Low Dose Low-Molecular-Weight Heparin for Thrombosis Prophylaxis: Systematic Review with Meta-Analysis and Trial Sequential Analysis

Ruben J Eck, Wouter Bult, Jørn Wetterslev, Reinold O B Gans, Karina Meijer, Iwan C C van der Horst, Frederik Keus, Ruben J Eck, Wouter Bult, Jørn Wetterslev, Reinold O B Gans, Karina Meijer, Iwan C C van der Horst, Frederik Keus

Abstract

International guidelines recommend low-molecular-weight heparin (LMWH) as first-line pharmacological option for the prevention of venous thromboembolism (VTE) in many patient categories. Guidance on the optimal prophylactic dose is lacking. We conducted a systematic review with meta-analysis and trial sequential analysis (TSA) of randomized controlled trials to assess benefits and harms of low-dose LMWH versus placebo or no treatment for thrombosis prophylaxis in patients at risk of VTE. PubMed, Cochrane Library, Web of Science, and Embase were searched up to June 2019. Results were presented as relative risk (RR) with conventional and TSA-adjusted confidence intervals (CI). Forty-four trials with a total of 22,579 participants were included. Six (14%) had overall low risk of bias. Low-dose LMWH was not statistically significantly associated with all-cause mortality (RR 0.99; 95%CI 0.85-1.14; TSA-adjusted CI 0.89-1.16) but did reduce symptomatic VTE (RR 0.62; 95%CI 0.48-0.81; TSA-adjusted CI 0.44-0.89) and any VTE (RR 0.61; 95%CI 0.50-0.75; TSA-adjusted CI 0.49-0.82). Analyses on major bleeding (RR 1.07; 95%CI 0.72-1.59), as well as serious adverse events (SAE) and clinically relevant non-major bleeding were inconclusive. There was very low to moderate-quality evidence that low-dose LMWH for thrombosis prophylaxis did not decrease all-cause mortality but reduced the incidence of symptomatic and asymptomatic VTE, while the analysis of the effects on bleeding and adverse events remained inconclusive.

Keywords: low-molecular-weight heparin; meta-analysis; venous thromboembolism.

Conflict of interest statement

KM reports grants from Bayer, Sanquin, and Pfizer; speaker fees from Bayer, Sanquin, Boehringer Ingelheim, BMS, and Aspen; travel support from Bayer, and consulting fees from Uniqure outside the submitted work; JW is a member of the task force at the Copenhagen Trial Unit to develop theory and software of Trial Sequential Analysis; other authors have disclosed no potential conflicts of interest.

Figures

Figure 1
Figure 1
Preferred reporting items for systematic reviews and meta-analyses (PRISMA) flow-chart of study inclusion.
Figure 2
Figure 2
Forest plot of all-cause mortality. Forest plot of all-cause mortality at maximal follow-up of LMWH prophylaxis compared to placebo or no treatment, stratified according to population. The size of the squares reflects the weight of the trial in the pooled analysis. Horizontal bars represent 95% confidence intervals; LMWH, low-molecular-weight heparin; CI, confidence intervals.
Figure 3
Figure 3
Trial sequential analysis of symptomatic venous thromboembolism (VTE). Trial sequential analysis of symptomatic VTE at maximal follow-up of LMWH compared to placebo or no treatment. The required information size was calculated using α = 0.025, β = 0.90, relative risk reduction (RRR) = 20%, diversity (D2) as suggested by trials, and a control event rate of 1.81%. The cumulative Z-curve was constructed using a random-effects model, and each cumulative Z-value was calculated after inclusion of a new trial (represented by black dots). The dotted horizontal lines represent the conventional naïve boundaries for benefit. The etched lines represent the trial sequential boundaries for benefit (positive), harm (negative), or futility (middle triangular area). The cumulative Z-curve crosses the TSA boundary for benefit, indicating future trials are very unlikely to change the conclusions. Note: the two most recent trials were excluded from this TSA because inclusion would result in an incorrect graphical display of the LanDeMets boundary for benefit. The TSA-adjusted confidence interval remained similar.
Figure 4
Figure 4
Forest plot of symptomatic VTE. Forest plot of symptomatic VTE at maximal follow-up of LMWH prophylaxis compared to placebo or no treatment, stratified according to patient type. The size of the squares reflects the weight of the trial in the pooled analysis. Horizontal bars represent 95% confidence intervals.
Figure 5
Figure 5
Forest plot of major bleeding. Forest plot of major bleeding at maximal follow-up of LMWH prophylaxis compared to placebo or no treatment, stratified according to patient type. The size of the squares reflects the weight of the trial in the pooled analysis. Horizontal bars represent 95% confidence intervals.

References

    1. Cohen A.T., Alikhan R., Arcelus J.I., Bergmann J.-F., Haas S., Merli G.J., Spyropoulos A.C., Tapson V.F., Turpie A.G.G. Assessment of venous thromboembolism risk and the benefits of thromboprophylaxis in medical patients. Thromb. Haemost. 2005;94:750–759. doi: 10.1160/TH05-06-0385.
    1. Schünemann H.J., Cushman M., Burnett A.E., Kahn S.R., Beyer-Westendorf J., Spencer F.A., Rezende S.M., Zakai N.A., Bauer K.A., Dentali F., et al. American Society of Hematology 2018 guidelines for management of venous thromboembolism: Prophylaxis for hospitalized and nonhospitalized medical patients. Blood Adv. 2018;2:3198–3225. doi: 10.1182/bloodadvances.2018022954.
    1. NICE Guideline Venous Thromboembolism in over 16s: Reducing the Risk of Hospital-Acquired Deep Vein Thrombosis or Pulmonary Embolism. [(accessed on 31 January 2019)];2018 Available online: .
    1. Alhazzani W., Lim W., Jaeschke R.Z., Murad M.H., Cade J., Cook D.J. Heparin thromboprophylaxis in medical-surgical critically ill patients: A systematic review and meta-analysis of randomized trials. Crit. Care Med. 2013;41:2088–2098. doi: 10.1097/CCM.0b013e31828cf104.
    1. Ageno W., Bosch J., Cucherat M., Eikelboom J.W. Nadroparin for the prevention of venous thromboembolism in nonsurgical patients: A systematic review and meta-analysis. J. Thromb. Thrombolysis. 2016;42:90–98. doi: 10.1007/s11239-015-1294-3.
    1. Di Nisio M., Porreca E., Candeloro M., De Tursi M., Russi I., Rutjes A.W. Primary prophylaxis for venous thromboembolism in ambulatory cancer patients receiving chemotherapy. Cochrane Database Syst. Rev. 2016;12:CD008500. doi: 10.1002/14651858.CD008500.pub4.
    1. Kahale L.A., Tsolakian I.G., Hakoum M.B., Matar C.F., Barba M., Yosuico V.E., Terrenato I., Sperati F., Schünemann H., Akl E.A. Anticoagulation for people with cancer and central venous catheters. Cochrane Database Syst. Rev. 2018;6:CD006468. doi: 10.1002/14651858.CD006468.pub6.
    1. Beitland S., Sandven I., Kjaervik L.K., Sandset P.M., Sunde K., Eken T. Thromboprophylaxis with low molecular weight heparin versus unfractionated heparin in intensive care patients: A systematic review with meta-analysis and trial sequential analysis. Intensive Care Med. 2015;41:1209–1219. doi: 10.1007/s00134-015-3840-z.
    1. Sanford D., Naidu A., Alizadeh N., Lazo-Langner A. The effect of low molecular weight heparin on survival in cancer patients: An updated systematic review and meta-analysis of randomized trials. J. Thromb. Haemost. 2014;12:1076–1085. doi: 10.1111/jth.12595.
    1. Chapelle C., Rosencher N., Jacques Zufferey P., Mismetti P., Cucherat M., Laporte S. Prevention of venous thromboembolic events with low-molecular-weight heparin in the non-major orthopaedic setting: Meta-analysis of randomized controlled trials. Arthroscopy. 2014;30:987–996. doi: 10.1016/j.arthro.2014.03.009.
    1. Eck R.J., Bult W., Wetterslev J., Gans R.O.B., Meijer K., Keus F., Van der Horst I.C.C. Intermediate Dose low-molecular-weight heparin for thrombosis prophylaxis: Systematic review with meta-analysis and trial sequential analysis. Semin. Thromb. Hemost. 2019;45:810–824. doi: 10.1055/s-0039-1696965.
    1. The Cochrane Collaboration . In: Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. Higgins J.P.T., Green S., editors. The Cochrane Collaboration; Lonodn, UK: 2011.
    1. Liberati A., Altman D.G., Tetzlaff J., Mulrow C., Gotzsche P.C., Ioannidis J.P.A., Clarke M., Devereaux P.J., Kleijnen J., Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700. doi: 10.1136/bmj.b2700.
    1. Jakobsen J.C., Wetterslev J., Winkel P., Lange T., Gluud C. Thresholds for statistical and clinical significance in systematic reviews with meta-analytic methods. BMC Med. Res. Methodol. 2014;14:120. doi: 10.1186/1471-2288-14-120.
    1. Guyatt G.H., Oxman A.D., Kunz R., Vist G.E., Falck-Ytter Y., Schunemann H.J. What is “quality of evidence” and why is it important to clinicians? BMJ. 2008;336:995–998. doi: 10.1136/.
    1. International Conference on Harmonisation Expert Working Group International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use, ed. ICH Harmonised Tripartite Guideline; Guideline for Good Clinical Practice. [(accessed on 31 January 2019)]; Available online: .
    1. Sterne J., Savović J., Page M., Elbers R., Blencowe N., Boutron I., Cates C., Cheng H.-Y., Corbett M., Eldridge S., et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898. doi: 10.1136/bmj.l4898.
    1. Savović J., Turner R.M., Mawdsley D., Jones H.E., Beynon R., Higgins J.P.T., Sterne J.A.C. Association between risk-of-bias assessments and results of randomized trials in cochrane reviews: The ROBES meta-epidemiologic study. Am. J. Epidemiol. 2018;187:1113–1122. doi: 10.1093/aje/kwx344.
    1. Wetterslev J., Jakobsen J.C., Gluud C. Trial sequential analysis in systematic reviews with meta-analysis. BMC Med. Res. Methodol. 2017;17:1–18. doi: 10.1186/s12874-017-0315-7.
    1. Brok J., Thorlund K., Wetterslev J., Gluud C. Apparently conclusive meta-analyses may be inconclusive—Trial sequential analysis adjustment of random error risk due to repetitive testing of accumulating data in apparently conclusive neonatal meta-analyses. Int. J. Epidemiol. 2009;38:287–298. doi: 10.1093/ije/dyn188.
    1. Thorlund K., Devereaux P.J., Wetterslev J., Guyatt G., Ioannidis J.P.A., Thabane L., Gluud L.-L., Als-Nielsen B., Gluud C. Can trial sequential monitoring boundaries reduce spurious inferences from meta-analyses? Int. J. Epidemiol. 2009;38:276–286. doi: 10.1093/ije/dyn179.
    1. Pogue J.M., Yusuf S. Cumulating evidence from randomized trials: Utilizing sequential monitoring boundaries for cumulative meta-analysis. Control. Clin. Trials. 1997;18:580–586. doi: 10.1016/S0197-2456(97)00051-2.
    1. Pogue J., Yusuf S. Overcoming the limitations of current meta-analysis of randomised controlled trials. Lancet. 1998;351:47–52. doi: 10.1016/S0140-6736(97)08461-4.
    1. Wetterslev J., Thorlund K., Brok J., Gluud C. Estimating required information size by quantifying diversity in random-effects model meta-analyses. BMC Med. Res. Methodol. 2009;9:86. doi: 10.1186/1471-2288-9-86.
    1. Alikhan R., Bedenis R., Cohen A.T. Heparin for the prevention of venous thromboembolism in acutely ill medical patients (excluding stroke and myocardial infarction) Cochrane Database Syst. Rev. 2014;5:CD003747. doi: 10.1002/14651858.CD003747.pub4.
    1. Hostler D.C., Marx E.S., Moores L.K., Petteys S.K., Hostler J.M., Mitchell J.D., Holley P.R., Collen J.F., Foster B.E., Holley A.B. Validation of the international medical prevention registry on venous thromboembolism bleeding risk score. Chest. 2016;149:372–379. doi: 10.1378/chest.14-2842.

Source: PubMed

3
Předplatit