Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Publication year range
1.
N Engl J Med ; 372(15): 1419-29, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25853746

ABSTRACT

BACKGROUND: Some observational studies have reported that transfusion of red-cell units that have been stored for more than 2 to 3 weeks is associated with serious, even fatal, adverse events. Patients undergoing cardiac surgery may be especially vulnerable to the adverse effects of transfusion. METHODS: We conducted a randomized trial at multiple sites from 2010 to 2014. Participants 12 years of age or older who were undergoing complex cardiac surgery and were likely to undergo transfusion of red cells were randomly assigned to receive leukocyte-reduced red cells stored for 10 days or less (shorter-term storage group) or for 21 days or more (longer-term storage group) for all intraoperative and postoperative transfusions. The primary outcome was the change in Multiple Organ Dysfunction Score (MODS; range, 0 to 24, with higher scores indicating more severe organ dysfunction) from the preoperative score to the highest composite score through day 7 or the time of death or discharge. RESULTS: The median storage time of red-cell units provided to the 1098 participants who received red-cell transfusion was 7 days in the shorter-term storage group and 28 days in the longer-term storage group. The mean change in MODS was an increase of 8.5 and 8.7 points, respectively (95% confidence interval for the difference, -0.6 to 0.3; P=0.44). The 7-day mortality was 2.8% in the shorter-term storage group and 2.0% in the longer-term storage group (P=0.43); 28-day mortality was 4.4% and 5.3%, respectively (P=0.57). Adverse events did not differ significantly between groups except that hyperbilirubinemia was more common in the longer-term storage group. CONCLUSIONS: The duration of red-cell storage was not associated with significant differences in the change in MODS. We did not find that the transfusion of red cells stored for 10 days or less was superior to the transfusion of red cells stored for 21 days or more among patients 12 years of age or older who were undergoing complex cardiac surgery. (Funded by the National Heart, Lung, and Blood Institute; RECESS ClinicalTrials.gov number, NCT00991341.).


Subject(s)
Blood Preservation , Cardiac Surgical Procedures , Erythrocyte Transfusion , Adult , Aged , Blood Grouping and Crossmatching , Erythrocyte Transfusion/adverse effects , Female , Humans , Intention to Treat Analysis , Length of Stay , Male , Middle Aged , Mortality , Multiple Organ Failure/classification , Proportional Hazards Models , Severity of Illness Index , Time Factors
2.
Blood ; 125(1): 155-61, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25342714

ABSTRACT

Antibodies specific for platelet factor 4 (PF4)/heparin complexes are the hallmark of heparin-induced thrombocytopenia and thrombosis (HIT), but many antibody-positive patients have normal platelet counts. The basis for this is not fully understood, but it is believed that antibodies testing positive in the serotonin release assay (SRA) are the most likely to cause disease. We addressed this issue by characterizing PF4-dependent binding of HIT antibodies to intact platelets and found that most antibodies testing positive in the SRA, but none of those testing negative, bind to and activate platelets when PF4 is present without any requirement for heparin (P < .0001). Binding of SRA-positive antibodies to platelets was inhibited by chondroitinase ABC digestion (P < .05) and by the addition of chondroitin-4-sulfate (CS) or heparin in excess quantities. The findings suggest that although all HIT antibodies recognize PF4 in a complex with heparin, only a subset of these antibodies recognize more subtle epitopes induced in PF4 when it binds to CS, the major platelet glycosaminoglycan. Antibodies having this property could explain "delayed HIT" seen in some individuals after discontinuation of heparin and the high risk for thrombosis that persists for weeks in patients recovered from HIT.


Subject(s)
Antibodies/chemistry , Blood Platelets/immunology , Heparin/chemistry , Platelet Factor 4/chemistry , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology , Chondroitin Sulfates/chemistry , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Glycosaminoglycans/chemistry , Humans , Immunoglobulin G/chemistry , P-Selectin/chemistry , Platelet Activation , Polysaccharide-Lyases/chemistry , Protein Binding , Serotonin/chemistry , Thrombosis/chemically induced
3.
Blood ; 126(10): 1234-6, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26209661

ABSTRACT

Incompatibility of the human platelet antigen-1 (HPA-1) system is the most common cause of fetal/neonatal alloimmune thrombocytopenia (F/NAIT) and is thought to be mediated by accelerated clearance of antibody-opsonized fetal platelets. We evaluated the effect of maternal sera containing anti-HPA-1a antibodies (F/NAIT sera) on in vitro megakaryopoiesis. Compared with control maternal sera, 14 out of 17 F/NAIT sera significantly reduced megakaryocyte (MK) number. This finding was associated with increased apoptosis and cell death of early MKs/MK progenitors, but normal maturation and differentiation of surviving MKs. An analysis of platelet counts in infants born to mothers following antenatal intravenous immunoglobulin (IVIG) ± prednisone therapy demonstrated a significant and moderately strong correlation between the MK growth in cultures and the infants' platelet counts at birth. These findings suggest that maternal anti-HPA-1a antibodies can suppress fetal megakaryopoiesis by inducing early cell death and that this influences the neonatal platelet count. Thus, the ability of maternal antibodies to suppress MK growth is a potential predictive factor for the fetal response to maternal IVIG therapy.


Subject(s)
Antigens, Human Platelet/immunology , Hematopoiesis/immunology , Megakaryocytes/immunology , Thrombocytopenia, Neonatal Alloimmune/immunology , Female , Humans , In Vitro Techniques , Integrin beta3 , Pregnancy , Thrombocytopenia, Neonatal Alloimmune/physiopathology
4.
Blood ; 126(18): 2153-61, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26333778

ABSTRACT

High-dose granulocyte transfusion therapy has been available for 20 years, yet its clinical efficacy has never been conclusively demonstrated. We report here the results of RING (Resolving Infection in Neutropenia with Granulocytes), a multicenter randomized controlled trial designed to address this question. Eligible subjects were those with neutropenia (absolute neutrophil count <500/µL) and proven/probable/presumed infection. Subjects were randomized to receive either (1) standard antimicrobial therapy or (2) standard antimicrobial therapy plus daily granulocyte transfusions from donors stimulated with granulocyte colony-stimulating factor (G-CSF) and dexamethasone. The primary end point was a composite of survival plus microbial response, at 42 days after randomization. Microbial response was determined by a blinded adjudication panel. Fifty-six subjects were randomized to the granulocyte arm and 58 to the control arm. Transfused subjects received a median of 5 transfusions. Mean transfusion dose was 54.9 × 10(9) granulocytes. Overall success rates were 42% and 43% for the granulocyte and control groups, respectively (P > .99), and 49% and 41%, respectively, for subjects who received their assigned treatments (P = .64). Success rates for granulocyte and control arms did not differ within any infection type. In a post hoc analysis, subjects who received an average dose per transfusion of ≥0.6 × 10(9) granulocytes per kilogram tended to have better outcomes than those receiving a lower dose. In conclusion, there was no overall effect of granulocyte transfusion on the primary outcome, but because enrollment was half that planned, power to detect a true beneficial effect was low. RING was registered at www.clinicaltrials.gov as #NCT00627393.


Subject(s)
Granulocytes/cytology , Infections/complications , Leukocyte Transfusion/methods , Neutropenia/complications , Neutropenia/therapy , Anti-Infective Agents/therapeutic use , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/drug effects , Humans , Infections/drug therapy , Leukocyte Count , Treatment Outcome
5.
Am J Hematol ; 92(8): 730-738, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28388835

ABSTRACT

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a thrombotic disorder usually prompting treatment with non-heparin anticoagulants. The benefits and risks of such treatments have not been fully assessed. METHODS: We analyzed data for 442 patients having a positive heparin-platelet factor 4 antibody test and recent heparin exposure. The primary outcome was a composite endpoint (death, limb amputation/gangrene, or new thrombosis). Secondary outcomes included bleeding and the effect of anticoagulation. FINDINGS: Seventy-one patients (16%) had HIT with thrombosis (HIT-T); 284 (64%) had HIT without thrombosis (isolated HIT); 87 (20%) did not have HIT. An intermediate or high "4T" score was found in 85%, 58%, and 8% of the three respective groups. Non-heparin anticoagulation was begun in 80%, 56%, and 45%. The composite endpoint occurred in 48%, 36%, and 17% (P = .01) of which 61%, 38%, and 40% were receiving non-heparin anticoagulation. Compared with the no HIT group, the composite endpoint was significantly more likely in HIT-T [HR 2.48 (1.35-4.55), P = .003)] and marginally more likely in isolated HIT [HR 1.66 (0.96-2.85), P = .071]. Importantly, risk increased (HR 1.77, P = .02) after platelet transfusion. Major bleeding occurred in 48%, 36%, and 16% of the three groups (P = .005). Non-heparin anticoagulation was not associated with a reduction in composite endpoint events in either HIT group. INTERPRETATION: HIT patients have high risks of death, limb amputation/gangrene, thrombosis, and bleeding. Non-heparin anticoagulant treatment may not benefit all patients and should be considered only after careful assessment of the relative risks of thrombosis and bleeding in individual patients.


Subject(s)
Heparin/adverse effects , Thrombocytopenia/epidemiology , Thrombocytopenia/etiology , Adult , Aged , Anticoagulants/therapeutic use , Autoantibodies/blood , Autoantibodies/immunology , Enzyme-Linked Immunosorbent Assay , Female , Heparin/immunology , Humans , Male , Middle Aged , Mortality , Patient Outcome Assessment , Platelet Count , Platelet Factor 4/immunology , Proportional Hazards Models , Retrospective Studies , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy
6.
Transfusion ; 56(10): 2449-2454, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27611703

ABSTRACT

BACKGROUND: Incompatibility between parental platelet (PLT) antigens may lead to sensitization of mother and development of fetal and neonatal alloimmune thrombocytopenia (FNAIT) resulting in fetal thrombocytopenia. Intravenous immunoglobulin (IVIG) with or without prednisone is the most effective, evidence-based antenatal treatment for subsequent FNAIT-affected pregnancies. IVIG infusion causes hemolysis in other settings, the degree depending upon patient blood groups (BGs). STUDY DESIGN AND METHODS: In ClinicalTrials.gov NCT00194987, 102 pregnant women received randomized antenatal treatment: Arm A received 2 g/kg/week IVIG; Arm B received 1 g/kg/week IVIG + 0.5 mg/kg/day prednisone. This post hoc analysis explored BG and anemia in 69 FNAIT mothers treated with Arm A or Arm B without salvage treatment to explore the effects of IVIG and steroid treatment on development of anemia in these women. Mothers whose treatment changed, for example, those with insufficient or unknown fetal PLT response who received salvage therapy, were excluded. RESULTS: For Arm A, 17 of 21 (hemoglobin [Hb] < 10 g/dL) mothers with anemia but only three of 15 mothers without anemia had BG-A and/or BG-B (p = 0.0005). BG was unrelated to anemia in Arm B; only nine of 33 Arm B mothers became anemic during treatment. The mean decrease in Hb level in women with BG-non-O was 1.9 g/dL and in women with BG-O was 1.1 g/dL (p = 0.004). Anemia was not caused by iron deficiency; the lowest mean corpuscular volume was 79. CONCLUSION: FNAIT women with BG-non-O more frequently develop anemia secondary to high-dose IVIG infusion (2 g/kg/week), quite possibly from isohemagglutinin-mediated hemolysis; maternal Hb requires monitoring. IVIG at 1 g/kg/week did not cause anemia in women with BG-non-O; concomitant prednisone likely alleviated the IVIG effect. Maternal BG could influence selection of antenatal treatment for FNAIT.


Subject(s)
Anemia/etiology , Blood Group Antigens , Immunoglobulins, Intravenous/administration & dosage , Thrombocytopenia, Neonatal Alloimmune/drug therapy , Anemia/chemically induced , Anemia/immunology , Female , Hemoglobins/analysis , Humans , Immunoglobulins, Intravenous/adverse effects , Mothers , Prednisone/pharmacology , Prednisone/therapeutic use , Pregnancy , Steroids/pharmacology , Steroids/therapeutic use , Treatment Outcome
7.
Am J Obstet Gynecol ; 215(4): 471.e1-9, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27131591

ABSTRACT

BACKGROUND: Fetal-neonatal alloimmune thrombocytopenia affects approximately 1 of 1000 live births, most of which are not severely thrombocytopenic. Despite effective treatment with intravenous gammaglobulin and/or prednisone, antenatal management of a subsequent affected pregnancy is complicated by the risks associated with fetal blood sampling. Furthermore, there are no biomarker(s) of high risk other than the occurrence of intracranial hemorrhage in a previous sibling. Management of these high-risk pregnancies requires intensive treatment initiated at 12 weeks of gestation. OBJECTIVE: The objective of the study was to evaluate whether empiric escalation of therapy at 32 weeks allows the omission of fetal blood sampling in all fetal-neonatal alloimmune thrombocytopenia-affected patients. Specifically, we sought to determine whether intensive intravenous gammaglobulin-based regimens for the treatment of a subsequent fetal-neonatal alloimmune thrombocytopenia-affected pregnancy followed by empirically escalated intravenous gammaglobulin and prednisone treatment would increase the fetal platelet count and thus safely allow omission of fetal blood sampling in the antepartum management of these patients. STUDY DESIGN: In this prospective, multicenter, randomized controlled study, 99 women with fetal-neonatal alloimmune thrombocytopenia whose prior affected child did not have an intracranial hemorrhage were randomized to receive an intensive intravenous gammaglobulin-based regimen: 2 g/kg per week or intravenous gammaglobulin 1 g/kg per week plus prednisone 0.5 mg/kg per day, starting at 20-30 weeks of gestation. Escalated therapy (intravenous gammaglobulin 2 g/kg per week plus prednisone 0.5 mg/kg per day) was recommended and usually initiated at 32 weeks when fetal counts were <50,000/mL(3) or when fetal blood sampling was not performed. The preliminary report of this study from 2007 demonstrated the efficacy of both intravenous gammaglobulin-based regimens in most patients. Most patients who underwent fetal sampling had adequate fetal counts and therefore did not have their treatment escalated. This post hoc analysis describes the 29 fetuses who had their treatment escalated either because they had low counts at 32 weeks or when sampling was not performed. This study explored whether the empiric escalation of treatment at 32 weeks was sufficiently effective in increasing fetal platelet counts in these patients. RESULTS: Mean fetal and birth counts of fetuses randomized to each of the 2 initial treatment groups were all >100,000/mL(3). Three neonates had an intracranial hemorrhage; all 3 were grade 1 and all had birth platelet counts >130,000/mL(3). In a post hoc analysis, 19 fetuses undergoing fetal blood sampling at 32 weeks had fetal platelet counts <50,000/mL(3) despite their initial treatment. Of these 19, birth platelet counts were >50,000/mL(3) in 11 of 13 fetuses who received escalated treatment compared with only 1 of 6 of those who did not (P = .01); only 3 fetuses that received initial therapy followed by escalated treatment had birth platelet counts <50,000/mL(3) and none had an intracranial hemorrhage. The platelet counts of 14 of 15 fetuses that received empirically escalated treatment without sampling were >50,000/mL(3) at birth. In addition, none of these had an intracranial hemorrhage. CONCLUSION: The 2 recommended protocols of intensive initial treatment followed by empiric escalation of therapy at 32 weeks of gestation are reasonably safe, effective in increasing fetal platelet counts, and allow omission of fetal blood sampling by increasing the fetal platelet count in almost all cases.


Subject(s)
Immunoglobulins, Intravenous/administration & dosage , Immunologic Factors/administration & dosage , Pregnancy Complications/diagnosis , Pregnancy Complications/drug therapy , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Thrombocytopenia, Neonatal Alloimmune/drug therapy , Cordocentesis/adverse effects , Female , Fetal Blood , Gestational Age , Humans , Intracranial Hemorrhages/etiology , Platelet Count , Prednisone/administration & dosage , Pregnancy , Pregnancy Complications/blood , Prenatal Diagnosis , Prospective Studies , Thrombocytopenia, Neonatal Alloimmune/blood
8.
Blood ; 122(3): 303-4, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23869072

ABSTRACT

In this issue of Blood, Ghevaert et al propose to develop a therapeutic antibody for fetal and neonatal alloimmune thrombocytopenia (FNAIT) that would block the actual antibody in sensitized mothers from binding and therefore prevent, or at least ameliorate, fetal and neonatal thrombocytopenia in fetuses who would otherwise be affected.1 The goal of the group is to engineer an antibody reagent that would on the one hand not engage conventional activating Fc receptors and on the other hand interact normally with FcRn, allowing transplacental passage.


Subject(s)
Antibodies/therapeutic use , Recombinant Proteins/therapeutic use , Thrombocytopenia, Neonatal Alloimmune/drug therapy , Female , Humans , Male
9.
Transfusion ; 54(5): 1286-93, 2014 May.
Article in English | MEDLINE | ID: mdl-24128174

ABSTRACT

BACKGROUND: Twenty-four low-frequency human platelet antigens (LFHPAs) have been implicated as immunogens in neonatal alloimmune thrombocytopenia (NAIT). We performed studies to define more fully how often these antigens trigger maternal immunization leading to NAIT. STUDY DESIGN AND METHODS: In a Phase 1 study, fathers of selected NAIT cases not resolved by serologic testing but thought to have a high likelihood of NAIT on clinical and serologic grounds were typed for LFHPAs by DNA sequencing. In a Phase 2 study, high-throughput methods were used to type fathers of 1067 consecutive unresolved NAIT cases for LFHPAs. Mothers of 1338 unresolved cases were also typed to assess the prevalence of LFHPAs in a population racially/ethnically similar to the fathers. RESULTS: In Phase 1, LFHPAs were identified in 16 of 244 fathers (6.55%). In Phase 2, LFPAs were found in only 28 of 1067 fathers (2.62%). LFHPAs were identified in 27 of 1338 maternal samples (2.01%). HPA-9bw was by far the most common LFHPA identified in the populations studied and was the only LFHPA that was significantly more common in fathers than in mothers of affected infants (p = 0.02). CONCLUSIONS: Maternal immunization against recognized LFHPAs accounts for only a small fraction of the cases of apparent NAIT not resolved by standard serologic testing. Typing of the fathers of such cases for LFHPAs is likely to be rewarding only when a maternal antibody specific for a paternal platelet glycoprotein is demonstrated and/or there is compelling clinical evidence for NAIT.


Subject(s)
Antigens, Human Platelet/genetics , Thrombocytopenia, Neonatal Alloimmune/etiology , Alleles , Female , High-Throughput Screening Assays , Humans , Male , Polymorphism, Single Nucleotide
11.
Br J Haematol ; 161(1): 3-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23384054

ABSTRACT

Neonatal alloimmune thrombocytopenia, (NAIT) is caused by maternal antibodies raised against alloantigens carried on fetal platelets. Although many cases are mild, NAIT is a significant cause of morbidity and mortality in newborns and is the most common cause of intracranial haemorrhage in full-term infants. In this report, we review the pathogenesis, clinical presentation, laboratory diagnosis and prenatal and post-natal management of NAIT and highlight areas of controversy that deserve the attention of clinical and laboratory investigators.


Subject(s)
Thrombocytopenia, Neonatal Alloimmune/diagnosis , Antigens, Human Platelet/immunology , Biomarkers/blood , Female , Humans , Infant, Newborn , Perinatal Care/methods , Platelet Transfusion/methods , Pregnancy , Prenatal Care/methods , Thrombocytopenia, Neonatal Alloimmune/immunology , Thrombocytopenia, Neonatal Alloimmune/therapy
12.
N Engl J Med ; 362(7): 600-13, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20164484

ABSTRACT

BACKGROUND: We conducted a trial of prophylactic platelet transfusions to evaluate the effect of platelet dose on bleeding in patients with hypoproliferative thrombocytopenia. METHODS: We randomly assigned hospitalized patients undergoing hematopoietic stem-cell transplantation or chemotherapy for hematologic cancers or solid tumors to receive prophylactic platelet transfusions at a low dose, a medium dose, or a high dose (1.1x10(11), 2.2x10(11), or 4.4x10(11) platelets per square meter of body-surface area, respectively), when morning platelet counts were 10,000 per cubic millimeter or lower. Clinical signs of bleeding were assessed daily. The primary end point was bleeding of grade 2 or higher (as defined on the basis of World Health Organization criteria). RESULTS: In the 1272 patients who received at least one platelet transfusion, the primary end point was observed in 71%, 69%, and 70% of the patients in the low-dose group, the medium-dose group, and the high-dose group, respectively (differences were not significant). The incidences of higher grades of bleeding, and other adverse events, were similar among the three groups. The median number of platelets transfused was significantly lower in the low-dose group (9.25x10(11)) than in the medium-dose group (11.25x10(11)) or the high-dose group (19.63x10(11)) (P=0.002 for low vs. medium, P<0.001 for high vs. low and high vs. medium), but the median number of platelet transfusions given was significantly higher in the low-dose group (five, vs. three in the medium-dose and three in the high-dose group; P<0.001 for low vs. medium and low vs. high). Bleeding occurred on 25% of the study days on which morning platelet counts were 5000 per cubic millimeter or lower, as compared with 17% of study days on which platelet counts were 6000 to 80,000 per cubic millimeter (P<0.001). CONCLUSIONS: Low doses of platelets administered as a prophylactic transfusion led to a decreased number of platelets transfused per patient but an increased number of transfusions given. At doses between 1.1x10(11) and 4.4x10(11) platelets per square meter, the number of platelets in the prophylactic transfusion had no effect on the incidence of bleeding. (ClinicalTrials.gov number, NCT00128713.)


Subject(s)
Hematopoietic Stem Cell Transplantation , Hemorrhage/prevention & control , Platelet Transfusion , Thrombocytopenia/therapy , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hemorrhage/etiology , Hemostasis , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/therapy , Platelet Count , Platelet Transfusion/adverse effects , Platelet Transfusion/methods , Thrombocytopenia/etiology
13.
Transfusion ; 53(6): 1309-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23003125

ABSTRACT

BACKGROUND: Recent studies suggest that HPA-1a-specific, low-avidity maternal antibodies not detectable by conventional methods can cause neonatal alloimmune thrombocytopenia (NAIT). We performed studies to further define the incidence and clinical significance of this type of antibody. STUDY DESIGN AND METHODS: Surface plasmon resonance analysis was used to detect low-avidity antibodies in HPA-1a-negative, "antibody-negative" mothers of suspected NAIT cases. The ability of antibodies detected to promote immune destruction of human platelets (PLTs) was examined in a newly developed NOD/SCID mouse model. RESULTS: Among 3478 suspected cases of NAIT, 677 HPA-1a-negative mothers were identified. HPA-1a-specific antibodies were detected by conventional antibody testing in 616 cases (91%). Low-avidity HPA-1a-specific antibodies were identified in 18 of the remaining 61 cases (9%). Clinical follow-up on 13 cases showed that eight were referred because of suspected NAIT and five because the mother's sister had previously had an infant with NAIT. Only six infants born to the 13 sensitized mothers had clinically significant thrombocytopenia at birth. Three of four low-avidity antibodies tested in the mouse caused accelerated clearance of HPA-1a/a but not HPA-1b/b PLTs. Only 3 of 12 mothers with low-avidity HPA-1a antibodies were positive for HLA-DRB3*0101. CONCLUSIONS: The findings confirm previous reports that low-avidity HPA-1a antibodies can cause NAIT but show that the presence of such an antibody does not predict that an infant will be affected. The low incidence of HLA-DRB3*0101 in this cohort (p < 0.0001) suggests that women negative for DRB3*0101 may be predisposed to produce low-avidity HPA-1a antibodies.


Subject(s)
Antigens, Human Platelet/immunology , Pregnancy Complications/epidemiology , Pregnancy Complications/immunology , Thrombocytopenia, Neonatal Alloimmune/epidemiology , Thrombocytopenia, Neonatal Alloimmune/immunology , Animals , Antibody Affinity/immunology , Blood Platelets/immunology , Cohort Studies , Female , HLA-DRB3 Chains/immunology , Humans , Incidence , Infant , Infant, Newborn , Integrin beta3 , Male , Mice , Mice, Inbred NOD , Mice, SCID , Pregnancy , Prevalence , Seroepidemiologic Studies , Surface Plasmon Resonance
14.
Br J Haematol ; 158(4): 539-47, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22775462

ABSTRACT

Rituximab is widely used in autoimmune diseases including immune thrombocytopenia (ITP), although the mechanism of effect remains unclear. This study describes the effects of rituximab on platelet-associated antibodies (PA-APAs), B and T cell counts and clonality ( IGHV and TRG@ gene rearrangements), FCGR3A (FcγRIIIa) and FCGR2A (FcγRIIa) polymorphisms and correlation to anti-CD40 ligand (CD40L) response. PA-APA levels fell more frequently in responders (6/8) than in non-responders (2/10: P = 0·08-0·15). Two responders had no PA-APAs. Two non-responders with a fall in PA-APAs had very high CD8 levels. One non-responder had a B cell clone, one responder and one non-responder had a T cell clone. 15/16 patients had the same responses to rituximab and antiCD40L. Patients with FCGR3A V/V polymorphisms were more likely to respond to rituximab (P = 0·03). In summary, the fall in PA-APAs in responders confirms the humoural effect of rituximab. Failure to respond in patients with very high CD8 levels, despite PA-APA fall indicates a role for T cell-mediated platelet/megakaryocyte destruction. Concordance of response to anti-CD40L suggests autoantibody-producing cells are under T cell control. Finally, the effect of FCGR polymorphisms on response confirms the importance of FCGR-mediated depletion of B cells in autoimmunity. This has implications on the pathology of ITP as well as the immunological effect of B cell depletion.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Autoantibodies/blood , Blood Platelets/immunology , Immunosuppressive Agents/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptors, IgG/genetics , Adult , Aged , Antigens, CD20/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Drug Therapy, Combination , Female , Genotype , Humans , Immunity, Cellular , Immunophenotyping , Killer Cells, Natural/immunology , Leukocyte Count , Male , Middle Aged , Neutrophils/immunology , Polymorphism, Genetic , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Purpura, Thrombocytopenic, Idiopathic/genetics , Rituximab , Thrombopoietin/blood , Treatment Outcome
15.
Transfusion ; 52(5): 1117-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22070736

ABSTRACT

BACKGROUND: Maternal immunization against low-frequency, platelet (PLT)-specific antigens is being recognized with increasing frequency as a cause of neonatal alloimmune thrombocytopenia (NAIT). STUDY DESIGN AND METHODS: Serologic and molecular studies were performed on PLTs and DNA from two families in which an infant was born with severe thrombocytopenia not attributable to maternal immunization against known PLT-specific alloantigens. RESULTS: Antibodies reactive only with paternal PLTs were identified in each mother using flow cytometry and solid-phase assays. Unique mutations encoding amino acid substitutions K164T in glycoprotein (GP)IIb (Case 1) and R622W in GPIIIa (Case 2) were identified in paternal DNA and in DNA from the affected infants. Each maternal antibody recognized recombinant GPIIb/IIIa mutated to contain the polymorphisms identified in the corresponding father. None of 100 unselected normal subjects possessed these paternal mutations. CONCLUSIONS: Severe NAIT observed in the affected infants was caused by maternal immunization against previously unrecognized, low-frequency antigens created by amino acid substitutions in GPIIb/IIIa (α(IIb) /ß(3) integrin). A search should be conducted for novel paternal antigens in cases of apparent NAIT not explained on the basis of maternal-fetal incompatibility for known human PLT antigens.


Subject(s)
Antigens, Human Platelet/immunology , Platelet Membrane Glycoproteins/genetics , Polymorphism, Genetic , Thrombocytopenia, Neonatal Alloimmune/etiology , Female , Humans , Infant, Newborn , Male , Mutation , Pregnancy
16.
Am J Hematol ; 87(8): 776-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22641378

ABSTRACT

Heparin induced thrombocytopenia (HIT) is a serious complication of heparin therapy. The PF4 ELISA is a serologic assay that provides laboratory support for the clinical diagnosis of HIT, but it is often positive in patients who do not have the syndrome. We examined whether the specificity of the PF4 ELISA can be improved by 1) taking antibody potency into consideration, 2) by measuring only IgG antibodies, and 3) by utilizing a high concentration heparin inhibition step. We reviewed clinical information on 116 patients whose samples were referred for HIT antibody testing and assigned each a clinical score related to the likelihood of the patient having HIT. The scores were then correlated with serologic findings. Patients with strongly positive PF4ELISA results (OD ≥ 1.0) using both versions of the assay (IgG/A/M and IgG only) had clinical scores and SRA activity that were significantly higher than those having reactive or negative results. When the IgG-only PF4 ELISA was used, only the strongly positive result group had significantly higher clinical scores and SRA release, and fewer samples were classified as weakly positive or reactive, suggesting that detection of IgG only in the PF4 ELISA improves the assay's specificity. The heparin inhibition step identified "reactive" samples that were associated with clinical scores and SRA release indistinguishable from the "negative" result groups, confirming that this step further improves specificity of the test. This study supports utilizing these 3 modifications of the PF4 ELISA to improve specificity in supporting the clinical diagnosis of HIT.


Subject(s)
Anticoagulants/adverse effects , Autoantibodies , Heparin/adverse effects , Immunoglobulin G , Platelet Factor 4/immunology , Thrombocytopenia , Adolescent , Adult , Aged , Aged, 80 and over , Anticoagulants/therapeutic use , Autoantibodies/blood , Autoantibodies/immunology , Child , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Female , Heparin/therapeutic use , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Male , Middle Aged , Sensitivity and Specificity , Thrombocytopenia/blood , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis , Thrombocytopenia/immunology
17.
N Engl J Med ; 356(9): 904-10, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17329697

ABSTRACT

BACKGROUND: Vancomycin has only rarely been implicated as a cause of thrombocytopenia, and there is only limited evidence that this complication is caused by immune mechanisms. We conducted a study to determine whether thrombocytopenia is caused by vancomycin-dependent antibodies in patients being treated with vancomycin. METHODS: We identified and characterized vancomycin-dependent, platelet-reactive antibodies in patients who had been referred for testing during a 5-year period because of a clinical suspicion of vancomycin-induced thrombocytopenia. We obtained clinical information about the patients from their referring physicians. RESULTS: Drug-dependent, platelet-reactive antibodies of the IgG class, the IgM class, or both were identified in 34 patients, and clinical follow-up information was obtained from 29 of these patients. The mean nadir platelet count in these patients was 13,600 per cubic millimeter, and severe bleeding occurred in 10 patients (34%). Platelet levels returned to baseline in all 26 surviving patients after vancomycin was stopped. In 15 patients, the drug was continued for 1 to 14 days while other possible causes of thrombocytopenia were investigated. Vancomycin-dependent antibodies were not found in 25 patients who had been given vancomycin and in whom thrombocytopenia did not develop. CONCLUSIONS: Severe bleeding can occur in patients with vancomycin-induced immune thrombocytopenia. The detection of vancomycin-dependent antiplatelet antibodies in patients receiving the antibiotic in whom thrombocytopenia develops, and the absence of antibodies in patients given the drug in whom platelet counts remain stable, indicate that these antibodies are the cause of the thrombocytopenia.


Subject(s)
Anti-Bacterial Agents/immunology , Blood Platelets/immunology , Immunoglobulin G/blood , Immunoglobulin M/blood , Thrombocytopenia/chemically induced , Vancomycin/immunology , Acute Disease , Adolescent , Adult , Aged , Anti-Bacterial Agents/adverse effects , Antibodies/blood , Female , Humans , Male , Middle Aged , Platelet Count , Reference Values , Thrombocytopenia/immunology , Vancomycin/adverse effects
19.
Transfusion ; 50(2): 324-33, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19821948

ABSTRACT

BACKGROUND: Recent reports suggest that maternal immunization against low-frequency, platelet (PLT)-specific glycoprotein (GP) polymorphisms is a more common cause of neonatal alloimmune thrombocytopenia (NATP) than previously thought. STUDY DESIGN AND METHODS: Serologic and molecular studies were performed on PLTs and DNA from three families in which an infant was born with apparent NATP not attributable to maternal immunization against known PLT-specific alloantigens. RESULTS: Antibodies reactive only with paternal PLTs were identified in each mother. In Cases 2 (Kno) and 3 (Nos), but not Case 1 (Sta), antibody recognized paternal GPIIb/IIIa in solid-phase assays. Unique mutations encoding amino acid substitutions in GPIIb (Case 2) or GPIIIa (Cases 1 and 3) were identified in paternal DNA and in DNA from two of the affected infants. Antibody from all three cases recognized recombinant GPIIIa (Case 1 [Sta] and Case 3 [Nos]) and GPIIb (Case 2, Kno) mutated to contain the polymorphisms identified in the respective fathers. None of 100 unselected normal subjects possessed the paternal mutations. Enzyme-linked immunosorbent assay and flow cytometric studies suggested that failure of maternal serum from Case 1 (Sta) to react with paternal GPIIIa in solid-phase assays resulted from use of a monoclonal antibody AP2, for antigen immobilization that competed with the maternal antibody for binding to the Sta epitope. CONCLUSION: NATP in the three cases was caused by maternal immunization against previously unreported, low-frequency GP polymorphisms. Maternal immunization against low-frequency PLT-specific alloantigens should be considered in cases of apparent NATP not resolved by conventional serologic and molecular testing.


Subject(s)
Antigens, Human Platelet/genetics , Integrin alpha2/genetics , Integrin beta3/genetics , Polymorphism, Genetic , Thrombocytopenia, Neonatal Alloimmune/genetics , Adult , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Antigens, Human Platelet/immunology , CHO Cells , Cricetinae , Cricetulus , Epitopes/genetics , Epitopes/immunology , Female , Gene Frequency , Humans , Immunoglobulin G/immunology , Infant, Newborn , Integrin alpha2/immunology , Integrin beta3/immunology , Isoantibodies/immunology , Male , Maternal-Fetal Exchange , Models, Molecular , Pregnancy , Recombinant Fusion Proteins/immunology , Thrombocytopenia, Neonatal Alloimmune/immunology
20.
Pediatr Blood Cancer ; 53(3): 459-61, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19459189

ABSTRACT

Neonatal alloimmune thrombocytopenia (NAIT) is one of the most frequent causes of both severe thrombocytopenia and intracranial hemorrhage (ICH) in fetuses and term neonates. The diagnosis is established by demonstrating antibodies against human platelet antigens (HPA) and discordance in platelet antigen typing between parents or between the mother and neonate. We report a case of NAIT that was likely due to maternal sensitization to HPA-9b (Max(a)), a recently recognized, rare platelet-specific antigen.


Subject(s)
Antigens, Human Platelet/immunology , Thrombocytopenia, Neonatal Alloimmune/etiology , Female , Humans , Infant, Newborn , Male , Maternal-Fetal Exchange/immunology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL