Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cochrane Database Syst Rev ; 2: CD014544, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38411279

ABSTRACT

BACKGROUND: Management of congenital hemophilia A and B is by prophylactic or on-demand replacement therapy with clotting factor concentrates. The effects of newer non-clotting factor therapies such as emicizumab, concizumab, marstacimab, and fitusiran compared with existing standards of care are yet to be systematically reviewed. OBJECTIVES: To assess the effects (clinical, economic, patient-reported, and adverse outcomes) of non-clotting factor therapies for preventing bleeding and bleeding-related complications in people with congenital hemophilia A or B compared with prophylaxis with clotting factor therapies, bypassing agents, placebo, or no prophylaxis. SEARCH METHODS: We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Coagulopathies Trials Register, electronic databases, conference proceedings, and reference lists of relevant articles and reviews. The date of the last search was 16 August 2023. SELECTION CRITERIA: Randomized controlled trials (RCTs) evaluating people with congenital hemophilia A or B with and without inhibitors, who were treated with non-clotting factor therapies to prevent bleeds. DATA COLLECTION AND ANALYSIS: Two review authors independently reviewed studies for eligibility, assessed risk of bias, and extracted data for the primary outcomes (bleeding rates, health-related quality of life (HRQoL), adverse events) and secondary outcomes (joint health, pain scores, and economic outcomes). We assessed the mean difference (MD), risk ratio (RR), 95% confidence interval (CI) of effect estimates, and evaluated the certainty of the evidence using GRADE. MAIN RESULTS: Six RCTs (including 397 males aged 12 to 75 years) were eligible for inclusion. Prophylaxis versus on-demand therapy in people with inhibitors Four trials (189 participants) compared emicizumab, fitusiran, and concizumab with on-demand therapy in people with inhibitors. Prophylaxis using emicizumab likely reduced annualized bleeding rates (ABR) for all bleeds (MD -22.80, 95% CI -37.39 to -8.21), treated bleeds (MD -20.40, 95% CI -35.19 to -5.61), and annualized spontaneous bleeds (MD -15.50, 95% CI -24.06 to -6.94), but did not significantly reduce annualized joint and target joint bleeding rates (AjBR and AtjBR) (1 trial; 53 participants; moderate-certainty evidence). Fitusiran also likely reduced ABR for all bleeds (MD -28.80, 95% CI -40.07 to -17.53), treated bleeds (MD -16.80, 95% CI -25.80 to -7.80), joint bleeds (MD -12.50, 95% CI -19.91 to -5.09), and spontaneous bleeds (MD -14.80, 95% CI -24.90 to -4.71; 1 trial; 57 participants; moderate-certainty evidence). No evidence was available on the effect of bleed prophylaxis using fitusiran versus on-demand therapy on AtjBR. Concizumab may reduce ABR for all bleeds (MD -12.31, 95% CI -19.17 to -5.45), treated bleeds (MD -10.10, 95% CI -17.74 to -2.46), joint bleeds (MD -9.55, 95% CI -13.55 to -5.55), and spontaneous bleeds (MD -11.96, 95% CI -19.89 to -4.03; 2 trials; 78 participants; very low-certainty evidence), but not target joint bleeds (MD -1.00, 95% CI -3.26 to 1.26). Emicizumab prophylaxis resulted in an 11.31-fold increase, fitusiran in a 12.5-fold increase, and concizumab in a 1.59-fold increase in the proportion of participants with no bleeds. HRQoL measured using the Haemophilia Quality of Life Questionnaire for Adults (Haem-A-QoL) physical and total health scores was improved with emicizumab, fitusiran, and concizumab prophylaxis (low-certainty evidence). Non-serious adverse events were higher with non-clotting factor therapies versus on-demand therapy, with injection site reactions being the most frequently reported adverse events. Transient antidrug antibodies were reported for fitusiran and concizumab. Prophylaxis versus on-demand therapy in people without inhibitors Two trials (208 participants) compared emicizumab and fitusiran with on-demand therapy in people without inhibitors. One trial assessed two doses of emicizumab (1.5 mg/kg weekly and 3.0 mg/kg bi-weekly). Fitusiran 80 mg monthly, emicizumab 1.5 mg/kg/week, and emicizumab 3.0 mg/kg bi-weekly all likely resulted in a large reduction in ABR for all bleeds, all treated bleeds, and joint bleeds. AtjBR was not reduced with either of the emicizumab dosing regimens. The effect of fitusiran prophylaxis on target joint bleeds was not assessed. Spontaneous bleeds were likely reduced with fitusiran (MD -20.21, 95% CI -32.12 to -8.30) and emicizumab 3.0 mg/kg bi-weekly (MD -15.30, 95% CI -30.46 to -0.14), but not with emicizumab 1.5 mg/kg/week (MD -14.60, 95% CI -29.78 to 0.58). The percentage of participants with zero bleeds was higher following emicizumab 1.5 mg/kg/week (50% versus 0%), emicizumab 3.0 mg/kg bi-weekly (40% versus 0%), and fitusiran prophylaxis (40% versus 5%) compared with on-demand therapy. Emicizumab 1.5 mg/kg/week did not improve Haem-A-QoL physical and total health scores, EQ-5D-5L VAS, or utility index scores (low-certainty evidence) when compared with on-demand therapy at 25 weeks. Emicizumab 3.0 mg/kg bi-weekly may improve HRQoL measured by the Haem-A-QoL physical health score (MD -15.97, 95% CI -29.14 to -2.80) and EQ-5D-5L VAS (MD 9.15, 95% CI 2.05 to 16.25; 1 trial; 43 participants; low-certainty evidence). Fitusiran may result in improved HRQoL shown as a reduction in Haem-A-QoL total score (MD -7.06, 95% CI -11.50 to -2.62) and physical health score (MD -19.75, 95% CI -25.76 to -11.94; 1 trial; 103 participants; low-certainty evidence). The risk of serious adverse events in participants without inhibitors also likely did not differ following prophylaxis with either emicizumab or fitusiran versus on-demand therapy (moderate-certainty evidence). Transient antidrug antibodies were reported in 4% (3/80) participants to fitusiran, with no observed effect on antithrombin lowering. A comparison of the different dosing regimens of emicizumab identified no differences in bleeding, safety, or patient-reported outcomes. No case of treatment-related cancer or mortality was reported in any study group. None of the included studies assessed our secondary outcomes of joint health, clinical joint function, and economic outcomes. None of the included studies evaluated marstacimab. AUTHORS' CONCLUSIONS: Evidence from RCTs shows that prophylaxis using non-clotting factor therapies compared with on-demand treatment may reduce bleeding events, increase the percentage of individuals with zero bleeds, increase the incidence of non-serious adverse events, and improve HRQoL. Comparative assessments with other prophylaxis regimens, assessment of long-term joint outcomes, and assessment of economic outcomes will improve evidence-based decision-making for the use of these therapies in bleed prevention.


Subject(s)
Hemophilia A , Male , Adult , Humans , Hemophilia A/complications , Hemophilia A/drug therapy , Blood Coagulation Factors/therapeutic use , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Hemarthrosis/etiology , Hemarthrosis/prevention & control , Heme/therapeutic use
2.
Front Pediatr ; 11: 1155853, 2023.
Article in English | MEDLINE | ID: mdl-37082707

ABSTRACT

The introduction of emicizumab into the treatment regime of persons with hemophilia A has dramatically reduced frequency of bleeding in patients with and without inhibitors. However, in children with Hemophilia A (CwHA) who require surgical or other invasive procedures, additional treatment with factor replacement or other hemostatic agents may still be needed to prevent intraoperative or postoperative bleeding. This review will look at the reported outcomes in CwHA on emicizumab who have had surgery and propose recommendations for the best perioperative management of major and minor procedures.

3.
Pediatr Res ; 93(6): 1463-1469, 2023 05.
Article in English | MEDLINE | ID: mdl-36127405

ABSTRACT

Nephrotic syndrome (NS) is a common kidney disease of childhood, affecting 2-7 children per 100,000. A potentially life-threatening complication affecting children with NS is thromboembolism (TE). However, there remains a paucity of information regarding the burden of TE and its associated risk factors in this population. A systematic review was performed on observational studies examining TE events in children with NS, published in Medline, Embase, CINAHL, and CENTRAL, until May 2021. Meta-analyses were separately conducted on the prevalence of TE within articles exclusively studying children with congenital NS and among articles including all forms of NS. Out of 13,626 articles, 22 were included (14,290 children). The pooled prevalence of symptomatic TE among articles including patients with all forms of NS was 3.60% (95% CI 1.95-5.63), which increased to 8.70% (95% CI 5.11-12.96) in articles with exclusively congenital NS patients. Children with steroid-resistant NS were at a higher risk of TE compared to steroid-sensitive children (OR 4.40, 95% CI 1.34-15.59, p = 0.013). Focal segmental glomerulosclerosis was the most common histology present in patients with TE (51.2%). Children diagnosed with NS have a significant risk of TE, particularly in patients with congenital NS and steroid resistance. IMPACT: The prevalence of symptomatic thromboembolic (TE) events in children with nephrotic syndrome (NS) was 3.60% (95% CI 1.95-5.63), which increased more than two-fold in children with congenital NS to 8.70% (95% CI 5.11-12.96). Potential risk factors for TE events in this population include congenital forms of NS and steroid resistance. This review provides a better estimate of the prevalence of TE in children with NS, while identifying potentially higher-risk populations who may benefit from TE screening and thromboprophylaxis.


Subject(s)
Nephrotic Syndrome , Venous Thromboembolism , Child , Humans , Nephrotic Syndrome/complications , Nephrotic Syndrome/epidemiology , Nephrotic Syndrome/drug therapy , Anticoagulants/therapeutic use , Venous Thromboembolism/complications , Steroids/therapeutic use
6.
Front Pediatr ; 10: 855162, 2022.
Article in English | MEDLINE | ID: mdl-35463883

ABSTRACT

Venous thromboembolism (VTE) occurs in 2.1 to up to 50% of children with cancer and contributes to long term morbidity as well as early mortality in this population. Pediatric patients with malignancy are predisposed to VTE due to the prothrombotic nature of cancer and its associated coagulopathies as well as chemotherapeutic agents, use of central venous catheters, surgery, radiotherapy, and concomitant thrombophilia. Management of thrombosis in this population is challenging due to concomitant thrombocytopenia, associated bleeding risks, concurrent co-morbidities, and toxicities of therapy. The aim of this paper is to highlight clinically relevant issues and management dilemmas using clinical vignettes. We review the clinical significance of asymptomatic and symptomatic thrombosis, examine the various options for asparaginase-associated thrombosis, address the role and controversies of direct oral anticoagulants, and describe our approach to managing anticoagulation therapy in the context of chemotherapy-induced thrombocytopenia.

7.
J Pediatr Hematol Oncol ; 44(5): e888-e891, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35398856

ABSTRACT

BACKGROUND: Severe neonatal aortic thrombosis is rare but can lead to significant morbidity or death if inadequately treated. Thrombolytic therapy is indicated for thrombi which are life-threatening, organ-threatening, or limb-threatening, but dosing consensus has not been established. OBSERVATION: We report a case of a 700 g preterm neonate with spontaneous intestinal perforation who developed an occlusive aortic thrombus with signs of limb ischemia. He was treated successfully with tissue plasminogen activator without hemorrhagic complications. He was started at a dose of 0.06 mg/kg/h and received a maximum dose of 0.3 mg/kg/h. Long-term follow-up at 3 years and 3 months showed no negative sequelae. CONCLUSION: Alteplase may be considered in premature, extremely low-birth weight infants with careful assessment of risk and benefits, along with frequent surveillance and supportive care.


Subject(s)
Infant, Premature, Diseases , Thrombosis , Fibrinolytic Agents/therapeutic use , Humans , Infant , Infant, Extremely Low Birth Weight , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/drug therapy , Male , Thrombolytic Therapy , Thrombosis/diagnosis , Thrombosis/drug therapy , Thrombosis/etiology , Tissue Plasminogen Activator/therapeutic use
8.
Blood Coagul Fibrinolysis ; 33(4): 193-200, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35285449

ABSTRACT

Among children, neonates have the highest incidence of thrombosis. Thrombolytic agents are used for the management of life and/or organ-threatening thrombosis. Literature on the efficacy and safety of thrombolytic agents in neonates is limited. We reviewed the evidence on dosing, administration, monitoring and treatment duration of tissue plasminogen activator (tPA), streptokinase and urokinase (URK) in neonates (≤ 28days). A systematic literature search was conducted of current databases from inception until 31 March 2021. The initial search yielded 6881 articles and 18 were retained for review. tPA, streptokinase and URK was utilized in 12, seven and four studies on 115, 51 and 16 patients, respectively. The dose range for tPA, streptokinase and URK was 0.01 -0.6 mg/kg/h, 50-2000 and 1000-0 000 units/kg/h, respectively, and treatment duration ranged from 30 min to 30 days. This is the first study to objectively summarize the efficacy and safety of thrombolytic agents in neonates. Overall, thrombolysis was associated with 87.9% complete or partial thrombus resolution and 7.4% recurrence risk. The bleeding risk associated with thrombolytic agents was 23.1% on pooled analysis, which is higher than other anticoagulants. Larger prospective studies are required to determine effective dosing regimens of these therapeutic drugs and further clarify their efficacy and safety. Blood Coagul Fibrinolysis 33:000-000 Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.


Subject(s)
Fibrinolytic Agents , Thrombosis , Child , Fibrinolytic Agents/therapeutic use , Humans , Infant, Newborn , Streptokinase/therapeutic use , Thrombolytic Therapy , Thrombosis/drug therapy , Tissue Plasminogen Activator/therapeutic use , Urokinase-Type Plasminogen Activator
9.
Pediatr Blood Cancer ; 69(6): e29668, 2022 06.
Article in English | MEDLINE | ID: mdl-35289485

ABSTRACT

Among children, neonates have the highest incidence of thrombosis. We conducted a retrospective review of neonatal thrombosis, in a single intensive care unit (ICU) over 4.5 years. Among 4860 ICU admissions to our center, identified through the Canadian Neonatal Network database, 186 were associated with arterial and venous thrombosis involving 195 thrombotic sites. The neonatal thrombosis incidence was 38 per 1000 neonatal ICU admissions. We assessed patient characteristics and compared the association between risk factors and thrombosis. In the multivariate analysis, central venous catheters, sepsis, and respiratory distress syndrome were significant predictors of neonatal thrombosis.


Subject(s)
Catheterization, Central Venous , Thrombosis , Canada/epidemiology , Catheterization, Central Venous/adverse effects , Child , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Retrospective Studies , Risk Factors , Thrombosis/epidemiology , Thrombosis/etiology
10.
Blood Coagul Fibrinolysis ; 33(2): 83-89, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35001043

ABSTRACT

The incidence of neonatal venous and arterial thrombosis ranges from 6.9 to 15/1000 neonatal ICU (NICU) admissions, and is likely an underestimate based on population demographics, frequency of surveillance and vascular catheterization. This retrospective study involving 234 infants reviewed the epidemiology, diagnosis, and management of neonatal thrombosis in a single, tertiary care institution over more than 10 years. The incidence of thrombosis was 25/1000 NICU admissions, with a preterm to term infant ratio of 1.5 : 1 and a slightly higher proportion of male sex (55.1%). The mean (range) gestational age and birth weight was 33.8 weeks (23-41.6) and 2360 g (512-5890). The median age (IQR) of thrombus diagnosis was 7 (3-17) days. Portal vein thrombosis was most prevalent (59.4%) compared with other sites of thrombosis. Almost three-quarter (171/234; 73.1%) of the thrombotic episodes were line-related, while infection and surgery were associated with 19.7% (46/234) and 10.7% (25/234), respectively. Twenty patients (8.3%) were screened for thrombophilia and 3 were positive; 2 for antithrombin deficiency, 1 for factor V Leiden gene mutation. Subjects were followed with imaging for 3 months with a treatment duration, mean (IQR) of 33.5 (10.8-42.5) days. Complete clot resolution was significantly higher in the anticoagulation group (48%; 17%; P = 0.03) compared with untreated patients. No group difference was noted for partial thrombus resolution (33.3%; 12.4%; P = 0.313). Anticoagulation halted thrombus progression (2.6 versus 12.4%; P = 0.025) and fewer treated patients failed to attend follow-up visits (6.5 versus 18.6%; P = 0.022). Well designed, multicenter prospective studies with larger sample sizes are required to confirm these findings.


Subject(s)
Thrombosis , Venous Thrombosis , Cohort Studies , Humans , Infant , Infant, Newborn , Male , Prospective Studies , Retrospective Studies , Thrombosis/diagnosis , Thrombosis/drug therapy , Thrombosis/epidemiology
11.
J Pediatr Hematol Oncol ; 44(2): e442-e443, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34486549

ABSTRACT

Hereditary protein S (PS) deficiency is a rare autosomal dominant disorder with increased risk of venous thromboembolism. The PS Heerlen polymorphism at codon 501 of the PROS1 gene is considered a variant of uncertain significance. It has since been shown that PS Heerlen has a reduced half-life, resulting in reduced levels of free PS. We report a case of an adolescent female with May Thurner syndrome and heterozygous PS Heerlen mutation resulting in a mild PS deficiency and venous thromboembolism. With this nonmodifiable risk factor, the patient received prolonged anticoagulation with strong consideration for lifelong prophylaxis.


Subject(s)
Protein S Deficiency , Venous Thromboembolism , Venous Thrombosis , Adolescent , Child , Female , Humans , Protein S/genetics , Protein S/metabolism , Protein S Deficiency/complications , Protein S Deficiency/genetics , Thrombophilia , Venous Thromboembolism/drug therapy , Venous Thromboembolism/genetics , Venous Thrombosis/drug therapy , Venous Thrombosis/genetics
12.
Paediatr Child Health ; 26(7): 385-387, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34777652
13.
Cardiol Young ; 31(8): 1213-1219, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34344494

ABSTRACT

Children with cardiac diseases suffer from significant morbidity and mortality secondary to thromboembolic complications. Anticoagulant agents currently used for thromboprophylaxis have many limitations, including subcutaneous administration (low molecular weight heparins) and requirement for frequent monitoring via venipuncture (vitamin K antagonists). Edoxaban is an oral direct factor Xa inhibitor without need of monitoring. In the treatment of venous thromboembolism in adults, edoxaban has shown to be effective and safe.This manuscript summarises the rationale and design of a phase 3, open-label, randomised controlled trial to evaluate and compare the safety and efficacy of edoxaban against standard of care (namely, vitamin K antagonist and low molecular weight heparin) in children with cardiac diseases.A goal of 150 children with cardiac diseases at risk of thromboembolic complications who need primary or secondary anticoagulant prophylaxis will be recruited. Eligible children between 6 months and <18 years of age will be randomised in a ratio of 2 to 1 for edoxaban versus standard of care. Randomisation will be stratified based on underlying cardiac disease and concomitant use of aspirin for patients other than Kawasaki disease. The primary outcome will be safety, comprised of major and clinically relevant non-major bleeding in first 3 months of treatment. Bleeding beyond 3 months, symptomatic and asymptomatic thromboembolic events, and pharmacokinetic and pharmacodynamic parameters will be evaluated as secondary outcomes.Randomised controlled anticoagulation trials are challenging in children. This study will evaluate a potentially valuable alternative of oral anticoagulant prophylactic use in children with cardiac diseases.


Subject(s)
Heart Diseases , Venous Thromboembolism , Adult , Anticoagulants/adverse effects , Child , Heart Diseases/complications , Humans , Pyridines , Thiazoles , Venous Thromboembolism/drug therapy , Venous Thromboembolism/prevention & control
14.
Blood Coagul Fibrinolysis ; 32(8): 531-538, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34102656

ABSTRACT

Among children, neonates have the highest incidence of thrombosis due to risk factors such as catheter instrumentation, an evolving coagulation system and congenital heart disease. Low-molecular-weight heparins (LMWHs) are the most commonly used anticoagulants in neonates. Published guidelines delineate dosing and monitoring protocols for LMWH therapy in newborns. However, challenging clinical situations frequently present that warrant healthcare providers to think critically beyond the range of guidelines, and judiciously resolve specific problems. This review describes the use of LMWH in the neonatal population, including practical aspects such as route and site of administration, preparation from concentrated formulations and methods to minimize pain of subcutaneous injection. It is followed by a discussion on dosing, monitoring and outcomes of LMWH therapy in neonates. The risk of recurrence of thrombosis in neonates after LMWH therapy is approximately 3% based on a pooled analysis of studies reporting this outcome over the last 24 years. The article concludes with an overview of the side-effects of LMWH, including the risk of bleeding which is around 4% based on pooled analyses of more than 30 studies.


Subject(s)
Heparin, Low-Molecular-Weight , Thrombosis , Anticoagulants/therapeutic use , Child , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Heparin , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Infant, Newborn , Thrombosis/drug therapy
15.
Fac Rev ; 10: 20, 2021.
Article in English | MEDLINE | ID: mdl-33718937

ABSTRACT

The incidence of thrombotic disorders in neonates and children is increasing with advances in diagnostic modalities, supportive care, and management of many health conditions. The developing coagulation system, need for intensive care, including catheterization, and co-morbid conditions are responsible for the relatively high risk of thrombosis in neonates compared to older children. This review addresses the advances over the last 3 years in neonatal thrombosis, with a focus on epidemiology, cerebral sinovenous thrombosis (CSVT), renal vein thrombosis (RVT), and portal vein thrombosis (PVT). The incidence of neonatal thrombosis in the contemporary era is reported to be 6.9-15 per 1,000 neonatal intensive care unit (NICU) admissions, compared to 2.4 per 1,000 NICU admissions reported in older registry data. The majority of recently published studies are small, retrospective, and from single centers, albeit they emphasize the need for definitive data to support the efficacy and safety of anticoagulation therapy (ACT) in the management of CSVT, RVT, and PVT. We highlight two important international initiatives geared towards improving the evidence base for these conditions. The International Pediatric Thrombosis Network (IPTN) is a collaboration of 74 centers across 27 countries (as of January 2021) which has started important projects like the international neonatal RVT registry, while the International Pediatric Stroke Study (IPSS) group is in the planning stages of a randomized controlled trial to evaluate the utility of ACT in the management of neonatal CSVT.

16.
J Pediatr Hematol Oncol ; 43(2): e237-e239, 2021 03 01.
Article in English | MEDLINE | ID: mdl-31688638

ABSTRACT

Treatment options exist for patients with severe hemophilia and high titer factor VIII inhibitors but is often inadequate. Few studies have been conducted to evaluate the utility of short-term corticosteroid therapy for improvement in bleeding complications and temporary or sustained resolution of inhibitors in these patients. We describe 2 patients with acute muscular hematomas successfully treated with 4 to 5 days of oral adjuvant corticosteroid therapy resulting in improvement in their acute bleeds and temporary reduction of inhibitors. Thus, the addition of corticosteroids to traditional therapy of hemophilia with inhibitors may be beneficial in some patients. In those with impending compartment syndrome steroids may improve edema and bleeding symptoms preventing the need for surgical interventions.


Subject(s)
Adrenal Cortex Hormones/administration & dosage , Chemotherapy, Adjuvant/methods , Factor VIII/antagonists & inhibitors , Hemophilia A/complications , Hemorrhage/drug therapy , Acute Disease , Administration, Oral , Adolescent , Adrenal Cortex Hormones/therapeutic use , Child , Hemorrhage/etiology , Hemorrhage/pathology , Humans , Male , Prognosis
17.
Acta Paediatr ; 110(3): 1001-1008, 2021 03.
Article in English | MEDLINE | ID: mdl-32815198

ABSTRACT

AIM: To establish the incidence and characteristics of paediatric thrombosis (PT) in a Canadian tertiary care centre during the era of low molecular weight heparin (LMWH). METHODS: A retrospective observational case study of all patients <18 years of age evaluated for arterial and venous thrombosis from May 2008 to July 2018 at McMaster Children's Hospital was conducted through the electronic medical record. RESULTS: The incidence of PT was 52.2 per 10 000 hospital admissions (n = 477/91 462). Provoked thrombosis was more prevalent (88.9%, n = 424/477) than unprovoked (2.9%, n = 14/477) or idiopathic thrombosis (4%, n = 19/477). Half of PT were in children <2 years (51.2%, n = 244/477). Central vascular catheterisation was a contributory factor in more than half of thrombotic events (56.2%, n = 268/477), while trauma (1.1%, n = 5/477), oral contraceptives (4%, n = 19/477), infection (4%, n = 19/477), surgery (6.9%, n = 33/477) and malignancy (8.4%, n = 40/477) were also risk factors. Arterial ischaemic stroke was diagnosed in 11.1% of cases (n = 53/477), while pulmonary embolism was identified in 7.1% (n = 34/477) and 1.7% of cases were fatal (n = 8/477). LMWH was the first-line therapeutic of choice (47.8%, n = 228/477), with 28.1% (n = 134/477) requiring no intervention. CONCLUSION: These data reiterate the elevated thrombosis risk to which infants and children with central vascular access are exposed.


Subject(s)
Brain Ischemia , Central Venous Catheters , Stroke , Thrombosis , Anticoagulants , Canada/epidemiology , Central Venous Catheters/adverse effects , Child , Heparin, Low-Molecular-Weight/adverse effects , Humans , Infant , Retrospective Studies , Thrombosis/epidemiology , Thrombosis/etiology
18.
J Pediatr Hematol Oncol ; 43(7): e1040-e1044, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-33369999

ABSTRACT

BACKGROUND: Low-molecular-weight heparin is cleared through the kidneys and is commonly used for anticoagulation in the pediatric population. OBSERVATION: We present 3 challenging cases of children requiring anticoagulation in the context of acute kidney injury, nephrotic syndrome, and hemodialysis. CONCLUSIONS: A significant change in anti-factor Xa (anti-Xa) levels-used for drug monitoring-should prompt an assessment of renal function. In nephrotic syndrome, anti-Xa levels should be closely monitored when there is a change in the status of nephrotic disease activity. In hemodialysis patients, enoxaparin at once daily reduced dosing should be considered with trough and peak anti-Xa levels monitoring.


Subject(s)
Acute Kidney Injury/complications , Algorithms , Enoxaparin/administration & dosage , Factor Xa Inhibitors/administration & dosage , Nephrotic Syndrome/complications , Renal Dialysis/adverse effects , Thromboembolism/drug therapy , Anticoagulants/administration & dosage , Anticoagulants/blood , Child , Child, Preschool , Drug Monitoring/methods , Enoxaparin/blood , Factor Xa Inhibitors/blood , Female , Humans , Male , Prognosis , Thromboembolism/blood , Thromboembolism/etiology , Thromboembolism/pathology
19.
Pediatr Blood Cancer ; 67(4): e28114, 2020 04.
Article in English | MEDLINE | ID: mdl-31876366

ABSTRACT

Neonatal inferior vena cava syndrome (IVCS), though uncommon, is associated with significant morbidity and mortality. Information on risk factors, diagnosis, treatment, and outcomes is limited. This review comprised 61 neonates across 33 reports. Thrombosis occurred in 98% and 42% involved a central venous catheter. Diagnosis was mainly established by ultrasound in 82%. Therapeutically, heparin was employed in 36% and thrombolysis in 18% of the cases. The overall mortality was 23%. An algorithm of clinical signs, investigation, and management is presented. Well-designed prospective studies are needed to establish a concrete investigational approach to neonatal IVCS and institute safe, evidence-based treatment.


Subject(s)
Vena Cava, Inferior/pathology , Venous Thrombosis/diagnosis , Venous Thrombosis/etiology , Venous Thrombosis/therapy , Female , Humans , Infant, Newborn , Male , Risk Factors , Syndrome
20.
Pediatr Blood Cancer ; 66(4): e27572, 2019 04.
Article in English | MEDLINE | ID: mdl-30520242

ABSTRACT

BACKGROUND: Neonatal portal vein thrombosis (PVT) is uncommon with potentially serious complications that may manifest in infancy and childhood. OBJECTIVE: The primary aim of our study was to describe the short-term and long-term outcomes of neonatal PVT. METHODS: A retrospective chart review was conducted from 2008 to 2016 of neonates diagnosed with PVT. A systematic review was also performed from 2000 to 2018 to evaluate anticoagulant therapy (ACT) in neonatal PVT. RESULTS: Forty-four premature and 30 term infants (mean gestational age 30.7 vs 39.1 weeks, respectively) had PVT. Sixty-eight involved the left portal vein, one involved only the main portal vein, and 5 involved ≥1 vein. PVT was catheter associated in 46 (62%); none of the 7 neonates tested had thrombophilia. Of 74 neonates, 19 (26%) received ACT and 55 (74%) were untreated. The mean follow-up duration was 16.6 months (SD = 17.62; range, 0-89.6); 59.5% were followed for ≥6 months. On last ultrasound examination, thrombus resolution was documented in treated (ACT; n = 19) and nontreated (n = 55) neonates: 12 (63%) versus 32 (58%) with complete resolution, 1 (5%) versus 6 (11%) partial, 0 versus 1 (2%) extension, and 6 (32%) versus 16 (29%) had nonprogressive lesions, respectively. Seventy-one (96%) had no complications. Seventy-one articles met inclusion criteria for the systematic review and 19 were retained for analysis after assessment. CONCLUSIONS: PVT resolution rate was similar to previous reports. Although a low complication rate was detected, longer follow-up is necessary to determine the need for early treatment and the precise incidence of outcomes such as portal hypertension.


Subject(s)
Infant, Newborn, Diseases , Portal Vein/diagnostic imaging , Thrombosis , Female , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases/diagnostic imaging , Infant, Newborn, Diseases/therapy , Male , Retrospective Studies , Thrombosis/diagnostic imaging , Thrombosis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...