Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 63
Filtrer
1.
Clinicoecon Outcomes Res ; 16: 225-232, 2024.
Article de Anglais | MEDLINE | ID: mdl-38623087

RÉSUMÉ

Background: Paroxysmal nocturnal hemoglobinuria is a rare, acquired disease characterized by hemolytic episodes and associated with significant clinical burden. The introduction of C5 inhibitory monoclonal antibodies (C5i) represented a major breakthrough in PNH treatment, effectively reducing intravascular hemolysis (IVH) but showing limited impact on extravascular hemolysis (EVH). In 2021, the C3 inhibitor pegcetacoplan was approved by EMA and recently reimbursed in Italy, which also has the advantages in the reduction of both IVH and EVH, increasing hemoglobin values and simultaneously improving the quality of life and fatigue of patients. A cost-utility analysis was developed to compare pegcetacoplan to C5i (eculizumab and ravulizumab) in the PNH population who remain anemic after treatment with C5i for at least 3 months. Materials and Methods: The analysis employed a Markov model with a 5-year time horizon whereby patients can transition among 3 PNH health states, adopting the perspective of the Italian NHS. Efficacy data were sourced from the PEGASUS study, with drug prices reflecting ex-factory costs. Additionally, costs associated with resource utilization, adverse events, and complications were estimated based on outpatient and hospital care rates, excluding indirect expenses. Utility and disutility values related to transfusions were also considered, with pegcetacoplan allowing for dose escalation. Results: The cumulative cost of treatment per individual patient at 5 years was estimated to be €1,483,454 for pegcetacoplan, €1,585,763 for eculizumab, and €1,574,826 for ravulizumab. Pegcetacoplan demonstrated a superior increase in quality-adjusted life years (QALYs) compared to both eculizumab (0.51 increase) and ravulizumab (0.27 increase). Furthermore, pegcetacoplan showed a reduction in complication management costs (€22,891 less compared to eculizumab and €22,611 less compared to ravulizumab) and lower transfusion-related expenses (€14,147 less than both C5i treatments). Conclusion: Pegcetacoplan emerged as the dominant strategy in this analysis, being more effective, less expensive and improves quality of life in the analyzed population affected by PNH.

2.
N Engl J Med ; 390(11): 994-1008, 2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38477987

RÉSUMÉ

BACKGROUND: Persistent hemolytic anemia and a lack of oral treatments are challenges for patients with paroxysmal nocturnal hemoglobinuria who have received anti-C5 therapy or have not received complement inhibitors. Iptacopan, a first-in-class oral factor B inhibitor, has been shown to improve hemoglobin levels in these patients. METHODS: In two phase 3 trials, we assessed iptacopan monotherapy over a 24-week period in patients with hemoglobin levels of less than 10 g per deciliter. In the first, anti-C5-treated patients were randomly assigned to switch to iptacopan or to continue anti-C5 therapy. In the second, single-group trial, patients who had not received complement inhibitors and who had lactate dehydrogenase (LDH) levels more than 1.5 times the upper limit of the normal range received iptacopan. The two primary end points in the first trial were an increase in the hemoglobin level of at least 2 g per deciliter from baseline and a hemoglobin level of at least 12 g per deciliter, each without red-cell transfusion; the primary end point for the second trial was an increase in hemoglobin level of at least 2 g per deciliter from baseline without red-cell transfusion. RESULTS: In the first trial, 51 of the 60 patients who received iptacopan had an increase in the hemoglobin level of at least 2 g per deciliter from baseline, and 42 had a hemoglobin level of at least 12 g per deciliter, each without transfusion; none of the 35 anti-C5-treated patients attained the end-point levels. In the second trial, 31 of 33 patients had an increase in the hemoglobin level of at least 2 g per deciliter from baseline without red-cell transfusion. In the first trial, 59 of the 62 patients who received iptacopan and 14 of the 35 anti-C5-treated patients did not require or receive transfusion; in the second trial, no patients required or received transfusion. Treatment with iptacopan increased hemoglobin levels, reduced fatigue, reduced reticulocyte and bilirubin levels, and resulted in mean LDH levels that were less than 1.5 times the upper limit of the normal range. Headache was the most frequent adverse event with iptacopan. CONCLUSIONS: Iptacopan treatment improved hematologic and clinical outcomes in anti-C5-treated patients with persistent anemia - in whom iptacopan showed superiority to anti-C5 therapy - and in patients who had not received complement inhibitors. (Funded by Novartis; APPLY-PNH ClinicalTrials.gov number, NCT04558918; APPOINT-PNH ClinicalTrials.gov number, NCT04820530.).


Sujet(s)
Anémie hémolytique , Facteur B du complément , Inhibiteurs du complément , Hémoglobines , Hémoglobinurie paroxystique , Humains , Administration par voie orale , Anémie hémolytique/complications , Complément C5/antagonistes et inhibiteurs , Facteur B du complément/antagonistes et inhibiteurs , Inhibiteurs du complément/administration et posologie , Inhibiteurs du complément/effets indésirables , Inhibiteurs du complément/usage thérapeutique , Transfusion d'érythrocytes , Céphalée/induit chimiquement , Hémoglobines/analyse , Hémoglobinurie paroxystique/traitement médicamenteux , Hémoglobinurie paroxystique/étiologie , Essais cliniques de phase III comme sujet , Essais contrôlés randomisés comme sujet
3.
Am J Hematol ; 99(3): 505-507, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38264841

RÉSUMÉ

C3 binding on PNH red blood cells after in vitro complement activation in normal and terminal complement depleted (∆5, ∆6, ∆9) sera: C3 binding, and the subsequent extravascular hemolysis, happens anytime there is a block of a component of the terminal complement pathway.


Sujet(s)
Hémoglobinurie paroxystique , Humains , Complément C3 , Érythrocytes/métabolisme , Activation du complément , Hémolyse , Protéines du système du complément/physiologie
5.
Clin Cancer Res ; 29(24): 5217-5226, 2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-37888299

RÉSUMÉ

PURPOSE: In estrogen receptor-positive (ER+) breast cancer, single-nucleotide polymorphisms (SNP) in the aromatase gene might affect aromatase inhibitors (AI) metabolism and efficacy. Here, we assessed the impact of SNP on prognosis and toxicity of patients receiving adjuvant letrozole. EXPERIMENTAL DESIGN: We enrolled 886 postmenopausal patients in the study. They were treated with letrozole for 2 to 5 years after taking tamoxifen for 2 to 6 years, continuing until they completed 5 to 10 years of therapy. Germline DNA was genotyped for SNP rs4646, rs10046, rs749292, and rs727479. Log-rank test and Cox model were used for disease-free survival (DFS) and overall survival (OS). Cumulative incidence (CI) of breast cancer metastasis was assessed through competing risk analysis, with contralateral breast cancer, second malignancies and non-breast cancer death as competing events. CI of skeletal and cardiovascular events were assessed using DFS events as competing events. Subdistribution HR (sHR) with 95% confidence intervals were calculated through Fine-Gray method. RESULTS: No SNP was associated with DFS. Variants rs10046 [sHR 2.03, (1.04-2.94)], rs749292 [sHR 2.11, (1.12-3.94)], and rs727479 [sHR 2.62, (1.17-5.83)] were associated with breast cancer metastasis. Three groups were identified on the basis of the number of these variants (0, 1, >1). Variant-based groups were associated with breast cancer metastasis (10-year CI 2.5%, 7.6%, 10.7%, P = 0.035) and OS (10-year estimates 96.5%, 93.0%, 89.6%, P = 0.030). Co-occurrence of rs10046 and rs749292 was negatively associated with 10-year CI of skeletal events (3.2% vs. 10%, P = 0.033). A similar association emerged between rs727479 and cardiovascular events (0.3% vs. 2.1%, P = 0.026). CONCLUSIONS: SNP of aromatase gene predict risk of metastasis and AI-related toxicity in ER+ early breast cancer, opening an opportunity for better treatment individualization.


Sujet(s)
Inhibiteurs de l'aromatase , Tumeurs du sein , Femelle , Humains , Aromatase/génétique , Inhibiteurs de l'aromatase/effets indésirables , Inhibiteurs de l'aromatase/toxicité , Marqueurs biologiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Maladies cardiovasculaires/induit chimiquement , Maladies cardiovasculaires/génétique , Traitement médicamenteux adjuvant , Létrozole/effets indésirables , Polymorphisme de nucléotide simple , Tamoxifène/usage thérapeutique
6.
Sci Rep ; 13(1): 5267, 2023 03 31.
Article de Anglais | MEDLINE | ID: mdl-37002241

RÉSUMÉ

ETV4, one of ETS proteins overexpressed in prostate cancer, promotes migration, invasion, and proliferation in prostate cells. This study identifies a series of previously unknown ETV4 alternatively spliced transcripts in human prostate cell lines. Their expression has been validated using several unbiased techniques, including Nanopore sequencing. Most of these transcripts originate from an in-frame exon skipping and, thus, are expected to be translated into ETV4 protein isoforms. Functional analysis of the most abundant among these isoforms shows that they still bear an activity, namely a reduced ability to promote proliferation and a residual ability to regulate the transcription of ETV4 target genes. Alternatively spliced genes are common in cancer cells: an analysis of the TCGA dataset confirms the abundance of these novel ETV4 transcripts in prostate tumors, in contrast to peritumoral tissues. Since none of their translated isoforms have acquired a higher oncogenic potential, such abundance is likely to reflect the tumor deranged splicing machinery. However, it is also possible that their interaction with the canonical variants may contribute to the biology and the clinics of prostate cancer. Further investigations are needed to elucidate the biological role of these ETV4 transcripts and of their putative isoforms.


Sujet(s)
Tumeurs de la prostate , Protéines proto-oncogènes c-ets , Humains , Mâle , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate/anatomopathologie , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-ets/génétique , Protéines proto-oncogènes c-ets/métabolisme
8.
Blood Rev ; 58: 101013, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36117056

RÉSUMÉ

This article presents the results of group discussion among an ad hoc constituted panel of experts aimed at identifying and addressing unmet clinical needs (UCNs) in the management of infectious risk associated with eculizumab or new terminal complement inhibitors (CIs) in paroxysmal nocturnal hemoglobinuria (PNH). With the Delphi technique, the most clinically relevant UCNs in PNH patients candidate to or on terminal CI were selected. They resulted to be: optimizing the infection prevention measures; developing non pharmacological infectious risk-mitigation strategies; improving the management of disease exacerbation during infectious complications. For each of these issues consensus opinions were provided and, when appropriate, proposals for advancement in clinical practice were addressed. The hope is that this comprehensive overview will serve to improve the practice of CIs therapy and inform the design and implementation of new studies in the field.


Sujet(s)
Hémoglobinurie paroxystique , Humains , Hémoglobinurie paroxystique/complications , Hémoglobinurie paroxystique/traitement médicamenteux , Inhibiteurs du complément/pharmacologie , Inhibiteurs du complément/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Consensus
11.
Blood ; 138(20): 1928-1938, 2021 11 18.
Article de Anglais | MEDLINE | ID: mdl-34314483

RÉSUMÉ

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by uncontrolled terminal complement activation and subsequent intravascular hemolysis (IVH). C5 inhibitors prevent membrane attack complex formation, but patients may experience extravascular hemolysis (EVH) and continue to require blood transfusions. Danicopan, an oral proximal complement inhibitor of alternative pathway factor D (FD), is designed to control IVH and EVH. In a phase 2 dose-finding trial, eculizumab-treated transfusion-dependent patients with PNH (n = 12) received danicopan, 100 to 200 mg thrice daily, in addition to their eculizumab regimen for 24 weeks. End points included hemoglobin (Hgb) change vs baseline at week 24 (primary), reduction in blood transfusions, and patient-reported outcomes. Safety, tolerability, and pharmacokinetics/pharmacodynamics were measured. Twelve patients received ≥1 danicopan dose; 1 patients discontinued from a serious adverse event deemed unlikely related to danicopan. Eleven patients completed the 24-week treatment period. Addition of danicopan resulted in a mean Hgb increase of 2.4 g/dL at week 24. In the 24 weeks prior to danicopan, 10 patients received 31 transfusions (50 units) compared with 1 transfusion (2 units) in 1 patient during the 24-week treatment period. Mean Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue score increased by 11 points from baseline to week 24. The most common adverse events were headache, cough, and nasopharyngitis. Addition of danicopan, a first-in-class FD inhibitor, led to a meaningful improvement in Hgb and reduced transfusion requirements in patients with PNH who were transfusion-dependent on eculizumab. These benefits were associated with improvement of FACIT-Fatigue. This trial was registered at www.clinicaltrials.gov as #NCT03472885.


Sujet(s)
Aminopyridines/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Inhibiteurs du complément/usage thérapeutique , Hémoglobinurie paroxystique/traitement médicamenteux , Indazoles/usage thérapeutique , Proline/usage thérapeutique , Pyrimidines/usage thérapeutique , Adulte , Sujet âgé , Aminopyridines/effets indésirables , Anticorps monoclonaux humanisés/effets indésirables , Inhibiteurs du complément/effets indésirables , Femelle , Humains , Indazoles/effets indésirables , Mâle , Adulte d'âge moyen , Proline/effets indésirables , Pyrimidines/effets indésirables , Résultat thérapeutique , Jeune adulte
14.
Haematologica ; 106(12): 3188-3197, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-33121236

RÉSUMÉ

Paroxysmal nocturnal hemoglobinuria (PNH) is characterised by complement-mediated intravascular hemolysis (IVH) due to absence of complement regulators CD55 and CD59 on affected erythrocytes. Danicopan is a first-in-class oral proximal, complement alternative pathway factor D (FD) inhibitor. Therapeutic FD inhibition was designed to control IVH and prevent C3-mediated extravascular hemolysis (EVH). In this open-label, phase 2, dose-finding trial, 10 untreated hemolytic PNH patients received danicopan monotherapy (100-200 mg thrice daily). Endpoints included change in lactate dehydrogenase (LDH) at day 28 (primary) and day 84 and hemoglobin. Safety, pharmacokinetics/pharmacodynamics, and patient-reported outcomes were measured. Ten patients reached the primary endpoint; two later discontinued: one for a serious adverse event (elevated aspartate aminotransferase/alanine aminotransferase coincident with breakthrough hemolysis, resolving without sequelae) and one for personal reasons unrelated to safety. Eight patients completed treatment. IVH was inhibited, demonstrated by mean decreased LDH (5.7 times upper limit of normal [ULN] at baseline vs 1.8 times ULN [day 28] and 2.2 times ULN [day 84]; both p.


Sujet(s)
Facteur D du complément , Hémoglobinurie paroxystique , Anticorps monoclonaux humanisés , Complément C3 , Inhibiteurs du complément , Protéines du système du complément , Érythrocytes , Hémoglobinurie paroxystique/traitement médicamenteux , Hémolyse , Humains
16.
J Hematol Oncol ; 13(1): 112, 2020 08 13.
Article de Anglais | MEDLINE | ID: mdl-32791988

RÉSUMÉ

BACKGROUND: ETV4 is one of the ETS proteins overexpressed in prostate cancer (PC) as a result of recurrent chromosomal translocations. In human prostate cell lines, ETV4 promotes migration, invasion, and proliferation; however, its role in PC has been unclear. In this study, we have explored the effects of ETV4 expression in the prostate in a novel transgenic mouse model. METHODS: We have created a mouse model with prostate-specific expression of ETV4 (ETV4 mice). By histochemical and molecular analysis, we have investigated in these engineered mice the expression of p21, p27, and p53. The implications of our in vivo findings have been further investigated in human cells lines by chromatin-immunoprecipitation (ChIP) and luciferase assays. RESULTS: ETV4 mice, from two independent transgenic lines, have increased cell proliferation in their prostate and two-thirds of them, by the age of 10 months, developed mouse prostatic intraepithelial neoplasia (mPIN). In these mice, cdkn1a and its p21 protein product were reduced compared to controls; p27 protein was also reduced. By ChIP assay in human prostate cell lines, we show that ETV4 binds to a specific site (-704/-696 bp upstream of the transcription start) in the CDKN1A promoter that was proven, by luciferase assay, to be functionally competent. ETV4 further controls CDKN1A expression by downregulating p53 protein: this reduction of p53 was confirmed in vivo in ETV4 mice. CONCLUSIONS: ETV4 overexpression results in the development of mPIN but not in progression to cancer. ETV4 increases prostate cell proliferation through multiple mechanisms, including downregulation of CDKN1A and its p21 protein product: this in turn is mediated through direct binding of ETV4 to the CDKN1A promoter and through the ETV4-mediated decrease of p53. This multi-faceted role of ETV4 in prostate cancer makes it a potential target for novel therapeutic approaches that could be explored in this ETV4 transgenic model.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Régulation de l'expression des gènes tumoraux , Protéines tumorales/physiologie , Protéines de fusion oncogènes/physiologie , Tumeur intraépithéliale prostate/génétique , Tumeurs de la prostate/génétique , Protéine de liaison aux androgènes/génétique , Animaux , Division cellulaire , Lignée cellulaire tumorale , Inhibiteur p21 de kinase cycline-dépendante/biosynthèse , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p27 de kinase cycline-dépendante/biosynthèse , Inhibiteur p27 de kinase cycline-dépendante/génétique , Régulation négative , Cellules HEK293 , Humains , Mâle , Matrix metalloproteinases/biosynthèse , Matrix metalloproteinases/génétique , Souris , Souris transgéniques , Régions promotrices (génétique)/génétique , Prostate/métabolisme , Tumeur intraépithéliale prostate/métabolisme , Tumeur intraépithéliale prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Liaison aux protéines , Rats , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéine p53 suppresseur de tumeur/physiologie
17.
Blood ; 136(11): 1225-1240, 2020 09 10.
Article de Anglais | MEDLINE | ID: mdl-32702756

RÉSUMÉ

Glucose 6-phosphate dehydrogenase (G6PD) deficiency is 1 of the commonest human enzymopathies, caused by inherited mutations of the X-linked gene G6PD. G6PD deficiency makes red cells highly vulnerable to oxidative damage, and therefore susceptible to hemolysis. Over 200 G6PD mutations are known: approximately one-half are polymorphic and therefore common in various populations. Some 500 million persons with any of these mutations are mostly asymptomatic throughout their lifetime; however, any of them may develop acute and sometimes very severe hemolytic anemia when triggered by ingestion of fava beans, by any of a number of drugs (for example, primaquine, rasburicase), or, more rarely, by infection. Approximately one-half of the G6PD mutations are instead sporadic: rare patients with these mutations present with chronic nonspherocytic hemolytic anemia. Almost all G6PD mutations are missense mutations, causing amino acid replacements that entail deficiency of G6PD enzyme activity: they compromise the stability of the protein, the catalytic activity is decreased, or a combination of both mechanisms occurs. Thus, genotype-phenotype correlations have been reasonably well clarified in many cases. G6PD deficiency correlates remarkably, in its geographic distribution, with past/present malaria endemicity: indeed, it is a unique example of an X-linked human polymorphism balanced through protection of heterozygotes from malaria mortality. Acute hemolytic anemia can be managed effectively provided it is promptly diagnosed. Reliable diagnostic procedures are available, with point-of-care tests becoming increasingly important where primaquine and its recently introduced analog tafenoquine are required for the elimination of malaria.


Sujet(s)
Déficit en glucose-6-phosphate-déshydrogénase , Infections à VIH , Donneurs de sang , Sécurité transfusionnelle , Glucose 6-phosphate dehydrogenase/génétique , Déficit en glucose-6-phosphate-déshydrogénase/épidémiologie , Déficit en glucose-6-phosphate-déshydrogénase/génétique , Humains , Primaquine/usage thérapeutique
18.
Front Immunol ; 10: 1157, 2019.
Article de Anglais | MEDLINE | ID: mdl-31258525

RÉSUMÉ

The treatment of paroxysmal nocturnal hemoglobinuria has been revolutionized by the introduction of the anti-C5 agent eculizumab; however, eculizumab is not the cure for Paroxysmal nocturnal hemoglobinuria (PNH), and room for improvement remains. Indeed, the hematological benefit during eculizumab treatment for PNH is very heterogeneous among patients, and different response categories can be identified. Complete normalization of hemoglobin (complete and major hematological response), is seen in no more than one third of patients, while the remaining continue to experience some degree of anemia (good and partial hematological responses), in some cases requiring regular red blood cell transfusions (minor hematological response). Different factors contribute to residual anemia during eculizumab treatment: underlying bone marrow dysfunction, residual intravascular hemolysis and the emergence of C3-mediated extravascular hemolysis. These two latter pathogenic mechanisms are the target of novel strategies of anti-complement treatments, which can be split into terminal and proximal complement inhibitors. Many novel terminal complement inhibitors are now in clinical development: they all target C5 (as eculizumab), potentially paralleling the efficacy and safety profile of eculizumab. Possible advantages over eculizumab are long-lasting activity and subcutaneous self-administration. However, novel anti-C5 agents do not improve hematological response to eculizumab, even if some seem associated with a lower risk of breakthrough hemolysis caused by pharmacokinetic reasons (it remains unclear whether more effective inhibition of C5 is possible and clinically beneficial). Indeed, proximal inhibitors are designed to interfere with early phases of complement activation, eventually preventing C3-mediated extravascular hemolysis in addition to intravascular hemolysis. At the moment there are three strategies of proximal complement inhibition: anti-C3 agents, anti-factor D agents and anti-factor B agents. These agents are available either subcutaneously or orally, and have been investigated in monotherapy or in association with eculizumab in PNH patients. Preliminary data clearly demonstrate that proximal complement inhibition is pharmacologically feasible and apparently safe, and may drastically improve the hematological response to complement inhibition in PNH. Indeed, we envision a new scenario of therapeutic complement inhibition, where proximal inhibitors (either anti-C3, anti-FD or anti-FB) may prove effective for the treatment of PNH, either in monotherapy or in combination with anti-C5 agents, eventually leading to drastic improvement of hematological response.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Inhibiteurs du complément/usage thérapeutique , Érythrocytes/immunologie , Hémoglobinurie paroxystique/traitement médicamenteux , Activation du complément/effets des médicaments et des substances chimiques , Complément C3/antagonistes et inhibiteurs , Complément C5/antagonistes et inhibiteurs , Hémolyse/effets des médicaments et des substances chimiques , Humains
20.
Semin Hematol ; 55(3): 130-135, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-30032749

RÉSUMÉ

The introduction of eculizumab, a human monoclonal antibody against the C5 component of complement, has changed radically the management of paroxysmal nocturnal hemoglobinuria (PNH). The blockade of the terminal complement pathway by eculizumab abrogates intravascular hemolysis, reduces the transfusion requirement and the risk of thrombosis in most of hemolytic PNH patients. However, in almost all PNH patients on eculizumab arises a fraction of PNH red cells that bind fragments of C3 and become a potential target of phagocytosis by macrophages. Eventually, this phagocytosis results in a variable degree of extravascular hemolysis that may reduce clinical benefits of eculizumab and, in fact, about one-fourth of patients remain transfusion-dependent. The treatment of the few PNH patients in which this de novo extravascular hemolysis become clinically relevant is still unsatisfactory. Nevertheless, the investigations of the mechanisms responsible of the extravascular hemolysis on eculizumab have resulted in the development of novel strategies for complement blockade that could overcome this condition.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Complément C3/métabolisme , Hémoglobinurie paroxystique/traitement médicamenteux , Anticorps monoclonaux humanisés/pharmacologie , Hémolyse , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE