Scolaris Content Display Scolaris Content Display

Trasplante intramuscular local de células mononucleares autólogas de médula ósea para la isquemia crítica de los miembros inferiores

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Barc 2006 {published data only}

Barc P, Skora J, Pupka A, Turkiewicz D, Dorobisz AT, Garcarek J, et al. Bone-marrow cells in therapy of critical limb ischaemia of lower extremities - own experience. Acta Angiologica 2006;12(4):155-66. CENTRAL

Li 2013 {published data only}

Li M, Zhou H, Jin X, Wang M, Zhang S, Xu L. Autologous bone marrow mononuclear cells transplant in patients with critical leg ischemia: preliminary clinical results. Experimental and Clinical Transplantation 2013;11(5):435-9. CENTRAL

Lindeman 2018 {published data only}

Lindeman JH, Zwaginga JJ, Kallenberg-Lantrua G, van Wissen RC, Schepers A, van Bockel HJ, et al. No clinical benefit of intramuscular delivery of bone marrow-derived mononuclear cells in non-reconstructable peripheral arterial disease: results of a phase-III randomized-controlled trial. Annals of Surgery 2018;268(5):756-61. CENTRAL

Pignon 2017 {published data only}

NCT00904501. Bone marrow auto graft in limb ischemia (BALI). clinicaltrials.gov/ct2/show/study/NCT00904501 (first received 19 May 2009). CENTRAL
Pignon B, Sevestre MA, Kanagaratnam L, Pernod G, Stephan D, Emmerich J, et al. Autologous bone marrow mononuclear cell implantation and its impact on the outcome of patients with critical limb ischemia - results of a randomized, double-blind, placebo-controlled trial. Circulation Journal 2017;81(11):1713-20. CENTRAL

Referencias de los estudios excluidos de esta revisión

Benoit 2011 {published data only}

Benoit E, O'Donnell TF Jr, Iafrati MD, Asher E, Bandyk DF, Hallett JW, et al. The role of amputation as an outcome measure in cellular therapy for critical limb ischemia: implications for clinical trial design. Journal of Translational Medicine 2011;9(1):1-9. CENTRAL

BONMOT 2008 {published data only}

Amann B, Lüdemann C, Rückert R, Lawall H, Liesenfeld B, Schneider M, et al. Design and rationale of a randomized, double-blind, placebo-controlled phase III study for autologous bone marrow cell transplantation in critical limb ischemia: the bone marrow outcomes trial in critical limb ischemia (BONMOT-CLI). Vasa 2008;37(4):319-25. CENTRAL
Luedemann C, Amann B, Rueckert R, Ratei R, Schmidt-Lucke JA. Induction of arteriogenesis by autologous bone marrow transplantation (aBMT) in critical limb ischemia (CLI): the BONMOT 1 and 2 (bone marrow transplantation) studies. European Heart Journal 2008;29(Suppl 1):144. CENTRAL

Burt 2010 {published data only}

Burt RK, Testori A, Oyama Y, Rodriguez HE, Yaung K, Villa M, et al. Autologous peripheral blood CD133+ cell implantation for limb salvage in patients with critical limb ischemia. Bone Marrow Transplantation 2010;45(1):111–6. CENTRAL

Capiod 2009 {published data only}

Capiod JC, Tournois C, Vitry F, Sevestre MA, Daliphard S, Reix T, et al. Characterization and comparison of bone marrow and peripheral blood mononuclear cells used for cellular therapy in critical leg ischaemia: towards a new cellular product. Vox Sanguinis 2009;96(3):256-65. CENTRAL

Chen 2009 {published data only}

Chen B, Lu DB, Liang ZW, Jiang YZ, Wang FH, Wu QN, et al. Autologous bone marrow mesenchymal stem cell transplantation for treatment of diabetic foot following amplification in vitro. Journal of Clinical Rehabilitative Tissue Engineering Research 2009;13:6227-30. CENTRAL

Debin 2008 {published data only}

Debin L, Youzhao J, Ziwen L, Xiaoyan L, Zhonghui Z, Bing C. Autologous transplantation of bone marrow mesenchymal stem cells on diabetic patients with lower limb ischemia. Journal of Medical Colleges of PLA 2008;23:106-15. CENTRAL

Dong 2013 {published data only}

Dong Z, Chen B, Fu W, Wang Y, Guo D, Wei Z, et al. Transplantation of purified CD34+ cells in the treatment of critical limb ischemia. Journal of Vascular Surgery 2013;58(2):404-11. CENTRAL

Dong 2018 {published data only}

Dong Z, Pan T, Fang Y, Wei Z, Gu S, Fang G, et al. Purified CD34(+) cells versus peripheral blood mononuclear cells in the treatment of angiitis-induced no-option critical limb ischaemia: 12-month results of a prospective randomised single-blinded non-inferiority trial. EBioMedicine 2018;35:46-57. CENTRAL

Du 2017 {published data only}

Du JW, Wu T, Zhang K, Su BY, Lu CP, Wang WC, et al. Umbilical cord mesenchymal stem cells combined with bone marrow stem cells for treatment of lower limb ischemia. Chinese Journal of Tissue Engineering Research 2017;21(1):82–6. CENTRAL

Flugelman 2017 {published data only}

Flugelman MY, Halak M, Yoffe B, Schneiderman J, Rubinstein C, Bloom AI, et al. Phase Ib safety, two-dose study of MultiGeneAngio in patients with chronic critical limb ischemia. Molecular Therapy 2017;25(3):816–25. CENTRAL

Frogel 2017 {published data only}

Frogel M, Niven MJ, Galili O, Sivak G, Kafri E, Moshe M, et al. Adult stem/progenitor cells derived from peripheral blood as a personalized treatment for critical limb ischemia (CLI). Vascular 2017;25(2 Suppl 1):88. CENTRAL

Gupta 2013 {published data only}

Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, et al. A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. Journal of Translational Medicine 2013;11:143. CENTRAL

Gurunathan 2009 {published data only}

Gurunathan Mani S, Raju R, Kuppu Sampath V. The Vascular Society of Great Britain and Ireland Yearbook 2009: Multicentre randomised clinical trial on the role of autologous bone-marrow aspirate-concentrate (BMAC)/CD34/(EPC) in non-reconstructable critically ischaemic limbs. www.vascularsociety.org.uk/_userfiles/pages/files/Document%20Library/yearbook_09_web_no_members.pdf (accessed 8 August 2019). CENTRAL

Higashi 2010 {published data only}

Higashi Y, Miyazaki M, Goto C, Sanada H, Sueda T, Chayama K. Sarpogrelate hydrochloride, a selective 5-hydroxytryptamine 2A antagonist, augments autologous bone marrow mononuclear cell implantation-induced improvement in endothelium-dependent vasodilation in patients with critical limb ischemia. Journal of Cardiovascular Pharmacology 2010;55(1):56-61. CENTRAL

Holzinger 1994 {published data only}

Holzinger C, Zuckermann A, Kopp C, Schollhammer A, Imhof M, Zwolfer W, et al. Treatment of non-healing skin ulcers with autologous activated mononuclear cells. European Journal of Vascular Surgery 1994;8(3):351-6. CENTRAL

Horie 2018 {published data only}

Horie T, Yamazaki S, Hanada S, Kobayashi S, Tsukamoto T, Haruna T. Outcome from a randomized controlled clinical trial - improvement of peripheral arterial disease by granulocyte colony-stimulating factor-mobilized autologous peripheral-blood-mononuclear cell transplantation (IMPACT). Circulation Journal 2018;82(8):2165-74. CENTRAL

Huang 2005a {published data only}

Huang P, Li S, Han M, Xiao Z, Yang R, Han ZC. Autologous transplantation of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells improves critical limb ischemia in diabetes. Diabetes Care 2005;28(9):2155-60. CENTRAL

Iafrati 2011 {published data only}

Iafrati MD, Hallett JW, Geils G, Pearl G, Lumsden A, Peden E, et al. Early results and lessons learned from a multicenter, randomized, double-blind trial of bone marrow aspirate concentrate in critical limb ischemia. Journal of Vascular Surgery 2011;54(6):1650-8. CENTRAL
Iafrati MDB, Pearl A. Bone marrow aspirate concentrate in critical limb ischemia: results of a multicenter randomized double-blind trial. Journal of Vascular Surgery 2010;52:1123. CENTRAL

Iafrati 2016 {published data only}

Iafrati MD, O’Donnell TF Jr, Perler B, Illig KA, Hallett J, Woo K, et al. Bone marrow aspirate concentrate in critical limb ischemia: results of an abridged prospective randomized pivotal trial in no option CLI. Abstracts of the 2016 Vascular Annual Meeting, The Society for Vascular Surgery 2016;63(6 Suppl):47S. CENTRAL

JUVENTAS 2008 {published data only}NCT00371371

Han ZC, Huang P, Li S. Autologous peripheral blood stem cell implantation as therapeutic angiogenesis for severe limb ischemia. Blood 2004;104:130b. CENTRAL
NCT00371371. Intra-arterial stem cell therapy for patients with chronic limb ischemia (CLI) (JUVENTAS). clinicaltrials.gov/ct2/show/NCT00371371 (first received 4 September 2006). CENTRAL
Sprengers RW, Moll FL, Teraa M, Verhaar MC, JUVENTAS Study Group. Rationale and design of the JUVENTAS trial for repeated intra-arterial infusion of autologous bone marrow-derived mononuclear cells in patients with critical limb ischemia. Journal of Vascular Surgery 2010;51(6):1564-8. CENTRAL

Kirana 2007 {published data only}

Kirana S, Stratmann B, Lammers D, Minartz P, Negrean M, Stirban A, et al. Autologous tissue repair cells (TRC) in the treatment of ischemia induced chronic tissue ulcers of diabetic foot patients without option of revascularisation: first year experiences. Medizinische Klinik 2007;102:80. CENTRAL

Kirana 2012 {published data only}

Kirana S, Stratmann B, Prante C, Prohaska W, Koerperich H, Lammers D, et al. Autologous stem cell therapy in the treatment of limb ischaemia induced chronic tissue ulcers of diabetic foot patients. International Journal of Clinical Practice 2012;66(4):384-93. CENTRAL

Klepanec 2012 {published data only}

Klepanec A, Mistrik M, Altaner C, Valachovicova M, Olejarova I, Slysko R, et al. No difference in intra-arterial and intramuscular delivery of autologous bone marrow cells in patients with advanced critical limb ischemia. Cell Transplantation 2012;21(9):1909-18. CENTRAL

Korymasov 2009 {published data only}

Korymasov EA, Tiumina OV, Aiupov AM, Kazantsev AV, Rossiev VA, Mikheev GV, et al. Use of autologous progenitor cells of the bone marrow in treatment of patients with lower-limb atherosclerosis obliterans. Angiologiia iSosudistaia Khirurgiia 2009;15(3):28-31. CENTRAL

Lasala 2011 {published data only}

Lasala GP, Silva JA, Minguell JJ. Therapeutic angiogenesis in patients with severe limb ischemia by transplantation of an autologous bone marrow-derived combination stem cell product. Journal of the American College of Cardiology 2011;57:E2020. CENTRAL

Lu 2011 {published data only}

Lu D, Chen B, Liang Z, Deng W, Jiang Y, Li S, et al. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Research and Clinical Practice 2011;92(1):26-36. CENTRAL

Madaric 2011 {published data only}

Madaric J, Klepanec A, Mistrik M, Altaner C, Valachovicova M, Necpal R. Autologous bone marrow cells transplantation in patients with advanced critical limb ischemia: no difference in intra-arterial and intramuscular application. Journal of the American College of Cardiology 2011;57(14 Suppl 1):E1473. CENTRAL
Madaric JK. Higher CD34+ cells concentration and lower degree of inflammation are associated with better therapeutic response to autologous bone marrow cells application in patients with critical limb ischemia. Journal of the American College of Cardiology 2011;58:B150. CENTRAL

Majumdar 2015 {published data only}

Majumdar AS, Balasubramanian S, Thej C, Rajkumar M, Krishna M, Dutta S, et al. A first of its kind phase II clinical trial in critical limb ischemia patients using bone marrow derived, pooled, allogeneic mesenchymal stromal cells (Stempeucel). Cytotherapy 2015;17:S84. CENTRAL

Mohamed 2020 {published data only}10.1016/j.jcyt.2020.02.007

Mohamed S, Howard L, McInerney V, Hayat A, Krawczyk J, Naughton S, et al. Autologous bone marrow mesenchymal stromal cell therapy for "no-option" critical limb ischemia is limited by karyotype abnormalities. Cytotherapy 2020;22(6):313-21. CENTRAL [DOI: 10.1016/j.jcyt.2020.02.007]

Molavi 2016 {published data only}

Molavi B, Zafarghandi MR, Aminizadeh E, Hosseini SE, Mirzayi H, Arab L, et al. Safety and efficacy of repeated bone marrow mononuclear cell therapy in patients with critical limb ischemia in a pilot randomized controlled trial. Archives of Iranian Medicine 2016;19(6):388-96. CENTRAL

Murphy 2017 {published data only}

Murphy MP, Ross CB, Kibbe M, Kelso R, Sharafuddin M, Tzeng E, et al. Intramuscular injection of autologous bone marrow cells to prevent amputation in critical limb ischemia: the results of the phase III MOBILE trial. Abstracts of the 2017 Vascular Annual Meeting 2017;65(6 Suppl):131S–2S. CENTRAL

NCT00282646 {published data only}

NCT00282646. Safety and feasibility study of autologous bone marrow cell transplantation in patients with PAOD. clinicaltrials.gov/ct/show/NCT00282646 (first received 27 January 2006). CENTRAL

NCT00306085 {published data only}

NCT00306085. Autologous bone marrow cell treatment in peripheral atherosclerosis. clinicaltrials.gov/ct2/show/NCT00306085 (first received 22 March 2006). CENTRAL

NCT00498069 {published data only}

NCT00498069. Study of autologous bone marrow concentrate for the treatment of CLI. clinicaltrials.gov/ct2/show/NCT00498069 (first received 9 July 2007). CENTRAL

NCT00539266 {published data only}NCT00539266

NCT00539266. Autologous bone marrow-derived mononuclear cells for therapeutic arteriogenesis in patients with limb ischemia (ABC). clinicaltrials.gov/ct2/show/NCT00539266 (first received 4 October 2007). CENTRAL

NCT00595257 {published data only}

NCT00595257. Feasability study of autologous bone marrow aspirate concentrate for treatment of CLI. clinicaltrials.gov/ct2/show/NCT00595257 (first received 16 January 2008). CENTRAL

NCT00616980 {published data only}NCT00616980

NCT00616980. Injection of autologous CD34-positive cells for critical limb ischemia (ACT34-CLI). clinicaltrials.gov/ct2/show/NCT00616980 (first received 15 February 2008). CENTRAL

NCT00913900 {published data only}NCT00913900

NCT00913900. Safety study of adult stem cells to treat patients with severe leg artery disease (SCRIPT-CLI). clinicaltrials.gov/ct2/show/NCT00913900 (first received 4 June 2009). CENTRAL

NCT00922389 {published data only}

NCT00922389. A clinical trial on diabetic foot using peripheral blood derived stem cells for treating critical limb ischemia. clinicaltrials.gov/ct2/show/NCT00922389 (first received 17 June 2009). CENTRAL

NCT00955669 {published data only}

NCT00955669. Comparison of autologous mesenchymal stem cells and mononuclear cells on diabetic critical limb ischemia and foot ulcer. clinicaltrials.gov/ct2/show/NCT00955669 (first received 10 August 2009). CENTRAL

NCT01049919 {published data only}

NCT01049919. Safety and efficacy study of autologous concentrated bone marrow aspirate (cBMA) for critical limb ischemia (CLI). clinicaltrials.gov/ct2/show/NCT01049919 (first received 15 January 2010). CENTRAL

NCT01245335 {published data only}

NCT01245335. Bone marrow aspirate concentrate (BMAC) for treatment of critical limb ischemia (CLI). clinicaltrials.gov/ct2/show/NCT01245335 (first received 22 November 2010). CENTRAL

NCT01584986 {published data only}

NCT01584986. Autologous angiogenic cell precursors (ACPs) for the treatment of peripheral artery disease. clinicaltrials.gov/ct2/show/NCT01584986 (first received 25 April 2012). CENTRAL

NCT02336646 {published data only}

NCT02336646. Cell therapy with mesenchymal stem cell in ischemic limb disease. clinicaltrials.gov/ct2/show/NCT02336646 (first received 13 January 2015). CENTRAL

NCT03174522 {published data only}

NCT03174522. The efficacy and safety of Rexmyelocel-T to treat ischemic ulcers in subjects with CLI Rutherford category 5 and DM. clinicaltrials.gov/ct2/show/NCT03174522 (first received 2 June 2017). CENTRAL

NCT03214887 {published data only}

NCT03214887. Autologous BMMNC combined with HA therapy for PAOD. clinicaltrials.gov/ct2/show/NCT03214887 (first received 12 July 2017). CENTRAL

NCT03304821 {published data only}

NCT03304821. Granulocyte-macrophage stimulating factor (GM-CSF) in peripheral arterial disease. clinicaltrials.gov/ct2/show/NCT03304821 (first received 9 October 2017). CENTRAL

NCT03339973 {published data only}

NCT03339973. Allogeneic ABCB5-positive stem cells for treatment of PAOD. clinicaltrials.gov/ct2/show/NCT03339973 (first received 13 November 2017). CENTRAL

Niven 2017 {published data only}

Niven MJ, Sivak G, Kafri E, Moshe M, Galili O, Frogel M, et al. Adult stem/progenitor cells as a personalised treatment for peripheral vascular disease. Diabetologia 2017;60:S31. CENTRAL

Ohtake 2017 {published data only}

Ohtake T, Mochida Y, Ishioka K, Oka M, Maesato K, Moriya H, et al. Effect of autologous G-CSF-mobilized CD34+ cell transplantation in hemodialysis patients with critical limb ischemia. Nephrology Dialysis Transplantation 2017;Suppl 3:iii309. CENTRAL

Pawan 2012 {published data only}

Pawan K, Anoop CH, Seetharam RN, Das AK, Majumdar AS, Pherwani A. A randomized, double blind, placebo controlled, single dose, phase I/II study using adult bone marrow derived allogeneic mesenchymal stem cells in patients with critical limb ischemia. In: XXVI Annual Meeting of the European Society for Vascular Surgery; 2012 Sep 19-21; Bologna, Italy. 2012:154. CENTRAL

Perin 2011 {published data only}

NCT00392509. ALDHbr cells for critical limb ischemia, randomized trial (CLI-001). clinicaltrials.gov/ct2/show/NCT00392509 (first received 26 October 2006). CENTRAL
Perin EC, Silva G, Gahremanpour A, Canales J, Zheng Y, Cabreira-Hansen MG, et al. A randomized, controlled study of autologous therapy with bone marrow-derived aldehyde dehydrogenase bright cells in patients with critical limb ischemia. Catheterization and Cardiovascular Interventions 2011;78(7):1060-7. CENTRAL

Perin 2017 {published data only}

Perin EC, Murphy M, Cooke JP, Moye L, Henry TD, Bettencourt J, et al. Rationale and design for PACE: patients with intermittent claudication injected with ALDH bright cells. American Heart Journal 2014;168(5):667–73. CENTRAL
Perin EC, Murphy MP, March KL, Bolli R, Loughran J, Yang PC, et al. Evaluation of cell therapy on exercise performance and limb perfusion in peripheral artery disease: the CCTRN PACE Trial (patients with intermittent claudication injected with ALDH bright cells). Circulation 2017;135(15):1417–28. CENTRAL

Poole 2013 {published data only}

Poole J, Mavromatis K, Binongo JN, Khan A, Li Q, Khayata M, et al. Effect of progenitor cell mobilization with granulocyte-macrophage colony-stimulating factor in patients with peripheral artery disease: a randomized clinical trial. JAMA 2013;310(24):2631–9. CENTRAL

Prochazka 2010 {published data only}

Prochazka V, Gumulec J, Jaluvka F, Salounova D, Jonszta T, Czerny D, et al. Cell therapy, a new standard in management of chronic critical limb ischemia and foot ulcer. Cell Transplantation 2010;19(11):1413-24. CENTRAL

PROVASA 2011 {published data only}

Walter D, Krankenberg H, Balzer J, Kalka C, Baumgartner I, Schluter M. Intra-arterial administration of bone marrow mononuclear cells in patients with critical limb ischemia - a randomized-start, placebo-controlled pilot trial (PROVASA). Vasa 2010;39:37. CENTRAL
Walter DH, Krankenberg H, Balzer JO, Kalka C, Baumgartner I, Schluter M, et al. Intraarterial administration of bone marrow mononuclear cells in patients with critical limb ischemia: a randomized-start, placebo-controlled pilot trial (PROVASA). Circulation: Cardiovascular Interventions 2011;4(1):26-37. CENTRAL

RESTORE‐CLI 2012 {published data only}NCT00468000

NCT00468000. Use of vascular repair cells (VRC) in patients with peripheral arterial disease to treat critical limb ischemia (RESTORE-CLI). clinicaltrials.gov/ct2/show/NCT00468000 (first received 1 May 2007). CENTRAL
Powell RJ, Marston WA, Berceli SA, Guzman R, Henry TD, Longcore AT, et al. Cellular therapy with Ixmyelocel-T to treat critical limb ischemia: the randomized, double-blind, placebo-controlled RESTORE-CLI trial. Molecular Therapy: the Journal of the American Society of Gene Therapy 2012;20(6):1280-6. CENTRAL
Powell RJC. Interim analysis results from the RESTORE-CLI, a randomized, double-blind multicenter phase II trial comparing expanded autologous bone marrow-derived tissue repair cells and placebo in patients with critical limb ischemia. Journal of Vascular Surgery 2011;54(4):1032-41. CENTRAL

Sharma 2021 {published data only}

Sharma S, Pandey NN, Sinha M, Kumar S, Jagia P, Gulati GS, et al. Randomized, double-blind, placebo-controlled trial to evaluate safety and therapeutic efficacy of angiogenesis induced by intraarterial autologous bone marrow–derived stem cells in patients with severe peripheral arterial disease. Journal of Vascular and Interventional Radiology 2021;32(2):157-63. CENTRAL

Skóra 2015 {published data only}

Skóra J, Pupka A, Janczak D, Barć P, Dawiskiba T, Korta K, et al. Combined autologous bone marrow mononuclear cell and gene therapy as the last resort for patients with critical limb ischemia. Archives of Medical Science 2015;11(2):325-31. CENTRAL

Subramaniyam 2009 {published data only}

Subramaniyam V, Reddy U, Harris W, Sutcliffe D. Safety and efficacy of bone marrow mobilization with granulocyte-macrophage colony stimulating factor in patients with intermittent claudication. In: American College of Cardiology 55th Annual Scientific Session; 2006 Mar 11–14; Atlanta, GA. 2006. CENTRAL
Subramaniyam V, Waller EK, Murrow JR, Manatunga A, Lonial S, Kasirajan K, et al. Bone marrow mobilization with granulocyte macrophage colony-stimulating factor improves endothelial dysfunction and exercise capacity in patients with peripheral arterial disease. American Heart Journal 2009;158(1):53-60. CENTRAL

Szabo 2013 {published data only}

Szabo GV, Kovesd Z, Cserepes J, Daroczy J, Belkin M, Acsady G. Peripheral blood-derived autologous stem cell therapy for the treatment of patients with late-stage peripheral artery disease - results of the short- and long-term follow-up. Cytotherapy 2013;15(10):1245-52. CENTRAL

Teraa 2015 {published data only}

Sprengers RW, Moll FL, Teraa M, Verhaar MC. Rationale and design of the JUVENTAS trial for repeated intra arterial infusion of autologous bone marrow-derived mononuclear cells in patients with critical limb ischemia. Journal of Vascular Surgery 2010;51(6):1564–8. CENTRAL
Teraa M, Sprengers RW, Schutgens RE, Slaper-Cortenbach IC, van der Graaf Y, Algra A, et al. Effect of repetitive intra-arterial infusion of bone marrow mononuclear cells in patients with no-option limb ischemia: the randomized, double-blind, placebo-controlled rejuvenating endothelial progenitor cells via transcutaneous intra-arterial supplementation (JUVENTAS) trial. Circulation 2015;131(10):851-60. CENTRAL

Tournois 2015 {published data only}

Tournois C, Pignon B, Sevestre MA, Djerada Z, Capiod JC, Poitevin G, et al. Critical limb ischemia: thrombogenic evaluation of two autologous cell therapy products and biologic profile in treated patients. Transfusion 2015;55(11):2692–701. CENTRAL

Walter 2011 {published data only}

Walter D, Krankenberg H, Balzer J, Kalka C, Baumgartner I, Schluter M. Intra-arterial administration of bone marrow mononuclear cells in patients with critical limb ischemia - a randomized-start, placebo-controlled pilot trial (PROVASA). Vasa 2010;39:37. CENTRAL
Walter DH, Krankenberg H, Balzer JO, Kalka C, Baumgartner I, Schlüter M, et al. Intraarterial administration of bone marrow mononuclear cells in patients with critical limb ischemia a randomized-start, placebo-controlled pilot trial (PROVASA). Circulation: Cardiovascular Interventions 2011;4(1):26-37. CENTRAL

Wang 2014 {published data only}

Wang X, Jiang L, Wang X, Yin F, Li G, Feng X, et al. Combination of autologous transplantation of G-CSF mobilized peripheral blood mononuclear cells and Panaxnotoginseng saponins in the treatment of unreconstructable critical limb ischemia. Annals of Vascular Surgery 2014;28(6):1501–12. CENTRAL

Wang 2017 {published data only}

Wang SK, Green L, Babbey C, Wilson M, Motaganahalli R, Fajardo A, et al. Ethnic minorities with critical limb ischemia derive equal amputation risk reduction from autologous cell therapy compared to Caucasians. Journal of Vascular Surgery 2017;65(6):113S. CENTRAL
Wang SK, Green LA, Motaganahalli RL, Wilson MG, Fajardo A, Murphy MP. Rationale and design of the MarrowStim PAD Kit for the treatment of critical limb ischemia in subjects with severe peripheral arterial disease (MOBILE) trial investigating autologous bone marrow cell therapy for critical limb ischemia. Journal of Vascular Surgery 2017;65(6):1850–7.e2. CENTRAL

Wang 2018 {published data only}

Wang SK, Green LA, Drucker NA, Motaganahalli RL, Fajardo A, Murphy MP. Rationale and design of the clinical and histologic analysis of mesenchymal stromal cells in amPutations (CHAMP) trial investigating the therapeutic mechanism of mesenchymal stromal cells in the treatment of critical limb ischemia. Journal of Vascular Surgery 2018;68(1):176–81.e1. CENTRAL

Wijnand 2018 {published data only}

NCT03042572. Allogeneic mesenchymal stromal cells for angiogenesis and neovascularization in no-option ischemic limbs (SAIL). clinicaltrials.gov/ct2/show/NCT03042572 (first received 3 February 2017). CENTRAL
Wijnand JGJ, Teraa M, Gremmels H, van Rhijn-Brouwer FCC, de Borst GJ, Verhaar MC. Rationale and design of the SAIL trial for intramuscular injection of allogeneic mesenchymal stromal cells in no-option critical limb ischemia. Journal of Vascular Surgery 2018;67(2):656-61. CENTRAL

Zafarghandi 2010 {published data only}

Zafarghandi MR, Fazel AP, Baharvand H. Safety and efficacy of granulocyte colony-stimulating factor administration following autologous intramuscular implantation of bone marrow mononuclear cells: a randomized controlled trial in patients with advanced lower limb ischemia. Cytotherapy 2010;12:783-91. CENTRAL

Zhang 2010 {published data only}

Zhang HFZ. Endovascular transplantation of autologous bone marrow stem cells for the treatment of diabetic lower limb arterial occlusion. Journal of Clinical Rehabilitative Tissue Engineering Research 2010;14:6040-3. CENTRAL

Zhao 2008 {published data only}

Zhao ZG, Yuan HJ, Zhang HF, Zhang CL, Wang YF, Ma SP, et al. Combined transplantation of autologous peripheral blood and bone narrow stem cells for the treatment of diabetic lower limb ischaemia: randomized controlled trial. Journal of Clinical Rehabilitative Tissue Engineering Research 2008;12(8):1464-6. CENTRAL

Zhou 2017a {published data only}

Zhou HM, Liu F, Yang AG, Guo YQ, Zhou YR, Gu YQ, et al. Efficacy, safety and influencing factors of intra-calf muscular injection of bone marrow mononuclear cells in the treatment of type 2 diabetes mellitus-induced lower extremity vascular disease. Experimental and Therapeutic Medicine 2017;14(5):5177–85. CENTRAL

Zhou 2017b {published data only}

Zhou CH, Xu LL, Hao XX, Sun XJ, Guo MJ, Liu B. Autologous CD34+ cell transplantation promotes angiogenesis in older adult patients with atherosclerotic ischemia: study protocol for a prospective, single-center, open label, randomized controlled clinical trial. Chinese Journal of Tissue Engineering Research 2017;21(13):1998–2002. CENTRAL

NCT00753025 {published data only}

NCT00753025. Autologous bone marrow for lower extremity ischemia treating. clinicaltrials.gov/ct2/show/study/NCT00753025 (first received 16 September 2008). CENTRAL

NCT01446055 {published data only}

NCT01446055. Safety and efficacy study of autologous BM-MNC processed by two methods for treating patients with chronic limb ischemia. clinicaltrials.gov/ct2/show/study/NCT01446055 (first received 4 October 2011). CENTRAL

NCT02454231 {published data only}

NCT02454231. Monocentric trial: stem cell emergency life threatening limbs arteriopathy (SCELTA). clinicaltrials.gov/ct2/show/NCT02454231 (first received 27 May 2015). CENTRAL

Abdul Wahid 2018

Abdul Wahid SF, Ismail NA, Wan Jamaludin WF, Muhamad NA, Abdul Hamid MKA, Harunarashid H, et al. Autologous cells derived from different sources and administered using different regimens for 'no-option' critical lower limb ischaemia patients. Cochrane Database of Systematic Reviews 2018, Issue 8. Art. No: CD010747. [DOI: 10.1002/14651858.CD010747.pub2]

Franz 2009

Franz RW, Parks A, Shah KJ, Hankins T, Hartman JF, Wright ML. Use of autologous bone marrow mononuclear cell implantation therapy as a limb salvage procedure in patients with severe peripheral arterial disease. Journal of Vascular Surgery 2009;50(6):1378-90.

GRADE 2004

GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328:1490-4.

GRADEpro GDT [Computer program]

GRADEpro GDT. Version accessed 2 December 2021. Hamilton (ON): McMaster University (developed by Evidence Prime). Available at gradepro.org.

Higashi 2004

Higashi Y, Kimura M, Hara K, Noma K, Jitsuiki D, Nakagawa K, et al. Autologous bone-marrow mononuclear cell implantation improves endothelium-dependent vasodilation in patients with limb ischemia. Circulation 2004;109(10):1215-8.

Higgins 2011

Higgins JT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from training.cochrane.org/handbook/archive/v5.1/.

Higgins 2017

Higgins JP, Altman DG, Sterne JA, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Churchill R, Chandler J, Cumpston MS, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.2.0 (updated June 2017). Cochrane, 2017. Available from training.cochrane.org/handbook/archive/v5.2.

Higgins 2021

Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook/archive/v6.2.

Hirsch 2006

Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL, et al. ACC/AHA 2005 Practice Guidelines for the management of patients with peripheral arterial disease (lower extremity, renal, mesenteric, and abdominal aortic): a collaborative report from the American Association for Vascular Surgery/Society for Vascular Surgery, Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, Society of Interventional Radiology, and theACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease): endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease Foundation. Circulation 2006;113(11):e463-654.

Huang 2004

Huang PP, Li SZ, Han MZ, Xiao ZJ, Yang RC, Qiu LG, et al. Autologous transplantation of peripheral blood stem cells as effective therapeutic approach for severe arteriosclerosis obliterans of lower extremities. Thrombosis and Haemostasis 2004;91(3):606-9.

Huang 2005b

Huang P, Li S, Han M, Xiao Z, Yang R, Han ZC. Autologous transplantation of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells improves critical limb ischemia in diabetes. Diabetes Care 2005;28(9):2155-60.

Iso 2010

Iso Y, Soda T, Sato T, Sato R, Kusuyama T, Omori Y, et al. Impact of implanted bone marrow progenitor cell composition on limb salvage after cell implantation in patients with critical limb ischemia. Atherosclerosis 2010;209(1):167-72.

Lara‐Hernandez 2010

Lara-Hernandez R, Lozano-Vilardell P, Blanes P, Torreguitart-Mirada N, Galmés A, Besalduch J. Safety and efficacy of therapeutic angiogenesis as a novel treatment in patients with critical limb ischemia. Annals of Vascular Surgery 2010;24(2):287-94.

Lefebvre 2021

Lefebvre C, Glanville J, Briscoe S, Littlewood A, Marshall C, Metzendorf M-I, et al. Chapter 4: Searching for and selecting studies. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook/archive/v6.2.

Liu 2012

Liu FP, Dong JJ, Sun SJ, Gao WY, Zhang ZW, Zhou XJ, et al. Autologous bone marrow stem cell transplantation in critical limb ischemia: a meta-analysis of randomized controlled trials. Chinese Medical Journal 2012;125(23):4296–300.

Liu 2015

Liu Y, Xu Y, Fang F, Zhang J, Guo L, Weng Z. Therapeutic efficacy of stem cell-based therapy in peripheral arterial disease: a meta-analysis. PLOS ONE 2015;10(4):e0125032.

Norgren 2007

Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FGR, et al. Inter-society consensus for the management of peripheral arterial disease (TASC II). Journal of Vascular Surgery 2007;45 Suppl 1:5–67.

Rigato 2017

Rigato M, Monami M, Fadini GP. Autologous cell therapy for peripheral arterial disease: systematic review and meta-analysis of randomized, non-randomized, and non-controlled studies. Circulation Research 2017;120(8):1326-40.

Schünemann 2021

Schünemann HJ, Higgins JP, Vist GE, Glasziou P, Akl EA, Skoetz N, et al. Chapter 14: Completing ‘Summary of findings’ tables and grading the certainty of the evidence. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook/archive/v6.2.

Shintani 2001

Shintani S, Murohara T, Ikeda H. Augmentation of postnatal neovascularization with autologous bone marrow transplantation. Circulation 2001;103(6):897–903.

Takahashi 2006

Takahashi M, Li TS, Suzuki R, Kobayashi T, Ito H, Ikeda Y, et al. Cytokines produced by bone marrow cells can contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury. American Journal of Physiology-Heart and Circulatory Physiology 2006;291(2):H886-93.

Tateishi‐Yuyama 2002

Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, et al. Therapeutic angiogenesis for patients with limb ischemia by autologous transplantation of bone-marrow cells: a pilot study and a randomized controlled trial. Lancet 2002;360(9331):427-35.

Teraa 2013

Teraa M, Sprengers RW, van der Graaf Y, Peters CE, Moll FL, Verhaar MC. Autologous bone marrow-derived cell therapy in patients with critical limb ischemia: a meta analysis of randomized controlled clinical trials. Annals of Surgery 2013;258(6):922-9.

Wen 2011

Wen Y, Meng L, Gao Q. Autologous bone marrow cell therapy for patients with peripheral arterial disease: a meta analysis of randomized controlled trials. Expert Opinion on Biological Therapy 2011;11(12):1581-9.

Referencias de otras versiones publicadas de esta revisión

Moazzami 2011

Moazzami K, Majdzadeh R, Nedjat S. Local intramuscular transplantation of autologous mononuclear cells for critical lower limb ischaemia. Cochrane Database of Systematic Reviews 2011, Issue 12. Art. No: CD008347. [DOI: 10.1002/14651858.CD008347.pub2]

Moazzami 2014

Moazzami K, Moazzami B, Roohi A, Nedjat S, Dolmatova E. Local intramuscular transplantation of autologous mononuclear cells for critical lower limb ischaemia. Cochrane Database of Systematic Reviews 2014, Issue 12. Art. No: CD008347. [DOI: 10.1002/14651858.CD008347.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Barc 2006

Study characteristics

Methods

Study design: stated as randomised
Method of randomisation: not stated
Blinding: not stated
Exclusions postrandomisation: not stated
Losses to follow‐up: not stated

Participants

Country: Poland
Participants: 29 randomised
Mean age: not stated
Sex: not stated
Inclusion criteria:

  • patients with CLI with risk of amputation

  • presence of rest pain and/or necrosis lasting > 12 weeks

  • no progress after 8 weeks of conventional therapy

  • ABI < 0.5 in 2 independent examinations performed during the 7‐day interval pre‐inclusion

  • peripheral type of atherosclerosis and no possibility for operative therapy, confirmed in angiogram

Exclusion criteria:

  • age < 18 years

  • need for urgent amputation

  • reasons for ischaemia other than atherosclerosis

  • cancer

  • absence of conscious consent

  • lack of understanding of the idea of the therapy by patient

  • poor general condition (life expectancy < 6 months)

Interventions

Treatment group:

  • type of cells: BMMNC

  • cell isolation: not mentioned

  • dose of cells: not reported

  • route and frequency of delivery: multiple IM injections

Control group: not stated in the paper, but described by the authors as standard conservative therapy

Outcomes

Primary outcomes:

  • ABI

  • photographic documentation of ischaemic ulcerations and necrosis

  • subjective parameters (feeling of pain in VAS, QoL in WHO scale)

Secondary outcomes:

  • not stated

Outcome assessment points: baseline and months 1, 3, and 6

Funding

Not reported

Declarations of interest

Not reported

Notes

It is unclear who injects monocytes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study is described as randomised, but details are unclear.

Allocation concealment (selection bias)

Unclear risk

Not mentioned

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not mentioned

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not mentioned

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

All participants are accounted for.

Selective reporting (reporting bias)

High risk

The ABI and pain data were not reported in sufficient detail to include in the analysis.

Other bias

Low risk

We did not identify any other potential sources of bias.

Li 2013

Study characteristics

Methods

Study design: stated as randomised
Method of randomisation: not stated
Blinding: single‐blind
Exclusions postrandomisation: stated that 16 patients could not be evaluated for haemodynamic assessment because of extensive ulceration that made ankle pressure assessment not feasible
Losses to follow‐up: not stated

Participants

Country: China
Participants: 58 randomised
Mean age: 61 years in treatment group, 63 years in control group
Sex: male and female
Inclusion criteria:

  • evidence of CLI including rest pain and/or non‐healing ischaemic ulcers for a minimum of 4 weeks without improvement in response to conventional therapies

Exclusion criteria:

  • history of malignancy

  • evidence of possible malignancies after evaluation with carcinoembryonic antigen levels, chest radiographs, CT scans, and mammography in women or prostate examination in men

Interventions

Treatment group:

  • type of cells: BMMNC

  • cell isolation: cells were extracted from the posterior superior iliac spine. BMMNCs were isolated from the bone marrow by density gradient centrifugation with lymphocyte separating fluid. Cell counting was performed. Isolated BMMNCs were diluted into 50 to 120 mL suspensions to be transplanted.

  • dose of cells: 1 × 107 piece [sic]/mL BMMNC transplant

  • route and frequency of delivery: multiple IM injections

Control group: 0.9% sodium chloride (saline)

Outcomes

Primary outcomes:

  • safety assessments included adverse events, physical examination, cancer screening, ECG, blood chemistry, haematology, and urinalysis for fever, allergies, myocardial, stoke, malignancy, and death

Secondary outcomes:

  • haemodynamic improvement (an absolute increase of > 15% in the ABI)

  • skin ulcers improvement using photographic documentation of ischaemic ulcerations

  • pain score improvement using VAS

  • limb survival

Outcome assessment points: baseline and months 1, 3, and 6

Funding

Not reported

Declarations of interest

Not reported

Notes

It is unclear who injects monocytes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The study is described as randomised, but details are unclear.

Allocation concealment (selection bias)

Unclear risk

There was no information regarding allocation.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "This was a single blinded study aimed at analyzing the effect of..."

Comment: not stated who was blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "This was a single blinded study aimed at analyzing the effect of..."

Comment: not stated who was blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "Sixteen patients could not be evaluated for hemodynamic assessment because of extensive ulceration that made ankle pressure assessment not feasible" and "Fifteen patients were not evaluated for pain scores owing to minor surgical intervention just before the transplant or they could not understand the VAS scale"

Selective reporting (reporting bias)

Low risk

We did not identify any reporting bias.

Other bias

Low risk

We did not identify any other potential sources of bias.

Lindeman 2018

Study characteristics

Methods

Study design: stated as randomised
Method of randomisation: computer‐generated randomisations and allocation
Blinding: fully blinded
Exclusions postrandomisation: not stated
Losses to follow‐up: 1 participant in the placebo group

Participants

Country: the Netherlands
Participants: 54 randomised
Mean age: 58.5 years in treatment group, 57.8 years in control group
Sex: male and female
Inclusion criteria:

  • persistent disabling claudication (Fontaine’s stages IIb/III or Rutherford’s categories 3/4) or CLI (Fontaine’s stages IV or Rutherford’s categories 5/6) despite > 6 months optimal medical therapy (including exercise therapy, and revascularisation attempts in accordance with prevailing guidelines)

Exclusion criteria:

  • diabetes

  • immune suppressive therapy

  • age < 18 years

  • compromised life expectancy (anticipated survival < 1 year)

  • severe tissue loss or non‐manageable pain with an anticipated need for amputation within the first month of therapy

Interventions

Treatment group:

  • type of cells: BMMNC

  • cell isolation: BMMNCs were collected from multiple locations from the posterior iliac crests. The obtained bone marrow suspension was then filtered and concentrated to a final volume of 40 mL on the COBE Spectra Apheresis System (Gambro, Stockholm, Sweden) in the LUMC GMP‐facility without any further treatment.

  • dose of cells: 1.7 × 109 total

  • route and frequency of delivery: IM injections in 40 locations (1 mL per injection site)

Control group: diluted autologous peripheral blood, which was visually indistinguishable from the BMMNC

Outcomes

Primary outcomes:

  • PFWD or limb salvage

  • full wound recovery

Secondary outcomes:

  • ABI

  • SF‐36

  • Pain (BPI‐SF)

Time points for assessment: months 1, 6, and 12

Funding

Not reported

Declarations of interest

Not reported

Notes

It is unclear who injects monocytes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Computer randomization and allocation were done after bone marrow collection, by the stem laboratory in permuted blocks of size 4 in 2 strata (disabling claudication or CLI) and secured until all data entry was completed and the database was locked"

Allocation concealment (selection bias)

Low risk

Allocation was adequately concealed.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "All patients, clinicians, and trial investigators remained blinded for the treatment allocation until reaching the secondary 12 month end point."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "All patients, clinicians, and trial investigators remained blinded for the treatment allocation until reaching the secondary 12 month end point."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were analysed, except for 1 participant in the placebo group. Quote: "One patient in the placebo group was unable to comply with the follow‐up (language barrier), hence 25 patients could be evaluated."

Selective reporting (reporting bias)

Low risk

The clinical measures prespecified in the protocols were adequately reported in the results.

Other bias

Low risk

None suspected.

Pignon 2017

Study characteristics

Methods

Study design: stated as randomised
Method of randomisation: website‐generated randomisation
Blinding: double‐blind
Exclusions postrandomisation: not stated
Intention‐to‐treat analysis: used
Losses to follow‐up: 1 participant in placebo group and 1 participant in treatment group

Participants

Country: France
Participants: 38 randomised
Mean age: 72 years in treatment group, 65 years in control group
Sex: male and female
Inclusion criteria:

  • age ≥ 18 years

  • presented with atherosclerosis‐related CLI with no sign of improvement following previous appropriate medical treatment

Exclusion criteria:

  • Buerger’s disease

  • active infection

  • uncontrolled diabetes mellitus

  • prior history of neoplasm or malignancy

  • contraindication for general anaesthesia

  • prothrombin time < 50%

  • myocardial or brain infarction within 3 months

  • any medical condition contraindicating the initiation of anticoagulation

  • unexplained haematological abnormality

  • HIV

  • hepatitis B or C virus infection

  • any concomitant disease associated with a life expectancy of < 1 year

Interventions

Treatment group:

  • type of cells: BMMNC

  • cell isolation: 500 mL of bone marrow collected from the posterior iliac crests. 8 centres used a blood‐cell separator (COBE Spectra, version 4, Bone Marrow Processing Program, Gambro BCT, Lakewood, CO, USA); 2 centres used a blood‐cell separator requiring a Ficoll density‐gradient for isolation of the BMMNC.

  • dose of cells: 1.3 × 109 total (not clear in report)

  • route and frequency of delivery: IM injections (30 intramuscular injections of 1 mL)

Control group: 30 mL saline with 4 mL autologous peripheral blood

Outcomes

Primary outcomes:

  • major amputation rate

  • mortality

Secondary outcomes:

  • ABI

  • TcPO2

  • ulcers

  • pain

Time points for assessment: baseline, days 1, 3, 15, 28, and then months 6 and 12

Funding

The reported work was supported by the French Ministry of Health (Programme Hospitalier de Recherche Clinique 2007) and by the French Blood Agency.

Declarations of interest

Not reported

Notes

It is unclear who injects monocytes.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was performed by the local investigator in charge of the patient, through a dedicated website, with stratification by center."

Allocation concealment (selection bias)

Low risk

Allocation was adequately concealed.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The steering committee, the investigators, the observers, and the patients were unaware of the treatment allocation."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The steering committee, the investigators, the observers, and the patients were unaware of the treatment allocation."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 participants (1 in placebo group and 1 in treatment group) dropped out of study due to adverse events. Quote: "Therefore, the modified ITT analysis excluded these 2 patients and the analysis was conducted on 19 patients in the placebo group and 17 in the BMMNC group."

Selective reporting (reporting bias)

High risk

The data for ABI and pain were not reported in sufficient detail to include in the analysis.

Other bias

Low risk

None suspected.

ABI: ankle‐brachial index
BMMNC: bone marrow mononuclear cell
BPI‐SF: Brief Pain Inventory‐Short Form
CLI: critical limb ischaemia
CT: computed tomography
ECG: echocardiographs
IM: intramuscular
PFWD: pain‐free walking distance
QoL: quality of life
SF‐36: 36‐Item Short‐Form Health Survey
TcPO2: transcutaneous pressure of oxygen
VAS: visual analogue scale
VEGF: vascular endothelial growth factor
WHO: World Health Organization

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Benoit 2011

This study used bone marrow from the iliac crest, but after aspiration, BMMNC was processed into 40 mL of concentrate using the SmartPReP2 Bone Marrow Aspirate Concentrate (BMAC) system.

BONMOT 2008

The study is an ongoing RCT investigating the effects of autologous bone marrow concentrate, not BMMNCs.

Burt 2010

This study assessed the safety and feasibility of autologous CD133+ cells and included no control group.

Capiod 2009

The study compared the effect of transplantation of BMMNCs or G‐CSF‐mobilised PBMNCs, and included no control group.

Chen 2009

The study included people with diabetic foot not CLI, and participants received autologous bone marrow MSC, not mononuclear cells.

Debin 2008

The study investigated the effects of bone marrow MSC, not BMMNCs.

Dong 2013

The study investigated the effects of purified CD34+ cells, not BMMNCs.

Dong 2018

The study compared the effect of transplantation of PBMNCs or purified CD34+ cells, and included no control group.

Du 2017

The study assessed the clinical efficacy of implanted umbilical cord MSCs combined with bone marrow stem cells compared with implanted bone marrow stem cells for treatment of lower limb ischaemia.

Flugelman 2017

The study assessed venous endothelial cells combined with venous smooth muscle cells.

Frogel 2017

This is a pilot open‐label study of treatment progenitor cells derived from peripheral blood for people with CLI.

Gupta 2013

The study investigated the effects of allogeneic bone marrow‐derived MSC, not BMMNC.

Gurunathan 2009

The study is an ongoing RCT investigating the effects of autologous bone marrow concentrate, not BMMNCs.

Higashi 2010

The study investigated the combined effect of BMMNC implantation and sarpogrelate, a selective 5‐HT(2A) antagonist.

Holzinger 1994

The study investigated people with chronic skin ulcers caused by chronic arterial occlusive disease or venous post‐thrombotic syndrome, not CLI.

Horie 2018

This RCT evaluated the efficacy and safety of G‐CSF‐mobilised PBMNC transplantation in people with PAD.

Huang 2005a

The study investigated PBMNC, not BMMNC.

Iafrati 2011

The study investigated BMA concentrate, not BMMNC.

Iafrati 2016

The study investigated BMA concentrate, not BMMNC.

JUVENTAS 2008

The study is an ongoing RCT investigating the effects of IA injection of BMMNCs.

Kirana 2007

The study investigated people with diabetic foot ulcers, not CLI.

Kirana 2012

The study investigated people with diabetic foot ulcers, not CLI.

Klepanec 2012

The study compared IA and IM MSC therapy with no control group.

Korymasov 2009

The study is a randomised triple‐arm study investigating the safety and effectiveness of highly purified CD133+ autologous stem cells in CLI.

Lasala 2011

The study investigated the transplantation of an autologous bone marrow‐derived combination stem cell product.

Lu 2011

The study compared bone marrow MSC with BMMNCs with no control group.

Madaric 2011

The study compared IA and IM application of autologous BMC transplantation.

Majumdar 2015

This phase 2 study assessed allogeneic MSC in CLI.

Mohamed 2020

The study included bone marrow mesenchymal stromal cell therapy.

Molavi 2016

This RCT investigated the safety and efficacy of repeated BMMNC injections in comparison with a single BMMNC injection in CLI patients.

Murphy 2017

This double‐blinded, placebo‐controlled trial assessed the safety and efficacy of autologous concentrated BMA versus placebo.

NCT00282646

This is an ongoing RCT investigating the effects of IA injection of autologous BMMNCs versus placebo.

NCT00306085

The study investigated patients with peripheral atherosclerosis, not CLI.

NCT00498069

This is an ongoing RCT investigating the effects of BMCs, not BMMNCs.

NCT00539266

This is an ongoing RCT investigating the effects of BMCs, not BMMNCs.

NCT00595257

This is an ongoing RCT investigating the effects of autologous bone marrow concentrate, not BMMNCs.

NCT00616980

This is an ongoing RCT investigating the effects of peripheral blood‐derived stem cells, not BMMNCs.

NCT00913900

The study is an ongoing RCT investigating the effects of adult stem cells.

NCT00922389

This is an ongoing RCT investigating the effects of CD34 positive cells, not BMMNCs.

NCT00955669

This is an ongoing RCT comparing autologous MSC and mononuclear cells, not BMMNCs.

NCT01049919

This is an ongoing RCT investigating the effects of autologous concentrated BMA, not BMMNCs.

NCT01245335

This is an ongoing RCT investigating the effects of BMA concentrate, not BMMNCs.

NCT01584986

This RCT assessed the safety and efficacy of autologous ACPs compared with SMT.

NCT02336646

This RCT compared allogeneic MSCs versus placebo in people with CLI.

NCT03174522

This clinical trial compared Rexmyelocel‐T versus placebo in people with CLI.

NCT03214887

This clinical trial investigated the safety and efficacy of hyaluronan combined with BMMNCs for PAD.

NCT03304821

This double‐blind, placebo‐controlled RCT examined whether 3 weeks of 3‐times‐a week injection of GM‐CSF would improve measures of ischaemia in people with IC compared with placebo.

NCT03339973

This clinical trial is investigating the efficacy and safety of 1 dose of allo‐APZ2‐PAOD administered IM into the affected lower leg of people with PAD.

Niven 2017

This pilot open‐label study examined treatment with progenitor cells for PAD.

Ohtake 2017

This prospective phase 1/2 interventional clinical trial examined autologous G‐CSF‐mobilised CD34+ cell transplantation in haemodialysis patients with CLI.

Pawan 2012

The study investigated adult bone marrow‐derived allogeneic MSC.

Perin 2011

The study investigated autologous therapy with bone marrow‐derived aldehyde dehydrogenase bright cells. These cells are derived from CD15 and glycophorin A‐expressing cells from autologous bone marrow via immunomagnetic beads and cell sorters.

Perin 2017

The study investigated autologous therapy with bone marrow‐derived aldehyde dehydrogenase bright cells. These cells are derived from CD15 and glycophorin A‐expressing cells from autologous bone marrow via immunomagnetic beads and cell sorters.

Poole 2013

This trial investigated the effects of GM‐CSF, not cell therapy, in people with IC.

Prochazka 2010

This RCT investigated major limb amputation in participants given autologous BMC compared to those given standard care for CLI and foot ulcer.

PROVASA 2011

This is a multicentre, phase II, double‐blind RCT comparing IA administration of BMMNC or placebo followed by active treatment with BMMNC (open‐label) after 3 months.

RESTORE‐CLI 2012

This is an ongoing RCT investigating the effects of vascular repair cells, not BMMNCs.

Sharma 2021

In this study, BMMNC was injected IA, not IM.

Skóra 2015

This study investigated autologous BMMNC plus VEGF, not BMMNC alone.

Subramaniyam 2009

The study investigated patients with IC, not CLI.

Szabo 2013

The study investigated the effects of autologous stem cell therapy, not BMMNCs.

Teraa 2015

This double‐blind, placebo‐controlled RCT compared BMMNCs versus placebo in people with limb ischaemia, which was administered IA.

Tournois 2015

This phase 1 and 2 clinical trial examined BMC therapy products and cell therapy products obtained by cytapheresis (peripheral blood‐cell therapy products).

Walter 2011

The study compared the effect of IA route of transplantation.

Wang 2014

This trial assessed the efficacy and safety of the combination of peripheral blood mononuclear cells and Panax notoginseng saponins for treatment of CLI.

Wang 2017

This multicentre, double‐blind, placebo‐controlled RCT assessed the efficacy of IM injections of concentrated BMA for promoting amputation‐free survival in people with poor‐option CLI.

Wang 2018

This study compared IM injections of allogeneic MSCs or autogenous concentrated BMA.

Wijnand 2018

This study used allogeneic MSC for CLI.

Zafarghandi 2010

The study compared the effect of transplantation of BMMNCs with G‐CSF‐mobilised PBMNCs and included no control group.

Zhang 2010

The study only included people with diabetes.

Zhao 2008

The study investigated the effect of combined PBMNCs and BMMNCs injection versus placebo.

Zhou 2017a

This open, parallel‐control RCT compared BMMNC with SMT.

Zhou 2017b

This prospective, single‐centre, open‐label RCT compared peripheral blood CD34+ cells transfected with VEGF‐165 versus SMT.

ACPs: angiogenic cell precursors
BMA: bone marrow aspirate
BMC: bone marrow cell
BMMNC: bone marrow mononuclear cell
CLI: critical limb ischaemia
G‐CSF: granulocyte colony‐stimulating factor
GM‐CSF: granulocyte‐macrophage colony‐stimulating factor
IA: intra‐arterial
IC: intermittent claudication
IM: intramuscular
MSC: mesenchymal stem cells
PAD: peripheral arterial disease
PBMNC: peripheral blood‐derived mononuclear cell
RCT: randomised controlled trial
SMT: standard medical treatment
VEGF: vascular endothelial growth factor

Characteristics of ongoing studies [ordered by study ID]

NCT00753025

Study name

Autologous bone marrow for lower extremity ischaemia treating

Methods

Allocation: randomised
Endpoint classification: safety/efficacy study
Intervention model: parallel assignment
Masking: quadruple (participant, care provider, investigator, outcomes assessor)
Primary purpose: treatment

Participants

Inclusion criteria:

  • obliterating lower extremity atherosclerosis IIB stage (on Fontaine classification)

  • PFWD of 10 to 50 m

  • pulse absence on dorsalis pedis, tibialis posterior, poplitea

  • absence of ischaemia in rest and necrotic changes

  • mainly distal form of disease (lesion of a superficial femoral artery, a popliteal artery, anticnemion (anterior edge of the tibia) arteries) according to an angiography that testifies to impossibility of reconstructive operation performance

  • after lumbar sympathectomy and tibial bone osteoperforations executed previously

  • heavy smokers

Exclusion criteria:

  • insulin‐dependent diabetes

  • myocardial infarction or stroke within the past year

  • idiopathic hypertension III stage

  • anaemia and other diseases of blood

  • decompensation of chronic diseases that are contraindications to any surgical operation

  • HIV infection

  • A virus hepatitis

  • oncological disease

  • prior history of chemotherapy

Interventions

Biological: injection of isolated CD133+ cells

Outcomes

Primary outcome measure: increase in PFWD

Starting date

September 2008

Contact information

Andrey Toropovskiy, Clinical Center of Cellular Technologies, Russia

Notes

Primary completion date: September 2008

NCT01446055

Study name

Safety and efficacy study of autologous BMMNC processed by two methods for treating patients with CLI

Methods

Allocation: randomised
Endpoint classification: safety/efficacy study
Intervention model: parallel assignment
Masking: single‐blind (participant)
Primary purpose: treatment

Participants

Inclusion criteria:

  • Fontaine stage 2 to 4 or resting ABI < 0.7

  • age between 20 and 80 years

  • signed informed consent, voluntary participants

  • diagnosis of lower extremity arterial occlusive disease, or diabetic lower limb ischaemia, or Buerger’s disease

Exclusion criteria:

  • poorly controlled diabetes (glycated haemoglobin > 7.0%) and proliferative retinopathy (III to IV stage)

  • malignancy history in the past 5 years or serum level of tumour markers elevated more than doubled

  • severe heart, liver, kidney, respiratory failure or poor general condition and cannot tolerate BMMNC implantation

  • serious infections (such as cellulitis, osteomyelitis, etc.) or gangrene such that a major amputation cannot be avoided

  • aortic or iliac or common femoral artery occlusion

  • pregnant female, or reproductive age female, who wants to give birth throughout the course of the study

  • life expectancy less than 1 year

Interventions

Experimental: autologous BMMNC is enriched with ResQ process (an automatic cell separator). Then the cell product is implanted into the ischaemic limbs of a participant.
Active comparator: a conventional method based on Ficoll cell separation is used to process bone marrow

Outcomes

Primary outcomes:

  • cell treatment‐related adverse event: temperature, pulse, respiration, blood pressure, routine analysis of blood and urine, liver function (ALT, AST), renal function (blood urea nitrogen, creatinine), function of coagulation (APTT, prothrombin time, fibrinogen, thrombin time), ECG, local inflammatory response, cell treatment‐related death, cell treatment‐related unexpected amputation

Secondary outcomes:

  • ulcer size

  • rest pain score

  • cold sensation score

  • claudication distance (m)

  • resting ABI

  • resting TcO (mmHg)

  • collateral vessel score

  • amputation rate

  • skin microcirculation measurement

  • resting TBI

Starting date

October 2011

Contact information

Contact: Yongquan Gu, MD, Xuanwu Hospital, Beijing; gu‐[email protected]

Notes

The recruitment status of this study is unknown because the information has not been verified recently on ClinicalTrials.gov.

NCT02454231

Study name

Monocentric trial: stem cell emergency life threatening limbs arteriopathy (SCELTA)

Methods

Allocation: randomised
Endpoint classification: safety/efficacy study
Intervention model: parallel assignment
Masking: open‐label
Primary purpose: treatment

Participants

Inclusion criteria:

  • men and women older than 40 years of age with a diagnosis of CLI due to atherosclerosis of the lower extremities, as defined by the presence of persistent rest pain requiring systemic and continued analgesic treatment in the past 15 days and/or the presence of trophic lesions imputable to the occluding arteriopathy, ABI < 0.40 (with systolic ankle pressure < 50 to 70 mmHg), TBI < 0.40 (with big toe systolic pressure < 30 to 50 mmHg), and TcO < 30 mmHg

  • eligible for treatment and enrolled only after demonstration that intravascular or surgical revascularisation was not possible, as revealed by echography and angio‐CAT, or when the patient refused to undergo surgical treatments and after written informed consent was obtained

Exclusion criteria:

  • age < 40

  • not atherosclerotic CLI

  • myocardial infarction occurrence within 6 months

  • cardiac failure of III‐IV class NYHA

  • ejection fraction < 40%

  • arterial hypertension (> 160/100 mmHg) uncontrolled despite usage of 2 antihypertensive drugs

  • presence of current or chronic severe infectious disease

  • osteomyelitis

  • diabetes with glycated haemoglobin > 7.5

  • proliferative diabetic retinopathy

  • haemorrhagic disorders

  • non‐atherosclerotic arteriopathy

  • chronic airway insufficiency (pO2 < 65 mmHg, pCO2 > 0.50 mmHg)

  • renal failure (creatinine > 2 mg/dL)

  • contraindications or intolerance to contrast media for radiological imaging

Interventions

Experimental: peripheral blood EPC injection
Active comparator: BMMNC injection

Outcomes

Primary outcomes:

  • safety as measured by evaluation of any adverse event temporarily correlated with treatment

  • changes in ischaemic leg perfusion from baseline

Secondary outcomes:

  • improvement in mean values of TcO

  • improvement in mean values of ABI

  • improvement in vessel anatomical status

  • improvement in leg perfusion

  • improvement in vessel anatomical status

  • quality of life improvement

  • improvement in rest pain

  • improvement in trophic limb lesions

  • reduction in number of major amputations

  • improvement in microvascular anatomy

Starting date

September 2009

Contact information

Contacts:

Enrico Maggi, professor, University of Florence, Italy; [email protected]
Francesco Annunziato, professor; [email protected]

Notes

Primary completion date: May 2015

ABI: ankle‐brachial index
Angio‐CAT: computerised tomography (CT) coronary angiogram
APTT: activated partial thromboplastin time
ALT: alanine aminotransferase
AST: aspartate aminotransferase
BMI: body mass index
BMMNC: bone marrow mononuclear cells
CLI: critical limb ischaemia
DVT: deep vein thrombosis
ECG: electrocardiography
EPC: endothelial progenitor cells
G‐CSF: granulocyte colony‐stimulating factor
IC: intermittent claudication
IM: intramuscular
NYHA: New York Heart Association
PAD: peripheral arterial disease
pCO2: partial pressure of carbon dioxide
PB‐MNC: peripheral blood mononuclear cells
PFWD: pain‐free walking distance
pO2: partial pressure of oxygen
TASC: TransAtlantic Intersociety Consensus
TBI: toe brachial index
TcO: transcutaneous oxygen pressure

Data and analyses

Open in table viewer
Comparison 1. BMMNC versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 All‐cause mortality Show forest plot

3

123

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.15, 6.63]

Analysis 1.1

Comparison 1: BMMNC versus control, Outcome 1: All‐cause mortality

Comparison 1: BMMNC versus control, Outcome 1: All‐cause mortality

1.2 Amputation Show forest plot

4

176

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.27, 0.99]

Analysis 1.2

Comparison 1: BMMNC versus control, Outcome 2: Amputation

Comparison 1: BMMNC versus control, Outcome 2: Amputation

1.3 Amputation ‐ sensitivity analysis Show forest plot

2

89

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.19, 1.39]

Analysis 1.3

Comparison 1: BMMNC versus control, Outcome 3: Amputation ‐ sensitivity analysis

Comparison 1: BMMNC versus control, Outcome 3: Amputation ‐ sensitivity analysis

1.4 Side effects and complications Show forest plot

4

176

Risk Ratio (M‐H, Random, 95% CI)

2.13 [0.50, 8.97]

Analysis 1.4

Comparison 1: BMMNC versus control, Outcome 4: Side effects and complications

Comparison 1: BMMNC versus control, Outcome 4: Side effects and complications

1.5 Side effects and complications ‐ sensitivity analysis Show forest plot

2

89

Risk Ratio (M‐H, Random, 95% CI)

2.69 [0.11, 63.18]

Analysis 1.5

Comparison 1: BMMNC versus control, Outcome 5: Side effects and complications ‐ sensitivity analysis

Comparison 1: BMMNC versus control, Outcome 5: Side effects and complications ‐ sensitivity analysis

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1: BMMNC versus control, Outcome 1: All‐cause mortality

Figuras y tablas -
Analysis 1.1

Comparison 1: BMMNC versus control, Outcome 1: All‐cause mortality

Comparison 1: BMMNC versus control, Outcome 2: Amputation

Figuras y tablas -
Analysis 1.2

Comparison 1: BMMNC versus control, Outcome 2: Amputation

Comparison 1: BMMNC versus control, Outcome 3: Amputation ‐ sensitivity analysis

Figuras y tablas -
Analysis 1.3

Comparison 1: BMMNC versus control, Outcome 3: Amputation ‐ sensitivity analysis

Comparison 1: BMMNC versus control, Outcome 4: Side effects and complications

Figuras y tablas -
Analysis 1.4

Comparison 1: BMMNC versus control, Outcome 4: Side effects and complications

Comparison 1: BMMNC versus control, Outcome 5: Side effects and complications ‐ sensitivity analysis

Figuras y tablas -
Analysis 1.5

Comparison 1: BMMNC versus control, Outcome 5: Side effects and complications ‐ sensitivity analysis

Summary of findings 1. Intramuscular injection of bone marrow mononuclear cells (BMMNC) compared to control for treatment of critical lower limb ischaemia (CLI)

Intramuscular injection of bone marrow mononuclear cells (BMMNC) compared to control for treatment of critical lower limb ischaemia (CLI)

Patient or population: people with CLI who were not suitable for revascularisation and showed no improvement in response to the best standard therapy
Setting: hospital
Intervention: BMMNCa
Comparison: controlb

Outcomes

Anticipated absolute effects (95% CI)*

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control

Risk with BMMNC

All‐cause mortality

(follow‐up: 6 or 12 months)

Study population

RR 1.00 (0.15 to 6.63)

123

(3 RCTs)

⊕⊝⊝⊝
VERY

LOWc

No deaths were reported for 2 studies (Barc 2006; Pignon 2017). Li 2013 reported 4 deaths (2 in each group) during 6 months follow‐up; these deaths were not considered to be related to treatment. Information regarding mortality was not available in 1 study (Lindeman 2018).

32 per 1000

32 per 1000

(5 to 210)

Reduction in pain

Various scales including VAS, PISQ; scales ranged from 0 to 4/10/100, where 0 = no pain

(follow‐up: 3 to 12 months)

All studies reported on pain, but due to different measurements and incomplete information we were unable to pool the data. See comment

176
(4 RCTs)

⊕⊝⊝⊝
VERY

LOWd

Barc 2006 assessed pain with VAS, where 0 was no pain at all and 10 was the most severe pain experienced. Pain levels decreased in both BMMNC and control groups.

Li 2013 assessed pain with VAS, but reported the improvement of pain defined as a > 50% decrease in VAS during study follow‐up. Study authors reported a greater reduction in pain in the BMMNC group (P = 0.045).

Lindeman 2018 used the PISQ, and reported that no differences were observed in average pain reduction between BMMNC and control group (P = 0.23).

Pignon 2017 assessed pain with VAS, where 0 was no pain at all and 100 was the most severe pain experienced. Study authors reported that no differences in pain reduction were observed between the BMMNC and control group.

Incidence of amputation

(follow‐up: 6 or 12 months)

Study population

RR 0.52 (0.27 to 0.99)

176
(4 RCTs)

⊕⊝⊝⊝
VERY LOWe

All studies reported the incidence of amputation. A possible small benefit was no longer seen after removal of 2 studies at high risk of bias in sensitivity analysis (RR 0.52, 95% CI 0.19 to 1.39; 89 participants, 2 studies) (Barc 2006; Li 2013).

250 per 1000

130 per 1000

(68 to 248)

Angiographic analysis

See comment

None of the studies reported angiographic analysis.

Increase in ABI

An ABI ratio of 0.9 or less indicates PAD. Values between 0.9 and 1.0 are borderline, and above 1.0 is considered normal

(follow‐up: range 1 month to 12 months)

All studies measured ABI, but due to incomplete information we were unable to pool the data. See comment

176
(4 RCTs)

⊕⊝⊝⊝
VERY LOWf

Barc 2006 reported that ABI did not change from baseline during study follow‐up in both BMMNC and control groups.

Li 2013 used an absolute increase of > 15% ABI to quantify haemodynamic improvement. Study authors reported that this was greater in the BMMNC group compared to control (P = 0.002).

Lindeman 2018 reported no difference in mean ABI between the BMMNC and control groups after 12 months follow‐up (P = 0.50).

Pignon 2017 did not show any changes in median ABI value between groups.

Increase in PFWD (m)

(follow‐up: 12 months)

See comment

53
(1 RCT)

⊕⊕⊝⊝
LOWg

Only Lindeman 2018 reported PFWD, finding no clear difference between groups. The mean PFWD for the treatment and control groups at 12 months follow‐up was 128 ± 71 m vs 160 ± 11 m, P = 0.87.

Side effects and complications

(follow‐up: 6 or 12 months)

Study population

RR 2.13 (0.50 to 8.97)

176

(4 RCTs)

⊕⊝⊝⊝
VERY LOWh

All trials reported adverse events. 2 studies reported that no identifiable treatment‐related adverse events were observed, and that the therapy was well‐tolerated (Barc 2006; Pignon 2017).

Li 2013 reported adverse events in both BMMNC and control groups (3 vs 1 fever, 1 vs 0 MI, 0 vs 1 stroke). There were no differences in the incidence of adverse events between groups, and the therapy was well‐tolerated

Lindeman 2018 reported that leukaemia occurred in 1 participant during the follow‐up period. It is unclear if this was related to the procedure.

After applying the sensitivity analysis by removing 2 studies at high risk of bias (Barc 2006; Li 2013), the significance of the results did not change (RR 2.69, 95% CI 0.11 to 63.18).

23 per 1000

48 per 1000

(11 to 204)

*The risk in the intervention group (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).

ABI: ankle‐brachial index;BMMNC: bone marrow mononuclear cells; CI: confidence interval; CLI: critical limb ischaemia;MI: myocardial infarction; PAD: peripheral arterial disease; PFWD: pain‐free walking distance;PISQ: pain inventory score questionnaire; RCT: randomised controlled trial; RR: risk ratio; VAS: visual analogue scale

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aThe stem cells used in the included RCTs originate from bone marrow. All four trials used mononuclear cells collected during the harvesting procedure from bone marrow and implanted into affected limbs (Barc 2006; Li 2013; Lindeman 2018; Pignon 2017).
bThe control arms varied across all included RCTs: conventional therapy (Barc 2006), 0.9% sodium chloride (saline) (Li 2013), diluted autologous peripheral blood (Lindeman 2018), 30 mL saline with 4 mL autologous peripheral blood (Pignon 2017).
cWe downgraded a total of three levels due to concerns related to risk of bias, imprecision (few participants and events), and inconsistency (wide CIs and clinical heterogeneity).
dWe downgraded one level for concerns related to risk of bias, one level for imprecision (few participants and events), and one level for inconsistency (heterogeneity due to different control arms).
eWe downgraded one level for concerns related to risk of bias, one level for imprecision (few participants and events), and one level for inconsistency (heterogeneity due to different control arms).
fWe downgraded one level for concerns related to risk of bias, one level for imprecision (few participants and events), and one level for inconsistency (wide CIs, and heterogeneity due to different control arms).
gWe downgraded one level for imprecision (few participants and events) and one level for inconsistency (wide CIs, heterogeneity due to different control arms).
hWe downgraded one level for concerns related to risk of bias, one level for imprecision (few participants and events), and one level for inconsistency (wide CIs).

Figuras y tablas -
Summary of findings 1. Intramuscular injection of bone marrow mononuclear cells (BMMNC) compared to control for treatment of critical lower limb ischaemia (CLI)
Comparison 1. BMMNC versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 All‐cause mortality Show forest plot

3

123

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.15, 6.63]

1.2 Amputation Show forest plot

4

176

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.27, 0.99]

1.3 Amputation ‐ sensitivity analysis Show forest plot

2

89

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.19, 1.39]

1.4 Side effects and complications Show forest plot

4

176

Risk Ratio (M‐H, Random, 95% CI)

2.13 [0.50, 8.97]

1.5 Side effects and complications ‐ sensitivity analysis Show forest plot

2

89

Risk Ratio (M‐H, Random, 95% CI)

2.69 [0.11, 63.18]

Figuras y tablas -
Comparison 1. BMMNC versus control