Scolaris Content Display Scolaris Content Display

Compresión neumática intermitente de la pierna y profilaxis farmacológica combinadas para la profilaxis de la tromboembolia venosa en pacientes de alto riesgo

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Arabi 2019 {published and unpublished data}

Arabi Y, Al-Hameed F, Burns KE, Mehta S, Alsolamy S, Almaani M, et al.Statistical analysis plan for the Pneumatic CompREssion for PreVENting Venous Thromboembolism (PREVENT) trial: a study protocol for a randomized controlled trial. Trials 2018;19(1):182. CENTRAL [DOI: 10.1186/s13063-018-2534-6]
Arabi YM, Al-Hameed F, Burns KE, Mehta S, Alsolamy SJ, Alshahrani MS, et al.Adjunctive intermittent pneumatic compression for venous thromboprophylaxis. New England Journal of Medicine 2019;380(14):1305-15. CENTRAL
Arabi YM, Alsolamy S, Al-Dawood A, Al-Omari A, Al-Hameed F, Burns KE, et al.Erratum to: Thromboprophylaxis using combined intermittent pneumatic compression and pharmacologic prophylaxis versus pharmacologic prophylaxis alone in critically ill patients: study protocol for a randomized controlled trial [Trials,17 (2016) (390)]. Trials 2016;17:420. CENTRAL [DOI: 10.1186/s13063-016-1556-1]
Arabi YM, Alsolamy S, Al-Dawood A, Al-Omari A, Al-Hameed F, Burns KE, et al.Thromboprophylaxis using combined intermittent pneumatic compression and pharmacologic prophylaxis versus pharmacologic prophylaxis alone in critically ill patients: study protocol for a randomized controlled trial. Trials  2016;17(1):390. CENTRAL [DOI: 10.1186/s13063-016-1520-0]
ISRCTN44653506.The PREVENT trial: pneumatic compression for preventing venous thromboembolism. who.int/trialsearch/Trial2.aspx?TrialID=ISRCTN44653506 (first received 14 July 2014). CENTRAL
NCT02040103.Pneumatic compression for preventing venous thromboembolism (PREVENT). clinicaltrials.gov/show/NCT02040103 (first received 20 January 2014). CENTRAL

Bigg 1992 {published data only}

Bigg SW, Catalona WJ.Prophylactic mini-dose heparin in patients undergoing radical retropubic prostatectomy. A prospective trial. Urology 1992;39(4):309-13. CENTRAL

Borow 1983 {published data only}

Borow M, Goldson HJ.Prevention of postoperative deep venous thrombosis and pulmonary emboli with combined modalities. American Surgeon 1983;49(11):599-605. CENTRAL

Bradley 1993 {published data only}

Bradley JG, Krugener GH, Jager HJ.The effectiveness of intermittent plantar venous compression in prevention of deep venous thrombosis after total hip arthroplasty. Journal of Arthroplasty 1993;8(1):57-61. CENTRAL

Cahan 2000 {published data only}

Cahan MA, Hanna DJ, Wiley LA, Cox DK, Killewich LA.External pneumatic compression and fibrinolysis in abdominal surgery. Journal of Vascular Surgery 2000;32(3):537-43. CENTRAL

Dickinson 1998 {published data only}

Dickinson LD, Miller LD, Patel CP, Gupta SK.Enoxaparin increases the incidence of postoperative intracranial hemorrhage when initiated preoperatively for deep venous thrombosis prophylaxis in patients with brain tumors. Neurosurgery 1998;43(5):1074–81. CENTRAL

Dong 2018 {published data only}

Dong J, Wang J, Feng Y, Qi LP, Fang H, Wang GD, et al.Effect of low molecular weight heparin on venous thromboembolism disease in thoracotomy patients with cancer. Journal of Thoracic Disease 2018;10(3):1850-6. CENTRAL [DOI: 10.21037/jtd.2018.03.13]

Edwards 2008 {published data only}

Edwards JZ, Pulido PA, Ezzet KA, Copp SN, Walker RH, Colwell CW Jr.Portable compression device and low-molecular-weight heparin compared with low-molecular-weight heparin for thromboprophylaxis after total joint arthroplasty. Journal of Arthroplasty 2008;23(8):1122-7. CENTRAL

Eisele 2007 {published data only}

Eisele R, Kinzl L, Koelsch T.Rapid-inflation intermittent pneumatic compression for prevention of deep venous thrombosis. Journal of Bone and Joint Surgery - American Volume 2007;89(5):1050-6. CENTRAL

Hata 2019 {published data only}

Hata T, Yasui M, Ikeda M, Miyake M, Ide Y, Okuyama M, et al.Efficacy and safety of anticoagulant prophylaxis for prevention of postoperative venous thromboembolism in Japanese patients undergoing laparoscopic colorectal cancer surgery. Annals of Gastroenterological Surgery 2019;3(5):568-75. CENTRAL

Jung 2018 {published data only}NCT01448746

Jung YJ, Seo HS, Park CH, Jeon HM, Kim JI, Yim HW, et al.Venous thromboembolism incidence and prophylaxis use after gastrectomy among Korean patients with gastric adenocarcinoma: the PROTECTOR randomized clinical trial. JAMA Surgery 2018;153(10):939-46. CENTRAL [DOI: 10.1001/jamasurg.2018.2081]
Jung YJ, Song KY, Seo HS, Park CH.Optimal prophylactic method of venous thromboembolism for gastrectomy in Korean patients: prospective randomized controlled trial. Journal of Clinical Oncology 2017;35(4 Suppl):7. CENTRAL
Song KY, Yoo HM, Kim EY, Kim JI, Yim HW, Jeon HM, et al.Optimal prophylactic method of venous thromboembolism for gastrectomy in Korean patients: an interim analysis of prospective randomized trial. Annals of Surgical Oncology 2014;21(13):4232-8. CENTRAL

Kamachi 2020 {published data only}

Kamachi H, Homma S, Kawamura H, Yoshida T, Ohno Y, Ichikawa N, et al.Intermittent pneumatic compression versus additional prophylaxis with enoxaparin for prevention of venous thromboembolism after laparoscopic surgery for gastric and colorectal malignancies: multicentre randomized clinical trial. BJS Open  2020;4(5):804-10. CENTRAL [DOI: 10.1002/bjs5.50323]

Kurtoglu 2003 {published data only}

Kurtoglu M, Aksoy M, Genc FA, Baktiroglu S.The efficacy of intermittent pneumatic compression devices in combination with or without low molecular weight heparin in the prophylaxis of venous thromboembolism in patients with trauma. Phlebology 2003;18(3):148. CENTRAL
Yanar H, Kurtoglu M, Taviloglu K, Guloglu R, Ertekin C.Does intermittent pneumatic compression make low molecular weight heparin more efficient in the prophylaxis of venous thromboembolism in trauma patients. European Journal of Trauma and Emergency Surgery 2007;33 Suppl II:79-80. CENTRAL

Liu 2017a {published data only}

Liu JW, Jiang WP.Prevention of the perioperative deep venous thrombosis in hip arthroplasty. Chinese Journal of Tissue Engineering Research 2017;21(31):4927-32. CENTRAL [DOI: 10.3969/j.issn.2095-4344.2017.31.002]

Liu 2017b {published data only}

Liu P, Liu J, Chen L, Xia K, Wu X.Intermittent pneumatic compression devices combined with anticoagulants for prevention of symptomatic deep vein thrombosis after total knee arthroplasty: a pilot study. Therapeutics and Clinical Risk Management 2017;13:179-83. CENTRAL [DOI: 10.2147/TCRM.S129077]

Lobastov 2021 {published data only}

Lobastov K, Sautina E, Alencheva E, Bargandzhiya A, Schastlivtsev I, Barinov V, et al.Intermittent pneumatic compression in addition to standard prophylaxis of postoperative venous thromboembolism in extremely high-risk patients (IPC SUPER): a randomized controlled trial. Annals of Surgery 2021;274(1):63–9. CENTRAL [DOI: 10.1097/SLA.0000000000004556]

Nakagawa 2020 {published data only}

Nakagawa K, Watanabe J, Ota M, Suwa Y, Suzuki S, Suwa H, et al.Efficacy and safety of enoxaparin for preventing venous thromboembolic events after laparoscopic colorectal cancer surgery: a randomized-controlled trial (YCOG 1404). Surgery Today 2020;50(1):68-75. CENTRAL [DOI: 10.1007/s00595-019-01859-w]

Obitsu 2020 {published data only}

Obitsu T, Tanaka N, Oyama A, Ueno T, Saito M, Yamaguchi T, et al, Tohoku Surgical Clinical Research Promotion Organization Study Group.Efficacy and safety of low-molecular-weight heparin on prevention of venous thromboembolism after laparoscopic operation for gastrointestinal malignancy in Japanese patients: a multicenter, open-label, prospective, randomized controlled trial. Journal of the American College of Surgeons 2020;231(5):501-10. CENTRAL

Patel 2020 {published data only}

Patel H, Faisal F, Trock BB, Joice G, Schwen Z, Pierorazio P, et al.Effect of pharmacologic prophylaxis on venous thromboembolism after radical prostatectomy: the PREVENTER randomized clinical trial. European Urology 2020;78(3):360-8. CENTRAL

Ramos 1996 {published data only}

Ramos R, Salem BI, De Pawlikowski MP, Coordes C, Eisenberg S, Leidenfrost R.The efficacy of pneumatic compression stockings in the prevention of pulmonary embolism after cardiac surgery. Chest 1996;109(1):82-5. CENTRAL

Sakai 2016 {published data only}

ISRCTN18090286.Effects of a foot pump on the incidence of deep vein thrombosis after total knee arthroplasty in patients given edoxaban. who.int/trialsearch/Trial2.aspx?TrialID=ISRCTN18090286 (first received 8 July 2016). CENTRAL
Sakai T, Izumi M, Kumagai K, Kidera K, Yamaguchi T, Asahara T, et al.Effects of a foot pump on the incidence of deep vein thrombosis after total knee arthroplasty in patients given edoxaban: a randomized controlled study. Medicine (Baltimore) 2016;95(1):e2247. CENTRAL

Sang 2018 {published data only}

CHICTR-IPR-15007324.The mechanical and medical prevention of lower extremity deep venous thrombosis formation post gynecologic pelvic surgery, a multiple center randomized case control study. who.int/trialsearch/Trial2.aspx?TrialID=ChiCTR-IPR-15007324 (first received 30 October 2015). CENTRAL
Sang CQ, Zhao N, Zhang J, Wang SZ, Guo SL, Li SH, et al.Different combination strategies for prophylaxis of venous thromboembolism in patients: a prospective multicenter randomized controlled study. Scientific Reports 2018;8(1):8277. CENTRAL [DOI: 10.1038/s41598-018-25274-2]

Sieber 1997 {published data only}

Sieber PR, Rommel FM, Agusta VE, Breslin JA, Harpster LE, Huffnagle HW, et al .Is heparin contraindicated in pelvic lymphadenectomy and radical prostatectomy? Journal of Urology 1997;158(3 Pt 1):869-71. CENTRAL

Silbersack 2004 {published data only}

Silbersack Y, Taute BM, Hein W, Podhaisky H.Prevention of deep-vein thrombosis after total hip and knee replacement. Low-molecular-weight heparin in combination with intermittent pneumatic compression. Journal of Bone and Joint Surgery - British Volume 2004;86(6):809-12. CENTRAL

Siragusa 1994 {published data only}

Siragusa S, Vicentini L, Carbone S, Barone M, Beltrametti C, Piovella F.Intermittent pneumatic leg compression (IPLC) and unfractionated heparin (UFH) in the prevention of post-operative deep vein thrombosis in hip surgery: a randomized clinical trial. British Journal of Haematology 1994;87 Suppl 1:186. CENTRAL

Stannard 1996 {published data only}

Stannard JP, Harris RM, Bucknell AL, Cossi A, Ward J, Arrington ED.Prophylaxis of deep venous thrombosis after total hip arthroplasty by using intermittent compression of the plantar venous plexus. American Journal of Orthopedics 1996;25(2):127–34. CENTRAL

Tsutsumi 2012 {published data only}

Tsutsumi S, Yajima R, Tabe Y, Takaaki T, Fujii F, Morita H, et al.The efficacy of fondaparinux for the prophylaxis of venous thromboembolism after resection for colorectal cancer. Hepatogastroenterology 2012;59(120):2477-9. CENTRAL

Turpie 2007 {published data only}

Del Priore G, Herzog TJ, Bauer KA, Caprini JA, Comp P, Gent M, et al.Fondaparinux combined with intermittent pneumatic compression (IPC) versus IPC alone in the prevention of venous thromboembolism after major abdominal surgery: results of the APOLLO study. Gynecologic Oncology 2006;101:S21. CENTRAL
Turpie AG, Bauer KA, Caprini JA, Comp PC, Gent M, Muntz JE, on behalf of the APOLLO Investigators.Fondaparinux combined with intermittent pneumatic compression vs. intermittent pneumatic compression alone for prevention of venous thromboembolism after abdominal surgery: a randomized, double-blind comparison. Journal of Thrombosis and Haemostasis 2007;5(9):1854-61. CENTRAL
Turpie AG, Bauer KA, Caprini JA, Comp PP, Gent M, Muntz JE.Fondaparinux combined with intermittent pneumatic compression (IPC) versus IPC alone in the prevention of venous thromboembolism after major abdominal surgery: the randomized APOLLO study. Blood 2005;106(11):Abstract 279. CENTRAL

Westrich 2005 {published data only}

Westrich GH, Rana AJ, Terry MA, Taveras NA, Kapoor K, Helfet DL.Thromboembolic disease prophylaxis in patients with hip fracture: a multimodal approach. Journal of Orthopaedic Trauma 2005;19(4):234-40. CENTRAL

Westrich 2006 {published data only}

Westrich GH, Bottner F, Windsor RE, Laskin RS, Haas SB, Sculco TP.VenaFlow plus Lovenox vs VenaFlow plus aspirin for thromboembolic disease prophylaxis in total knee arthroplasty. Journal of Arthroplasty 2006;21(6 Suppl 2):139-43. CENTRAL

Windisch 2011 {published data only}

Windisch C, Kolb W, Kolb K, Grützner P, Venbrocks R, Anders J.Pneumatic compression with foot pumps facilitates early postoperative mobilisation in total knee arthroplasty. International Orthopaedics 2011;35(7):995-1000. CENTRAL

Woolson 1991 {published data only}

Woolson ST, Watt JM.Intermittent pneumatic compression to prevent proximal deep venous thrombosis during and after total hip replacement. A prospective, randomized study of compression alone, compression and aspirin, and compression and low-dose warfarin. Journal of Bone and Joint Surgery - American Volume 1991;73(4):507-12. CENTRAL

Yokote 2011 {published data only}

Yokote R, Matsubara M, Hirasawa N, Hagio S, Ishii K, Takata C.Is routine chemical thromboprophylaxis after total hip replacement really necessary in a Japanese population? Journal of Bone and Joint Surgery. British Volume 2011;93(2):251-6. CENTRAL

Zhou 2020 {published data only}

Zhou X, Gong S, Liang W, Hu J.Elastic stockings plus enoxaparin and intermittent pneumatic compression in preventing postoperative deep venous thrombosis in patients with ovarian cancer. International Journal of Clinical and Experimental Medicine 2020;13(1):2717-23. CENTRAL

Referencias de los estudios excluidos de esta revisión

Ailawadi 2001 {published data only}

Ailawadi M, Del Priore G.A comparison of thromboembolic prophylaxis in gynecologic oncology patients. International Journal of Gynecological Cancer 2001;11(5):354-8. CENTRAL

Eskander 1997 {published data only}

Eskander MB, Limb D, Stone MH, Furlong AJ, Shardlow D, Stead D, et al.Sequential mechanical and pharmacological thromboprophylaxis in the surgery of hip fractures: a pilot study. International Orthopaedics 1997;21(4):259-61. CENTRAL

Frim 1992 {published data only}

Frim DM, Barker FG 2nd, Poletti CE, Hamilton AJ.Postoperative low-dose heparin decreases thromboembolic complications in neurosurgical patients. Neurosurgery 1992;30(6):830-2. CENTRAL

Gagner 2012 {published data only}

Gagner M, Selzer F, Belle SH, Bessler M, Courcoulas AP, Dakin GF, et al.Adding chemoprophylaxis to sequential compression might not reduce risk of venous thromboembolism in bariatric surgery patients. Surgery for Obesity and Related Diseases 2012;8(6):663-70. CENTRAL

Gelfer 2006 {published data only}

Gelfer Y, Tavor H, Oron A, Peer A, Halperin N, Robinson D.Deep vein thrombosis prevention in joint arthroplasties: continuous enhanced circulation therapy vs low molecular weight heparin. Journal of Arthroplasty 2006;21(2):206-14. CENTRAL

Kamran 1998 {published data only}

Kamran SI, Downey D, Ruff RL.Pneumatic sequential compression reduces the risk of deep vein thrombosis in stroke patients. Neurology 1998;50(6):1683-8. CENTRAL

Kiudelis 2010 {published data only}

Kiudelis M, Gerbutavicius R, Gerbutaviciene R, Griniūte R, Mickevicius A, Endzinas Z, et al.A combinative effect of low-molecular-weight heparin and intermittent pneumatic compression device for thrombosis prevention during laparoscopic fundoplication. Medicina 2010;46:18-23. CENTRAL

Kumaran 2008 {published data only}

Kumaran R, Sokolovic M, Ashame E, George EM, Bernstein L, George L.Prevention of venous thromboembolism in critically ill medical patients. Chest2008;134(4):36003s. CENTRAL

Lieberman 1994 {published data only}

Lieberman JR, Huo MM, Hanway J, Salvati EA, Sculco TP, Sharrock NE.The prevalence of deep venous thrombosis after total hip arthroplasty with hypotensive epidural anesthesia. Journal of Bone and Joint Surgery 1994;76-A(3):341–8. CENTRAL

Macdonald 2003 {published data only}

Macdonald RL, Amidei C, Baron J, Weir B, Brown F, Erickson RK, et al.Randomized, pilot study of intermittent pneumatic compression devices plus dalteparin versus intermittent pneumatic compression devices plus heparin for prevention of venous thromboembolism in patients undergoing craniotomy. Surgical Neurology 2003;59(5):363-72; discussion 372-4. CENTRAL

Mehta 2010 {published data only}

Mehta KV, Lee HC, Loh JS.Mechanical thromboprophylaxis for patients undergoing hip fracture surgery. Journal of Orthopaedic Surgery (Hong Kong) 2010;18(3):287-9. CENTRAL
Mehta KV, Lee HC, Loh JS.Reply to a letter to the Editor re: mechanical thromboprophylaxis for patients undergoing hip fracture surgery. Journal of Orthopaedic Surgery (Hong Kong) 2011;19(2):260-2. CENTRAL

Nathan 2006 {published data only}

Nathan SS, Simmons KA, Lin PP, Hann LE, Morris CD, Athanasian EA, et al.Proximal deep vein thrombosis after hip replacement for oncologic indications. Journal of Bone and Joint Surgery - American Volume 2006;88(5):1066-70. CENTRAL

Patel 2010 {published data only}

Patel AR, Crist MK, Nemitz J, Mayerson JL.Aspirin and compression devices versus low-molecular-weight heparin and PCD for VTE prophylaxis in orthopedic oncology patients. Journal of Surgical Oncology 2010;102:276–81. CENTRAL

Roberts 1975 {published data only}

Roberts VC, Cotton LT.Failure of low-dose heparin to improve efficacy of perioperative intermittent calf compression in preventing postoperative deep vein thrombosis. British Medical Journal 1975;3(5981):458-60. CENTRAL

Spinal cord injury investigators {published data only}

Spinal Cord Injury Thromboprophylaxis Investigators.Prevention of venous thromboembolism in the acute treatment phase after spinal cord injury: a randomized, multicenter trial comparing low-dose heparin plus intermittent pneumatic compression with enoxaparin. Journal of Trauma - Injury Infection and Critical Care 2003;54(6):1116-24; discussion 1125-6. CENTRAL

Stannard 2006 {published data only}

Stannard JP, Lopez-Ben RR, Volgas DA, Anderson ER, Busbee M, Karr DK, et al.Prophylaxis against deep-vein thrombosis following trauma: a prospective, randomized comparison of mechanical and pharmacologic prophylaxis. Journal of Bone and Joint Surgery - American Volume 2006;88(2):261-6. CENTRAL

Tsutsumi 2000 {published data only}

Tsutsumi K, Udagawa H, Kajiyama Y, Kinoshita Y, Ueno M, Nakamura T, et al.Pulmonary thromboembolism after surgery for esophageal cancer: its features and prophylaxis. Surgery Today 2000;30(5):416-20. CENTRAL

Wan 2015 {published data only}

Wan B, Fu H-Y, Yin J-T, Ren G-Q.Low‑molecular‑weight heparin and intermittent pneumatic compression for thromboprophylaxis in critical patients. Experimental and Therapeutic Medicine 2015;10(6):2331-6. CENTRAL

Westrich 1996 {published data only}

Westrich GH, Sculco TP.Prophylaxis against deep venous thrombosis after total knee arthroplasty. Pneumatic plantar compression and aspirin compared with aspirin alone. Journal of Bone and Joint Surgery 1996;78-A(6):826-34. CENTRAL

Whitworth 2011 {published data only}

Whitworth JM, Schneider KE, Frederick PJ, Finan MA, Reed E, Fauci JM, et al.Double prophylaxis for deep venous thrombosis in patients with gynecologic oncology who are undergoing laparotomy: does preoperative anticoagulation matter? International Journal of Gynecological Cancer 2011;21(6):1131-4. CENTRAL

Winemiller 1999 {published data only}

Winemiller MH, Stolp-Smith KA, Silverstein MD, Therneau TM.Prevention of venous thromboembolism in patients with spinal cord injury: effects of sequential pneumatic compression and heparin. Journal of Spinal Cord Medicine 1999;22(3):182-91. CENTRAL

ChiCTR1800014257 {published data only}

ChiCTR1800014257.The strategies of risk-stratified prophylaxis of deep venous thrombosis after gynecologic pelvic surgery in patients at different levels of risk: a prospective multicenter randomized controlled trial. who.int/trialsearch/Trial2.aspx?TrialID=ChiCTR1800014257 (first received 1 February 2018). CENTRAL

EUCTR2007‐006206‐24 {published data only}

EUCTR2007-006206-24.A blind randomized trial to compare the efficacy of intermittent pneumatic compression (IPC) with and without early anticoagulant treatment for prevention of venous thromboembolism (VTE) in patients with acute primary intracerebral hemorrhage (ICH) including comparison of the American and European guideline recommendations. who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2007-006206-24-FI (first received 14 November 2007). CENTRAL

NCT00740987 (CIREA 2) {published data only}

NCT00740987.Efficacy of the association mechanical prophylaxis plus anticoagulant prophylaxis on venous thromboembolism incidence in intensive care unit (ICU). clinicaltrials.gov/ct2/show/NCT00740987 (first received 25 August 2008). CENTRAL

NCT02271399 {published data only}

NCT02271399.Comparative study of prophylactic agent for venous thromboembolism after total knee arthroplasty. clinicaltrials.gov/show/NCT02271399 (first received 22 October 2014). CENTRAL

NCT03559114 (PROTEST) {published data only}

NCT03559114.PROphylaxis for Venous ThromboEmbolism in Severe Traumatic Brain Injury (PROTEST). clinicaltrials.gov/ct2/show/NCT03559114 (first received 15 June 2018). CENTRAL

ASH 2019

Anderson DR,  Morgano GP,  Bennett C,  Dentali F,  Francis CW,  Garcia DA, et al.American Society of Hematology 2019 guidelines for management of venous thromboembolism: prevention of venous thromboembolism in surgical hospitalized patients. Blood Advances 2019;3(23):3898-944.

Atkins 2004

Atkins D, Best D, Briss PA, Eccles M, Falck-Ytter Y, Flottorp S, et al.Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490-4.

Chouhan 1999

Chouhan VD, Comerota AJ, Sun L, Harada R, Gaughan JP, Rao AK.Inhibition of tissue factor pathway during intermittent pneumatic compression: a possible mechanism for antithrombotic effect. Arteriosclerosis, Thrombosis and Vascular Biology 1999;19(11):2812-7.

Deeks 2021

Deeks JJ, Higgins JP, Altman DG editor(s).Chapter 10: Analysing data and undertaking meta-analyses. 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 www.training.cochrane.org/handbook.

Egger 1997

Egger M, Davey SG, Schneider M, Minder C.Bias in meta-analysis detected by a simple, graphical test. British Medical Journal (Clinical Research Ed.) 1997;315(7109):629-34.

Eriksson 2001

Eriksson BI, Bauer KA, Lassen MR, Turpie AG, for the Steering Committee of the Pentasaccharide in Hip-Fracture Surgery Study.Fondaparinux compared with enoxaparin for the prevention of venous thromboembolism after hip-fracture surgery. New England Journal of Medicine 2001;345(18):1298-304.

Eriksson 2008

Eriksson BI, Borris LC, Friedman RJ, Haas S, Huisman MV, Kakkar AK, et al.Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. New England Journal of Medicine 2008;358(26):2765-75.

Goldhaber 2001

Goldhaber SZ.Prophylaxis of venous thrombosis. Current Treatment Options in Cardiovascular Medicine 2001;3(3):225-35.

Gould 2012

Gould MK, Garcia DA, Wren SM, Karanicolas PJ, Arcelus JI, Heit JA, et al.Prevention of VTE in nonorthopedic surgical patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012;141:e227S-e277S.

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime)GRADEpro GDT.Version accessed 12 October 2021. Hamilton (ON): McMaster University (developed by Evidence Prime). Available at gradepro.org.

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG.Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60.

Higgins 2011

Higgins JP, Altman DG, Sterne JA (editors).Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration. Available from training.cochrane.org/handbook/archive/v5.1/2011.

Ho 2013

Ho KM, Tan JA.Stratified meta-analysis of intermittent pneumatic compression of the lower limbs to prevent venous thromboembolism in hospitalized patients. Circulation 2013;128:1003-20.

Kakkos 2005a

Kakkos SK, Griffin M, Geroulakos G, Nicolaides AN.The efficacy of a new portable sequential compression device (SCD Express) in preventing venous stasis. Journal of Vascular Surgery 2005;42(2):296-303.

Kakkos 2012

Kakkos SK, Warwick D, Nicolaides AN, Stansby GP, Tsolakis IA.Combined (mechanical and pharmacological) modalities for VTE prevention in joint arthroplasty. Journal of Bone and Joint Surgery. British Volume 2012;94-B:729-34.

King 2007

King CS, Holley AB, Jackson JL, Shorr AF, Moores LK.Twice vs three times daily heparin dosing for thromboembolism prophylaxis in the general medical population: a meta-analysis. Chest 2007;131(2):507-16.

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 www.training.cochrane.org/handbook.

McLeod 2001

McLeod RS, Geerts WH, Sniderman KW, Greenwood C, Gregoire RC, Taylor BM, et al Canadian Colorectal Surgery DVT Prophylaxis Trial investigators.Subcutaneous heparin versus low-molecular-weight heparin as thromboprophylaxis in patients undergoing colorectal surgery: results of the Canadian colorectal DVT prophylaxis trial: a randomized, double-blind trial. Annals of Surgery 2001;233(3):438-44.

NICE 2009

National Clinical Guideline Centre - Acute and chronic conditions.Venous thromboembolism: reducing the risk of venous thromboembolism (deep vein thrombosis and pulmonary embolism) in patients admitted to hospital. nice.org.uk/guidance/cg92/ (accessed April 2016).

NICE 2018

NICE guidelines.Venous thromboembolism in over 16s: reducing the risk of hospital-acquired deep vein thrombosis or pulmonary embolism. nice.org.uk/guidance/ng89 (accessed November 2021).

Nicolaides 2013

Nicolaides AN, Fareed J, Kakkar AK, Comerota AJ, Goldhaber SZ, Hull R, et al.Prevention and treatment of venous thromboembolism. International Consensus Statement (guidelines according to scientific evidence). International Angiology 2013;32:111-260.

NRSMG

Cochrane Non-Randomised Studies Methods Group.Draft chapters for the Guidelines on non-randomised studies in Cochrane reviews. cochrane.dk/nrsmg/guidelines.htm 2001 (accessed August 2007).

Page 2021

Page MJ, Higgins JP, Sterne JA.Chapter 13: Assessing risk of bias due to missing results in a synthesis. 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 www.training.cochrane.org/handbook.

Piazza 2007

Piazza G, Seddighzadeh A, Goldhaber SZ.Double trouble for 2,609 hospitalized medical patients who developed deep vein thrombosis: prophylaxis omitted more often and pulmonary embolism more frequent. Chest 2007;132(2):554-61.

Rosendaal 1999

Rosendaal FR.Venous thrombosis: a multicausal disease. Lancet 1999;353(9159):1167-73.

Schulman 2010

Schulman S, Angerås U, Bergqvist D, Eriksson B, Lassen MR, Fisher W, on behalf of the Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis.Definition of major bleeding in clinical investigations of antihemostatic medicinal products in surgical patients. Journal of Thrombosis and Haemostasis 2010;8(1):202-4.

Sobieraj 2013

Sobieraj DM, Coleman CI, Tongbram V, Chen W, Colby J, Lee S, et al.Comparative effectiveness of combined pharmacologic and mechanical thromboprophylaxis versus either method alone in major orthopedic surgery: a systematic review and meta-analysis. Pharmacotherapy 2013;33(3):275–83.

Song 2014

Song KY, Yoo HM, Kim EY, Kim JI, Yim HW, Jeon HM, et al.Optimal prophylactic method of venous thromboembolism for gastrectomy in Korean patients: an interim analysis of prospective randomized trial. Annals of Surgical Oncology  2014;21(13):4232-8.

Torrejon Torres 2019

Torrejon Torres R, Saunders R, Ho KM.A comparative cost-effectiveness analysis of mechanical and pharmacological VTE prophylaxis after lower limb arthroplasty in Australia. Journal of Orthopaedic Surgery and Research 2019;14(1):93.

Virchow 1856

Virchow RL.Thrombosis and emboli [Phlogose und thrombose im gefässsystem]. In: Gesammelte Adhandlungen zur Wissenschaftlichen Medicine. Frankfurt am Main: von Meidinger & Sohn, 1856:458-636.

Zareba 2014

Zareba P, Wu C, Agzarian J, Rodriguez D, Kearon C.Meta-analysis of randomized trials comparing combined compression and anticoagulation with either modality alone for prevention of venous thromboembolism after surgery. British Journal of Surgery 2014;101:1053–62.

Referencias de otras versiones publicadas de esta revisión

Kakkos 2005b

Kakkos SK, Geroulakos G, Caprini J, Nicolaides AN, Stansby G.Combined intermittent pneumatic leg compression and pharmacological prophylaxis for prevention of venous thromboembolism in high risk patients. Cochrane Database of Systematic Reviews 2005, Issue 2. Art. No: CD005258. [DOI: 10.1002/14651858.CD005258]

Kakkos 2008

Kakkos SK, Caprini JA, Geroulakos G, Nicolaides AN, Stansby G, Reddy DJ.Combined intermittent pneumatic leg compression and pharmacological prophylaxis for prevention of venous thromboembolism in high-risk patients. Cochrane Database of Systematic Reviews 2008, Issue 4. Art. No: CD005258. [DOI: 10.1002/14651858.CD005258.pub2]

Kakkos 2016

Kakkos SK, Caprini JA, Geroulakos G, Nicolaides AN, Stansby G, Reddy DJ, et al.Combined intermittent pneumatic leg compression and pharmacological prophylaxis for prevention of venous thromboembolism. Cochrane Database of Systematic Reviews 2016, Issue 9. Art. No: CD005258. [DOI: 10.1002/14651858.CD005258.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Arabi 2019

Study characteristics

Methods

Study design: RCT
Method of randomisation: centralised computer‐generated randomisation system with variable block size. Randomisation was stratified according to study site and type of heparin used
Concealment of allocation: centralised computer‐generated randomisation system with variable block size
Exclusions: 1 site was terminated by the study sponsor because some participants had been enrolled without full adherence to the approved informed‐consent process, and all data from this site were excluded from the analyses
Losses to follow‐up: none
ITT analysis: yes, modified

Participants

Countries: Saudi Arabia, Canada, Australia, and India
Number of participants: 2003, intervention group 991; control group 1012
Age (mean, years): intervention group 57.6; control group 58.7
Sex: 1148 (57.3%) male
Inclusion criteria: medical‐surgical ICU patients ≥ 14 years old at participating ICUs. ICUs that use other age cut‐off for adult patients will adhere to their local standard (16 or 18 years); patient weight ≥ 45 kg; expected ICU LOS ≥ 72 h; eligible for pharmacologic thromboprophylaxis with UFH and LMWH
Exclusion criteria: patient treated with IPC for > 24 h in this current ICU admission; patient in the ICU for > 48 h; patient treated with pharmacologic VTE prophylaxis with medications other than UFH or LMWH; inability or contraindication to applying IPC to both legs (burns in the lower extremities, lacerations, active skin infection and ischaemic lower limb at the site of IPC placement, acute ischaemia in the lower extremities, amputated foot or leg on 1 or both sides, compartment syndrome, severe PAD, vein ligation, gangrene, recent vein grafts and draining incisions, evidence of bone fracture in lower extremities); therapeutic dose of anticoagulation with UFH or LMWH; pregnancy; limitation of life support; life expectancy ≤ 7 days or palliative care; allergy to the sleeve material; patients with inferior VCF

Interventions

Intervention group: IPC and heparin (any type)
Control group: heparin (any type)

Outcomes

Primary outcome: incident (i.e. new) proximal lower‐limb DVT, as detected on twice‐weekly lower‐limb US after the third calendar day since randomisation until ICU discharge, death, attainment of full mobility, or study day 28, whichever occurred first. DVT that were detected on study days 1 to 3 were considered to be prevalent (i.e. pre‐existing) and were not included in the primary outcome analysis

Secondary outcomes: percentage of participants who had prevalent proximal DVT, the occurrence of any lower limb DVT thromboses (proximal, distal, prevalent, or incident), the occurrence of PE, a composite outcome of VTE that included PE or all prevalent and incident lower‐limb DVT, a composite outcome of VTE or death from any cause at 28 days, and safety outcomes

Funding

Grants from King Abdulaziz City for Science and Technology (AT 34‐65) and King Abdullah International Medical Research Center (RC12/045/R)

Declarations of interest

MH reports receiving payment for patient cost, US and start‐up fees

Notes

The modified ITT population comprised all the participants who underwent randomisation with the exception of those who withdrew consent for both the intervention and the collection of data and those who were identified as ineligible after randomisation.

The single case of fatal PE was observed in the control group (personal communication).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation system

Allocation concealment (selection bias)

Low risk

Centralised computer‐generated randomisation system with variable block size

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo IPC was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unclear if outcome assessors of adjudicating committee (if any) were blinded to allocation group, with the exception of radiologists who interpreted US findings

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Relatively few exclusions

Selective reporting (reporting bias)

Low risk

None detected

Other bias

Low risk

None detected

Bigg 1992

Study characteristics

Methods

Study design: CCT
Method of randomisation: study was planned to be randomised and method of planned randomisation was stated as patient order
Concealment of allocation: none stated
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 68, intervention group 32; control group 36
Age (mean, years): intervention group 67; control group 65
Sex: male
Inclusion criteria: retropubic RP with bilateral pelvic prostatectomy for clinically localised prostate cancer
Exclusion criteria: none stated

Interventions

Intervention group: SCDs with elastic stockings and UFH (5000 IU twice daily, sc)
Control group: SCDs with elastic stockings

Outcomes

Symptomatic PE, confirmed with ventilation‐perfusion scan

Funding

Not reported

Declarations of interest

Not reported

Notes

The study was planned to be randomised but due to administrative errors the randomisation protocol was violated

SCDs were started in the operating room and discontinued when the participants were ambulatory, usually 18 h postoperatively

Heparin was started 2 h before the operation and was continued for 7 days or the time of discharge

Study was discontinued because of bleeding complications associated with heparin use. No specific bleeding definitions were provided.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The study was planned to be randomised but due to administrative errors the randomisation protocol was violated. Method of planned randomisations was stated as patient order

Allocation concealment (selection bias)

High risk

Alternating patients received the study medication and in most cases the surgeon was aware of which participants received heparin

Blinding of participants and personnel (performance bias)
All outcomes

High risk

In most cases the surgeon was aware of which participants received heparin, and the same perhaps applies to the anaesthesia personnel

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing the pulmonary ventilation‐perfusion scans or angiograms were aware of which participants received heparin

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participants were lost to follow‐up

Selective reporting (reporting bias)

Low risk

PE was the only VTE event stated in methodology and was reported

Other bias

Unclear risk

No baseline characteristics, apart from age, were provided

Borow 1983

Study characteristics

Methods

Study design: CCT
Method of randomisation: none
Concealment of allocation: not reported
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 272, but only 237 of them were eligible for inclusion based on type of prophylaxis
Age (mean, years): not reported
Sex: not reported
Inclusion criteria: general, surgery, orthopaedics, gynaecology, and vascular surgery
Exclusion criteria: genitourinary surgery

Interventions

Intervention group: SCDs and pharmacological prophylaxis (UFH or coumadin)
Control group: SCDs or pharmacological prophylaxis (UFH or coumadin)

Outcomes

DVT diagnosed with I‐125 fibrinogen scanning, IPG, Doppler US and venography

Funding

Not reported

Declarations of interest

Not reported

Notes

Participants who received aspirin or dextran as an exclusive pharmacological modality or elastic stockings as an exclusive mechanical modality were not included in our review.

All modalities were started with the preoperative medication and continued until the participants were well ambulatory.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Patients were placed into each category in rotation by the vascular technicians.

Allocation concealment (selection bias)

High risk

No details on the allocation procedure were provided.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Placebo medications or devices were not used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing DVT testing were aware of which participants received heparin.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants had an event reported.

Selective reporting (reporting bias)

Low risk

DVT was the only VTE event stated in methodology and was reported.

Other bias

Unclear risk

No baseline characteristics were provided.

Bradley 1993

Study characteristics

Methods

Study design: CCT
Method of randomisation: states that patients with an even date of birth were randomised to receive the plantar arteriovenous impulse system on the side to be operated on.
Concealment of allocation: not reported other than the radiologist who read the venograms was blinded to patient allocation.
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: UK
Number of participants: 74
Age (mean, years): 70
Sex: not reported
Inclusion criteria: unilateral primary THA for osteoarthritis
Exclusion criteria: non‐consenting patients

Interventions

Intervention group: UFH (5000 IU twice daily, sc) with GCS (TEDs), and pneumatic foot compression on the side to be operated on
Control group: UFH (5000 IU twice daily, sc) and GCS (TEDs)

Outcomes

DVT on bilateral lower extremity venography performed postoperative day 12

Funding

IPC devices provided by the Novamedix Ltd

Declarations of interest

Not reported

Notes

The foot pump started at the beginning of surgery and continued until discharge from the hospital. No details were provided for heparin

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Patients with an even date of birth were randomised to receive IPC

Allocation concealment (selection bias)

High risk

Patients with an even date of birth were allocated to receive IPC ‐ allocation therefore predictable

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo device was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The radiologist who read the venograms was blinded to participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT was the only VTE outcome event stated in methodology and was reported

Other bias

Low risk

Baseline characteristics were comparable

Cahan 2000

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: not reported
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 48
Age (mean, years): 67
Sex: 47 male, 1 female
Inclusion criteria: major intra‐abdominal surgical procedures
Exclusion criteria: pre‐existing venous disease, history of VTE, preoperative or postoperative requirement for systemic anticoagulation (with the exception of the 12 participants undergoing aortic aneurysm repair, who did receive systemic doses of heparin intraoperatively)

Interventions

Intervention group: UFH (5000 IU twice daily) and thigh‐length sequential pneumatic compression (Kendall Health Care, Manchester, Mass, USA)
Control groups:
1. UFH (5000 IU twice daily)
2. thigh‐length sequential pneumatic compression device (Kendall Health Care, Manchester, Mass, USA)

Outcomes

DVT on DUS and also clinically evident DVT and PE

Funding

Not reported

Declarations of interest

None

Notes

Investigation on the effect of study interventions on fibrinolytic activity, but also reported VTE outcomes

DVT prophylaxis was initiated in the operating room after induction of anaesthesia and continued until postoperative day 5 (or discharge, if this occurred sooner). If the participant remained hospitalised after postoperative day 5, DVT prophylaxis was left to the discretion of the primary surgeon, and the participant was no longer participating in the research study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo anticoagulants or IPC devices were used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel who performed the DVT screening were blinded to the treatment regimens

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT was the only VTE outcome event stated in methodology and was reported

Other bias

Low risk

No significant baseline imbalances

Dickinson 1998

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: not reported
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 66
Age (mean, years): 47.4 (calculated)
Sex: not reported
Inclusion criteria: patients undergoing surgical treatment of intracranial neoplasms
Exclusion criteria: history of DVT or PE, allergy to heparin or other anticoagulant agents, history of surgery or major trauma to the lower extremities, a concurrent condition requiring anticoagulation therapy, cranial base neoplasms and pituitary adenomas

Interventions

Intervention group: LMWH enoxaparin (Lovenox; Rhône‐Poulenc Rorer Pharmaceuticals) sc at a dose of 30 mg in the anaesthesia holding room, and continued at a dose of 30 mg twice daily combined with IPC (thigh‐high SCDs (Kendall), functioning on the patient before induction of anaesthesia
Control groups:
1. LMWH enoxaparin sc at a dose of 30 mg and continued at a dose of 30 mg twice daily
2. IPC (thigh‐high SCDs ‐ Kendall)

Outcomes

DVT on DUS between days 1 and 3, between days 5 and 7, at the wound check appointment between days 10 and 14, and at the 1‐month follow‐up appointment

Incidence of adverse events (including bleeding) was assessed by principal investigator by thorough review of medical records. No specific bleeding definitions were provided

Funding

Not reported

Declarations of interest

Not reported

Notes

The IPC devices functioned throughout the surgical procedure and remained on the participant during the postoperative period, until the participant was walking without assistance. If the participant remained nonambulatory, the devices were discontinued at the time of discharge from the Neurosurgery Service.

Enoxaparin was started in the anaesthesia holding room and was discontinued at the time of discharge from the Neurosurgery Service.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo anticoagulants or IPC devices were used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel who performed the DVT screening were blinded to the treatment regimens

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT was the only VTE outcome event stated in methodology and was reported

Other bias

High risk

The study stopped early (enrolment was planned for 120 participants)

Dong 2018

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: unclear
Exclusions: 12 participants in intervention group and 6 participants in the control group
Losses to follow‐up: 3 participants in intervention group
ITT analysis: yes

Participants

Country: China
Number of participants: 111, intervention group 55; control group 56
Age (mean, years): intervention group 62.20; control group 59.96
Sex: 63 (56.8%) male
Inclusion criteria: age between 18 and 80 years old; pathological diagnosis of malignant tumours; scheduled for thoracotomy under general anaesthesia; all anticoagulant treatment stopped for at least 7 days preoperatively; normal coagulation function or mild coagulation dysfunction (PT < 3 seconds above the upper limit, APTT < 10 seconds above the upper limit); preoperative VTE excluded by CTPA or extremity venous US
Exclusion criteria: postoperative therapeutic anticoagulant requirement such as for heart valve replacement; active bleeding or transfusion RBC was > 2 units within 24 h; active bleeding was defined as bloody chest drainage more than 200 mL/h for 5 h; or patients had symptom of hypovolaemic shock; tumour metastasis; PLT count of < 10 × 109/L

Interventions

Intervention group: IPC with elastic stockings and LMWH nadroparin
Control group: IPC with elastic stockings 

Outcomes

Primary end points: incidence of PE, DVT, and the PESI of PE patients. CTPA and extremity venous Doppler US were performed in all patients on POD 7. Simultaneously, patients diagnosed with PE by radiologist through CTPA were assessed using the PESI

Secondary end points: haemoglobin, PLT, D‐dimer (mcg/L), the PO2/FiO2 ratio (P/F) at POD 7 and the chest drainage time and the LOS in hospital after operation

Funding

Not reported

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No use of placebo. Exception for PE (low risk)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Large number of exclusions

Selective reporting (reporting bias)

Low risk

Results were provided for all outcomes

Other bias

Low risk

None detected

Edwards 2008

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: not reported
Exclusions: 10 consented participants cancelled their surgery; 33 participants were excluded for protocol violations, such as missed US (n = 9), surgery other than THA or TKA (n = 1), previous history of thrombosis (n = 12), prophylaxis other than LMWH (n = 8), and other protocol deviations (n = 3)
Losses to follow‐up: none
ITT analysis: no

Participants

Country: USA
Number of participants: 320
Age (mean, years): 67.3 (calculated)
Sex: 162 female, 115 male
Inclusion criteria: patients undergoing THR or TKR
Exclusion criteria: not provided

Interventions

Intervention group: LMWH enoxaparin (30 mg twice daily, starting the morning after surgery for 7‐8 days) combined with IPC (CECT device, ActiveCare DVT; Medical Compression Systems, Or Akiva, Israel) with calf sleeves
Control group: LMWH enoxaparin (30 mg twice daily, starting the morning after surgery for 7‐8 days)

Outcomes

DVT on DUS before discharge and also clinically evident DVT and PE at 3 months

Funding

Received from Medical Compression Active Care DVT, Medical Compression Systems

Declarations of interest

Benefits received from Medical Compression Active Care DVT, Medical Compression Systems

Notes

IPC was placed on the calves of the participant in the operating room and continued during hospitalisation
Enoxaparin was started the morning after surgery and continued for 7‐8 days

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo devices were used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel who performed the DVT screening were blinded to the treatment regimens

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT and PE were VTE events stated in methodology and were reported

Other bias

Low risk

None detected

Eisele 2007

Study characteristics

Methods

Study design: RCT
Method of randomisation: not stated
Concealment of allocation: not reported
Exclusions post randomisation: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Germany
Number of participants: 1803
Age (mean, years): not reported
Sex: not reported
Inclusion criteria: total joint arthroplasty (24%); knee ligamentous and meniscal repair; tumour resection; open fixation of traumatic fractures; elective osteotomies to correct deformities of the femur, tibia, foot and ankle; and to treat high‐impact contusion injuries of the lower extremity, pelvis, abdomen, spine, and chest
Exclusion criteria: a surgery location that would interfere with the application of the pneumatic compression calf cuff and existing acute DVT

Interventions

Intervention group: LMWH certoparin (3000 IU 12 h pre‐op, 12 post‐op then daily, sc), compression stockings (18‐20 mmHg), and rapid‐inflation IPC
Control group: LMWH certoparin (3000 IU 12 h pre‐op, 12 post‐op then daily, sc), and compression stockings (18‐20 mmHg)

Outcomes

Symptomatic DVT and DVT on colour‐coded DUS performed on the day of discharge

Funding

Received from Aircast Europe

Declarations of interest

Funding received from the manufacturer by "one or more of the authors" 

Notes

Quote: "The DVT prophylaxis regimen was randomly assigned in the operating theatre at the time of completion of surgery and the randomisation was stratified by age."

No information on PE was provided

Participants in the IPC group had the IPC system applied to both calves in the recovery room shortly after the completion of surgery. IPC therapy was applied daily during the time that the participant was confined to bed postoperatively, and it was terminated at the time that the participant was able to walk

LMWH was started 12 h preoperatively and continued throughout hospitalisation

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided, apart from the information that it was stratified by patient age, so that we can make an assumption that they used a computer‐generated sequence or the sealed envelope method

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo device was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel who performed the DVT screening were blinded to the treatment regimens

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT was the only VTE event stated in methodology and was reported

Other bias

Low risk

Baseline number of risk factors for DVT per participant were comparable

Hata 2019

Study characteristics

Methods

Study design: RCT
Method of randomisation: central randomisation
Concealment of allocation: central concealment
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Japan
Number of participants: 302, intervention group 145; control group 157
Age (mean, years): intervention group 65.4; control group 64.9
Sex: 168 (55.6%) male
Inclusion criteria: patients who were undergoing laparoscopic colorectal surgery who had an additional risk factor for VTE were included. As noted in the Japanese VTE guidelines, these additional risk factors include "thrombotic disorder, history of VTE, malignant disease, cancer chemotherapy, serious infection, central venous catheterisation, long‐term bed rest (more than 24 h after surgery), leg paralysis, leg cast fixation, hormone therapy, obesity (body mass index 25kg/m2 or more), and varicose veins of the lower extremities." Other inclusion criteria were as follows: confirmed colorectal cancer by endoscopic examination; age ≥ 20 years; sufficient organ function, per laboratory data showing white blood cell count ≥ 3000/mm3, PLT ≥ 100000/mm3, total bilirubin ≤ 2.0 mg/dL, liver enzymes ≤ 100 IU/L, and serum creatinine ≤ 1.5 mg/dL; preoperative D‐dimer < 1 μg/mL or less than twice the institution limit for excluding asymptomatic DVT; symptomatic DVT; and provision of written informed consent
Exclusion criteria: active bleeding or with thrombocytopenia (PLT < 10 × 104/μL); risk of bleeding, including gastrointestinal ulcers, diverticulitis, colitis, acute bacterial endocarditis, uncontrolled severe hypertension, or uncontrolled diabetes mellitus; severe liver dysfunction (Child C); known hypersensitivity to UFH, LMWH, or heparinoids; history of intracranial bleeding; having undergone central cranial surgery, spine surgery, or ophthalmic surgery within 3 months before registration in the study; severe renal dysfunction (creatine clearance < 20 mL/min); known hypersensitivity to contrast media; or any condition that made the patient unfit for the study, as determined by the attending physician

Interventions

Intervention group: IPC with elastic stockings and anticoagulation (fondaparinux or enoxaparin)
Control group: IPC with elastic stockings 

Outcomes

Primary endpoint: incidence of VTE

Secondary endpoint: incidence of major bleeding

Funding

None

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation

Allocation concealment (selection bias)

Low risk

Central concealment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study without use of placebo, with the exception of PE (the radiologist interpreted the CT scans without any identifying information about the patients)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Jung 2018

Study characteristics

Methods

Study design: RCT
Method of randomisation: computer‐randomised treatment assignments
Concealment of allocation: sequential sealed envelopes
Exclusions: 16 participants (2.3%) were excluded from the per‐protocol analysis: 1 experienced HIT, 8 failed curative surgery, 3 had concurrent cancer diagnoses, and 4 withdrew from the study
Losses to follow‐up: none
ITT analysis: no

Participants

Country: Korea
Number of participants: 682, intervention group 341; control group 341
Age (mean, years): intervention group 57.9; control group 57.4
Sex: 435 (65.3%) of the participants were male
Inclusion criteria: gastric adenocarcinoma, confirmed by pathologic result; aged 20‐75; ECOG ≤ 2; ASA ≤ 3; patients who signed the written consent of the institutional ethics review committee to participate in this study with full understanding of the purpose and contents of the research prior to the participation
Exclusion criteria: active status of other cancer; diagnosed or treated with DVT or PE within 1 year; preoperative prolonged immobilisation or being wheelchair‐dependent; disease with hemorrhagic tendency; currently taking anticoagulants; underwent surgery under general anaesthesia for > 4 h within the last 6 months; history of stroke within the last 3 months; allergy to heparin or history of HIT; varicose veins or CVI, peripheral vascular disease, skin ulcer; history of anticancer or radiation therapy in the past; BMI ≤ 18.5 kg/m2; pregnant patients

Interventions

Intervention group: IPC and LMWH enoxaparin
Control group: IPC

Outcomes

DVT, on DUS but also clinically evident DVT, and PE

Bleeding: major and minor

Funding

Funded by Covidien and Medtronic

Declarations of interest

None

Notes

An interim analysis was published in 2014 (Song 2014), which was included in the previous version of this review

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐randomised treatment assignments

Allocation concealment (selection bias)

High risk

Sequential sealed envelopes, but no statement that these were opaque

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo injection was given

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

A relatively large number of participants in the combined group did not have DUS

Selective reporting (reporting bias)

Low risk

DVT and PE were stated in methodology to be the outcome measures of the study and results were reported

Other bias

Low risk

None detected

Kamachi 2020

Study characteristics

Methods

Study design: RCT
Method of randomisation: central computer‐generated randomisation
Concealment of allocation: central computer‐generated randomisation
Exclusions: 37
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Japan
Number of participants: 448, intervention group 225; control group 223
Age (mean, years): intervention group 64.8; control group 65.0
Sex: 270 (60.2%) male
Inclusion criteria: patients with gastric or colorectal cancer scheduled for laparoscopic surgery; patients > 40 years with WHO performance status 0 or 1, who agreed to participate in the study
Exclusion criteria: history of HIT or heparin hypersensitivity, acute bacterial endocarditis, creatinine clearance < 50 mL/min, severe hepatic dysfunction (Child grade C); weight < 40 kg; pregnancy; prescription of antiplatelet or anticoagulant drugs, history of venous thromboembolic disease within 1 year; history of hypersensitivity for iodinated contrast agent, presence of CVC; treatment with oestrogen or progesterone within 4 weeks of the operation, and radiotherapy or chemotherapy within 2 weeks of surgery

Interventions

Intervention group: IPC and LMWH enoxaparin
Control group: IPC

Outcomes

Primary outcome: VTE (DVT and PE), both symptomatic and asymptomatic, diagnosed by multidetector CT on POD 7

Secondary outcome: bleeding

Funding

Reported as not funded, however, the primary investigator of the study received research support from Kaken Pharmaceutical, Tokyo, Japan, a company that had a co‐marketing agreement for the anticoagulant enoxaparin used in the experimental group of the study

Declarations of interest

AT reported receiving research support as the primary investigator from Kaken Pharmaceutical, Tokyo, Japan

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central computer‐generated randomisation 

Allocation concealment (selection bias)

Low risk

Central computer‐generated randomisation 

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding was considered to be impossible

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Blinding was considered to be impossible, however, the radiologist in each hospital evaluated the occurrence of VTE with no information on participant allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

Large number of exclusions

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Kurtoglu 2003

Study characteristics

Methods

Study design: quasi‐RCT
Method of randomisation: by the last digit of year of birth
Concealment of allocation: none
Exclusions: not reported
Losses to follow‐up: not reported
ITT analysis: yes

Participants

Country: Turkey
Number of participants: 80
Age (mean, years): not provided
Sex: not provided
Inclusion criteria: trauma patients, at high risk for bleeding
Exclusion criteria: low risk for bleeding

Interventions

Intervention group: LMWH (40 mg/day) combined with IPC
Control group: IPC

Outcomes

DVT on DUS and clinically evident DVT; and PE by CT scanning

Funding

Not reported

Declarations of interest

Not reported

Notes

Information on randomisation and blinding was obtained from the study authors. No information on start and discontinuation of IPC or LMWH

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quasi‐randomised trial, randomised by the last digit of year of birth

Allocation concealment (selection bias)

High risk

Quasi‐randomised trial so predictable

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo anticoagulants were used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The radiologist who performed the US tests was not aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All study participants were reported in the results section

Selective reporting (reporting bias)

Low risk

DVT and PE were the VTE events stated in methodology and results were provided

Other bias

Unclear risk

Insufficient details were provided to allow a conclusion to be made

Liu 2017a

Study characteristics

Methods

Study design: RCT
Method of randomisation: random numbers
Concealment of allocation: unclear
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: China
Number of participants: 350, intervention group 175; control group 175
Age (mean, years): intervention group 62.9; control group 58.4
Sex: 169 (48.3%) male
Inclusion criteria: patients who received hip replacement; age > 50, voluntary‐based, pre‐op US shown no DVT
Exclusion criteria: pre‐op DVT+ve; active bleeding or recent intracranial haemorrhage; have been taking anticoagulants with history of diseases related to bleeding tendency; with other diseases that might influence the result of the study

Interventions

Intervention group: IPC and LMWH nadroparin
Control group: LMWH nadroparin

Outcomes

Primary: operation time; blood loss during hip replacement; pre‐ and post‐op PLT count, APTT, PT, fibrinogen, total cholesterol; occurrence of DVT after joint replacement

Funding

The Research Project of Health and Family Planning Commission of Guangxi Zhuang Autonomous Region, No. Z2014459

Declarations of interest

Not reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number method

Allocation concealment (selection bias)

Unclear risk

No details were provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study, without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details were provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Results were provided for all outcomes

Other bias

Low risk

None detected

Liu 2017b

Study characteristics

Methods

Study design: RCT
Method of randomisation: randomisation was performed using consecutively numbered sealed envelopes produced by an independent specialist, which were opened after surgery
Concealment of allocation: randomisation was performed using consecutively numbered sealed envelopes produced by an independent specialist, which were opened after surgery
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: China
Number of participants: 120, intervention group 60; control group 60
Age (mean, years): both groups 64.0
Sex: 56 (46.7%) male
Inclusion criteria: patients aged > 45 years old, who had osteoarthritis of the knee and had undergone unilateral primary TKA surgery
Exclusion criteria: inability to give informed consent; history of lower extremity varicose vein; combined organ bleeding risk can not receive drug anticoagulation therapy; anticoagulation treatment (including high dose aspirin); planned follow‐up at another hospital; renal failure; heart failure with pitting oedema; thrombophlebitis; thromboembolic event during the previous 3 months; other surgery during the previous months; malignancy; haemophilia; and pregnancy

Interventions

Intervention group: IPC and rivaroxaban
Control group: rivaroxaban

Outcomes

Primary outcome: DVT in popliteal or femoral veins, detected on a screening compression DUS, or any symptomatic DVT in the proximal veins, confirmed by imaging, within 21 days of randomisation

Secondary outcomes: death, any DVTs, symptomatic DVTs, PE, skin breaks on the legs, falls with injury or fractures, and duration of IPC devices use occurring within 21 days of randomisation, symptomatic VTE, mean intraoperative blood loss, and length of initial hospital stay measured 1 month after randomisation

Funding

National Science Foundation of China (grant number 81371950)

Declarations of interest

None reported

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Consecutively numbered sealed envelopes

Allocation concealment (selection bias)

Low risk

Randomisation was performed using consecutively numbered sealed envelopes produced by an independent specialist, which were opened after surgery

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study, however technician blinded to patients’ allocation performed US

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition was reported

Selective reporting (reporting bias)

High risk

Results on symptomatic DVT were not reported

Other bias

Low risk

None reported

Lobastov 2021

Study characteristics

Methods

Study design: quasi‐RCT
Method of randomisation: based on the number of hospital medical records: those with an even last digit were assigned to the experimental group, and those with an odd last digit were assigned to the control group; if the last digit was zero, the penultimate digit was used
Concealment of allocation: as above
Exclusions: none
Losses to follow‐up: 6
ITT analysis: yes

Participants

Country: Russian Federation
Number of participants: 407, intervention group 204; control group 203
Age (mean, years): intervention group 68.8; control group 68.8
Sex: 160 (39.3%) male
Inclusion criteria: patients > 40 years of age, required major surgery; had a high risk for postoperative VTE, Caprini score of > 11, and provided their informed consent
Exclusion criteria: acute DVT at baseline; performed IVC plication or implanted IVC filter; regular preoperative anticoagulation; postoperative anticoagulation needed at therapeutic doses; absence of anticoagulation > 5 days after surgery; coagulopathy (not related to DIC syndrome); thrombocytopaenia; haemorrhagic diathesis; lower‐limb soft‐tissue infection; lower‐limb skin lesion; ABI < 0.6

Interventions

Intervention group: IPC with elastic stockings and LMWH
Control group: LMWH

Outcomes

Primary outcome: asymptomatic lower‐limb vein thrombosis, as detected by DUS repeated every 3–5 days after surgery until discharge from the hospital or death

Secondary outcomes: isolated calf muscle DVT; proximal DVT; symptomatic PE; fatal PE; total VTE events; postoperative mortality; leg skin injury; combination of major and clinically relevant non‐major bleeding; and compliance with IPC), and those obtained at 30th and 180th POD during the outpatient follow‐up (i.e. combination of symptomatic, asymptomatic venous thrombosis of the lower limbs and symptomatic PE; VTE‐related mortality; and non‐VTE‐related mortality

Funding

Research funding from Cardinal Health Ltd

Declarations of interest

KL reports receiving grant support/honoraria from Cardinal Health, Sanofi Aventis, Sigvaris; VicB reports receiving grant support/honoraria from Sigvaris; LL reports receiving grant support/honoraria from Sigvaris. The remaining authors declare no competing financial interests

Notes

DVT occurred in 29 participants of the control group per article supplementary materials. The main article reports on 34 participants developing thrombosis, but this figure included 5 participants with superficial vein thrombosis per supplementary materials

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Based on the number of hospital medical records: those with an even last digit were assigned to the experimental group, and those with an odd last digit were assigned to the control group; if the last digit was zero, the penultimate digit was used

Allocation concealment (selection bias)

High risk

Based on the number of hospital medical records: those with an even last digit were assigned to the experimental group, and those with an odd last digit were assigned to the control group; if the last digit was zero, the penultimate digit was used

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of a placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study. Exception is DVT (Quote: "the blinded expert who performed a duplex ultrasound scan (DUS) had no access to the original medical record or allocation list; he used only individual patients code for identification. Also, to achieve blindness, most duplex scans were performed in a separate room away from the patient’s bed. If it was impossible to transfer a patient, the DUS was performed at the bed, but the IPC device was removed before the blinded expert’s visit.")

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Results were provided for all outcomes

Other bias

Low risk

None detected

Nakagawa 2020

Study characteristics

Methods

Study design: RCT
Method of randomisation: computerised randomisation
Concealment of allocation: fax to the data centre
Exclusions: 5
Losses to follow‐up: none
ITT analysis: no

Participants

Country: Japan
Number of participants: 121, intervention group 61; control group 60
Age (mean, years): intervention group 69; control group 67
Sex: 55/116 (47.4%) male
Inclusion criteria: male or female patients were eligible for this study if they were ≥ 20 years of age (no upper age limit was applied); they were undergoing curative laparoscopic surgery for colorectal cancer (cecum, ascending colon, transverse colon, descending colon, sigmoid colon and rectosigmoid junction); and they had no signs of metastasis on preoperative diagnostic imaging and no DVT on screening lower extremity venous US within 28 days before registration
Exclusion criteria: patients were excluded from this study if they had received preoperative therapy such as radiotherapy or chemotherapy; they had evidence of thromboembolic disease, or they had hypersensitivity to heparin or HIT; history of VTE, anticoagulant or antiplatelet medication, active bleeding, impaired renal function, or signs of acute bacterial endocarditis; patients who were deemed unsuitable for participation in the study by the investigator

Interventions

Intervention group: IPC and LMWH enoxaparin
Control group: IPC

Outcomes

Primary endpoint: incidence of VTE (DVT or PE) 28 days after surgery in the efficacy analysis population

Secondary endpoint: incidence of all bleeding events, as a composite endpoint that consisted of the incidence of major or minor bleeding events

Funding

None

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computerised randomisation

Allocation concealment (selection bias)

Low risk

Fax to the data centre

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study without use of placebo

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Small number of exclusions

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Obitsu 2020

Study characteristics

Methods

Study design: RCT
Method of randomisation: interactive web response system
Concealment of allocation: interactive web response system
Exclusions: a large number of exclusions in both groups
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Japan
Number of participants: 400, intervention group 199; control group 201
Age (median, years): intervention group 67; control group 64
Sex: 214 (61.7%) male in the full analysis set following exclusions
Inclusion criteria: patients aged ≥ 40 years, with good performance status, who planned curative laparoscopic surgery for histologically diagnosed gastric and colorectal cancer; before recruitment for this study, patients diagnosed as not VTE, by contrast‐enhanced CT and venous sonography of the leg, were selected as candidates
Exclusion criteria: patients suffering from serious systemic diseases; patients with coexisting/clinical signs of VTE or past history of VTE within 1 year; patients who received administration of anticoagulants and/or antiplatelet agents such as warfarin, aspirin, and clopidogrel sulphate before operation; obese patients with BMI ≥ 30 kg/m2; patients who received neoadjuvant chemo/radiotherapy before the operation; and patients with abnormal high values of D‐dimer (≥ 10 mg/mL) within 4 weeks before the operation

Interventions

Intervention group: IPC with elastic stockings and LMWH enoxaparin
Control group: IPC with elastic stockings

Outcomes

Primary endpoint: incidence of VTE, including DVT and PE. The risk of adverse events, including hemorrhagic events was also investigated

Funding

Tohoku Surgical Clinical Research Promotion Organization study group

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Interactive web‐response system

Allocation concealment (selection bias)

Low risk

Interactive web‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study without use of placebo

Incomplete outcome data (attrition bias)
All outcomes

High risk

Large number of exclusions

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Patel 2020

Study characteristics

Methods

Study design: RCT
Method of randomisation: computer‐generated randomisation schedule
Concealment of allocation: a computer‐generated randomisation schedule was created by the blinded primary study biostatistician (BJT). Treatment groups were assigned in a 1:1 ratio with concealment until individual randomisation was performed on the day of surgery
Exclusions: 1 participant
Losses to follow‐up: 1 participant
ITT analysis: yes

Participants

Country: USA
Number of participants: 501, intervention group 251; control group 250
Age (median, years): intervention group 63; control group 61
Sex: 501 (100%) male
Inclusion criteria: men 18‐100 years of age with histologically confirmed prostate cancer of any stage undergoing RP; normal preoperative coagulation blood test (prothrombin time); patients who would have otherwise been eligible to receive routine post‐RP care

Exclusion criteria: active treatment for VTE; patients judged by their urologist or preoperative evaluation centre to be unsafe to forgo pharmacologic prophylaxis or systemic anticoagulation postoperatively (whether or not they are on systematic anticoagulation for indications other than VTE); known adverse reactions to heparin (HIT or any allergy); epidural analgesia; spinal anaesthesia; participation in a different study that increases a patient’s risk of VTE

Interventions

Intervention group: IPC and UFH
Control group: IPC

Outcomes

Primary outcome: incidence of symptomatic VTE

Secondary outcome: overall incidence of VTE (asymptomatic or symptomatic) determined from the screening US subcohort

Primary safety outcomes: incidence of symptomatic lymphocele, symptomatic haematoma, or bleeding after surgery

Secondary outcomes: estimated blood loss from surgery, total surgical drain output after surgery (for participants with surgical drains), complications, and surveillance imaging bias

Funding

James Buchanan Brady Urological Institute Minimally Invasive Urology Fund

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation schedule

Allocation concealment (selection bias)

Low risk

A computer‐generated randomisation schedule was created by the blinded primary study biostatistician (BJT). Treatment groups were assigned in a 1:1 ratio with concealment until individual randomisation was performed on the day of surgery

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study without use of placebo

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Ramos 1996

Study characteristics

Methods

Study design: RCT 
Method of randomisation: table of random numbers
Concealment of allocation: not reported
Exclusions post randomisation: intervention group 57; control group 178
Losses to follow‐up: yes
ITT analysis: no

Participants

Country: USA
Number of participants: randomised 2786, completed 2551
Age (mean, years): 63.9
Sex: male 1782; female 769
Inclusion criteria: open heart surgery
Exclusion criteria: known prior DVT; bleeding complications; intraoperative death; intolerance to IPC; or withdrawal of prophylaxis before full ambulation

Interventions

Intervention group: UFH (5000 IU twice daily, sc) and SCDs
Control group: UFH (5000 IU twice daily, sc)

Outcomes

Symptomatic PE, confirmed by ventilation perfusion scan and/or pulmonary angiography

Funding

Not reported

Declarations of interest

Not reported

Notes

Both prophylactic methods were started immediately after surgery and continued for 4‐5 days or until participants were fully ambulatory

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A table of random numbers was used

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo device was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing the pulmonary ventilation‐perfusion scans or angiograms were aware of which participants used a compression device

Incomplete outcome data (attrition bias)
All outcomes

High risk

A large number of participants were excluded after randomisation

Selective reporting (reporting bias)

Low risk

PE was the only VTE event stated in methodology and was reported

Other bias

Low risk

Baseline characteristics were comparable

Sakai 2016

Study characteristics

Methods

Study design: RCT
Method of randomisation: computer‐generated sequence
Concealment of allocation: sealed envelopes
Exclusions post randomisation: none
Losses to follow‐up: 2 participants
ITT analysis: no

Participants

Country: Japan
Number of participants: randomised 122, completed 120
Age (mean, years): 73.7
Sex: male 20; female 100
Inclusion criteria: patients (aged ≥ 20 years) undergoing knee replacement surgery for primary joint disease including osteoarthritis and rheumatoid arthritis
Exclusion criteria: the presence of predefined risk factors for bleeding, coagulation disorders, heart failure (NYHA class III or IV), significant renal dysfunction (creatinine clearance < 30 mL/min), and abnormalities in biochemical measurements (aspartate aminotransferase or alanine aminotransferase ≥ 5 times the upper limit of normal or total bilirubin ≥ 2 times the upper limit of normal); patients were also excluded if they were scheduled to undergo bilateral joint replacement or reoperation, were unable to walk, or had uncontrolled cardiovascular disease

Interventions

Intervention group: edoxaban (15 mg or 30 mg once daily) and a foot pump (A‐V Impulse System foot pump)
Control group: edoxaban (15 mg or 30 mg once daily)

Outcomes

Symptomatic VTE by postoperative day 28 and asymptomatic DVT on compression US on the POD 10

Bleeding: major bleeding was defined as wound haematoma or haemorrhage occurring at a critical site and bleeding required for > 2 units of RBC concentrates. Minor bleeding was defined as bleeding that did not fulfil the criteria for major bleeding

Funding

National Hospital Organization (NHO), Japan

Declarations of interest

None declared

Notes

Both groups also used bilateral knee‐high antithromboembolic stockings

The foot pump was activated in the recovery room and used for 4 days

Edoxaban started 12 h postoperatively and was used for a mean of 11.5 days

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated sequence

Allocation concealment (selection bias)

High risk

Sealed enveloped contained the randomisation slip, but no statement that these were opaque

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo device was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information was provided on who performed the US and if that person was blinded to participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Minimal losses to follow‐up

Selective reporting (reporting bias)

Low risk

DVT and PE were VTE events stated in methodology and were reported

Other bias

High risk

Study stopped prematurely

Sang 2018

Study characteristics

Methods

Study design: quasi‐randomised trial
Method of randomisation: "..consecutive random numbers were selected from a random number table, assigned to each patient, and then divided by four. When the remainder was 0, 1, 2, and 3, the patient was allocated to groups A, B, C, and D, respectively. The patients were included sequentially according to the date of surgery."
Concealment of allocation: the patients were included sequentially according to the date of surgery
Exclusions: none stated
Losses to follow‐up: none stated
ITT analysis: yes

Participants

Country: China
Number of participants: 477, intervention group 162; control group 1 (LMWH, 162); control group 2 (IPC, 153). Excluding 159 patients receiving only elastic stockings
Age (mean, years): intervention group 53.3; control group 1 54.7; control group 2 52.6
Sex: 477 (100%) female
Inclusion criteria: age >18 years; carrying ≥ 1 risk factor for postoperative VTE; not taking any prophylactic measures before enrolment; willing to sign a written informed consent form; gynaecologic diseases may be malignant or benign (malignant diseases included malignancies of the ovary, uterine body, uterine cervix, vulva, and other parts of the pelvis; benign diseases included uterine myoma, uterine adenomyoma, ovarian benign tumours, pelvic floor prolapsed, and others such as hydrosalpinx, fallopian tube abscess, encapsulated effusion, and mesosalpinx cyst)
Exclusion criteria: preoperative thrombophlebitis or PE; preoperative acute lower extremity venous thrombosis; preoperative thrombocytopenia (PLT count < 100×109/L) or coagulation disorders; usage of anticoagulant drugs such as aspirin within 1 month; bleeding tendency as revealed by coagulation indexes or previous intracranial or gastrointestinal bleeding; congestive heart failure or pulmonary oedema; serious leg abnormalities (such as dermatitis, gangrene, or recent skin grafting), severe lower limb vascular atherosclerosis, lower limb ischaemic vascular disease, or severe leg deformity; imperception of dorsalis pedis artery pulse

Interventions

Intervention group: IPC with elastic stockings and LMWH dalteparin
Control group 1: dalteparin with elastic stockings
Control group 2: IPC with elastic stockings

Outcomes

Screening of VTE was performed within 7 days before and 3–5 days after surgery. Initially, colour DUS imaging of lower extremities was performed by an experienced professional using an LEGIQ E9 colour Doppler system with the probe frequency set at 8.4–9 MHz. If VTE was found or suspected, lower limb venography was performed to confirm the presence of DVT, and CTPA was performed to detect if PE was present.
Bleeding events, classified as major and minor haemorrhagic events

Funding

The Capital Health Research and Development of Special Project (No. 2011‐2003‐03)

Declarations of interest

None

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quote: "A simple randomization method was adopted for this study. Based on the calculated sample size (see below), 660 consecutive random numbers were selected from a random number table, assigned to each patient, and then divided by four. When the remainder was 0, 1, 2, and 3, the patient was allocated to groups A, B, C, and D, respectively. The patients were included sequentially according to the date of surgery."

Allocation concealment (selection bias)

High risk

The patients were included sequentially according to the date of surgery

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study without use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study without use of placebo

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Results provided for all outcomes

Other bias

Low risk

None detected

Sieber 1997

Study characteristics

Methods

Study design: CCT
Method of randomisation: none
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 579
Age (mean, years): 65
Sex: male
Inclusion criteria: patients who had pelvic lymphadenectomy with or without radical RP
Exclusion criteria: none

Interventions

Intervention group: UFH (5000 IU twice daily, sc) and IPC (SCDs)
Control group: IPC (SCDs)

Outcomes

Symptomatic DVT or PE

Funding

American Foundation for Urologic Diseases

Declarations of interest

Not reported

Notes

Participants were assigned to heparin and control groups by the primary surgeon

Sequential compressive stockings were placed at the time of surgery and left in place for 48 h after surgery for all participants

Heparin was started preoperatively and continued for 3 days

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Allocation concealment (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo injection for heparin was given

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No exclusions or withdrawals were reported

Selective reporting (reporting bias)

Low risk

DVT and PE were VTE events stated in methodology and were reported

Other bias

Unclear risk

Insufficient details were provided to allow a conclusion to be made

Silbersack 2004

Study characteristics

Methods

Study design: RCT
Method of randomisation: not reported
Concealment of allocation: not reported
Exclusions post‐randomisation: 8
Losses to follow‐up: none
ITT analysis: no

Participants

Country: Germany
Number of participants: 131 (139 randomised)
Age (mean, years): 64
Sex: male 47; female 84
Inclusion criteria: primary unilateral THR or TKR
Exclusion criteria: heart failure NYHA class III/IV; stage III chronic renal insufficiency; severe PAD; acute thrombophlebitis; neurological disorders or arthrodeses of the lower limbs; recent anticoagulation; haemorrhagic diathesis; allergy to heparins; or active malignant disease

Interventions

Intervention group: LMWH enoxaparin (40 mg daily, sc) and pneumatic sequential compression
Control group: LMWH enoxaparin (40 mg daily, sc) and class‐I GCS

Outcomes

Symptomatic and asymptomatic DVT (on US)

Funding

Aircast Europa GmbH

Declarations of interest

Not reported

Notes

The calf cuffs were applied to both lower limbs directly after the operation in the recovery room and the system was activated. The use of the IPC was continued until POD 10 whenever the participant was in bed

Enoxaparin was started the evening before surgery and continued for 30 days

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

A placebo device was not used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Colour DUS was performed by an independent angiologist who was unaware of the patients' participation in the study or of the method of prophylaxis, but only to confirm the findings of compression US, which was not reported to be performed by a blinded or not observer, hence unclear risk of bias

Incomplete outcome data (attrition bias)
All outcomes

Low risk

8 patients who were randomised were subsequently excluded (2 from the LMWH/IPC and 6 from the LMWH/GCS group) for various reasons, but they represent a small percentage of the total participant number, unlikely to change the results and conclusions whatever their outcome might have been

Selective reporting (reporting bias)

Low risk

Thromboembolic (VTE) events were stated in methodology to be the outcome measures of the study and they were reported as such

Other bias

Low risk

Baseline characteristics were comparable

Siragusa 1994

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: unclear
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Italy
Number of participants: 70
Age (mean, years): not provided
Sex: not provided
Inclusion criteria: elective hip replacement
Exclusion criteria: not provided

Interventions

Intervention group: UFH + IPC
Control group: UFH

Outcomes

DVT on venography

Funding

Not reported

Declarations of interest

Not reported

Notes

No information on start and discontinuation of IPC or UFH

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

A placebo device was not used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No exclusions or withdrawals were reported

Selective reporting (reporting bias)

Low risk

DVT was the only VTE event stated in methodology and was reported

Other bias

Unclear risk

Insufficient details were provided to allow a conclusion to be made

Stannard 1996

Study characteristics

Methods

Study design: RCT
Method of randomisation and concealment of allocation: unclear
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 75
Age (mean, years): 67.4
Sex: not reported
Inclusion criteria: patients undergoing elective uncemented hip arthroplasty
Exclusion criteria: not provided

Interventions

Intervention group: UFH/aspirin and IPC (foot pump)
Control groups: UFH/aspirin or IPC (foot pump)

Outcomes

Asymptomatic DVT, symptomatic DVT, any DVT, PE

Funding

Not reported

Declarations of interest

Not reported

Notes

The pumps were started in the recovery room immediately after surgery and used until the end of the study, with the exact time not specified

Heparin was started 8 h before the operation and after 3 days of use it was replaced with 325 mg aspirin twice daily for an undefined duration

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo for compression, heparin and aspirin

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All duplex results and venograms were read by one of the study authors who was blinded to the prophylactic modality used

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participants were lost to follow‐up

Selective reporting (reporting bias)

Low risk

DVT was stated in methodology to be the outcome measure of the study and results were reported

Other bias

Low risk

Baseline characteristics were comparable

Tsutsumi 2012

Study characteristics

Methods

Study design: CCT
Method of randomisation: none
Concealment of allocation: not reported
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Japan
Number of participants: 137
Age (mean, years): 66.1 (calculated)
Sex: 83 men, 54 women
Inclusion criteria: patients with colorectal cancer undergoing elective resection surgery under general anaesthesia, regardless of tumour stage
Exclusion criteria: clinical signs of DVT, active bleeding, active GI ulceration, haemorrhagic stroke, contraindication for anticoagulation, indwelling epidural catheter, renal failure and inability to receive IPC

Interventions

Intervention group: IPC (stopped 24 h after surgery) combined with fondaparinux (sc injections of fondaparinux at 2.5 mg once daily)
Control group: IPC (stopped 24 h after surgery)

Outcomes

Clinically evident DVT and PE

Bleeding: major bleeding was defined as bleeding that was fatal, retroperitoneal, intracranial, involving any other critical organ, led to intervention being discontinued, or was associated with a need for transfusion of > 3 units of packed RBC. Other types of bleeding was included and defined as bleeding that did not fulfil the criteria for major bleeding

Funding

Not reported

Declarations of interest

Not reported

Notes

IPC was used for 24 h after surgery, but no information on when it was started

Fondaparinux was started 24 h after surgery and was continued until days 5‐7

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Allocation concealment (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo for fondaparinux

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All enrolled participants had results reported

Selective reporting (reporting bias)

Low risk

Thromboembolic events (DVT and PE) were stated in methodology to be the outcome measures of the study and they were reported as such

Other bias

Low risk

Baseline characteristics were comparable

Turpie 2007

Study characteristics

Methods

Study design: randomised, double‐blind, placebo‐controlled, superiority trial
Method of randomisation: centralised computer‐generated schedule (1:1 randomisation in blocks of 4 and stratified by centre)
Concealment of allocation: yes
Exclusions post‐randomisation: 24
Losses to follow‐up: none
ITT analysis: no

Participants

Country: USA
Number of participants: 1309 randomised, 1285 randomised and treated
Age: median age 59 and 60 years in the control and treatment groups, respectively
Sex: male 635; female 650
Inclusion criteria: abdominal surgery expected to last > 45 min in patients aged over 40 years; or patients weighing > 50 kg
Exclusion criteria: vascular surgery with evidence of leg ischaemia caused by peripheral vascular disease; unable to receive IPC or elastic stockings; pregnant women and women of childbearing age not using effective contraception; life‐expectancy < 6 months; clinical signs of DVT and/or history of VTE within the previous 3 months; active bleeding; documented congenital or acquired bleeding disorder; active ulcerative GI disease (unless it was the reason for the present surgery); haemorrhagic stroke or surgery on the brain, spine or eyes within the previous 3 months; bacterial endocarditis or other contraindications for anticoagulant therapy; planned indwelling intrathecal or epidural catheter for > 6 h after surgical closure; unusual difficulty in achieving epidural or spinal anaesthesia; known hypersensitivity to fondaparinux or iodinated contrast medium; current addictive disorders; serum creatinine concentration > 2.0 mg/dL in a well‐hydrated patient; PLT count below 100,000 mm; or patients requiring anticoagulant therapy or other pharmacologic prophylaxis besides IPC

Interventions

Intervention group: IPC and fondaparinux 
Control group: IPC

Outcomes

VTE (defined as DVT detected by mandatory screening and/or documented symptomatic DVT or PE, or both) and individual components up to day 10. Symptomatic VTE up to day 10 and day 32

Major bleeding (defined as bleeding that was fatal, retroperitoneal, intracranial, or involved any other critical organ, led to intervention being discontinued, or was associated with a bleeding index of ≥ 2.0) detected during the treatment period

Death during the treatment period and up to day 32

Funding

Sanofi‐Synthélabo and GlaxoSmithKline

Declarations of interest

Not reported

Notes

Study medications were packaged in boxes of identical appearance
Of the 1309 randomised participants, 842 (64.3%) had an evaluable venogram performed and were included in the primary efficacy analysis
Major bleeding occurred in 10 participants (1.6%) and 1 participant (0.2%) of the intervention and control groups, respectively (P = 0.006)
During the on‐study‐drug period of 5–9 days, all participants were to receive VTE prophylaxis with IPC using any type of device, except a foot pump, for a duration left to the investigator's discretion. The first injection of fondaparinux or placebo was scheduled 6–8 h after surgical closure, provided that haemostasis was achieved. The duration of the on‐study‐drug period was 5–9 days. If the participant was discharged from hospital before completing the on‐study‐drug period, a visiting nurse administered the remaining study injections

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centralised computer‐generated schedule randomisation (1:1 randomisation in blocks of four and stratified by centre)

Allocation concealment (selection bias)

Low risk

Centralised computer‐generated schedule randomisation

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Use of placebo injections

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Reports double‐blind (use of placebo injections) but it is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

A large number of exclusions in both study arms, around 35% of the total number of participants, mainly because of lack of mandatory or interpretable venography

Selective reporting (reporting bias)

Low risk

DVT and PE were the primary efficacy outcomes and they were reported in the results

Other bias

Low risk

Demographic variables and risk factors at baseline, type of anaesthesia, and type and duration of surgery were similar in the 2 groups among both randomised and treated participants (Tables 1 and 2) and among participants analysed for primary efficacy

Westrich 2005

Study characteristics

Methods

Study design: CCT
Method of randomisation: none
Concealment of allocation: none
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 200
Age (mean, years): 81.3
Sex: male 42; female 158
Inclusion criteria: patients > 60 years who sustained a fragility fracture to the hip; and an ability and willingness to comply with the mechanical and chemical prophylaxis protocol
Exclusion criteria: patients < 60 years; history of severe allergy to aspirin or warfarin; refusal to use the pneumatic compression device; multiple trauma injuries; or patients with a hip fracture that did not require surgical treatment

Interventions

Intervention group: IPC and warfarin
Control group: IPC and aspirin

Outcomes

DVT on US of the ipsilateral lower external iliac, common femoral, superficial femoral, deep femoral, and popliteal veins
Bleeding: all participants assessed for postoperative bleeding, no specific bleeding definition provided

Funding

Not reported

Declarations of interest

Not reported

Notes

No symptomatic VTE was observed

3 participants on warfarin developed bleeding complications

The IPC device was applied over the duration of the participant's preoperative and postoperative stay until the time of discharge. Participants sent to a rehabilitation centre were told to continue using the IPC until their final discharge home. Warfarin or aspirin started on the night before surgery but no duration of use was provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Allocation concealment (selection bias)

High risk

The type of the study (CCT) makes it high risk for selection bias

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not a double‐blind study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No exclusions/participants lost to follow‐up

Selective reporting (reporting bias)

Low risk

DVT and PE were the main study outcomes and they were reported in the results

Other bias

Unclear risk

Insufficient details were provided to allow a conclusion to be made

Westrich 2006

Study characteristics

Methods

Study design: RCT
Method of randomisation: not reported
Concealment of allocation: not reported
Exclusions post randomisation: 11
Losses to follow‐up: 73
ITT analysis: no

Participants

Country: USA
Number of participants: 275
Age (mean, years): 69
Sex: male 99; female 176
Inclusion criteria: unilateral TKA
Exclusion criteria: allergies to aspirin; congenital or acquired bleeding disorders; active ulcerative or angiodysplastic GI disease; multiple myeloma or other paraproteinemias; pheochromocytoma; hyperthyroidism; impaired renal function; known hepatic disease; past medical history of stroke; recent brain, spinal, or ophthalmologic surgery; hypersensitivity to enoxaparin; cardiac complications; severe peripheral vascular diseases; chronic heart failure; severe varicose veins; history of DVT and/or PE

Interventions

Intervention group: IPC and LMWH enoxaparin
Control group: IPC and aspirin

Outcomes

DVT on US before discharge on PODs 3‐5, and 4‐6 weeks after surgery

Funding

Aventis, Bridgewater, NJ, USA and Aircast, Summit, NJ, USA

Declarations of interest

Not reported

Notes

Bleeding complications were documented, no specific bleeding definitions provided

Upon their arrival in the recovery room, the participants received a VenaFlow calf compression device that was placed on both of their lower extremities. The compression device was used during each participant's entire hospital stay

Enoxaparin was initiated 2 h after epidural catheter removal (approximately 48 h postoperatively). Participants received 30 mg of enoxaparin twice daily until their hospital discharge; upon discharge, their dosage was changed to 40 mg once daily for 3 weeks. Aspirin started on the night of their surgery in the recovery room and was continued for 4 weeks postoperatively

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not a blinded study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

A large number of participants were lost to follow‐up, likely to affect outcome results

Selective reporting (reporting bias)

Low risk

DVT was the main study outcome and was reported in the results

Other bias

Low risk

Baseline characteristics were comparable

Windisch 2011

Study characteristics

Methods

Study design: RCT
Method of randomisation: not provided
Concealment of allocation: not provided
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: Germany
Number of participants: 80
Age (mean, years): 68.5 (calculated)
Sex: not provided
Inclusion criteria: patients undergoing TKR (primary diagnosis of knee "arthritis")
Exclusion criteria: patients aged < 60 years, BMI > 40 or < 25, existing acute DVT, thrombophlebitic varicosis (stages II – IV acc. Marshall), venous insufficiency (stages 2–3 according to Widmer)

Interventions

Intervention group: LMWH enoxaparin (40 mg once daily, beginning 24 h prior to the operation) and IPC (foot pump)
Control group: LMWH enoxaparin (40 mg once daily, beginning 24 h prior to the operation)

Outcomes

DVT on DUS, but also clinically evident DVT and PE

Funding

Not reported

Declarations of interest

Not reported

Notes

Reports none of the participants needed to be operated upon for hemarthrosis, no other details regarding bleeding were provided

The A‐V Impulse System foot pump was attached in the recovery room to both feet of the participants only shortly after completion of the operation; participants were free to discontinue its use at will

Enoxaparin was started 24 h before surgery, duration was not provided

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

A placebo device was not used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Sonographers were unaware of treatment allocations

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No exclusions/participants lost to follow‐up

Selective reporting (reporting bias)

Low risk

DVT and PE were VTE events stated in methodology and were reported

Other bias

Low risk

There were no baseline imbalances

Woolson 1991

Study characteristics

Methods

Study design: RCT
Method of randomisation: sealed envelopes
Concealment of allocation: sealed envelopes
Exclusions post randomisation: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: USA
Number of participants: 196 patients who had 217 procedures
Age (mean, years): 65
Sex: male 95 procedures; female 122 procedures
Inclusion criteria: primary or revision THA
Exclusion criteria: allergy to aspirin or warfarin; recent peptic ulcer or other bleeding diathesis; receiving any drug that affects PLT function within 2 weeks before the operation; or patients expected to remain in bed for > 4 days after the operation

Interventions

Intervention group: IPC, thigh‐high graduated elastic compression stockings, and warfarin (first group); or IPC, thigh‐high graduated elastic compression stockings and aspirin (second group)
Control group: IPC with thigh‐high graduated elastic compression stockings

Outcomes

Proximal DVT on venography, B‐mode US, or both, on discharge
Symptomatic DVT or PE, objectively diagnosed

Funding

None

Declarations of interest

None

Notes

Warfarin dose was 7.5 or 10 mg orally on the evening before the operation, then titrated to maintain the prothrombin time at 1.2 to 1.3 times the control value. Aspirin started the evening before surgery and continued at a dose of 650 mg twice daily. For both agents duration of use was not reported

IPC was started in the operating theatre, as soon as the participant was draped and used until discharge
Follow‐up was at least 3 months for all participants

Bleeding: 1 participant in each of the 3 groups had a wound haematoma, that required evacuation in the 2 intervention group participants but not in the control group. No specific definition of bleeding provided
No complications related to the use of the elastic stockings or pneumatic compression were reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Use of sealed envelope method

Allocation concealment (selection bias)

Unclear risk

Does not mention if the sealed envelopes were sequentially numbered and opaque

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not a blinded study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It is unclear if the personnel performing diagnostic testing were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participants were lost to follow‐up

Selective reporting (reporting bias)

Low risk

DVT was the main study outcome and was reported in the results

Other bias

Low risk

There were no baseline imbalances

Yokote 2011

Study characteristics

Methods

Study design: RCT
Method of randomisation: not provided
Concealment of allocation: not provided
Exclusions: none
Losses to follow‐up: 3 participants withdrawn after randomisation
ITT analysis: yes

Participants

Country: Japan
Number of participants: 255 randomised
Age (mean, years): 63.3 (calculated)
Sex: 204 female and 46 male
Inclusion criteria: elective primary unilateral THR
Exclusion criteria: bilateral and revision procedures, patients who were < 20 years of age, long‐term anticoagulation treatment such as UFH, LMWH, vitamin K antagonists, antiplatelet agents for pre‐existing cardiac or cerebrovascular disease, a history of VTE, a coagulation disorder including antiphospholipid syndrome, the presence of a solid malignant tumour or a peptic ulcer, and major surgery in the preceding three months. White patients were also excluded

Interventions

Intervention groups:
1. IPC and LMWH enoxaparin (20 mg twice daily)
2. IPC and fondaparinux (2.5 mg once daily)
Control group: IPC

Outcomes

DVT on DUS and also clinically evident DVT and PE

Any bleeding, both major or minor. Major bleeding: retro‐peritoneal, intracranial or intraocular, or if associated with either death, transfusion of > 2 units of packed RBC or whole blood (except autologous), a reduction in the level of haemoglobin of > 2 g/dL, or a serious or life‐threatening clinical event requiring medical intervention. Suspected intra‐abdominal or intracranial bleeding was confirmed by US, CT or MRI. Minor bleeding: epistaxis lasting for > 5 min or requiring intervention, ecchymosis or haematoma with a maximum size of > 5 cm, haematuria not associated with trauma from the urinary catheter, GI haemorrhage not related to intubation or the passage of a nasogastric tube, a wound haematoma or haemorrhagic wound complications not associated with major haemorrhage or subconjunctival haemorrhage, requiring cessation of medication

Funding

None

Declarations of interest

None

Notes

The pneumatic devices were initiated in the operating theatre (before surgery for the contralateral leg and just after surgery for the operated leg) and removed on the "second post‐operative day when the day of surgery was defined as post‐operative day 1"

Pharmacological prophylaxis was started postoperatively

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details provided

Allocation concealment (selection bias)

Unclear risk

No details provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Use of placebo

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Scans were read by experienced radiologist blinded to randomisation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

A small percentage of exclusions (5/255, 2%)

Selective reporting (reporting bias)

Low risk

DVT and PE were the main study outcomes and they were reported in the results

Other bias

Low risk

Baseline characteristics were comparable

Zhou 2020

Study characteristics

Methods

Study design: RCT
Method of randomisation: unclear
Concealment of allocation: unclear
Exclusions: none
Losses to follow‐up: none
ITT analysis: yes

Participants

Country: China
Number of participants: 92, intervention group 46; control group 46
Age (mean, years): intervention group 46.62; control group 46.52
Sex: 92 (100%) female
Inclusion criteria: patients diagnosed with ovarian cancer by 2 senior pathologists through postoperative pathological sections; patients receiving no adjuvant chemotherapy and radiotherapy; patients with surgery times of 2‐5 h; patients with clear minds; patients who voluntarily signed the consent form
Exclusion criteria: patients with mental diseases; patients with language communication impairments; patients with severe cardiovascular or cerebrovascular diseases; patients with poor compliance; patients allergic to LMWH; patients with complications such as coagulation dysfunction or varicose veins; patients with previous hyperlipidaemia, hypertension, or diabetes; patients with previous DVT or other high‐risk diseases; patients with other malignant tumours

Interventions

Intervention group: IPC with elastic stockings and LMWH enoxaparin
Control group: LMWH enoxaparin

Outcomes

DVT, PE, plasma D‐dimer levels, PLT count, PT, APTT, postoperative lower‐limb pain and swelling, patient satisfaction

Funding

Not reported

Declarations of interest

None

Notes

Although labelled as "double‐blind", this study did not use a placebo device to qualify as such

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details were provided

Allocation concealment (selection bias)

Unclear risk

No details were provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unclear if outcome assessors were aware of participant allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition

Selective reporting (reporting bias)

Low risk

Resuts reported for all outcomes 

Other bias

Low risk

None detected

ABI: ankle‐brachial index; APTT: activated partial thromboplastin time; ASA: American Society of Anesthesiology score; BMI: body mass index; CCT: controlled clinical trial; CT: computed tomography; CTPA: computed tomographic pulmonary angiography; CVC: central venous catheter; CVI: chronic venous insufficiency; DIC: disseminated intravascular coagulation; DUS: duplex ultrasound scan; DVT: deep vein thrombosis; ECOG: Eastern Cooperative Oncology Group; GCS: graduated compression stockings; GI: gastrointestinal; h: hours; HIT: heparin‐induced thrombocytopenia; ICU: intensive care unit; IPC: intermittent pneumatic compression; IPG: impedence plethysmography; ITT: intention‐to‐treat; IU: international units;IVC: inferior vena cava; LMWH: low molecular weight heparin; LOS: length of stay; MRI: magnetic resonance imaging; NYHA: New York Hospital Association; PAD: peripheral arterial disease; PE: pulmonary embolism; PESI: pulmonary embolism severity index; POD: postoperative day; PLT: platelet; PT: prothrombin time; RBC: red blood cells; RCT: randomised controlled trial; RP: radical prostatectomy; sc: subcutaneously; SCD: sequential compression device; THA: total hip arthroplasty; THR: total hip replacement; TKA: total knee arthroplasty; TKR: total knee replacement; UFH: unfractionated heparin; US: ultrasonography; VCF: vena cava filter; VTE: venous thromboembolism; WHO: World Health Organization; wk: weeks

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Ailawadi 2001

Retrospective case‐control study

Eskander 1997

Use of combined modalities was not concurrent in the intervention group

Frim 1992

Controlled before and after study

Gagner 2012

Registry study, non‐randomised

Gelfer 2006

Pharmacological prophylaxis was not the same in the 2 study groups

Kamran 1998

Controlled before and after study

Kiudelis 2010

Investigation restricted to intraoperative period up to 10 min after extubation

Kumaran 2008

The control (single modality) group included participants who were allocated to heparin or pneumatic compression

Lieberman 1994

Pharmacological prophylaxis consisted of aspirin, which has limited thromboprophylactic properties

Macdonald 2003

Pharmacological prophylaxis was not the same in the 2 study groups

Mehta 2010

Only aggregated VTE rates and not separate DVT and PE rates were provided and the study authors did not reply when we requested individual data

Nathan 2006

Prospective case‐control study

Patel 2010

Retrospective study

Roberts 1975

Pneumatic compression was used only intraoperatively

Spinal cord injury investigators

Pharmacological prophylaxis was not the same in the 2 study groups

Stannard 2006

Use of enoxaparin was not concurrent in the 2 study groups

Tsutsumi 2000

Controlled before and after study

Wan 2015

Retrospective study

Westrich 1996

Pharmacological prophylaxis consisted of aspirin, which has limited thromboprophylactic properties

Whitworth 2011

Retrospective case‐control study investigating preoperative anticoagulation in patients on postoperative LMWH and SCDs

Winemiller 1999

Retrospective case‐control study

DVT: deep vein thrombosis; LMWH: low molecular weight heparin; PE: pulmonary embolism; SCD: sequential compression device; VTE: venous thromboembolism

Characteristics of ongoing studies [ordered by study ID]

ChiCTR1800014257

Study name

ChiCTR1800014257

Methods

Multicenter parallel RCT

Participants

Women undergoing gynaecologic pelvic surgery

Interventions

High‐risk patients: GCS (control group) vs GCS + LMWH

Very high‐risk patients: GCS+ IPC (control group) vs GCS + IPC + LMWH

Outcomes

Lower extremity venous DUS findings, haematology laboratory measures and CTPA findings

Starting date

1 January 2018

Contact information

Zhengyu Zhang

Notes

EUCTR2007‐006206‐24

Study name

EUCTR2007‐006206‐24

Methods

IPC with and without early anticoagulant treatment

Participants

Patients with acute primary intracerebral haemorrhage

Interventions

Blind randomised trial of IPC with and without early anticoagulant treatment

Outcomes

Not provided

Starting date

17 December 2008

Contact information

Not provided

Notes

Study ended on 30 June 2016, no results are available

NCT00740987 (CIREA 2)

Study name

NCT00740987 (CIREA 2)

Methods

RCT in ICU patients without high risk of bleeding

Participants

621 ICU patients

Interventions

Patients were randomised to use IPC or not

Outcomes

Primary outcome measures: combined criterion evaluated at day 6 ± 2 days after randomisation:
symptomatic VTE event, non‐fatal, objectively confirmed; death related to PE;
asymptomatic DVT of the lower limbs detected by CUS on day 6 (time frame: 6 ± 2 days)
Secondary outcome measures: symptomatic thromboembolic events occurred between day 6 and
day 90; total mortality evaluated at 1 month and 3 months (time frame: 6 days to 3 months)

Starting date

October 2007

Contact information

Karine Lacut, MD. CHU Brest France, Univ Brest, EA 3878

Notes

Study completed in January 2015, with no results being presented or published at the time of writing this review

NCT02271399

Study name

NCT02271399

Methods

Double‐blind, parallel group, RCT

Participants

Patients undergoing TKA

Interventions

Aspirin and IPC vs  rivaroxaban and IPC

Outcomes

VTE, bleeding

Starting date

October 2014

Contact information

Jin Kyu Lee

Notes

Competed on February 2016, no results have been published

NCT03559114 (PROTEST)

Study name

NCT03559114 (PROTEST)

Methods

Phase III, multi‐centre, double blind, RCT

Participants

Patients with traumatic brain Injury

Interventions

SCD versus SCS and dalteparin

Outcomes

Clinically important VTE, clinically‐important intracranial bleeding progression, objectively confirmed new or progressing intracranial bleeding on radiology, mortality at 7 days, 30 days, 180 days, delayed VTE after day 7, functional neurological outcome at day 30 as measured by Glasgow Outcome Scale Extended, functional neurological outcome at day 180 as measured by Glasgow Outcome Scale Extended, quality of life outcome at 30 days as measured by the EuroQol5D, quality of life outcome at 180 days as measured by the EuroQol5D

Starting date

19 July 2018

Contact information

Farhad Pirouzmand, MD, MSc, FRCSC

Notes

CTPA: computed tomography pulmonary angiogram; CUS: colour ultrasound; DUS: duplex ultrasound; DVT: deep vein thrombosis; GCS: graduated compression stockings; ICU: intensive care unit; IPC: intermittent pneumatic compression; LMWH: low molecular weight heparin; PE: pulmonary embolism; RCT: randomised controlled trial; SCD: sequential compression device; SCS: sequential compression stockings; TKA: total knee arthroplasty; VTE: venous thromboembolism

Data and analyses

Open in table viewer
Comparison 1. IPC plus pharmacological prophylaxis versus IPC alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Incidence of PE ‐ orthopaedic and non‐orthopaedic patients Show forest plot

19

5462

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

Analysis 1.1

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

1.1.1 Orthopaedic patients

3

445

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.1.2 Non‐orthopaedic patients

16

5017

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.2 Incidence of PE ‐ foot IPC or other IPC Show forest plot

19

5462

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

Analysis 1.2

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

1.2.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.2.2 Other IPC

18

5412

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.3 Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

18

5394

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

Analysis 1.3

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

1.3.1 Orthopaedic patients

3

445

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.38, 1.69]

1.3.2 Non‐orthopaedic patients

15

4949

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.31, 0.68]

1.4 Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

10

4089

Odds Ratio (M‐H, Fixed, 95% CI)

0.48 [0.21, 1.10]

Analysis 1.4

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

1.4.1 Orthopaedic patients

1

250

Odds Ratio (M‐H, Fixed, 95% CI)

1.50 [0.06, 37.33]

1.4.2 Non‐orthopaedic patients

9

3839

Odds Ratio (M‐H, Fixed, 95% CI)

0.44 [0.19, 1.04]

1.5 Incidence of DVT ‐ by foot IPC or other IPC Show forest plot

18

5395

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

Analysis 1.5

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 5: Incidence of DVT ‐ by foot IPC or other IPC

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 5: Incidence of DVT ‐ by foot IPC or other IPC

1.5.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.5.2 Other IPC

17

5345

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

1.6 Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

13

4634

Odds Ratio (M‐H, Fixed, 95% CI)

6.02 [3.88, 9.35]

Analysis 1.6

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

1.6.1 Orthopaedic patients

2

400

Odds Ratio (M‐H, Fixed, 95% CI)

2.79 [0.77, 10.18]

1.6.2 Non‐orthopaedic patients

11

4234

Odds Ratio (M‐H, Fixed, 95% CI)

6.61 [4.14, 10.56]

1.7 Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

12

4133

Odds Ratio (M‐H, Fixed, 95% CI)

5.77 [2.81, 11.83]

Analysis 1.7

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

1.7.1 Orthopaedic patients

2

400

Odds Ratio (M‐H, Fixed, 95% CI)

3.35 [0.13, 83.62]

1.7.2 Non‐orthopaedic patients

10

3733

Odds Ratio (M‐H, Fixed, 95% CI)

5.91 [2.83, 12.36]

Open in table viewer
Comparison 2. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Incidence of PE ‐ orthopaedic and non‐orthopaedic patients Show forest plot

15

6737

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.30, 0.71]

Analysis 2.1

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

2.1.1 Orthopaedic patients

8

1202

Odds Ratio (M‐H, Fixed, 95% CI)

0.58 [0.08, 4.49]

2.1.2 Non‐orthopaedic patients

7

5535

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.29, 0.71]

2.2 Incidence of PE ‐ foot IPC or other IPC Show forest plot

15

6737

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.30, 0.71]

Analysis 2.2

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

2.2.1 Foot IPC

4

324

Odds Ratio (M‐H, Fixed, 95% CI)

0.32 [0.01, 8.25]

2.2.2 Other IPC

11

6413

Odds Ratio (M‐H, Fixed, 95% CI)

0.47 [0.30, 0.72]

2.3 Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

17

6151

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.21, 0.70]

Analysis 2.3

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

2.3.1 Orthopaedic patients

10

3075

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.18, 0.68]

2.3.2 Non‐orthopaedic patients

7

3076

Odds Ratio (M‐H, Random, 95% CI)

0.46 [0.13, 1.61]

2.4 Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

7

3032

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.34, 2.01]

Analysis 2.4

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

2.4.1 Orthopaedic patients

3

477

Odds Ratio (M‐H, Fixed, 95% CI)

2.09 [0.38, 11.50]

2.4.2 Non‐orthopaedic patients

4

2555

Odds Ratio (M‐H, Fixed, 95% CI)

0.55 [0.18, 1.66]

2.5 Incidence of DVT ‐ foot IPC or other IPC Show forest plot

16

4148

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.18, 0.63]

Analysis 2.5

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 5: Incidence of DVT ‐ foot IPC or other IPC

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 5: Incidence of DVT ‐ foot IPC or other IPC

2.5.1 Foot IPC

4

324

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.05, 3.47]

2.5.2 Other IPC

12

3824

Odds Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.54]

2.6 Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

6

1314

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.56, 1.35]

Analysis 2.6

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

2.6.1 Orthopaedic patients

3

550

Odds Ratio (M‐H, Fixed, 95% CI)

0.64 [0.20, 2.07]

2.6.2 Non‐orthopaedic patients

3

764

Odds Ratio (M‐H, Fixed, 95% CI)

0.91 [0.57, 1.47]

2.7 Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

5

908

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.35, 4.18]

Analysis 2.7

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

2.7.1 Orthopaedic patients

3

551

Odds Ratio (M‐H, Fixed, 95% CI)

1.07 [0.21, 5.54]

2.7.2 Non‐orthopaedic patients

2

357

Odds Ratio (M‐H, Fixed, 95% CI)

1.42 [0.21, 9.49]

Open in table viewer
Comparison 3. IPC plus pharmacological prophylaxis versus IPC plus aspirin

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Incidence of PE Show forest plot

3

605

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.19]

Analysis 3.1

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 1: Incidence of PE

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 1: Incidence of PE

3.2 Incidence of DVT Show forest plot

3

605

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.48, 1.42]

Analysis 3.2

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 2: Incidence of DVT

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 2: Incidence of DVT

3.3 Incidence of symptomatic DVT Show forest plot

1

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 3: Incidence of symptomatic DVT

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 3: Incidence of symptomatic DVT

3.4 Incidence of bleeding Show forest plot

3

616

Odds Ratio (M‐H, Fixed, 95% CI)

1.23 [0.27, 5.53]

Analysis 3.4

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 4: Incidence of bleeding

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 4: Incidence of bleeding

3.5 Incidence of major bleeding Show forest plot

3

616

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.15, 4.17]

Analysis 3.5

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 5: Incidence of major bleeding

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 5: Incidence of major bleeding

Open in table viewer
Comparison 4. IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Incidence of PE Show forest plot

13

4159

Odds Ratio (M‐H, Fixed, 95% CI)

0.54 [0.28, 1.07]

Analysis 4.1

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 1: Incidence of PE

4.2 Incidence of DVT Show forest plot

13

4159

Odds Ratio (M‐H, Fixed, 95% CI)

0.53 [0.36, 0.77]

Analysis 4.2

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 2: Incidence of DVT

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 2: Incidence of DVT

4.3 Incidence of symptomatic DVT Show forest plot

7

3064

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.15, 9.63]

Analysis 4.3

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

4.4 Incidence of DVT by foot IPC or other IPC Show forest plot

17

5085

Odds Ratio (M‐H, Fixed, 95% CI)

0.52 [0.36, 0.74]

Analysis 4.4

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

4.4.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

4.4.2 Other IPC

16

5035

Odds Ratio (M‐H, Fixed, 95% CI)

0.52 [0.36, 0.74]

4.5 Incidence of bleeding Show forest plot

10

4120

Odds Ratio (M‐H, Fixed, 95% CI)

5.45 [3.38, 8.80]

Analysis 4.5

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 5: Incidence of bleeding

4.6 Incidence of major bleeding Show forest plot

9

3619

Odds Ratio (M‐H, Fixed, 95% CI)

5.90 [2.82, 12.33]

Analysis 4.6

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Open in table viewer
Comparison 5. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Incidence of PE Show forest plot

11

5758

Odds Ratio (M‐H, Fixed, 95% CI)

0.45 [0.29, 0.70]

Analysis 5.1

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 1: Incidence of PE

5.2 Incidence of DVT Show forest plot

13

5172

Odds Ratio (M‐H, Random, 95% CI)

0.44 [0.22, 0.87]

Analysis 5.2

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 2: Incidence of DVT

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 2: Incidence of DVT

5.3 Incidence of symptomatic DVT Show forest plot

5

2312

Odds Ratio (M‐H, Fixed, 95% CI)

1.02 [0.29, 3.54]

Analysis 5.3

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

5.4 Incidence of DVT by foot IPC or other IPC Show forest plot

15

5431

Odds Ratio (M‐H, Random, 95% CI)

0.42 [0.23, 0.80]

Analysis 5.4

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

5.4.1 Foot IPC

4

324

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.05, 3.47]

5.4.2 Other IPC

11

5107

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.20, 0.81]

5.5 Incidence of bleeding Show forest plot

4

594

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.30, 2.14]

Analysis 5.5

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 5: Incidence of bleeding

5.6 Incidence of major bleeding Show forest plot

4

595

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.35, 4.18]

Analysis 5.6

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Open in table viewer
Comparison 6. IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Incidence of PE Show forest plot

2

405

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.19]

Analysis 6.1

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 1: Incidence of PE

6.2 Incidence of DVT Show forest plot

2

405

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.44, 1.39]

Analysis 6.2

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 2: Incidence of DVT

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 2: Incidence of DVT

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: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 1.1

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Figuras y tablas -
Analysis 1.2

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 1.3

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 1.4

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 5: Incidence of DVT ‐ by foot IPC or other IPC

Figuras y tablas -
Analysis 1.5

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 5: Incidence of DVT ‐ by foot IPC or other IPC

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 1.6

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 1.7

Comparison 1: IPC plus pharmacological prophylaxis versus IPC alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 2.1

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 1: Incidence of PE ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Figuras y tablas -
Analysis 2.2

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 2: Incidence of PE ‐ foot IPC or other IPC

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 2.3

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 3: Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 2.4

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 4: Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 5: Incidence of DVT ‐ foot IPC or other IPC

Figuras y tablas -
Analysis 2.5

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 5: Incidence of DVT ‐ foot IPC or other IPC

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 2.6

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 6: Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Figuras y tablas -
Analysis 2.7

Comparison 2: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone, Outcome 7: Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 1: Incidence of PE

Figuras y tablas -
Analysis 3.1

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 1: Incidence of PE

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 2: Incidence of DVT

Figuras y tablas -
Analysis 3.2

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 2: Incidence of DVT

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 3: Incidence of symptomatic DVT

Figuras y tablas -
Analysis 3.3

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 3: Incidence of symptomatic DVT

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 4: Incidence of bleeding

Figuras y tablas -
Analysis 3.4

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 4: Incidence of bleeding

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 5: Incidence of major bleeding

Figuras y tablas -
Analysis 3.5

Comparison 3: IPC plus pharmacological prophylaxis versus IPC plus aspirin, Outcome 5: Incidence of major bleeding

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 1: Incidence of PE

Figuras y tablas -
Analysis 4.1

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 2: Incidence of DVT

Figuras y tablas -
Analysis 4.2

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 2: Incidence of DVT

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Figuras y tablas -
Analysis 4.3

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Figuras y tablas -
Analysis 4.4

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Figuras y tablas -
Analysis 4.5

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Figuras y tablas -
Analysis 4.6

Comparison 4: IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 1: Incidence of PE

Figuras y tablas -
Analysis 5.1

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 2: Incidence of DVT

Figuras y tablas -
Analysis 5.2

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 2: Incidence of DVT

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Figuras y tablas -
Analysis 5.3

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 3: Incidence of symptomatic DVT

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Figuras y tablas -
Analysis 5.4

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 4: Incidence of DVT by foot IPC or other IPC

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Figuras y tablas -
Analysis 5.5

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 5: Incidence of bleeding

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Figuras y tablas -
Analysis 5.6

Comparison 5: IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only, Outcome 6: Incidence of major bleeding

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 1: Incidence of PE

Figuras y tablas -
Analysis 6.1

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 1: Incidence of PE

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 2: Incidence of DVT

Figuras y tablas -
Analysis 6.2

Comparison 6: IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only, Outcome 2: Incidence of DVT

Summary of findings 1. IPC plus pharmacological prophylaxis versus IPC alone

Does combined intermittent pneumatic compression (IPC) plus pharmacological prophylaxis increase prevention of venous thromboembolism compared with IPC alone?

Patient or population: people undergoing surgery or at risk of developing VTE due to surgery, trauma or ICU stay

Settings: hospital 

Intervention: combined modalities ‐  IPC plus pharmacological prophylaxis

Comparison: IPC alone

Outcomes

Anticipated absolute effects * (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with IPC alone

Risk with combined modalities

Incidence of PEa

(early postoperative period)

16 per 1000

7 per 1000 (4 to 12)

OR 0.51 (0.29 to 0.91)

5462 (19)

⊕⊕⊝⊝
Lowb

 

Incidence of DVTc

(early postoperative period)

38 per 1000

20 per 1000 (14 to 28)

OR 0.51 (0.36 to 0.72)

5394 (18)

⊕⊕⊝⊝
Lowb

 

Incidence of bleedingd

(early postoperative period)

10 per 1000

55 per 1000 (36 to 83)

OR 6.02 (3.88 to 9.35)

4634 (13)

⊕⊝⊝⊝
Very lowe

 

Incidence of major bleedingf

(early postoperative period)

3 per 1000

19 per 1000 (10 to 39)

OR 5.77 (2.81 to 11.83)

4133 (12)

⊕⊝⊝⊝

Very lowe

 

* 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)
CI: confidence interval;DVT: deep vein thrombosis;ICU: intensive care unit; IPC: intermittent pneumatic compression; OR: odds ratio; PE: pulmonary embolism; VTE: venous thromboembolism

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. 

aPulmonary embolism assessed by pulmonary angiography or scintigraphy, computed tomography (CT), angiography, or autopsy.
bDowngraded by two levels due to risk of bias concerns (high in one or more domains regarding all but one study) and due to imprecision as a result of a small number of overall events.
cDeep vein thrombosis assessed by ascending venography, I‐125 fibrinogen uptake test, and ultrasound scanning.
d Any type of bleeding as described by the study authors.
eDowngraded by three levels due to risk of bias concerns (high in one or more domains regarding all but one study), due to imprecision as a result of a small number of events overall, and indirectness because bleeding definitions were not uniform across the studies.
fMajor bleeding as defined by the study authors, but usually located at the surgical site or in a critical organ or site, requiring intervention or transfusion of at least units of blood, or leading to death.

Figuras y tablas -
Summary of findings 1. IPC plus pharmacological prophylaxis versus IPC alone
Summary of findings 2. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone

Does combined intermittent pneumatic compression (IPC) plus pharmacological prophylaxis increase prevention of venous thromboembolism compared with pharmacological prophylaxis alone?

Patient or population: people undergoing surgery or at risk of developing VTE because of surgery, trauma or ICU stay

Settings: hospital 

Intervention: combined modalities ‐ IPC plus pharmacological prophylaxis

Comparison: pharmacological prophylaxis alone

Outcomes

Anticipated absolute effects * (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with pharmacological prophylaxis alone

 

Risk with combined modalities

Incidence of PEa

(early postoperative period)

18 per 1000

9 per 1000 (6 to 13)

OR 0.46 (0.30 to 0.71)

6737 (15)

⊕⊕⊝⊝
Lowb

 

Incidence of DVTc

(early postoperative period)

93 per 1000

37 per 1000 (21 to 67)

OR 0.38 (0.21 to 0.70)

6151 (17)

⊕⊕⊕⊕
Highd

 

Incidence of bleedinge

(early postoperative period)

74 per 1000

65 per 1000 (43 to 98)

OR 0.87 (0.56 to 1.35

1314 (6)

⊕⊝⊝⊝

Very lowf

 

Incidence of major bleedingg

(early postoperative period)

11 per 1000

13 per 1000 (4 to 44)

OR 1.21 (0.35 to 4.18)

908 (5)

⊕⊝⊝⊝

Very lowf

 

* 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)
CI: confidence interval;DVT: deep vein thrombosis;ICU: intensive care unit; IPC: intermittent pneumatic compression; OR: odds ratio; PE: pulmonary embolism; VTE: venous thromboembolism

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.

aPulmonary embolism assessed by pulmonary angiography or scintigraphy, computed tomography (CT), angiography, single photon emission CT (SPECT) or autopsy.
bDowngraded by two levels due to risk of bias (which was high in one or more domains regarding all studies) and due to imprecision as a result of a small number of events overall.
cDeep vein thrombosis assessed predominantly by ascending venography, I‐125 fibrinogen uptake test, and ultrasound scanning.
dDowngraded by one level due to risk of bias (which was high in one or more domains regarding all but one study) and upgraded by one level because of a large magnitude of the effect.
eAny type of bleeding as described by the study authors.
fDowngraded by three levels due to risk of bias (which was high in one or more domains regarding all studies), due to imprecision as a result of a small number of events overall and a wide confidence interval, and indirectness because bleeding definitions were not uniform across the studies.
gMajor bleeding as defined by the study authors, but usually located at the surgical site or in a critical organ or site, requiring intervention or transfusion of at least units of blood, or leading to death.

Figuras y tablas -
Summary of findings 2. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone
Comparison 1. IPC plus pharmacological prophylaxis versus IPC alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Incidence of PE ‐ orthopaedic and non‐orthopaedic patients Show forest plot

19

5462

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.1.1 Orthopaedic patients

3

445

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.1.2 Non‐orthopaedic patients

16

5017

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.2 Incidence of PE ‐ foot IPC or other IPC Show forest plot

19

5462

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.2.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.2.2 Other IPC

18

5412

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.91]

1.3 Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

18

5394

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

1.3.1 Orthopaedic patients

3

445

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.38, 1.69]

1.3.2 Non‐orthopaedic patients

15

4949

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.31, 0.68]

1.4 Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

10

4089

Odds Ratio (M‐H, Fixed, 95% CI)

0.48 [0.21, 1.10]

1.4.1 Orthopaedic patients

1

250

Odds Ratio (M‐H, Fixed, 95% CI)

1.50 [0.06, 37.33]

1.4.2 Non‐orthopaedic patients

9

3839

Odds Ratio (M‐H, Fixed, 95% CI)

0.44 [0.19, 1.04]

1.5 Incidence of DVT ‐ by foot IPC or other IPC Show forest plot

18

5395

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

1.5.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

1.5.2 Other IPC

17

5345

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.36, 0.72]

1.6 Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

13

4634

Odds Ratio (M‐H, Fixed, 95% CI)

6.02 [3.88, 9.35]

1.6.1 Orthopaedic patients

2

400

Odds Ratio (M‐H, Fixed, 95% CI)

2.79 [0.77, 10.18]

1.6.2 Non‐orthopaedic patients

11

4234

Odds Ratio (M‐H, Fixed, 95% CI)

6.61 [4.14, 10.56]

1.7 Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

12

4133

Odds Ratio (M‐H, Fixed, 95% CI)

5.77 [2.81, 11.83]

1.7.1 Orthopaedic patients

2

400

Odds Ratio (M‐H, Fixed, 95% CI)

3.35 [0.13, 83.62]

1.7.2 Non‐orthopaedic patients

10

3733

Odds Ratio (M‐H, Fixed, 95% CI)

5.91 [2.83, 12.36]

Figuras y tablas -
Comparison 1. IPC plus pharmacological prophylaxis versus IPC alone
Comparison 2. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 Incidence of PE ‐ orthopaedic and non‐orthopaedic patients Show forest plot

15

6737

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.30, 0.71]

2.1.1 Orthopaedic patients

8

1202

Odds Ratio (M‐H, Fixed, 95% CI)

0.58 [0.08, 4.49]

2.1.2 Non‐orthopaedic patients

7

5535

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.29, 0.71]

2.2 Incidence of PE ‐ foot IPC or other IPC Show forest plot

15

6737

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.30, 0.71]

2.2.1 Foot IPC

4

324

Odds Ratio (M‐H, Fixed, 95% CI)

0.32 [0.01, 8.25]

2.2.2 Other IPC

11

6413

Odds Ratio (M‐H, Fixed, 95% CI)

0.47 [0.30, 0.72]

2.3 Incidence of DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

17

6151

Odds Ratio (M‐H, Random, 95% CI)

0.38 [0.21, 0.70]

2.3.1 Orthopaedic patients

10

3075

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.18, 0.68]

2.3.2 Non‐orthopaedic patients

7

3076

Odds Ratio (M‐H, Random, 95% CI)

0.46 [0.13, 1.61]

2.4 Incidence of symptomatic DVT ‐ orthopaedic and non‐orthopaedic patients Show forest plot

7

3032

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.34, 2.01]

2.4.1 Orthopaedic patients

3

477

Odds Ratio (M‐H, Fixed, 95% CI)

2.09 [0.38, 11.50]

2.4.2 Non‐orthopaedic patients

4

2555

Odds Ratio (M‐H, Fixed, 95% CI)

0.55 [0.18, 1.66]

2.5 Incidence of DVT ‐ foot IPC or other IPC Show forest plot

16

4148

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.18, 0.63]

2.5.1 Foot IPC

4

324

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.05, 3.47]

2.5.2 Other IPC

12

3824

Odds Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.54]

2.6 Incidence of bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

6

1314

Odds Ratio (M‐H, Fixed, 95% CI)

0.87 [0.56, 1.35]

2.6.1 Orthopaedic patients

3

550

Odds Ratio (M‐H, Fixed, 95% CI)

0.64 [0.20, 2.07]

2.6.2 Non‐orthopaedic patients

3

764

Odds Ratio (M‐H, Fixed, 95% CI)

0.91 [0.57, 1.47]

2.7 Incidence of major bleeding ‐ orthopaedic and non‐orthopaedic patients Show forest plot

5

908

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.35, 4.18]

2.7.1 Orthopaedic patients

3

551

Odds Ratio (M‐H, Fixed, 95% CI)

1.07 [0.21, 5.54]

2.7.2 Non‐orthopaedic patients

2

357

Odds Ratio (M‐H, Fixed, 95% CI)

1.42 [0.21, 9.49]

Figuras y tablas -
Comparison 2. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone
Comparison 3. IPC plus pharmacological prophylaxis versus IPC plus aspirin

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Incidence of PE Show forest plot

3

605

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.19]

3.2 Incidence of DVT Show forest plot

3

605

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.48, 1.42]

3.3 Incidence of symptomatic DVT Show forest plot

1

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.4 Incidence of bleeding Show forest plot

3

616

Odds Ratio (M‐H, Fixed, 95% CI)

1.23 [0.27, 5.53]

3.5 Incidence of major bleeding Show forest plot

3

616

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.15, 4.17]

Figuras y tablas -
Comparison 3. IPC plus pharmacological prophylaxis versus IPC plus aspirin
Comparison 4. IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Incidence of PE Show forest plot

13

4159

Odds Ratio (M‐H, Fixed, 95% CI)

0.54 [0.28, 1.07]

4.2 Incidence of DVT Show forest plot

13

4159

Odds Ratio (M‐H, Fixed, 95% CI)

0.53 [0.36, 0.77]

4.3 Incidence of symptomatic DVT Show forest plot

7

3064

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.15, 9.63]

4.4 Incidence of DVT by foot IPC or other IPC Show forest plot

17

5085

Odds Ratio (M‐H, Fixed, 95% CI)

0.52 [0.36, 0.74]

4.4.1 Foot IPC

1

50

Odds Ratio (M‐H, Fixed, 95% CI)

Not estimable

4.4.2 Other IPC

16

5035

Odds Ratio (M‐H, Fixed, 95% CI)

0.52 [0.36, 0.74]

4.5 Incidence of bleeding Show forest plot

10

4120

Odds Ratio (M‐H, Fixed, 95% CI)

5.45 [3.38, 8.80]

4.6 Incidence of major bleeding Show forest plot

9

3619

Odds Ratio (M‐H, Fixed, 95% CI)

5.90 [2.82, 12.33]

Figuras y tablas -
Comparison 4. IPC plus pharmacological prophylaxis versus IPC alone ‐ RCTs only
Comparison 5. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Incidence of PE Show forest plot

11

5758

Odds Ratio (M‐H, Fixed, 95% CI)

0.45 [0.29, 0.70]

5.2 Incidence of DVT Show forest plot

13

5172

Odds Ratio (M‐H, Random, 95% CI)

0.44 [0.22, 0.87]

5.3 Incidence of symptomatic DVT Show forest plot

5

2312

Odds Ratio (M‐H, Fixed, 95% CI)

1.02 [0.29, 3.54]

5.4 Incidence of DVT by foot IPC or other IPC Show forest plot

15

5431

Odds Ratio (M‐H, Random, 95% CI)

0.42 [0.23, 0.80]

5.4.1 Foot IPC

4

324

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.05, 3.47]

5.4.2 Other IPC

11

5107

Odds Ratio (M‐H, Random, 95% CI)

0.40 [0.20, 0.81]

5.5 Incidence of bleeding Show forest plot

4

594

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.30, 2.14]

5.6 Incidence of major bleeding Show forest plot

4

595

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.35, 4.18]

Figuras y tablas -
Comparison 5. IPC plus pharmacological prophylaxis versus pharmacological prophylaxis alone ‐ RCTs only
Comparison 6. IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Incidence of PE Show forest plot

2

405

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.19]

6.2 Incidence of DVT Show forest plot

2

405

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.44, 1.39]

Figuras y tablas -
Comparison 6. IPC plus pharmacological prophylaxis versus IPC plus aspirin ‐ RCTs only