

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 3 Grip strength (kPa) at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 4 Morning stiffness (hours) at 6 weeks.
Figuras y tablas -
Analysis 13.4

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 4 Morning stiffness (hours) at 6 weeks.

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 5 ACR20 responders at 6 weeks.
Figuras y tablas -
Analysis 13.5

Comparison 13 Tripterygium wilfordii (topical) versus placebo, Outcome 5 ACR20 responders at 6 weeks.

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 1 ACR20 responders.
Figuras y tablas -
Analysis 14.1

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 1 ACR20 responders.

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 2 Adverse events.
Figuras y tablas -
Analysis 14.2

Comparison 14 Ganoderma lucidum and SMS versus placebo, Outcome 2 Adverse events.

Evening primrose oil, borage seed oil, blackcurrent seed oil (containing gamma‐linolenic acid) for rheumatoid arthritis

Patient or population: patients with treating rheumatoid arthritis

Settings: community

Intervention: Evening primrose oil, borage seed oil, blackcurrent seed oil (with gamma‐linolenic acid) versus placebo

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

Evening primrose oil, borage seed oil, blackcurrent seed oil (with gamma‐linolenic acid) versus placebo

ACR 50% improvement ‐ not measured

See comment

See comment

Not estimable

See comment

Not measured

Pain
Visual Analogue Scale. Scale from: 0 to 100.
(follow‐up: 6 months)

The mean pain in the control groups was
60.6 points1

The mean Pain in the intervention groups was
32.83 lower
(56.25 to 9.42 lower)

82
(3)

⊕⊕⊕⊝
moderate2

Absolute risk difference 33 points lower (9 to 56 points lower); relative % change 54% (15 to 92%); NNTB 3 (2 to 12)3

Disability (HAQ score)
percent change. Scale from: 0 to 100.
(follow‐up: 6 months)

The mean disability (haq score) in the control groups was
42.54

The mean Disability (HAQ score) in the intervention groups was
16 lower
(27 to 4 lower)

41
(1)

⊕⊕⊝⊝
low2,5

Absolute risk difference 16% lower (4 to 27% lower); relative % change 38% (9% to 64%); NNTB 3 (95% CI 2 to 11)

Participants (n) reported adverse events
(follow‐up: 6 months)

Medium risk population

RR 4.24
(0.78 to 22.99)

61
(2)

⊕⊕⊕⊝
moderate2

Absolute risk difference 15% (0 to 30%); relative percent change 324% (‐22% to 2199 %); NNT=n/a 3

39 per 1000

165 per 1000
(30 to 897)

Change in radiographic progression ‐ not measured

See comment

See comment

Not estimable

See comment

Not measured

Achievement of low disease state (DAS 28) ‐ not measured

See comment

See comment

Not estimable

See comment

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; RR: Risk ratio;

GRADE Working Group grades of evidance
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 From Zurier 1996, mean (SD) pain at baseline in placebo = 60.6 (21.0)

2 Unclear if randomisation was concealed or outcome assessor blinded

3 NOTE: Number needed to treat (NNT)=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).

4 From Zurier 1996, mean (SD) HAQ score on 0‐100 scale at baseline in placebo = 42.5 (11.25)

5 Results based on one small trial nly

Figuras y tablas -

Triptrygium wilfordii Hook F 360 mg versus placebo for Rheumatoid arthritis

Patient or population: patients with Rheumatoid arthritis

Settings: Community

Intervention: Triptrygium wilfordii Hook F 360 mg versus placebo

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

Triptrygium wilfordii Hook F 360 mg versus placebo

ACR 50% improvement
(follow‐up: 20 weeks)

Medium risk population1

RR 11.92
(0.73 to 193.38)

23
(1)

⊕⊕⊝⊝
low2,3

Absolute risk difference = 45% (15% to 76%) increase; relative percent change = 1090% (‐27% to 19238%); NNTB n/a4

1 per 1000

12 per 1000
(1 to 193)

Change in pain ‐ not measured

See comment

See comment

Not estimable

See comment

Not measured

HAQ disability score ‐ not measured

See comment

See comment

Not estimable

See comment

Not measured

Participants (n) reported adverse events
(follow‐up: 20 weeks)

Medium risk population

RR 1.36
(0.49 to 3.82)

23
(1)

⊕⊕⊝⊝
low2,3

Absolute risk difference = 12% increase (‐28% to 52%); relative percent change = 36% (‐51% to 282%); NNTH = n/a
4

333 per 1000

453 per 1000
(163 to 1272)

Change in radiographic progression ‐ not measured

See comment

See comment

Not estimable

See comment

Not measured

Achievement of low disease state (DAS 28) ‐ not measured

See comment

See comment

Not estimable

See comment

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; RR: Risk ratio;

GRADE Working Group grades of evidance
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 Only one study available, with control event rate of 0%; thus assumed 1% control event rate for purposes of calculations; control event rates for 5 trials in review (different therapies) ranged from 0 to 35%

2 Unclear if randomisation was concealed

3 Results based on one small trial

4 NOTE: Number needed to treat (NNT)=n/a when result is not statistically significant. NNT for dichotomous outcomes calculated using Cates NNT calculator (http://www.nntonline.net/visualrx/); or for single studies as 1/RD. NNT for continuous outcomes calculated using Wells Calculator (CMSG editorial office)

Figuras y tablas -
Table 1. Details of the herbal medicinal products used for the treatment of RA

Botanical name

Plant part

Tradename

Preparation

Drug/Extract

mg/day

Constituent marker

Marker mg/day

References

Populus tremula + Fraxinus excelsior + Solidago virgaurea

bark, herb, leaf

Phytodolor

fresh plant ethanolic (45,6%) extract

3 : 1 : 1

5‐8 ml

total flavonoids

0.34‐0.56

Eberl 1988; Meier 1987

Populus tremula

bark, leaf

fresh plant ethanolic (45,6%) extract

salicin

4.8‐8.0

Solidago virgaurea

herb

fresh plant ethanolic (45,6%) extract

salicyl alcohol

0.48‐0.8

Fraxinus excelsior

bark

fresh plant ethanolic (45,6%) extract

isofraxidin

0.67‐1.1

Salix daphnoides

bark

SM

$ ethanolic (70%) extract

8‐14:1

1573

salicin

240

Biegert 2004

Tripterygium wilfordii Hook F

root

SM

ethanol / ethyl acetate extract

45:1

180

triptolide
tripdiolide
triptonide
triptophenolide

0.194, 0.056, 0.0142, 0.746

Tao 1995, Goldbach‐Mansky 2009

Tripterygium wilfordii Hook F

root

SM

ethanol / ethyl acetate extract

45:1

360

triptolide
tripdiolide
triptonide
triptophenolide

0.389, 0.112, 0.284, 1.472

Tao 1995, Goldbach‐Mansky 2009

Tripterygium wilfordii Hook F

root

T2

chloroform / methanol extract

not stated

60

tripdiolide
triptdiolide
triptonide
triptophenolide

0.021, 0.041, 0.002,

0.002

Tao 1995

Tripterygium wilfordii (local)

root

Thunder God vine

not stated

not stated

topical 5‐6 times per day

not stated

not stated

Cibere 2003

Tripterygium wilfordii Hook F

root

TwHF extract

ethanol / ethyl acetate extract

not stated

180

triptolide and tripdiolide

not stated

Goldbach‐Mansky 2009

Withania somnifera,
Boswellia serrata,
Zingiberis officinale,
Curcuma longa

RA‐1

not stated

not stated

444 ‐ 592

not stated

not stated

Chopra 2000

Clematis mandshurica,
Prunella vulgaris,
Trichosanthes kirilowii

root, flower, root; 1:1:2

SKI‐306X

ethanol 30% extracts
thereafter
butanol extraction

7:1

oleanolic acid 4%, rosmarinic acids 0.2%, ursolic acids 0.5%, hydroxybenzoic acid 0.03%,
hydroxymethoxybenzoic acid 0.03%, trans‐cinnamic
acid 0.05%

Song 2007

Uncaria tomentosa

bark

Krallendorn

aqueous acid axtract

not stated

60

pentacyclic
oxindole alkaloids

0.88

Mur 2002

Tanacetum parthenium

leaf

SM

powder

76

parthenolide

2‐3 micromol

Pattrick 1989

Capsicum (local)

fruit

Zostrix

not stated

0.025%

topical QID

Deal 1991

fruit

Arlacel 165

not stated

0.075%

topical QID

McCarthy 1992

Oenothera biennis

semen

SM

oil

not stated

540

gamma‐linolenic acid (GLA)

540

Belch 1988

semen

SM

oil

9% GLA

6000

GLA

540

Brzeski 1991

semen

SM

oil

not stated

20‐30 ml

GLA

not stated

Jantti 1989

Ribes nigrum

semen

SM

oil

17% GLA

3000

GLA

525

Watson 1993

semen

SM

oil

19% GLA

10500

GLA

2000

Leventhal 1994

Borago officinalis

semen

SM

oil

23% GLA

7.2 ml

GLA

1400

Leventhal 1993

semen

SM

oil

70% GLA

GLA

2800

Zurier 1996

Ganoderma lucida (4g) + San Miao San (Atractylodes macrocephala root, Phellodendron chinense cortex, Achyranthes bidentatae root)

not stated

not stated

aqueous extract

not stated

Rhizoma atractylodis 2.4g; Cotex phellodendri 2.4g; Radix achyranthes Bidentatae 2.4g

not stated

not stated

Li 2007

SM = study medication

$ ethanolic extract stated in the thesis (University of Tübingen)
$ 50g/100 g gel, details from Bioforce AG/Schweiz

Figuras y tablas -
Table 1. Details of the herbal medicinal products used for the treatment of RA
Comparison 1. Gamma‐linolenic acid 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

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 At 12 weeks‐ VAS score

1

18

Mean Difference (IV, Random, 95% CI)

6.00 [‐16.36, 28.36]

1.2 At 6 months‐ change from baseline

3

82

Mean Difference (IV, Random, 95% CI)

‐32.83 [‐56.25, ‐9.42]

2 Morning stiffness (minutes) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 At 12 weeks‐ minutes

1

18

Mean Difference (IV, Random, 95% CI)

‐5.0 [‐41.68, 31.68]

2.2 At 6 months‐ change from baseline

3

82

Mean Difference (IV, Random, 95% CI)

‐55.07 [‐76.87, ‐33.27]

3 68 tender joint count percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐53.80 [‐95.61, ‐12.00]

4 66 swollen joint count percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Fixed, 95% CI)

‐14.43 [‐31.43, 2.56]

5 Joint tenderness (0 to 3) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐56.64 [‐98.10, ‐15.17]

6 Joint swelling (0 to 3) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐24.02 [‐70.80, 22.76]

7 HAQ disability score percentage change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

8 Patient global (0 to 4) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐20.87 [‐39.43, ‐2.31]

9 Physician global (0 to 4) percentage change from baseline Show forest plot

3

82

Mean Difference (IV, Random, 95% CI)

‐21.28 [‐70.52, 27.95]

10 Participants (n) reported reduced NSAID use Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 At 6 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 At 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Participants (n) reported adverse events Show forest plot

2

61

Risk Ratio (M‐H, Random, 95% CI)

4.24 [0.78, 22.99]

11.1 GLA 540mg

1

34

Risk Ratio (M‐H, Random, 95% CI)

5.59 [0.29, 108.38]

11.2 GLA 1400mg+

1

27

Risk Ratio (M‐H, Random, 95% CI)

3.71 [0.47, 29.06]

12 Participants (n) withdrawn due to worsening disease Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 1. Gamma‐linolenic acid versus placebo
Comparison 2. Tripterygium wilfordii Hook F 60 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Joint tenderness (0 to 3) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 60 swollen joint count Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Morning stiffness (hours) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Grip strength (mmHg) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5 15 metre walking time (seconds) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 2. Tripterygium wilfordii Hook F 60 mg versus placebo
Comparison 3. Tripterygium wilfordii Hook F 180 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, 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

Figuras y tablas -
Comparison 3. Tripterygium wilfordii Hook F 180 mg versus placebo
Comparison 4. Triptrygium wilfordii Hook F 360 mg versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, 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

Figuras y tablas -
Comparison 4. Triptrygium wilfordii Hook F 360 mg versus placebo
Comparison 5. Tripterygium wilfordii Hook F 180 mg versus sulfasalazine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

3 Improvement more than 0.3 units on HAQ Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

4 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

5 Participants (n) withdrawn due to adverse events Show forest plot

1

121

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.19, 0.94]

Figuras y tablas -
Comparison 5. Tripterygium wilfordii Hook F 180 mg versus sulfasalazine
Comparison 6. Phytodolor N versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain (0 to 3) at 2 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 Joint swelling (0 to 3) at 2 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3 Morning stiffness (minutes) at 12 months Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4 Ritchie index at 12 months Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

5 Cumulative NSAID use (diclofenac) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5.1 At 1 month (tablets)

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 At 12 months (tablets)

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 6. Phytodolor N versus placebo
Comparison 7. SKI306X 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

Mean Difference (IV, Random, 95% CI)

Totals not selected

1.1 At 3 weeks

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

1.2 At 6 weeks

1

Mean Difference (IV, Random, 95% CI)

0.0 [0.0, 0.0]

2 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 At 3 weeks

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 At 6 weeks

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 7. SKI306X versus celecoxib
Comparison 8. Salix purpurea x daphnoides (willow bark) 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)

Totals not selected

2 28 tender joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 28 swollen joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Patient assessment of efficacy VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5 Physician assessment of effiacy VAS 0‐100 change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

6 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

7 HAQ disability index change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

8 SF‐36 physical component summary score change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

9 SF‐36 mental component summary score change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 8. Salix purpurea x daphnoides (willow bark) versus placebo
Comparison 9. Feverfew versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Grip strength (mmHg) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 Participants (n) reported adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 9. Feverfew versus placebo
Comparison 10. RA‐1 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)

Totals not selected

2 68 tender joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 66 swollen joint count change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Modified HAQ (Pune) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5 Patient global (1 to 5) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

6 Physician global (1 to 5) change from baseline Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

7 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

8 ACR50 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 10. RA‐1 versus placebo
Comparison 11. Boswellia serrata 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

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 11. Boswellia serrata versus placebo
Comparison 12. Capsaicin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain VAS 0‐100 percentage change at 4 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 Physician global (‐1 to 3) change from baseline at 4 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 12. Capsaicin versus placebo
Comparison 13. Tripterygium wilfordii (topical) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 42 tender joint count at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 40 swollen joint count at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

3 Grip strength (kPa) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

4 Morning stiffness (hours) at 6 weeks Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

5 ACR20 responders at 6 weeks Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 13. Tripterygium wilfordii (topical) versus placebo
Comparison 14. Ganoderma lucidum and SMS versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 ACR20 responders Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2 Adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Figuras y tablas -
Comparison 14. Ganoderma lucidum and SMS versus placebo