Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 140(9): 980-991, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35687757

RESUMO

Sutimlimab, a first-in-class humanized immunoglobulin G4 (IgG4) monoclonal antibody that selectively inhibits the classical complement pathway at C1s, rapidly halted hemolysis in the single-arm CARDINAL study in recently transfused patients with cold agglutinin disease (CAD). CADENZA was a 26-week randomized, placebo-controlled phase 3 study to assess safety and efficacy of sutimlimab in patients with CAD without recent (within 6 months prior to enrollment) transfusion history. Forty-two patients with screening hemoglobin ≤10 g/dL, elevated bilirubin, and ≥1 CAD symptom received sutimlimab (n = 22) or placebo (n = 20) on days 0 and 7 and then biweekly. Composite primary endpoint criteria (hemoglobin increase ≥1.5 g/dL at treatment assessment timepoint [mean of weeks 23, 25, 26], avoidance of transfusion, and study-prohibited CAD therapy [weeks 5-26]) were met by 16 patients (73%) on sutimlimab, and 3 patients (15%) on placebo (odds ratio, 15.9 [95% confidence interval, 2.9, 88.0; P < .001]). Sutimlimab, but not placebo, significantly increased mean hemoglobin and FACIT-Fatigue scores at treatment assessment timepoint. Sutimlimab normalized mean bilirubin by week 1. Improvements correlated with near-complete inhibition of the classical complement pathway (2.3% mean activity at week 1) and C4 normalization. Twenty-one (96%) sutimlimab patients and 20 (100%) placebo patients experienced ≥1 treatment-emergent adverse event. Headache, hypertension, rhinitis, Raynaud phenomenon, and acrocyanosis were more frequent with sutimlimab vs placebo, with a difference of ≥3 patients between groups. Three sutimlimab patients discontinued owing to adverse events; no placebo patients discontinued. These data demonstrate that sutimlimab has potential to be an important advancement in the treatment of CAD. This trial was registered at www.clinicaltrials.gov as #NCT03347422.


Assuntos
Anemia Hemolítica Autoimune , Anticorpos Monoclonais Humanizados , Anemia Hemolítica Autoimune/sangue , Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais Humanizados/uso terapêutico , Bilirrubina/sangue , Método Duplo-Cego , Hemoglobinas/análise , Humanos , Resultado do Tratamento
2.
Am J Hematol ; 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38884175

RESUMO

Crovalimab is a novel C5 complement inhibitor that enables rapid and sustained C5 inhibition with subcutaneous, low-volume self-administration every 4 weeks. COMMODORE 2 (NCT04434092) is a global, randomized, open-label, multicenter, phase 3 trial evaluating the non-inferiority of crovalimab versus eculizumab in patients with paroxysmal nocturnal hemoglobinuria not previously treated with C5 inhibition. C5 inhibitor-naive patients with lactate dehydrogenase (LDH) ≥2 × upper limit of normal (ULN) were randomized 2:1 to crovalimab or eculizumab. Co-primary efficacy endpoints were proportion of patients with hemolysis control (centrally assessed LDH ≤1.5 × ULN) and proportion with transfusion avoidance. Secondary efficacy endpoints were proportions of patients with breakthrough hemolysis, stabilized hemoglobin, and change in FACIT-Fatigue score. The primary treatment period was 24 weeks. Two hundred and four patients were randomized (135 crovalimab; 69 eculizumab). Crovalimab was non-inferior to eculizumab in the co-primary endpoints of hemolysis control (79.3% vs. 79.0%; odds ratio, 1.0 [95% CI, 0.6, 1.8]) and transfusion avoidance (65.7% vs. 68.1%; weighted difference, -2.8 [-15.7, 11.1]), and in the secondary efficacy endpoints of breakthrough hemolysis (10.4% vs. 14.5%; weighted difference, -3.9 [-14.8, 5.3]) and hemoglobin stabilization (63.4% vs. 60.9%; weighted difference, 2.2 [-11.4, 16.3]). A clinically meaningful improvement in FACIT-Fatigue score occurred in both arms. Complete terminal complement activity inhibition was generally maintained with crovalimab. The safety profiles of crovalimab and eculizumab were similar with no meningococcal infections. Most patients who switched from eculizumab to crovalimab after the primary treatment period preferred crovalimab. These data demonstrate the positive benefit-risk profile of crovalimab.

3.
Br J Haematol ; 203(3): 468-476, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37654088

RESUMO

To determine the prevalence and clinical relevance of glycosylphosphatidylinositol-anchored protein-deficient (GPI[-]) cell populations (paroxysmal nocturnal haemoglobinuria [PNH]-type cells) in patients with acquired aplastic anaemia (AA) or myelodysplastic syndrome (MDS), we prospectively studied peripheral blood samples of 2402 patients (1075 with AA, 900 with MDS, 144 with PNH, and 283 with other anaemia) using a high-sensitivity flow cytometry assay in a nationwide multi-centre observational study. PNH-type cells were detected in 52.6% of AA and 13.7% of MDS patients. None of the 35 patients with refractory anaemia (RA) with ringed sideroblasts or the 86 patients with RA with excess of blasts carried PNH-type cells. Among the 317 patients possessing PNH-type granulocytes, the percentage of PNH-type granulocytes increased by ≥10% in 47 patients (14.8%), remained unchanged in 240 patients (75.7%), and decreased by ≥10% in 30 patients (9.5%) during 3 years of follow-up. PNH-type granulocyte expansion occurred more frequently (27.1%) in the 144 patients who originally carried PNH-type granulocytes ≥1% than in the 173 patients with PNH-type granulocytes <1% (4.6%). This study confirmed that PNH-type cells are undetectable in authentic clonal MDS patients, and the presence of ≥1% PNH-type granulocytes predicts a higher likelihood of PNH-type cell expansion than with <1% PNH-type granulocytes.

4.
Eur J Haematol ; 111(2): 300-310, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37321625

RESUMO

OBJECTIVES: This study reports long-term outcomes from the open-label extension (OLE) period of the Phase I/II COMPOSER trial (NCT03157635) that evaluated crovalimab in patients with paroxysmal nocturnal haemoglobinuria, who were treatment-naive or switched from eculizumab at enrolment. METHODS: COMPOSER consists of four sequential parts followed by the OLE. The primary OLE objective was to assess long-term crovalimab safety, with a secondary objective to assess crovalimab pharmacokinetics and pharmacodynamics. Exploratory efficacy endpoints included change in lactate dehydrogenase (LDH), transfusion avoidance, haemoglobin stabilisation and breakthrough haemolysis (BTH). RESULTS: A total 43 of 44 patients entered the OLE after completing the primary treatment period. Overall, 14 of 44 (32%) experienced treatment-related adverse events. Steady state exposure levels of crovalimab and terminal complement inhibition were maintained over the OLE. During the OLE, mean normalised LDH was generally maintained at ≤1.5× upper limit of normal, transfusion avoidance was achieved in 83%-92% of patients and haemoglobin stabilisation was reached in 79%-88% of patients across each 24-week interval. Five BTH events occurred with none leading to withdrawal. CONCLUSIONS: Over a 3-year median treatment duration, crovalimab was well tolerated and sustained C5 inhibition was achieved. Intravascular haemolysis control, haemoglobin stabilisation and transfusion avoidance were maintained, signifying long-term crovalimab efficacy.


Assuntos
Hemoglobinúria Paroxística , Humanos , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Transfusão de Sangue , Hemoglobinas , Duração da Terapia , Hemólise , L-Lactato Desidrogenase
5.
Rinsho Ketsueki ; 64(6): 466-473, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37407469

RESUMO

The anti-C5 antibody eculizumab was approved in 2007 as the first anti-complement agent for the treatment of paroxysmal nocturnal hemoglobinuria (PNH). While eculizumab's indication has been expanded to include other diseases, the development of new anti-complement agents has been aggressively pursued for various diseases. In PNH, the anti-C5 recycling antibody ravulizumab, which is an improved version of eculizumab, has been developed, with an extended dosing interval of 2 to 8 weeks, vastly improving convenience. The treatment of PNH with terminal complement inhibitors such as eculizumab and ravulizumab presents a new challenge-extravascular hemolysis. To address this issue, the proximal complement inhibitor, a C3 inhibitor called pegcetacoplan, was approved in the United States of America. Furthermore, the amplification loop inhibitors-a factor B inhibitor iptacopan, and a factor D inhibitor danicopan-are being developed. Recently, the anti-C1s antibody sutimlimab was approved for the treatment of cold agglutinin disease, a type of autoimmune hemolytic anemia. This article discusses novel anti-complement therapies for hemolytic anemia.


Assuntos
Anemia Hemolítica Autoimune , Anemia Hemolítica , Hemoglobinúria Paroxística , Humanos , Proteínas do Sistema Complemento , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Inativadores do Complemento/uso terapêutico , Anemia Hemolítica Autoimune/tratamento farmacológico
6.
Rinsho Ketsueki ; 64(9): 884-891, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37793862

RESUMO

In recent years, it has become clear that various diseases are caused by complement (related molecule) abnormalities (complementopathies) or are exacerbated by complement (complement-related diseases), and novel therapeutic agents targeting complement (anti-complement agents) are now being developed. Paroxysmal nocturnal hemoglobinuria (PNH) is a hematopoietic stem cell disorder characterized by complement-mediated intravascular hemolysis due to a deficiency of complement regulatory factors, making it a perfect candidate for anti-complement agents. In 2007, the anti-C5 monoclonal antibody eculizumab was approved for PNH, as the first anti-complement agent. The indications for eculizumab are expanding, and aggressive development is underway for new anti-complement agents, not only for PNH but also a variety of other diseases. In addition, the anti-C1s antibody sutimlimab was approved last year for the treatment of cold agglutinin disease, a form of autoimmune hemolytic anemia. This presentation overviews novel anti-complement agents for these hemolytic anemias.


Assuntos
Anemia Hemolítica Autoimune , Anemia Hemolítica , Hemoglobinúria Paroxística , Humanos , Anemia Hemolítica/tratamento farmacológico , Proteínas do Sistema Complemento/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Anemia Hemolítica Autoimune/tratamento farmacológico , Anemia Hemolítica Autoimune/complicações
7.
Rinsho Ketsueki ; 64(1): 9-17, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-36775313

RESUMO

Ravulizumab is the first long-acting complement inhibitor approved for paroxysmal nocturnal hemoglobinuria (PNH) treatment. We evaluated patient preference for ravulizumab or eculizumab among Japanese adults with PNH. The ALXN1210-PNH-301 (NCT02946463) and ALXN1210-PNH-302 (NCT03056040) studies included 23 Japanese adults who are enrolled in complement inhibitor treatment-naive and eculizumab (≥6 months) treatment. Patient preference was assessed using the PNH-specific patient preference questionnaire (PNH-PPQ©). Most patients preferred ravulizumab (19/23, 82.6%), none preferred eculizumab, and four (17.4%) reported no preference (χ2 test, p<0.005). The preference for ravulizumab was driven by its lower infusion frequency (every 8 weeks) compared with eculizumab (every 2 weeks). The included Japanese patients with PNH preferred ravulizumab because of its reduced infusion frequency, which increases activity planning ability, treatment convenience, and overall quality of life, as compared with eculizumab. These data provide useful insight into patient perspectives and may aid decision-making for PNH treatment.


Assuntos
Hemoglobinúria Paroxística , Adulto , Humanos , Hemoglobinúria Paroxística/tratamento farmacológico , Preferência do Paciente , Qualidade de Vida , População do Leste Asiático , Inativadores do Complemento/uso terapêutico , Hemólise
8.
Blood ; 135(12): 912-920, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-31978221

RESUMO

Complement C5 inhibition is the standard of care (SoC) for patients with paroxysmal nocturnal hemoglobinuria (PNH) with significant clinical symptoms. Constant and complete suppression of the terminal complement pathway and the high serum concentration of C5 pose challenges to drug development that result in IV-only treatment options. Crovalimab, a sequential monoclonal antibody recycling technology antibody was engineered for extended self-administered subcutaneous dosing of small volumes in diseases amenable for C5 inhibition. A 3-part open-label adaptive phase 1/2 trial was conducted to assess safety, pharmacokinetics, pharmacodynamics, and exploratory efficacy in healthy volunteers (part 1), as well as in complement blockade-naive (part 2) and C5 inhibitor-treated (part 3) PNH patients. Twenty-nine patients were included in part 2 (n = 10) and part 3 (n = 19). Crovalimab concentrations exceeded the prespecified 100-µg/mL level and resulted in complete and sustained terminal complement pathway inhibition in treatment-naive and C5 inhibitor-pretreated PNH patients. Hemolytic activity and free C5 levels were suppressed below clinically relevant thresholds (liposome assay <10 U/mL and <50 ng/mL, respectively). Safety was consistent with the known profile of C5 inhibition. As expected, formation of drug-target-drug complexes was observed in all 19 patients switching to crovalimab, manifesting as transient mild or moderate vasculitic skin reactions in 2 of 19 participants. Both events resolved under continued treatment with crovalimab. Subcutaneous crovalimab (680 mg; 4 mL), administered once every 4 weeks, provides complete and sustained terminal complement pathway inhibition in patients with PNH, warranting further clinical development (ClinicalTrials.gov identifier, NCT03157635).


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais/farmacologia , Biomarcadores , Complemento C5/imunologia , Inativadores do Complemento/farmacologia , Monitoramento de Medicamentos , Feminino , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Resultado do Tratamento
9.
Eur J Haematol ; 109(3): 205-214, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35502600

RESUMO

OBJECTIVES: The complement component 5 (C5) inhibitor ravulizumab demonstrated non-inferiority to eculizumab following 26 weeks of treatment in complement inhibitor-naïve and complement inhibitor-experienced patients with paroxysmal nocturnal hemoglobinuria (PNH; studies 301 and 302, respectively). This study aims to describe the results of both studies from 27 weeks to 2 years. METHODS: Patients (N = 441) continued to receive ravulizumab throughout the extension period. Efficacy endpoints included lactate dehydrogenase (LDH) normalization, transfusion avoidance and fatigue score (FACIT-F). Safety analyses were also performed. RESULTS: From 27 weeks to 2 years, improvements in LDH levels were maintained in both study populations. Transfusion avoidance was maintained in 81.9% (study 301) and 85.6% (study 302) of patients, and FACIT-F scores remained stable. Ravulizumab was well tolerated, and the incidence of adverse events (AEs) were similar between patients of both studies. Incidence of serious AEs deemed related to ravulizumab treatment was low (<3%). CONCLUSIONS: This study reports, to date, the longest period of follow-up in over 400 patients with PNH treated with ravulizumab (662 patient-years). Long-term, ravulizumab demonstrated durable efficacy and was well tolerated, highlighting the importance of C5 inhibitors as the mainstay of PNH treatment.


Assuntos
Hemoglobinúria Paroxística , Anticorpos Monoclonais Humanizados , Complemento C5 , Inativadores do Complemento/efeitos adversos , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Humanos
10.
Ann Hematol ; 100(8): 1975-1982, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33095337

RESUMO

Small populations of glycosylphosphatidylinositol-anchored protein-deficient (GPI[-]) cells accounting for up to 0.01% of total granulocytes can be accurately detected by a high-sensitivity flow cytometry (FCM) assay established by the Clinical and Laboratory Standards Institute (CLSI method) and have a prognostic value in bone marrow failure (BMF); however, the significance of GPI(-) granulocytes accounting for 0.001-0.009% of granulocytes remains unclear. To clarify this issue, we examined the peripheral blood of 21 BMF patients in whom minor (around 0.01%) populations of GPI(-) granulocytes had been previously detected by a different high-resolution FCM method (OPTIMA method, which defines ≥ 0.003% GPI(-) granulocytes as an abnormal increase) using both the CLSI and OPTIMA methods simultaneously. These two methods detected an "abnormal increase" in GPI(-) granulocytes in 10 patients (48%) and 17 patients (81%), respectively. CLSI detected 0.002-0.005% (median, 0.004%) GPI(-) granulocytes in 7 patients who were deemed positive for PNH-type cells according to the OPTIMA method, which detected 0.003-0.012% (median 0.006%) GPI(-) granulocytes. The clone sizes of GPI(-) cells detected by each assay were positively correlated (r = 0.994, p < 0.001). Of the seven patients who were judged positive for PNH-type cells by OPTIMA alone, five received immunosuppressive therapy, and all of them achieved a partial or complete response. GPI(-) granulocytes detected in BMF patients by the CLSI method should thus be considered significant, even at percentages of < 0.01%.


Assuntos
Transtornos da Insuficiência da Medula Óssea/patologia , Proteínas Ligadas por GPI/análise , Granulócitos/patologia , Hemoglobinúria Paroxística/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Transtornos da Insuficiência da Medula Óssea/diagnóstico , Serviços de Laboratório Clínico , Feminino , Hemoglobinúria Paroxística/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Rinsho Ketsueki ; 62(5): 378-387, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34108318

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a hematopoietic stem cell disease whose main symptom is complement-mediated intravascular hemolysis as a result of the clonal expansion of hematopoietic stem cells having mutations in genes involved in glycosylphosphatidylinositol (GPI) anchor synthesis including PIGA. With the advent of a humanized anti-C5 monoclonal antibody (eculizumab), the inhibitory effect on hemolysis, improvement in its various complicating symptoms, and preventive effect on thrombus formation were observed. In addition, the QOL in patients with PNH was significantly improved. Subsequently, the technology of recycling antibodies (ravulizumab and crovalimab) significantly extended the treatment interval and improved convenience, although the poor improvement of anemia due to extravascular hemolysis has been a major issue in some patients. Several clinical trials using proximal complement inhibitors (C3, factor D, factor B) are being conducted to overcome this critical task. Not only efficacy but also safety and convenience will be evaluated, and the best therapeutic agent will be selected in the near future.


Assuntos
Hemoglobinúria Paroxística , Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Humanos , Qualidade de Vida
12.
Rinsho Ketsueki ; 62(5): 463-471, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34108330

RESUMO

Following the reports of paroxysmal nocturnal hemoglobinuria (PNH) due to PIGT and PIGB gene mutations, the definition of PNH was changed to a hematopoietic stem cell disease with complement-mediated intravascular hemolysis as a result of clonal expansion of hematopoietic stem cells with mutations in genes involved in glycosylphosphatidylinositol (GPI) anchor synthesis, including PIGA. Treatment with eculizumab (Soliris®), a humanized anti-C5 monoclonal antibody, significantly improved the quality of life in patients with PNH, remarkably reduced hemolysis, improved symptoms associated with hemolysis, and prevented thrombosis. Although the administration interval has been extended and convenience has been greatly improved using the technique of recycling antibodies (ravulizumab and crovalimab), extravascular hemolysis has become another issue. Although attempts have been made to overcome this issue with proximal complement (C3, factor D, and factor B) inhibitors, the optimal therapeutic agent is expected to be selected after evaluating for not only efficacy and safety but also convenience.


Assuntos
Hemoglobinúria Paroxística , Glicosilfosfatidilinositóis , Células-Tronco Hematopoéticas , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Humanos , Manosiltransferases , Qualidade de Vida
13.
Eur J Haematol ; 105(5): 659-661, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32716556

RESUMO

Donor cell-derived hematological disorder (DCHD) is a rare complication of allogeneic hematopoietic stem cell transplantation (HSCT). The number of reports of DCHD has been increasing in the last decade, which likely reflects the growing number of HSCTs and the improved ability to identify the donor cell origin. Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hematological disorder arising in the context of clonal expansion of hematopoietic stem cells harboring a somatic mutation in phosphatidylinositol glycan anchor biosynthesis, class A. We report here a patient with adult T-cell leukemia/lymphoma, who developed PNH 7 years after umbilical cord blood transplantation. The patient has maintained complete remission with full-donor chimerism after HSCT. Thus, PNH was derived from stem cells of donor origin. The immature immune environment in the recipient after cord blood transplantation might have contributed to the rapid clonal expansion for neonatal stem cells in cord blood to develop typical symptomatic PNH in a short period. To the best of our knowledge, this is the first report in the literature of a case of PNH that developed in donor stem cells after HSCT.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/efeitos adversos , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/etiologia , Doadores de Tecidos , Biomarcadores , Evolução Clonal , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Humanos , Proteínas de Membrana/genética , Mutação , Transplante Homólogo
14.
Rinsho Ketsueki ; 61(8): 929-936, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32908057

RESUMO

Treatment with eculizumab (Soliris®), a humanized anti-C5 monoclonal antibody improves the quality of life of patients with paroxysmal nocturnal hemoglobinuria (PNH), remarkably reduces hemolysis, improves symptoms associated with hemolysis, and prevents thrombosis. Because eculizumab therapy is not a curative treatment, it is necessary to continue infusion every two weeks, which has been an issue from the viewpoint of convenience. In recent years, an improved version of eculizumab, ravulizumab (Ultomiris®), which relies on the technology of recycling antibodies has been developed and can be administered every 8 weeks. Crovalimab (SKY59), which can be administered subcutaneously every four weeks, is also under development, and therefore, the convenience for patients with PNH is improving. However, many issues still persist, and several new anti-complement drugs are currently under development. Hopefully, a better drug will be developed by thorough examination of what drug is best for the patient by considering not only its efficacy and safety but also its convenience.


Assuntos
Anticorpos/uso terapêutico , Hemoglobinúria Paroxística , Proteínas do Sistema Complemento , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Humanos , Qualidade de Vida , Radioimunoterapia
16.
Ann Hematol ; 97(12): 2289-2297, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30039297

RESUMO

Minor populations of glycosylphosphatidylinositol-anchored protein-deficient (GPI[-]) cells in the peripheral blood may have a prognostic value in bone marrow failure (BMF). Our objective is to establish the optimal flow cytometry (FCM) assay that can discriminate GPI(-) populations specific to BMF from those of healthy individuals. To identify a cut-off that discriminates GPI(-) rare cells from GPI(+) cells, we determined a position of the borderline that separates the GPI(-) from GPI(+) cells on a scattergram by testing more than 30 healthy individuals, such that no GPI(-) dot fell into the upper left quadrant where fluorescein-labeled aerolysin (FLAER)-CD11b+ granulocytes and CD55-CD59- glycophorin A+ erythrocytes were positioned. This method allowed us to define ≥ 0.003% CD11b+FLAER- granulocytes and ≥ 0.005% glycophorin A+CD55-CD59- erythrocytes to be specific to BMF patients. Longitudinal cross-validation studies showed minimal (< 0.02%) inter-laboratory differences in the GPI(-) cell percentage. An analysis of 1210 patients with BMF revealed a GPI(-) cell population in 56.3% of patients with aplastic anemia and 18.5% of patients with myelodysplastic syndrome. The GPI(-) granulocyte percentages was 0.003-0.01% in 3.7% of patients. This FCM assay effectively identified an increase in the percentage of GPI(-) rare cells that are specific to BMF patients and allowed different laboratories to accurately detect 0.003-0.01% of pathological GPI(-) cells.


Assuntos
Anemia Aplástica , Antígenos CD/sangue , Doenças da Medula Óssea , Eritrócitos , Citometria de Fluxo/métodos , Granulócitos , Hemoglobinúria Paroxística , Anemia Aplástica/sangue , Anemia Aplástica/patologia , Doenças da Medula Óssea/sangue , Doenças da Medula Óssea/patologia , Transtornos da Insuficiência da Medula Óssea , Eritrócitos/metabolismo , Eritrócitos/patologia , Feminino , Granulócitos/metabolismo , Granulócitos/patologia , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/patologia , Humanos , Masculino
17.
Eur J Haematol ; 101(1): 3-11, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29532535

RESUMO

OBJECTIVE: Paroxysmal nocturnal hemoglobinuria (PNH) is a severe, life-threatening disorder for which early diagnosis is essential. However, given the rarity of the disease and non-specificity of symptoms, correct diagnosis may be delayed or missed. While various hematologic guidelines note common signs and symptoms associated with PNH, international expert consensus based on real-world clinical experience and an actionable algorithm for non-specialists to facilitate screening and diagnosis are lacking. The objective of the study is to develop a clinically relevant, consensus-driven screening and diagnostic algorithm on PNH for non-specialist clinicians. METHODS: An expert advisory committee of PNH experts from North America, Europe, and Japan was convened, and a modified Delphi methodology was employed to develop an algorithm to assist non-specialist clinicians in identifying signs/symptoms of PNH and conducting appropriate differential diagnosis. Twelve globally representative Delphi panelists with clinical expertise in PNH were identified and recruited. Panelists provided their differential diagnosis for 5 blinded case studies via 2 rounds of online questionnaires. Responses mentioned by >50% of panelists in the first round were included in the second-round questionnaire, at which point consensus was attained if >80% of panelists agreed on an approach. RESULTS: Consensus was reached for 95% of screening and diagnostic decision points and 90% of tests required at decision points. CONCLUSION: These results facilitated development of a consensus-based, clinically relevant algorithm, providing non-specialist clinicians with actionable guidance on PNH screening and diagnosis.


Assuntos
Algoritmos , Anemia Hemolítica/diagnóstico , Consenso , Hemoglobinúria Paroxística/diagnóstico , Adulto , Anemia Hemolítica/fisiopatologia , Diagnóstico Diferencial , Diagnóstico Precoce , Prova Pericial , Feminino , Hemoglobinúria Paroxística/fisiopatologia , Humanos , Masculino , Guias de Prática Clínica como Assunto , Inquéritos e Questionários
18.
Rinsho Ketsueki ; 59(8): 1042-1047, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-30185704

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) arises as a consequence of clonal expansion of hematopoietic stem cells that have acquired a somatic mutation in the PIGA gene. The resulting hematopoietic cells have deficiencies in the GPI-anchored complement regulatory proteins CD55 and CD59, which account for the intravascular hemolysis that is the primary clinical manifestation of PNH. Thromboembolism is a major cause of morbidity and mortality in PNH, particularly in Caucasian patients. In a previous report on the clinical course of PNH patients in the United States and Japan, we showed that thrombosis was significantly more prevalent in white PNH patients than in Asian PNH patients. The pathophysiological mechanisms underlying thrombosis in PNH have not been fully clarified, and multiple factors are likely to be involved. Eculizumab, a humanized monoclonal antibody, targets the terminal complement protein C5 and inhibits terminal complement-mediated hemolysis associated with PNH. Brodsky et al. reported that eculizumab treatment reduces the risk of clinical thromboembolism in patients with PNH. These facts strongly suggest that the main cause of thrombosis in PNH is complement activation and/or hemolysis. In this review, the pathophysiology of thrombosis in PNH is discussed in the context of observations in PNH patients treated with eculizumab.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Hemoglobinúria Paroxística/complicações , Trombose/etiologia , Hemoglobinúria Paroxística/terapia , Humanos , Japão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA