Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Haemophilia ; 13(5): 560-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17880444

ABSTRACT

The most serious site of bleeding for patients with haemophilia is the central nervous system. Intracranial haemorrhage (ICH) in patients with haemophilia can occur spontaneously or following mild head trauma however no guidelines exist for the approach to these patients. The goal of this review was to determine the utility of screening computed tomography (CT) of the head for patients with haemophilia who experience head trauma and to determine if the use of clinical criteria could allow a selective approach to radiographic imaging. In a retrospective study we reviewed the management of head trauma in a cohort of paediatric patients with haemophilia in a single institution. The cohort included males, ages birth to 18 years with haemophilia A or B who were followed at the haemophilia treatment center at The Children's Hospital of Philadelphia from 1994 to 2005. Between the years of 1994 and 2005, 97 patients were evaluated for head trauma for a total of 374 emergency department visits. There were 295 head CT scans performed to identify 9 (3%) episodes of intracranial bleeding. Fifty-six per cent of the patients with intracranial bleeding had no clinical signs or symptoms. The clinical outcome was excellent in all cases with no deaths or reported morbidity. In this cohort, a lack of symptoms and a normal neurological exam did not exclude ICH, especially in patients with severe haemophilia who were evaluated soon after a mild head trauma event suggesting the utility of early head CT imaging.


Subject(s)
Hemophilia A/complications , Intracranial Hemorrhages/diagnostic imaging , Tomography, X-Ray Computed/statistics & numerical data , Child , Child, Preschool , Craniocerebral Trauma/complications , Humans , Infant , Intracranial Hemorrhages/etiology , Male , Retrospective Studies , Treatment Outcome
2.
Pediatr Crit Care Med ; 6(3): 348-51, 2005 May.
Article in English | MEDLINE | ID: mdl-15857537

ABSTRACT

OBJECTIVE: To describe the outcome and treatment of two patients with recombinant factor VIIa (rFVIIa) for severe hemorrhage associated with extracorporeal membrane oxygenation (ECMO). DESIGN: Case report. SETTING: A 38-bed pediatric intensive care unit and 20-bed pediatric cardiac intensive care unit at a tertiary care children's hospital. PATIENT: Two patients with life-threatening hemorrhagic complications associated with ECMO requiring massive transfusion of blood products. INTERVENTIONS: Administration of repeated doses of rFVIIa at 90 microg/kg/dose. MEASUREMENT AND MAIN RESULTS: PATIENT 1 was an 11-yr-old male with a dilated cardiomyopathy who had undergone an orthotopic heart transplant treated with venoarterial ECMO postoperatively for right ventricular dysfunction. PATIENT 2 was a 13-yr-old male treated with venoarterial ECMO for cardiopulmonary failure from necrotizing staphylococcal pneumonia. Both patients had severe hemorrhage from the cannulation sites and thoracostomy tubes requiring massive transfusion to maintain intravascular blood volume and replace clotting factors. Both patients were treated with rFVIIa every 2-4 hrs and attained hemostasis. PATIENT 1 was administered three doses and PATIENT 2 was administered ten doses. No evidence of abnormal thrombus formation was noted in their respective ECMO circuits. CONCLUSIONS: The efficacy of rFVIIa in reducing intractable bleeding postcardiac surgery and in other coagulopathic states is being investigated. Despite theoretical concerns of thrombosis, these cases illustrate that there may be a role for the cautious use of rFVIIa in treating severe and intractable hemorrhage associated with ECMO.


Subject(s)
Extracorporeal Membrane Oxygenation/adverse effects , Factor VII/therapeutic use , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Adolescent , Child , Factor VIIa , Hemorrhage/etiology , Humans , Intensive Care Units, Pediatric , Male , Recombinant Proteins/therapeutic use
3.
Haemophilia ; 10 Suppl 4: 47-54, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15479372

ABSTRACT

During the past two decades, the improvement of therapeutic agents for the management of haemophilia has created the opportunity for individuals with haemophilia to live normal lives. However, in some instances, the progress made has been accompanied by emergence of unexpected risks and other new complications. A number of viruses have either emerged, or become greater risks to people with haemophilia. In addition, the drive of many countries towards self-sufficiency in blood products may in fact be endangering people with haemophilia by restricting blood donation to a pool of donors with high infection risk, discouraging commercial interests from developing safer products, and discouraging use of 'foreign' products even where that may be the safer option. Gene therapy for haemophilia, although an encouraging new treatment, has brought with it a number of adverse events, including risk of virus infection and development of carcinomas. The risk of inhibitors is still the most important problem for people with haemophilia, and a recent report showed that the type of factor concentrate does not impact significantly on this risk. Despite the advent of new and promising treatments for haemophilia, heathcare providers must be alert to new risks posed by them.


Subject(s)
Hemophilia A/therapy , Blood Coagulation Factors/adverse effects , Blood Coagulation Factors/antagonists & inhibitors , Blood Coagulation Factors/supply & distribution , Blood Component Transfusion/methods , Consumer Product Safety , Genetic Therapy/adverse effects , Health Policy , Hemophilia A/complications , Hemophilia A/drug therapy , Humans , Risk Factors , Virus Diseases/complications
6.
J Pediatr Hematol Oncol ; 23(9): 620-2, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11902309

ABSTRACT

Two boys with severe factor VIII deficiency that initially presented with acute onset of joint pain and swelling consistent with an uncomplicated hemarthrosis are reported. When appropriate management failed to provide resolution of symptoms, alternate diagnoses were considered. Both boys ultimately had complex regional pain syndrome (CRPS) diagnosed. The delay in diagnosis contributed to prolonged patient discomfort and lack of appropriate therapy. Complex regional pain syndrome encompasses a group of disorders that are characterized by pain severity or duration disproportionate to that expected. It is uncommon in the pediatric population. Because early diagnosis and appropriate treatment may improve outcome, it is important for practitioners to consider CRPS in the differential diagnosis of persistent pain in children with hemophilia.


Subject(s)
Causalgia/etiology , Hemophilia A/complications , Amitriptyline/therapeutic use , Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Causalgia/diagnosis , Causalgia/drug therapy , Causalgia/therapy , Child , Combined Modality Therapy , Diagnosis, Differential , Hemarthrosis/diagnosis , Hemarthrosis/etiology , Humans , Male , Physical Therapy Modalities , Remission Induction , Transcutaneous Electric Nerve Stimulation
7.
J Pediatr Hematol Oncol ; 22(4): 351-4, 2000.
Article in English | MEDLINE | ID: mdl-10959907

ABSTRACT

A female, term newborn born to a mother with a history of idiopathic thrombocytopenic purpura and antinuclear antibodies, single-stranded A antibody, and IgM anticardiolipin antibodies presented with immune thrombocytopenia, disseminated intravascular coagulation (DIC), microangiopathic hemolytic anemia, and a characteristic lupus rash in the periorbital areas. She responded to combined treatment with dexamethasone and intravenous immunoglobulin (IVIG). At age 9 months, she was readmitted with severe thrombocytopenia, DIC, and microangiopathic hemolytic anemia. She again responded to IVIG. This suggests that microangiopathic hemolysis can be a presenting symptom in neonatal lupus erythematosus and that reoccurrence of the microangiopathic hemolysis may occur even after the disappearance of lupus antibodies.


Subject(s)
Anemia, Hemolytic, Congenital/therapy , Lupus Erythematosus, Systemic/therapy , Anemia, Hemolytic, Congenital/immunology , Dexamethasone/therapeutic use , Disseminated Intravascular Coagulation/congenital , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/therapy , Female , Glucocorticoids/therapeutic use , Humans , Immunoglobulins, Intravenous/therapeutic use , Infant, Newborn , Lupus Erythematosus, Systemic/congenital , Lupus Erythematosus, Systemic/immunology , Plasma , Platelet Transfusion , Thrombocytopenia/congenital , Thrombocytopenia/immunology , Thrombocytopenia/therapy
8.
Nat Genet ; 24(3): 257-61, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10700178

ABSTRACT

Pre-clinical studies in mice and haemophilic dogs have shown that introduction of an adeno-associated viral (AAV) vector encoding blood coagulation factor IX (FIX) into skeletal muscle results in sustained expression of F.IX at levels sufficient to correct the haemophilic phenotype. On the basis of these data and additional pre-clinical studies demonstrating an absence of vector-related toxicity, we initiated a clinical study of intramuscular injection of an AAV vector expressing human F.IX in adults with severe haemophilia B. The study has a dose-escalation design, and all patients have now been enrolled in the initial dose cohort (2 x 10(11) vg/kg). Assessment in the first three patients of safety and gene transfer and expression show no evidence of germline transmission of vector sequences or formation of inhibitory antibodies against F.IX. We found that the vector sequences are present in muscle by PCR and Southern-blot analyses of muscle biopsies and we demonstrated expression of F.IX by immunohistochemistry. We observed modest changes in clinical endpoints including circulating levels of F.IX and frequency of FIX protein infusion. The evidence of gene expression at low doses of vector suggests that dose calculations based on animal data may have overestimated the amount of vector required to achieve therapeutic levels in humans, and that the approach offers the possibility of converting severe haemophilia B to a milder form of the disease.


Subject(s)
Dependovirus/genetics , Factor IX/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Hemophilia B/therapy , Muscle, Skeletal/metabolism , Adult , Aged , Blood Coagulation Tests , Blotting, Southern , Factor IX/analysis , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemophilia B/genetics , Humans , Injections, Intramuscular , Male , Muscle, Skeletal/virology , Polymerase Chain Reaction , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Treatment Outcome
9.
Haemophilia ; 5 Suppl 3: 33-41, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10597386

ABSTRACT

Inhibitors to factor VIII develop in 4-20% of haemophilia A patients, with the percentage rising to 52% in certain subpopulations. The management of inhibitor patients is directed toward stopping acute haemorrhages, providing short-term haemostasis before and after surgery, and inducing immune tolerance to factor VIII (immune tolerance therapy or ITT). Several different protocols have been used for ITT, but they are all centred around ongoing exposure to high doses of factor VIII. High responders (those patients with a large increase in inhibitor level after exposure to factor VIII) are the prime candidates for ITT, but low responders may also benefit from this treatment. It is often necessary to treat bleeding episodes during ITT, because elimination of inhibitors may require many years of therapy. Treatment of haemorrhages in inhibitor patients is reviewed for both low and high responders during ITT and in the absence of ITT. The choice of clotting agent for inhibitor patients who have not yet responded to ITT depends on current and past inhibitor levels, the severity of the haemorrhage, the site of the haemorrhage or the setting in which it occurs (e.g. surgical), and the extent of inhibitor cross-reactivity with porcine factor VIII. Patients with high-titre inhibitors experiencing a critical haemorrhage are generally best managed with bypassing agents (AUTOPLEX T or FEIBA VH), porcine factor VIII or rFVIIa.


Subject(s)
Hemophilia A/drug therapy , Hemorrhage/drug therapy , Immune Tolerance , Blood Coagulation Factors/immunology , Costs and Cost Analysis , Humans , Immunosuppressive Agents/therapeutic use , Isoantibodies/adverse effects
10.
Transfus Sci ; 21(1): 63-72, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10724785

ABSTRACT

The majority of children who undergo open-heart surgery with cardiopulmonary bypass (CPB) require perioperative blood transfusion. Blood product requirements are affected by factors such as patient age, underlying cardiac disease, complexity of the surgical procedure, and hemostatic alterations induced by CPB. Transfusion support may include the use of whole blood and/or individual blood components with transfusion practices varying widely based on individual preferences and blood product availability. Approaches to limit allogeneic blood exposure include the use of modified ultrafiltration and smaller bypass circuits, preoperative autologous blood donation and intraoperative blood salvage, and adjunctive antifibrinolytic agents. Potential advantages and disadvantages of the different blood products and pharmacological agents must be considered in managing the pediatric cardiac surgery patient.


Subject(s)
Blood Transfusion/methods , Cardiovascular Surgical Procedures/methods , Blood Transfusion/instrumentation , Blood Transfusion, Autologous/instrumentation , Blood Transfusion, Autologous/methods , Child , Child, Preschool , Humans , Perioperative Care
11.
J Pediatr ; 133(6): 777-81, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9842043

ABSTRACT

OBJECTIVE: Resistance to activated protein C (APC) has been identified as a risk factor for thrombotic disease in adults. In over 90% of cases, the basis for the APC resistance is a mutation in the coagulation factor V gene (factor V Leiden) that renders the protein more resistant to inactivation by APC. We sought to determine the prevalence of the factor V Leiden (FVL) mutation in neonates and children who had experienced an arterial or venous thromboembolic event. STUDY DESIGN: We retrospectively analyzed the clinical records of 33 neonates and 52 children with thromboembolic disease. Screening for the FVL mutation was performed by DNA analysis, allowing for identification of patients as normal, heterozygous, or homozygous. RESULTS: Of the 85 patients studied, 12 (14.1%) were heterozygous for FVL; none were homozygous. Of the 47 patients who had arterial central nervous system events, 8 (17%) were positive for the FVL mutation, including 6 of 22 (27%) neonates. Of those patients who had a venous thrombosis, 4 of 32 (12.5%) were FVL positive. None of the 85 patients had protein C deficiency, 3.5% had protein S deficiency, 1.2% had antithrombin III deficiency, and 16.5% had anti-phospholipid antibodies. CONCLUSION: These data suggest that the FVL mutation plays a role in the development of arterial and venous thrombotic events in neonates and children.


Subject(s)
Factor V/genetics , Mutation , Thromboembolism/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Humans , Infant , Infant, Newborn , Retrospective Studies , Venous Thrombosis/genetics
12.
Ann N Y Acad Sci ; 850: 242-50, 1998 Jun 30.
Article in English | MEDLINE | ID: mdl-9668546

ABSTRACT

Patients with homozygous beta-thalassemia are chronically transfused and, if not assiduously chelated, are at risk for cardiac dysfunction. Available data suggest that even in optimally chelated patients, cardiac pathology is abnormal secondary to iron deposition, fibrosis, hypertrophy, and the structural effects of chronic anemia. Evidence of myopericarditis may also be found. Cardiac performance is usually only subtly affected, primarily with diastolic abnormalities not routinely detected on echocardiograms or nuclear scan. In poorly chelated patients, severe heart failure occurs and is easily predictable but invariably fatal, despite treatment with diuretics, vasodilators, inotropes, and antiarrhythmics. Based on successful prevention of heart failure with ACE inhibitors in other forms of cardiomyopathy, we suggest multicenter trials to explore methods to stabilize cardiac function in patients at risk for iron-induced heart disease. Long-term adverse effects of iron deposition, diastolic dysfunction, and abnormal hormone regulation need to be quantitated in patients reaching their third and fourth decades when the potential for ischemic cardiac disease could compound cardiac dysfunction.


Subject(s)
Heart Diseases/etiology , Heart Diseases/therapy , Iron/metabolism , beta-Thalassemia/complications , beta-Thalassemia/therapy , Anemia, Hemolytic/etiology , Anemia, Hemolytic/physiopathology , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Heart Diseases/physiopathology , Homozygote , Humans , Myocarditis/etiology , Myocarditis/physiopathology , Pericarditis/etiology , Pericarditis/physiopathology , beta-Thalassemia/metabolism
13.
Transfusion ; 37(7): 703-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9225933

ABSTRACT

BACKGROUND: Hemoglobin (Hb) Bryn Mawr is an unstable Hb variant resulting in congenital hemolytic anemia. This variant Hb also has an increased affinity for oxygen. The perioperative transfusion management of this disorder is described, and the first genomic analysis of this Hb variant is given. CASE REPORT: An 11-year-old boy, heterozygous for Hb Bryn Mawr, was referred for cholecystectomy. Sequence analysis of genomic DNA confirmed that the patients was heterozygous for a T-->C transition in the codon for amino acid 85, causing a substitution of serine for phenylalanine in the beta-globin chain. On the basis of whole-blood O2 dissociation studies, projected tissue O2 delivery would have been suboptimal during general anesthesia; therefore, a partial red cell exchange transfusion was performed to lower variant Hb and prevent tissue hypoxia during surgery. The red cell mass to be exchanged (50%) was determined from the calculated increase in O2 delivery capacity required to maintain an O2 extraction of 4 to 5 mL of O2 per dL of whole blood. The p50 of whole blood from the patients immediately after the exchange transfusion was 16.0 torr. At the time of surgery, the p50 was normal (25.9 torr). The patient's whole blood 2,3 DPG levels were 4.70 mmol per mL of red cells (before transfusion) (normal range = 4.8 +/- 0.3 mmol/mL red cells), 4.07 mmol per mL of red cells (immediately after transfusion), and 4.55 mmol per mL of red cells (48 hours after transfusion). CONCLUSION: This patient with Hb Bryn Mawr was prepared for surgery with a partial exchange transfusion to prevent tissue hypoxia during anesthesia. Decreased 2,3 DPG levels immediately after transfusion resulted in increased O2 affinity of whole blood; however, 48 hours after exchange transfusion, a normal p50 (due to both removal of variant Hb and regeneration of 2,3, DPG) was observed. Partial exchange transfusion is useful in the preoperative management of patients with Hb variants characterized by increased O2 affinity.


Subject(s)
Blood Component Removal , Erythrocyte Transfusion , Hemoglobins, Abnormal/genetics , 2,3-Diphosphoglycerate , Anemia, Hemolytic , Child , Cholelithiasis/surgery , DNA Mutational Analysis , Diphosphoglyceric Acids/blood , Genetic Variation , Hemoglobinopathies/congenital , Humans , Hypoxia/prevention & control , Male , Oxygen/blood , Preoperative Care
14.
J Pediatr ; 130(6): 957-60, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9202619

ABSTRACT

OBJECTIVE: To determine whether the mean corpuscular hemoglobin concentration (MCHC) or other erythrocyte indexes, as determined by automated cell counters, remains a useful screening test for identifying patients with hereditary spherocytosis (HS). METHODS: Erythrocyte indexes from 112 children with HS who had not undergone splenectomy were compared with those measured in an equal number of healthy, age-matched children. All indexes were derived from measurements obtained by aperture impedance. RESULTS: Mean corpuscular hemoglobin concentration in the HS group was 35.9 gm/dl, significantly higher than in normal control subjects (34.3 gm/dl; p < 0.001). Mean erythrocyte distribution width also was significantly higher in patients with HS (19.3 vs 12.6; p < 0.001). The MCHC distinguishes individuals with HS, with an area under the receiver operating characteristic curve of 0.86. Although not disease specific, an erythrocyte distribution width > 14 has 85% sensitivity and 97% specificity and an area under the receiver operating characteristic curve of 0.92. An MCHC > 35 gm/dl has a sensitivity of 70% and a specificity of 86%. Combining the MCHC and erythrocyte distribution width increases the area under the receiver operating characteristic curve to 0.97. Specificity is 100% and likelihood ratio is infinite when both the MCHC and erythrocyte distribution width are elevated. CONCLUSIONS: The automated MCHC is an effective screening test to identify children with HS. An elevated erythrocyte distribution width adds additional specificity and is itself a powerful screening tool. The combination of the two tests is an excellent predictor for the diagnosis of HS.


Subject(s)
Erythrocyte Indices , Spherocytosis, Hereditary/diagnosis , Adolescent , Adult , Child , Child, Preschool , Humans , Infant , Reticulocyte Count , Splenectomy
15.
J Pediatr ; 130(4): 537-40, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9108849

ABSTRACT

OBJECTIVE: Our previous experience with highly purified plasma-derived factor VIII (pdFVIII) concentrates showed that adult dosage recommendations were not applicable to children. In this study, we compared the in vivo response and recovery of recombinant factor VIII (rFVIII) with those of highly purified pdFVIII concentrate in children with hemophilia A. STUDY DESIGN: Ten boys with severe factor VIII deficiency and no concurrent bleeding episodes participated in a masked, prospective, crossover study comparing factor VIII coagulant activity after infusion of 50 units of pdFVIII and rFVIII products per kilogram of body weight. RESULTS: Mean peak factor VIII response with rFVIII was 1.91% +/- 0.14%, significantly better than the response observed with highly purified pdFVIII of 1.5% +/- 0.15% (p = 0.007). Mean peak factor VIII recovery was 100.5% with rFVIII versus 78.7% with pdFVIII (p = 0.007). Positive correlations between response to rFVIII and body surface area (r = 0.734, p = 0.015), body weight (r = 0.762, p = 0.01), and plasma volume (r = 0.659, p = 0.03) were observed. CONCLUSIONS: Infusion of rFVIII produced a significantly better response and recovery in vivo than infusion of highly purified pdFVIII in children. The response in children after infusion of rFVIII was similar to the response previously observed in adults.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/therapy , Recombinant Proteins/therapeutic use , Adult , Child , Cross-Over Studies , Factor VIII/analysis , Hemophilia A/blood , Humans , Male , Prospective Studies
17.
Am J Hematol ; 53(4): 248-50, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8948664

ABSTRACT

Go(a) (D(Cor)) is a low-frequency antigen in the Rh system found on red cells lacking part of the D mosaic (category IVa). Anti-Go(a) has not been previously reported to cause hemolytic transfusion reactions. A 27-year-old African American male with sickle-cell disease, maintained on chronic transfusion, was noted to have dark plasma during an erythrocytapheresis, procedure, and the pretransfusion hemoglobin was noted to be 1 g/dl lower than 4 weeks before (with hyperbilirubinemia and a significantly increased LDH). Polyspecific direct antiglobulin test (DAT) was weakly positive (C3-weak, IgG-weak), and indirect antiglobulin tests (IATs) performed on the serum (pre- and posttransfusion reaction) and a red blood cell (RBC) eluate from the postreaction sample were negative. A segment from one of the four implicated units from the prior month's transfusion was strongly reactive at 37 degrees C and using anti-human globulin (AHG) when crossmatched with the postreaction serum and the eluate. The postreaction serum, screened with a panel of red cells positive for low-prevalence antigens, reacted with three Go(a+) cells. The implicated unit was reactive with a previously identified anti-Go(a) serum.


Subject(s)
Anemia, Hemolytic/etiology , Blood Group Incompatibility/immunology , Rh-Hr Blood-Group System/immunology , Transfusion Reaction , Adult , Anemia, Hemolytic/immunology , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/immunology , Anemia, Sickle Cell/therapy , Blood Group Antigens/immunology , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/therapy , Humans , Immunization , Isoantibodies/immunology , Male
18.
J Pediatr ; 129(5): 656-60, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8917229

ABSTRACT

OBJECTIVE: To evaluate the effect of recombinant human erythropoietin (EPO) and iron supplementation on transfusion requirements in pediatric patients with sarcoma who were receiving chemotherapy, we performed a double-blind, placebo-controlled, randomized trial. METHODS: Twenty-four pediatric patients with malignant solid tumors were randomly assigned to receive either placebo (saline solution) or EPO for a 16-week study period. The starting dose was 150 IU/kg per dose three times a week and was escalated by 50 IU/kg per dose increments monthly until packed red blood cell (PRBC) transfusion independence was achieved or a dosage of 300 IU/kg per dose was reached. Iron supplementation was prescribed at a dose of 6 mg of elemental iron per kilogram daily. The primary study end point was the comparison of PRBC transfusion requirements in the two groups. RESULTS: Of 24 patients, 20 were evaluable for response. The median PRBC transfusion requirement during the 16-week period was 23 ml/kg in EPO-treated patients versus 80 ml/kg in placebo patients (p = 0.02). The median number of single-donor platelet units transfused was zero in the EPO-treated patients compared with four in the placebo group (p = 0.005). No statistical difference in the intensity of bone marrow suppression was seen, as measured by the median number of complete blood cell counts with an absolute neutrophil count of < 1000 cells/microliter. CONCLUSIONS: Treatment with EPO and iron significantly reduces PRBC transfusions in pediatric patients receiving concomitant chemotherapy for malignant sarcomas. A decrease in the number of platelet transfusions was also seen and deserves further study.


Subject(s)
Erythrocyte Transfusion , Erythropoietin/therapeutic use , Platelet Transfusion , Sarcoma/drug therapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Double-Blind Method , Female , Humans , Male , Recombinant Proteins/therapeutic use , Sarcoma/therapy , Treatment Outcome
19.
Blood ; 88(8): 3216-22, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8874223

ABSTRACT

Bone marrow transplantation (BMT) is now an option for some patients with sickle cell disease (SCD). Many SCD patients are multiply transfused with red blood cells (RBCs), and may be immunized to alloantigens other than erythrocyte antigens. Because platelet refractoriness is a significant complication during BMT, we wished to determine the prevalence of alloimmunization to platelets in transfused SCD patients. Sera collected from 47 transfused and 14 untransfused SCD patients were screened for HLA and platelet-specific antibodies. Transfusion and RBC antibody histories were reviewed. A subset of the patients were rescreened 1 year later. Eighty-five percent of patients with at least 50 RBC transfusions (22 of 26), 48% of patients with less than 50 transfusions (10 of 21), and none of 14 untransfused patients demonstrated platelet alloimmunization (P < .05). Platelet alloimmunization was more prevalent than RBC alloimmunization (20% to 30%). Half of the platelet reactivity was chloroquine-elutable. Eighteen of 22 patients (82%) on chronic RBC transfusion remained platelet-alloimmunized 11 to 22 months after initial testing. In summary, 85% of heavily transfused SCD patients are alloimmunized to HLA and/or platelet-specific antigens. These patients may be refractory to platelet transfusion, a condition that would increase their risk during BMT. Leukodepletion in the transfusion support of SCD patients should be considered to prevent platelet alloimmunization.


Subject(s)
Anemia, Sickle Cell/immunology , Antigens, Human Platelet/immunology , Blood Platelets/immunology , Blood Transfusion , Immunization , Isoantibodies/biosynthesis , Adolescent , Anemia, Sickle Cell/therapy , Bone Marrow Transplantation , Child , Child, Preschool , Chloroquine/pharmacology , Female , HLA Antigens/immunology , Humans , Isoantibodies/blood , Isoantibodies/immunology , Male
20.
Pediatr Clin North Am ; 43(3): 793-808, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8649910

ABSTRACT

The safety of the blood supply has increased tremendously in the past decade. Donor screening and improved infectious disease testing of units for transfusion have contributed to the decreased risk of transfusion-transmitted diseases. Reduction of the number of passenger leukocytes from RBC and platelet transfusions decreases the rate of febrile transfusion reactions and alloimmunization. Irradiation of cellular products helps prevent TA-GVHD. The practice of obtaining an informed consent prior to transfusion helps patients and families understand the risks and benefits of and alternatives to transfusion therapy.


Subject(s)
Blood Transfusion, Autologous , Blood Transfusion , Blood Donors , Graft vs Host Disease , Humans , Infant, Newborn , Platelet Transfusion
SELECTION OF CITATIONS
SEARCH DETAIL
...