51
|
Characterization of a Missense Mutation in the Catalytic Domain and a Splicing Mutation of Coagulation Factor X Compound Heterozygous in a Chinese Pedigree. Genes (Basel) 2021; 12:genes12101521. [PMID: 34680916 PMCID: PMC8535979 DOI: 10.3390/genes12101521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Congenital coagulation factor X (FX) deficiency is a rare bleeding disorder with an incidence of one in one million caused by mutations in the FX-coding gene(F10), leading to abnormal coagulation activity and a tendency for severe hemorrhage. Therefore, identifying mutations in FX is important for diagnosing congenital FX deficiency. Results: Genetic analysis of the proband identified two single-base substitutions: c.794T > C: p.Ile265Thr and c.865 + 5G > A: IVS7 + 5G > A. His FX activity and antigen levels were < 1% and 49.7%, respectively; aPTT and PT were prolonged to 65.3 and 80.5 s, respectively. Bioinformatics analysis predicted the two novel variants to be pathogenic. In-vitro expression study of the missense mutation c.794T > C: p.Ile265Thr showed normal synthesis and secretion. Activation of FXs by RVV, FVII/TF, and FVIII/FIX all showed no obvious difference between the variant and the reference. However, clotting activity by PT and aPTT assays and activity of thrombin generation in a TGA assay all indicated reduced activity of the mutant FX-Ile265Thr compared to FX-WT. Minigene assay showed a normal splicing mode c.865 + 5G > A: IVS7 + 5G > A, which is inconsistent with clinical phenotype. Conclusions: The heterozygous variants c.794T > C: p.Ile265Thr or c.865 + 5G > A: IVS7 + 5G > A indicate mild FX deficiency, but the compound heterozygous mutation of the two causes severe congenital FX deficiency. Genetic analysis of these two mutations may help characterize the bleeding tendency and confirm congenital FX deficiency. In-vitro expression and functional study showed that the low activity of the mutant FX-Ile265Thr is caused by decrease in its enzyme activity rather than self-activation. The minigene assay help us explore possible mechanisms of the splicing mutation. However, more in-depth mechanism research is needed in the future.
Collapse
|
52
|
Bernal S, Pelaez I, Alias L, Baena M, De Pablo-Moreno JA, Serrano LJ, Camero MD, Tizzano EF, Berrueco R, Liras A. High Mutational Heterogeneity, and New Mutations in the Human Coagulation Factor V Gene. Future Perspectives for Factor V Deficiency Using Recombinant and Advanced Therapies. Int J Mol Sci 2021; 22:9705. [PMID: 34575869 PMCID: PMC8465496 DOI: 10.3390/ijms22189705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 02/07/2023] Open
Abstract
Factor V is an essential clotting factor that plays a key role in the blood coagulation cascade on account of its procoagulant and anticoagulant activity. Eighty percent of circulating factor V is produced in the liver and the remaining 20% originates in the α-granules of platelets. In humans, the factor V gene is about 80 kb in size; it is located on chromosome 1q24.2, and its cDNA is 6914 bp in length. Furthermore, nearly 190 mutations have been reported in the gene. Factor V deficiency is an autosomal recessive coagulation disorder associated with mutations in the factor V gene. This hereditary coagulation disorder is clinically characterized by a heterogeneous spectrum of hemorrhagic manifestations ranging from mucosal or soft-tissue bleeds to potentially fatal hemorrhages. Current treatment of this condition consists in the administration of fresh frozen plasma and platelet concentrates. This article describes the cases of two patients with severe factor V deficiency, and of their parents. A high level of mutational heterogeneity of factor V gene was identified, nonsense mutations, frameshift mutations, missense changes, synonymous sequence variants and intronic changes. These findings prompted the identification of a new mutation in the human factor V gene, designated as Jaén-1, which is capable of altering the procoagulant function of factor V. In addition, an update is provided on the prospects for the treatment of factor V deficiency on the basis of yet-to-be-developed recombinant products or advanced gene and cell therapies that could potentially correct this hereditary disorder.
Collapse
Affiliation(s)
- Sara Bernal
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
- CIBERER. U-705, 18014 Barcelona, Spain
| | - Irene Pelaez
- Department of Pediatric and Oncohematology, University Hospital Virgen de las Nieves, 18014 Granada, Spain;
| | - Laura Alias
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
- CIBERER. U-705, 18014 Barcelona, Spain
| | - Manel Baena
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
| | - Juan A. De Pablo-Moreno
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis J. Serrano
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - M. Dolores Camero
- Association for the Investigation and Cure of Factor V Deficiency, 23002 Jaén, Spain;
| | - Eduardo F. Tizzano
- Department of Clinical and Molecular Genetics, University Hospital Vall d’Hebron and Medicine Genetics Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
| | - Ruben Berrueco
- Pediatric Hematology Department, Hospital Sant Joan de Déu, University of Barcelona and Research Institute Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Antonio Liras
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| |
Collapse
|
53
|
Batsuli G, Kouides P. Rare Coagulation Factor Deficiencies (Factors VII, X, V, and II). Hematol Oncol Clin North Am 2021; 35:1181-1196. [PMID: 34389198 DOI: 10.1016/j.hoc.2021.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although rare clotting factor deficiencies primarily referred to as rare bleeding disorders (RBD), including factors II, V, VII, and X, make up ∼5% of all inherited bleeding disorders worldwide, each of these clotting factors play a critical role in the coagulation cascade. Incomplete bleeding evaluation or misinterpretation of laboratory studies can result in delayed diagnoses that ultimately affect patient outcomes. Bleeding manifestations can range from mild to severe, but the most common are mucocutaneous bleeding. The ideal treatment in RBD is dedicated single-factor concentrates that can be used for acute bleeding events, surgical management, and prophylaxis.
Collapse
Affiliation(s)
- Glaivy Batsuli
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta; Department of Pediatrics, Emory University, Atlanta, GA, USA.
| | - Peter Kouides
- Mary M. Gooley Hemophilia Center, Rochester Regional Health, 1415 Portland Avenue, Rochester, NY 14621, USA
| |
Collapse
|
54
|
A homozygous loss-of-function mutation in GP1BB causing variable clinical phenotypes in a family with Bernard-Soulier syndrome. Blood Coagul Fibrinolysis 2021; 32:352-355. [PMID: 33657022 DOI: 10.1097/mbc.0000000000001027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bernard-Soulier syndrome is a rare autosomal recessive bleeding disorder and has a low incidence. Bernard-Soulier syndrome is caused by the deficiency of glycoprotein GPIb-V-IX complex, a receptor for von Willebrand factor and is characterized by thrombocytopenia, giant platelets and bleeding tendency. We are reporting three members of a same family with variable phenotypic clinical presentation. The index case is a 20-year-old boy who has a frequent presentation with epistaxis, and low platelet counts (25 × 109/l). He had been hospitalized multiple times and received platelet transfusions. His brother and cousin reported bleeding symptoms with less frequent medical intervention. Genetic analysis by next-generation sequencing identified a homozygous GP1BB variant (c.423C>A:p.Cys141Ter), which segregated amongst the family members. The results led us to an improved insight into the disease for this family with variable phenotypic expression, in addition to the identification of a variant for further structural and functional characterization.
Collapse
|
55
|
John MJ, Byreddy P, Modak K, Makkar M. Congenital Fibrinogen Deficiency in India and Role of Human Fibrinogen Concentrate. Acta Haematol 2021; 144:595-602. [PMID: 34091452 DOI: 10.1159/000516339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/25/2021] [Indexed: 11/19/2022]
Abstract
Congenital fibrinogen deficiency is an inherited disorder due to genetic mutations with diverse presentations arising from reduced fibrinogen levels (hypofibrinogenemia), absence of fibrinogen in circulation (afibrinogenemia), abnormal functioning (dysfibrinogenemia) or both reduced levels and abnormal functioning (hypodysfibrinogenemia) of fibrinogen. The decreased fibrinogen concentration in congenital fibrinogen deficiency necessitates fibrinogen replacement therapy with fresh frozen plasma, cryoprecipitate, or human fibrinogen concentrate. However, the use of fresh frozen plasma and cryoprecipitate is limited owing to their longer transfusion time, requirement of high doses, volume overload, risk of viral transmission, and other safety concerns. The availability of human fibrinogen concentrate has made it the preferred replacement alternative due to its reduced risk of viral transmission, smaller infusion volume, and accurate dosing. The hemostatic efficacy and safety of human fibrinogen concentrate in congenital fibrinogen deficiency is well established in the literature. We review the prevalence of congenital fibrinogen deficiency in India and the current role of human fibrinogen concentrate in its management.
Collapse
Affiliation(s)
- M Joseph John
- Department of Clinical Haematology, Haemato-Oncology and Bone Marrow (Stem Cell) Transplantation, Christian Medical College, Ludhiana, India
| | - Poojitha Byreddy
- Department of Clinical Haematology, Haemato-Oncology and Bone Marrow (Stem Cell) Transplantation, Christian Medical College, Ludhiana, India
| | - Ketan Modak
- Department of Clinical Haematology, Haemato-Oncology and Bone Marrow (Stem Cell) Transplantation, Christian Medical College, Ludhiana, India
| | - Mridul Makkar
- Department of Clinical Haematology, Haemato-Oncology and Bone Marrow (Stem Cell) Transplantation, Christian Medical College, Ludhiana, India
| |
Collapse
|
56
|
ElAlfy MS, Tantawy AAG, Eldin BEMB, Mekawy MA, Mohammad YAE, Ebeid FSE. Epistaxis in a Pediatric Outpatient Clinic: Could It be an Alarming Sign? Int Arch Otorhinolaryngol 2021; 26:e183-e190. [PMID: 35602268 PMCID: PMC9122762 DOI: 10.1055/s-0041-1726040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 11/24/2020] [Indexed: 11/01/2022] Open
Abstract
Abstract
Introduction Epistaxis is a common presentation among children.
Objective To investigate the suitability of a simple tool of assessment for patients with epistaxis that could guide in subgrouping those with possible bleeding tendencies who may need further assessment.
Methods Children who presented to a tertiary outpatient clinic with epistaxis of an unknown cause were recruited. They underwent thorough clinical assessment and answered the pediatric bleeding questionnaire and the epistaxis severity score. All patients underwent complete blood count as well as coagulation profile, and confirmatory diagnostic tests were performed as needed.
Results Among the 30,043 patients who presented to the outpatient clinic over a year, 100 children had epistaxis, with an estimated annual frequency of 1 in 300. A total of 84% of the patients were younger than 12, and nearly half of these were younger than 6 years. Seventy-six patients had recurrent epistaxis, and 12 had systemic comorbidities. A significant higher percentage of patients presented with epistaxis in the hot months of the year. A total of 90% of the patients presented anterior bleeding, and the majority were treated with nasal compression only. Forty-three patients presented with epistaxis only; 37 of them were diagnosed as idiopathic epistaxis, and 6 had local causes. Fifty-seven patients presented with other bleeding manifestations, 47 of whom had a definite bleeding disorder and the other 10 had undiagnosed bleeding tendency. Those with other bleeding manifestations showed a higher frequency of positive family history of epistaxis; of being referred from a primary care physician; of having alarming low platelet count, and of presenting less seasonal variability. A bleeding score ≥ 2 showed significant value in suspecting an underlying systemic pathology as a cause of epistaxis.
Conclusion The pediatric bleeding questionnaire is a useful and simple tool in the identification of pediatric patients who need further diagnostic testing to detect any underlying bleeding tendency.
Collapse
Affiliation(s)
- Mohsen Saleh ElAlfy
- Department of Pediatrics, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | | | | | - Mohamed Amin Mekawy
- Department of Clinical Pathology, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | | | | |
Collapse
|
57
|
de Azevedo Kinalski M, Sarkis-Onofre R, Dos Santos MBF. Inherited bleeding disorders in oral procedures. Assessment of prophylactic and therapeutic protocols: a scoping review. Aust Dent J 2021; 66:150-158. [PMID: 33336389 DOI: 10.1111/adj.12813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND This scoping review aimed to map available evidence regarding minor/major oral procedures in patients with inherited bleeding disorders (IBDs). METHODS Studies in medicine or dentistry that reported minor and major oral procedures in individuals presenting IBDs (e.g. haemophilia A or B, von Willebrand disease) were selected. Search and screening were performed in PubMed/Medline, Scopus, Web of Science and Cochrane Library by two independent researchers. RESULTS Initial search yielded 4152 citations, of which 257 were included in the final analysis. Most of the evidence for prophylaxis use was derived from observational studies and the most-commonly reported prophylactic protocols were fresh frozen plasma and factor VIII concentrate. A considerable number of studies reported postoperative complications and hospitalizations. CONCLUSION The present study identified that (1) there is room for new studies to assess the use of antifibrinolytic agents with no factor replacement; (2) to date, the use of factor replacement therapy is the most indicated approach when treating patients with IBDs with adjunct systemic or local antifibrinolytic agents to reduce post-operative complications; and (3) there is a critical need for high-quality evidence studies since much of the conclusions of the included studies are not supported by statistical analysis.
Collapse
Affiliation(s)
| | - Rafael Sarkis-Onofre
- Graduate Program in Dentistry, Meridional University (IMED), Passo Fundo, RS, Brazil
| | | |
Collapse
|
58
|
Abstract
Introduction: Rare bleeding disorders (RBDs) are a heterogeneous group of bleeding disorders. Consanguineous marriage is the favorite partnership in many communities among a population of more than a billion. Consanguineous marriages allow the defective recessive genes in RBDs to remain in the family. Clinical approach to RBDs is not as straightforward as typical bleeding disorders, which are secondary to scarcity and variation of RBDs. However, in many developing countries, the limitations of diagnostic facilities, the supply of therapeutic coagulation concentrates, health budgets, and optimal health-care infrastructures may intensify the clinical practice of RBDs.Areas covered: The author addresses the role of consanguineous marriage in the incidence of RBDs and what preventive steps can be considered to minimize the incidence of RBDs. A literature review of PubMed and google Scholar was conducted in November of 2020 using the relevant keywords.Expert opinion: This review addresses the prevalence of consanguineous marriages in Middle Eastern countries and the various reasons behind the coexistence of consanguineous marriage with RBDs. Finally, prevention steps that may be considered by health-care systems to minimize the incidence of rare bleeding disorders have been discussed. Health-care systems should train partners with the intention of consanguineous marriage on the downside of consanguinity.
Collapse
Affiliation(s)
- Hassan Mansouritorghabeh
- Central Diagnostic Laboratories, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
59
|
Zhang DL, Xue F, Dou XQ, Liu XF, Fu RF, Chen YF, Liu W, Jia YJ, Wang YH, Xiao ZJ, Zhang L, Yang RC. [Clinical and genetic analyses of hereditary factor Ⅴ deficiency cases]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:302-307. [PMID: 33979974 PMCID: PMC8120128 DOI: 10.3760/cma.j.issn.0253-2727.2021.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 11/05/2022]
Abstract
Objective: To analyze the clinical phenotype and molecular pathogenesis of nine patients with hereditary factor Ⅴ (FⅤ) deficiency. Methods: Nine patients with hereditary FⅤ deficiency who were admitted to the Institute of Hematology and Blood Diseases Hospital from April 1999 to September 2019 were analyzed. The activated partial thromboplastin time, prothrombin time, and FⅤ procoagulant activity (FⅤ∶C) were measured for phenotypic diagnosis. High-throughput sequencing was employed for the F5 gene mutation screening, Sanger sequencing was adopted to confirm candidate variants and parental carrying status, Swiss-model was used for three-dimensional structure analysis, and ClustalX v.2.1 was used for homologous analysis. Results: The FⅤ∶C of the nine patients ranged from 0.1 to 10.6. Among them, eight had a hemorrhage history, with kin/mucosal bleeding as the most common symptom (three cases, 37.5%) , whereas one case had no bleeding symptom. There were five homozygotes and four compound heterozygotes. A total of 12 pathogenic or likely pathogenic mutations were detected, of which c.6100C>A/p.Pro2034Thr, c.6575T>C/p.Phe2192Ser, c.1600_1601delinsTG/p. Gln534*, c.4713C>A/p.Tyr1571*, and c.952+5G>C were reported for the first time. Conclusion: The newly discovered gene mutations enriched the F5 gene mutation spectrum associated with hereditary FⅤ deficiency. High-throughput sequencing could be an effective method to detect F5 gene mutations.
Collapse
Affiliation(s)
- D L Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - F Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - X Q Dou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - X F Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - R F Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y F Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - W Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y J Jia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y H Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Z J Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - L Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - R C Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
60
|
Duque P, Chasco-Ganuza M, Ortuzar A, Almaraz C, Terradillos E, Perez-Rus G, Pascual C. Acquired FXIII Deficiency is Associated with High Morbidity. Thromb Haemost 2021; 122:48-56. [PMID: 33851388 DOI: 10.1055/a-1481-2733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND A factor XIII (FXIII) level >30% is considered necessary to prevent spontaneous bleeding. Bleeding is also a risk in patients with acquired FXIII deficiency, but the hemostatic level of FXIII in this context remains to be determined. METHODS We retrospectively analyzed all patients diagnosed with acquired FXIII deficiency at a large hospital over 3 years (study ID NCT04416594, http://www.clinicaltrials.gov) and assessed clinical data to identify the best cut-off point for FXIII activity to distinguish between low and high risk of major bleeding in a mixed medical and surgical population. RESULTS Of the 97 patients who experienced bleeding despite a normal coagulation test, 43.2% had FXIII activity <70%. FXIII activity was significantly lower in surgical patients and patients admitted to the intensive care unit (ICU). Low FXIII activity was significantly associated with long ICU stays and a high incidence of major bleeding. CONCLUSION Acquired FXIII deficiency is associated with high morbidity. The hemostatic level of FXIII in the setting of acquired FXIII deficiency might be above 30%.
Collapse
Affiliation(s)
- Patricia Duque
- Anesthesiology and Critical Care Department, Gregorio Marañon Hospital, Madrid, Spain.,Gregorio Marañon Health Research Institute, Gregorio Marañon Hospital, Madrid, Spain
| | - Maite Chasco-Ganuza
- Anesthesiology and Critical Care Department, Gregorio Marañon Hospital, Madrid, Spain
| | - Ariana Ortuzar
- Department of Hematology, Gregorio Marañon Hospital, Madrid, Spain
| | - Carolina Almaraz
- Anesthesiology and Critical Care Department, Gregorio Marañon Hospital, Madrid, Spain
| | - Estrella Terradillos
- Anesthesiology and Critical Care Department, Gregorio Marañon Hospital, Madrid, Spain
| | - Gloria Perez-Rus
- Department of Hematology, Gregorio Marañon Hospital, Madrid, Spain.,Internal Medicine Department, Complutense University, Madrid, Spain
| | - Cristina Pascual
- Department of Hematology, Gregorio Marañon Hospital, Madrid, Spain.,Gregorio Marañon Health Research Institute, Gregorio Marañon Hospital, Madrid, Spain
| |
Collapse
|
61
|
Andersen E, Chollet ME, Sletten M, Stavik B, Skarpen E, Backe PH, Thiede B, Glosli H, Henriksson CE, Iversen N. Molecular Characterization of Two Homozygous Factor VII Variants Associated with Intracranial Bleeding. Thromb Haemost 2021; 121:1588-1598. [PMID: 33742435 DOI: 10.1055/a-1450-8568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Clinical parameters have been extensively studied in factor (F) VII deficiency, but the knowledge of molecular mechanisms of this disease is scarce. We report on three probands with intracranial bleeds at an early age, one of which had concomitant high titer of FVII inhibitor. The aim of the present study was to identify the causative mutations and to elucidate the underlying molecular mechanisms. All nine F7 exons were sequenced in the probands and the closest family members. A homozygous deletion in exon 1, leading to a frame shift and generation of a premature stop codon (p.C10Pfs*16), was found in proband 1. Probands 2 and 3 (siblings) were homozygous for a missense mutation in exon 8, resulting in a glycine (G) to arginine (R) substitution at amino acid 240 (p.G240R). All probands had severely reduced FVII activity (FVII:C < 1 IU/dL). Treatment consisted of recombinant FVIIa and/or plasma concentrate, and proband 1 developed a FVII inhibitor shortly after initiation of treatment. The FVII variants were overexpressed in mammalian cell lines. No FVII protein was produced in cells expressing the p.C10Pfs*16 variant, and the inhibitor development in proband 1 was likely linked to the complete absence of circulating FVII. Structural analysis suggested that the G to R substitution in FVII found in probands 2 and 3 would destabilize the protein structure, and cell studies demonstrated a defective intracellular transport and increased endoplasmic reticulum stress. The molecular mechanism underlying the p.G240R variant could be reduced secretion caused by protein destabilization and misfolding.
Collapse
Affiliation(s)
- Elisabeth Andersen
- Department of Hematology, Oslo University Hospital, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Maria Eugenia Chollet
- Department of Hematology, Oslo University Hospital, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Marit Sletten
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Benedicte Stavik
- Department of Hematology, Oslo University Hospital, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ellen Skarpen
- Core Facility for Advanced Light Microscopy, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Paul Hoff Backe
- Department of Microbiology, Oslo University Hospital, Oslo, Norway.,Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Heidi Glosli
- Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway.,Centre for Rare Disorders, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Carola Elisabeth Henriksson
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nina Iversen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
62
|
Thau A, Saffren B, Zakrzewski H, Anderst JD, Carpenter SL, Levin A. Retinal hemorrhage and bleeding disorders in children: A review. CHILD ABUSE & NEGLECT 2021; 112:104901. [PMID: 33401159 DOI: 10.1016/j.chiabu.2020.104901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Retinal hemorrhages (RH) are a common manifestation of abusive head trauma (AHT) resulting from acceleration-deceleration injury with or without blunt impact. Evaluation of a child with RH requires careful consideration of these differential diagnoses. The extent to which coagulopathy alone can cause RH would be useful to understand as coagulopathy may accompany AHT. OBJECTIVE In this systematic review, we sought to identify whether coagulopathies have been reported with RH similar to those of AHT. METHODS We performed a literature search for ocular manifestations of bleeding disorders in children less than 18 years old. We included clotting factor deficiencies, vitamin K deficiency, platelet function abnormalities, thrombocytopenia, disseminated intravascular coagulation (DIC), and trauma induced coagulopathy (TIC). We included only pediatric reports of intraocular bleeding or documented eye examinations that indicated no hemorrhages. We then re-examined cases for ocular and systemic findings that could potentially mimic abuse. RESULTS Our initial search yielded 816 results. Sixty-one articles met our inclusion criteria. Of these, there were 32 children within the AHT age range (less than 5 years old) who had RH and concomitant coagulopathy. Only 5 cases might potentially be confused for abuse. Of these, no classic characteristics of RH from abuse such as retinoschisis or retinal folds were found. Systemic features were inconsistent with AHT. CONCLUSIONS The presence of coagulopathy alone does not rule out the possibility that the child has been abused. Coagulopathy alone has not been reported as an etiology of RH that are consistent with AHT, especially when other findings are present.
Collapse
Affiliation(s)
- Avrey Thau
- Thomas Jefferson University, Philadelphia, PA, USA
| | - Brooke Saffren
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Helena Zakrzewski
- Department of Experimental Surgery, McGill University, Montreal, QC, Canada
| | - James D Anderst
- Division of Child Adversity and Resilience, Children's Mercy Hospital, Kansas City, MO, USA
| | - Shannon L Carpenter
- Department of Hematology, Oncology, and Bone Marrow Transplantation, Children's Mercy Hospital, Kansas City, MO, USA
| | - Alex Levin
- Flaum Eye Institute and Golisano Children's Hospital, University of Rochester, New York, USA.
| |
Collapse
|
63
|
Pharmacokinetics, surrogate efficacy and safety evaluations of a new human plasma-derived fibrinogen concentrate (FIB Grifols) in adult patients with congenital afibrinogenemia. Thromb Res 2021; 199:110-118. [PMID: 33486319 DOI: 10.1016/j.thromres.2020.12.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/04/2020] [Accepted: 12/29/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS Congenital afibrinogenemia is a rare coagulation disorder resulting from a deficiency in fibrinogen. This study assessed the pharmacokinetics, surrogate efficacy and safety of FIB Grifols, a new human plasma-derived fibrinogen concentrate, to treat congenital afibrinogenemia. METHODS Eleven adult patients from a multinational, phase 1-2, prospective, open-label, single-arm, uncontrolled clinical study received a single infusion of FIB Grifols, 70 mg/kg bw. Fibrinogen pharmacokinetics (fibrinogen activity: Clauss method; antigen plasma concentrations: ELISA) and efficacy parameters were determined over 14 days after infusion. Efficacy endpoints were the mean change on plasma maximum clot firmness (MCF) on viscoelastic testing and coagulation tests 1-hour post-infusion, and correlation with fibrinogen levels throughout. Safety parameters were also assessed. RESULTS For the Clauss method, (mean [standard deviation]) baseline adjusted Cmax was 1.99 (0.40) g/L, reached 1.76 (1.00) h after infusion, and half-life was 76.94 (20.21) h. Using ELISA, Cmax after FIB Grifols infusion was 2.88 (0.86) mg/mL, with a tmax of 3.06 (2.24) h. Fibrinogen activity and antigen concentrations showed statistically significant correlation of 0.9120 (P < 0.001). Surrogate efficacy was demonstrated by a significant increase of 12.35 (3.85) mm in MCF. Prothrombin time, activated partial thromboplastin time and thrombin time, returned to normal ranges over time, indicating restoration of functionally active fibrinogen. There were no treatment-related adverse events, allergic reactions, serious adverse events, or discontinuations. CONCLUSIONS The pharmacokinetic profile of functionally active FIB Grifols was established, hemostasis was restored, and FIB Grifols was safe and well tolerated in fibrinogen-deficient patients.
Collapse
|
64
|
Djambas Khayat C, Lohade S, D'Souza F, Shamanur LG, Zekavat OR, Kruzhkova I, Schwartz B, Solomon C, Knaub S, Peyvandi F. Efficacy and safety of fibrinogen concentrate for on-demand treatment of bleeding and surgical prophylaxis in paediatric patients with congenital fibrinogen deficiency. Haemophilia 2020; 27:283-292. [PMID: 33326665 PMCID: PMC8049000 DOI: 10.1111/hae.14230] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Background Congenital fibrinogen deficiency (CFD) is a rare, inherited disorder affecting normal blood clotting function, where patients can experience severe and/or frequent bleeding episodes (BEs). Treatment with human fibrinogen concentrate (HFC) can prevent/arrest bleeding. There is a need for more data on the efficacy, pharmacokinetics (PK) and safety of HFC treatment in paediatric patients with CFD. Methods Haemostatic efficacy of HFC (Fibryga®, Octapharma AG) for on‐demand treatment of bleeding and surgical prophylaxis in patients <12 years old was assessed by investigators and an Independent Data Monitoring and Endpoint Adjudication Committee (IDMEAC) based on an objective 4‐point efficacy scale. Maximum clot firmness (MCF; surrogate marker of haemostatic efficacy), single‐dose PK and safety were also assessed. Results Of 14 patients receiving HFC (median [range] age 6.0 years [1.0–10.0]), eight received HFC for 10 BEs, three for surgical prophylaxis and 13 for PK. The IDMEAC rated haemostatic efficacy as 100% successful for on‐demand BE treatment (95% CI 69.15–100.00) and surgical prophylaxis (95% CI 29.24–100.00). After a mean first dose of 70.78 mg/kg for BEs, mean (±SD) MCF significantly increased from pre‐treatment to 1‐hour post‐infusion (3.3 mm [±1.77]; P = 0.0002), coinciding with haemostatic efficacy. PK parameters were favourable. Two possibly related adverse events occurred, including one serious (portal vein thrombosis). No allergic/hypersensitivity reactions or deaths were observed. Conclusion HFC treatment for on‐demand treatment of BEs and surgical prophylaxis was efficacious for this ultra‐rare paediatric population with congenital afibrinogenaemia and showed a favourable PK and safety profile.
Collapse
Affiliation(s)
| | | | | | | | - Omid Reza Zekavat
- Hematology Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Irina Kruzhkova
- Research & Development Department, Octapharma, Lachen, Switzerland
| | - Bruce Schwartz
- Clinical Research & Development, Octapharma, Paramus, NJ, USA
| | - Cristina Solomon
- Research & Development Department, Octapharma, Lachen, Switzerland
| | - Sigurd Knaub
- Research & Development Department, Octapharma, Lachen, Switzerland
| | - Flora Peyvandi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
| |
Collapse
|
65
|
Saes JL, Verhagen MJA, Meijer K, Cnossen MH, Schutgens REG, Peters M, Nieuwenhuizen L, van der Meer FJM, Kruis IC, van Heerde WL, Schols SEM. Bleeding severity in patients with rare bleeding disorders: real-life data from the RBiN study. Blood Adv 2020; 4:5025-5034. [PMID: 33064819 PMCID: PMC7594388 DOI: 10.1182/bloodadvances.2020002740] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/31/2020] [Indexed: 11/20/2022] Open
Abstract
Patients with hereditary rare bleeding disorders (RBDs) present with diverse hemorrhagic symptoms. Correlation between factor activity levels and clinical bleeding severity is poor for most RBDs. Threshold factor activity levels have been previously described in relation to bleeding severity but have not yet been validated. The Rare Bleeding Disorders in the Netherlands (RBiN) study is a nationwide cross-sectional study of patients registered in all 6 Dutch Haemophilia Treatment Centers with a known RBD and who are age 1 to 99 years. Bleeding scores were determined, and laboratory and clinical data were extracted from patient files. In all, 263 patients were included, of whom 202 (77%) attended the scheduled study visit. The median International Society of Thrombosis and Haemostasis (ISTH) bleeding assessment tool (BAT) score was 9. Correlations between baseline factor activity levels and ISTH BAT scores were strong for deficiencies in factor II (FII) (r = -0.792) and FX (r = -0.838) and were moderate for deficiencies of fibrinogen (r = -0.683), FV (r = -0.623), FVII (r = -0.516), FXIII (r = -0.516), and α2-antiplasmin (r = -0.594). There was no correlation for FXI deficiency (r = -0.218). The RBD BAT identified more women (94% vs 83%) and children (100% vs 71%) with an RBD than the ISTH BAT did. Importantly, 48% of patients had more severe bleeding than predicted for their baseline factor activity level. In addition, 34% of patients were predicted to be asymptomatic, but they actually had grade 2 (31%) or 3 (3%) bleeding. Bleeding severity in patients with RBDs is more pronounced than previously anticipated. The previously determined threshold factor activity levels to ensure no (spontaneous) bleeding in patients with an RBD are inaccurate. This trial was registered at www.clinicaltrials.gov as #NCT03347591.
Collapse
Affiliation(s)
- Joline L Saes
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Hemophilia Treatment Center Nijmegen, Eindhoven, Maastricht, The Netherlands
| | - Marieke J A Verhagen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Hemophilia Treatment Center Nijmegen, Eindhoven, Maastricht, The Netherlands
| | - Karina Meijer
- Department of Hematology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marjon H Cnossen
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Roger E G Schutgens
- Department of Hematology, van Creveldkliniek, University Medical Center Utrecht and University Utrecht, Utrecht, The Netherlands
| | - Marjolein Peters
- Pediatric-Hematology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Laurens Nieuwenhuizen
- Hemophilia Treatment Center Nijmegen, Eindhoven, Maastricht, The Netherlands
- Department of Hematology, Maxima Medical Center Eindhoven, Eindhoven, The Netherlands
| | - Felix J M van der Meer
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Ilmar C Kruis
- Netherlands Hemophilia Society, Nijkerk, The Netherlands; and
| | - Waander L van Heerde
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Hemophilia Treatment Center Nijmegen, Eindhoven, Maastricht, The Netherlands
- Enzyre BV, Novio Tech Campus, Nijmegen, The Netherlands
| | - Saskia E M Schols
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Hemophilia Treatment Center Nijmegen, Eindhoven, Maastricht, The Netherlands
| |
Collapse
|
66
|
Yoon HS, Han Y, Kim YJ, Kim MJ, Byun JM, Youk T, Lee JH, Park TS, Yoo J. Epidemiology of Congenital Bleeding Disorders: a Nationwide Population-based Korean Study. J Korean Med Sci 2020; 35:e350. [PMID: 33045773 PMCID: PMC7550232 DOI: 10.3346/jkms.2020.35.e350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/02/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Except for data in the Korea Hemophilia Foundation Registry, little is known of the epidemiology of congenital bleeding disorders in Korea. METHODS Data were obtained from the Korean Health Insurance Review and Assessment Service (HIRA) database. RESULTS From 2010 to 2015, there were 2,029 patients with congenital bleeding disorders in the Korean HIRA database: 38% (n = 775) of these patients had hemophilia A (HA), 25% (n = 517) had von Willebrand disease (vWD), 7% (n = 132) had hemophilia B (HB), and 25% (n = 513) had less common factor deficiencies. The estimated age-standardized incidence rate (ASR) of HA and HB was 1.78-3.15/100,000 and 0.31-0.51/100,000, respectively. That of vWD was 1.38-1.95/100,000. The estimated ASR of HA showed increase over time though the number of new patients did not increase. Most patients with congenital bleeding disorders were younger than 19 years old (47.8%), and most were registered in Gyeonggi (22.1%) and Seoul (19.2%). CONCLUSION This is the first nationwide population-based study of congenital bleeding disorders in Korea. This study provides data that will enable more accurate estimations of patients with vWD. This information will help advance the comprehensive care of congenital bleeding disorders. We need to continue to obtain more detailed information on patients to improve the management of these diseases.
Collapse
Affiliation(s)
- Hoi Soo Yoon
- Department of Pediatrics, Kyung Hee University School of Medicine, Seoul, Korea
| | - Yujin Han
- Department of Laboratory Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Young Jin Kim
- Department of Laboratory Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | | | - Ja Min Byun
- Department of Internal Medicine, Seoul National University College of Medicine, National University Hospital, Seoul, Korea
| | - Taemi Youk
- Research Institute, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jae Hee Lee
- Department of Pediatrics, Chosun University Hospital, Gwangju, Korea
| | - Tae Sung Park
- Department of Laboratory Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Jongha Yoo
- Department of Laboratory Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Korea.
| |
Collapse
|
67
|
Ghosh S, Gayen P, Jan S, Kishore AV, Kumar V, Mallick AM, Mukherjee A, Nandi SK, Sinha Roy R. Bioinspired Non-Immunogenic Multifunctional Sealant for Efficient Blood Clotting and Suture-Free Wound Closure. ACS Biomater Sci Eng 2020; 6:6378-6393. [PMID: 33449650 DOI: 10.1021/acsbiomaterials.0c01254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Engineering bioinspired peptide-based molecular medicine is an emerging paradigm for the management of traumatic coagulopathies and inherent bleeding disorder. A hemostat-based strategy in managing uncontrolled bleeding is limited due to the lack of adequate efficacy and clinical noncompliance. In this study, we report an engineered adhesive peptide-based hybrid regenerative medicine, sealant 5, which is designed integrating the structural and functional features of fibrin and mussel foot-pad protein. AFM studies have revealed that sealant 5 (55.8 ± 6.8 nN adhesive force) has higher adhesive force than fibrin (46.4 ± 7.3 nN adhesive force). SEM data confirms that sealant 5 retains its network-like morphology both at 37 and 60 °C, inferring its thermal stability. Both sealant 5 and fibrin exhibit biodegradability in the presence of trypsin, and sealant 5 also showed biocompatibility in the presence of fibroblast cells. Engineered sealant 5 efficiently promotes hemostasis with enhanced adhesiveness and less blood-loss than fibrin. In vivo data suggests that in heparinized conditions, sealant 5 ceases bleeding at 212.3 ± 15.1 s, whereas fibrin halts bleeding at 294.3 ± 21.4 s and blood-loss is ∼4-fold less in sealant 5 than in fibrin. In a heparinized system, sealant 5 facilitates faster blood-clotting than fibrin (∼82 s faster) and RADA-16, a reported peptide-based sealant (∼113 s faster). Additionally, in the case of sealant 5, the process of clotting mimicry-like fibrin is independent of the body's own coagulation system. Sealant 5 efficiently halts bleeding for both external and internal wounds, even for a heparinized system overcoming the bacterial infection. ELISA data and PMBC cell proliferation data support the non-immunogenic feature of sealant 5. Though fibrin and sealant 5 have exhibited comparable efficacy in suture-free wound closure, in vivo H&E staining images have revealed infiltration of very few immune cells as well as the presence of abundant collagen formation in the case of sealant 5-treated wound. Such nature-inspired non-immunogenic sealants offer exciting possibilities for the treatment of uncontrolled bleeding vis-à-vis wound closure.
Collapse
Affiliation(s)
- Snehasish Ghosh
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Paramita Gayen
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Somnath Jan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Anyam Vijay Kishore
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Vinod Kumar
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Argha M Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Asmita Mukherjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Rituparna Sinha Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India.,Centre for Advanced Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India.,Centre for Climate and Environmental Studies, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| |
Collapse
|
68
|
Zimowski KL, McGuinn CE, Abajas YL, Schultz CL, Kaicker S, Batsuli G. Use of plasma-derived factor X concentrate in neonates and infants with congenital factor X deficiency. J Thromb Haemost 2020; 18:2551-2556. [PMID: 32613702 DOI: 10.1111/jth.14985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/29/2020] [Accepted: 06/23/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Congenital factor X deficiency (FXD) is a rare bleeding disorder that often presents with severe bleeding in the neonatal period. Long-term prophylaxis with infusions of FX-containing products is recommended in patients with FXD and a personal or family history of severe bleeding. A plasma-derived FX concentrate (pdFX) is approved for on-demand and prophylactic therapy in adults and children with FXD. The safety and efficacy of pdFX has been demonstrated in patients <12 years of age, yet limited data exist regarding its use in infants. PATIENTS/METHODS This retrospective case series details clinical experience using pdFX in four neonates with moderate and severe FXD across four institutions. RESULTS AND CONCLUSIONS All four patients presented in the first week of life with severe bleeding. Following treatment of the acute bleed, prophylactic pdFX was initiated at an average of 29 days of life and a dose of 69 IU/kg every 48 hours. Incremental recovery (IR) in three infants averaged 1.42 IU/dL per IU/kg (min-max: 1.06-1.67 IU/dL per IU/kg). One patient experienced thrombotic complications in the setting of sepsis. After a median follow-up of 26.5 months, no patient has experienced breakthrough bleeding episodes. Our study supports the use of pdFX in neonates and infants and suggests that higher pdFX dosing of 70 to 80 IU/kg every 48 hours based on the smallest available vial size is feasible. Because of variability in IR, close monitoring of FX activity should be used to guide dosing in this age group.
Collapse
Affiliation(s)
- Karen L Zimowski
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Catherine E McGuinn
- Division of Pediatric Hematology and Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Yasmina L Abajas
- Pediatric Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Corinna L Schultz
- Nemours Center for Cancer & Blood Disorders, Nemours/AI duPont Hospital for Children, Wilmington, Delaware, USA
| | - Shipra Kaicker
- Division of Pediatric Hematology and Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Glaivy Batsuli
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
69
|
Gorkom BLV, Holme PA, Joch C, Rogosch T, Feussner A, McKeand W, Roberts J, van Heerde W. Pharmacokinetics and pharmacodynamics of a recombinant fusion protein linking activated coagulation factor VII with human albumin (rVIIa-FP) in patients with congenital FVII deficiency. ACTA ACUST UNITED AC 2020; 25:17-25. [PMID: 31852380 DOI: 10.1080/16078454.2019.1700329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: Recombinant fusion protein linking activated factor VIIa to human albumin (rVIIa-FP) is a therapeutic option designed to prevent and treat bleeding events in patients with congenital FVII deficiency with reduced infusion frequency compared to current FVII treatments. This study characterized the pharmacokinetics (PK) and pharmacodynamics (PD) of rVIIa-FP.Methods: A phase I multicenter, randomized, open-label, parallel-arm, single-dose study (NCT02470871) was conducted in nine patients with severe congenital FVII deficiency. Patients received their routine FVII product (30 IU/kg plasma-derived FVII [pdFVII] or 25 μg/kg recombinant activated FVII (rFVIIa) [eptacog alfa]), and were then randomly assigned to receive 100 or 300 μg/kg of rVIIa-FP. Blood samples for PK and PD assessments were drawn up to 48 hr after administration. FVIIa activity was determined using a one-stage clotting assay. PD parameters were derived from thrombin generation testing, using the Nijmegen hemostasis assay.Results: rVIIa-FP showed improved PK compared to rFVIIa, with 2- to 3-fold longer t1/2 and 4- to 8-fold lower clearance. Analysis of PD data showed a sustained suppression of lag time below 4.5 min (upper limit of healthy people) for rVIIa-FP compared to rFVIIa. AUEC and ECmax were similar across the two dose groups of rVIIa-FP and rFVIIa.Discussion: rVIIa-FP was well tolerated in patients with congenital FVII deficiency, showed a longer half-life and lower clearance compared to rFVIIa, and lag time remaining within healthy ranges for ≥8 hr.Conclusion: These results warrant further investigation into the efficacy of rVIIa-FP to control and prevent bleeding in patients with FVII deficiency.
Collapse
Affiliation(s)
- Britta Laros-van Gorkom
- Department of Hematology and Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pål André Holme
- Department of Hematology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | - Waander van Heerde
- Department of Hematology and Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Laboratory Medicine, Hematology Laboratory, Radboud University Medical Center, Nijmegen, Netherlands.,Enzyre, Novio Tech Campus, Nijmegen, The Netherlands
| |
Collapse
|
70
|
Abstract
Factor X deficiency is a severe inherited coagulation disorder, which is characterized by severe systemic bleeding manifestations in affected individuals. It is a rare disorder with a frequency of around 1:1,000,000 in the general population. We present the case of an infant with factor X deficiency who presented with complex febrile seizure. Although febrile seizures are very common in children, a closer scrutiny leads to neuroimaging and finding of intracranial bleed. Hematologic and genetic investigations confirmed the diagnosis. A high index of suspicion should be maintained to diagnose uncommon bleeding disorders in children.
Collapse
|
71
|
Shaw JR, Carrier M, Dowlatshahi D, Chakraborty S, Tokessy M, Buyukdere H, Castellucci LA. Activated prothrombin complex concentrates for direct oral anticoagulant-associated bleeding or urgent surgery: Hemostatic and thrombotic outcomes. Thromb Res 2020; 195:21-28. [PMID: 32645667 DOI: 10.1016/j.thromres.2020.06.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/28/2020] [Accepted: 06/24/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Studies evaluating the use of activated prothrombin complex concentrates (aPCCs) for DOAC-associated bleeding are sparse. MATERIALS AND METHODS We conducted a retrospective study of patients receiving aPCC for DOAC-associated bleeding or for pre-operative optimization of hemostasis prior to urgent surgery. The primary efficacy outcome was hemostatic efficacy, the primary safety outcome was the 30-day thromboembolic complication rate. RESULTS Eighty-two patients were included in the analysis; 14 patients on dabigatran, 39 patients on rivaroxaban and 29 patients on apixaban. Fifty-four patients received aPCC for major bleeding and 28 patients prior to urgent surgery. Mean aPCC dosing was 2974 IU (SD ± 857 IU). Hemostasis was deemed effective by ISTH criteria in 50% of cases and "Good" or "Moderate" by Sarode criteria in 45.2% and 14.3% of cases, respectively. Surgical hemostasis was rated as "Normal" in 84% of cases pre-operative administration. Median pre-aPCC INR was 1.6 (IQR 0.5) and median post-aPCC INR was 1.2 (IQR 0.2) (p < 0.00001). Median pre-aPCC aPTT was 36 s (IQR 12.8), median post-aPCC aPTT was 29 s (IQR 9.8) (p = 0.0001). The 30-day thromboembolic event rate was 6.1%. CONCLUSION Further study is needed to characterize the hemostatic effects and thromboembolic risk of aPCC among patients with DOAC-associated bleeding or for attempted normalization of hemostasis prior to urgent surgery.
Collapse
Affiliation(s)
- Joseph R Shaw
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Marc Carrier
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dar Dowlatshahi
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Canada
| | | | - Melanie Tokessy
- Department of Pathology and Laboratory Medicine, Division of Hematology and Transfusion Medicine, The Ottawa Hospital, Canada
| | - Hakan Buyukdere
- Department of Pathology and Laboratory Medicine, Division of Hematology and Transfusion Medicine, The Ottawa Hospital, Canada
| | - Lana A Castellucci
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, Ottawa, Canada.
| |
Collapse
|
72
|
Lu S, Lin W, Ji H, Su M, Zhao X, Wang C. A Compound Heterozygosis of Two Novel Mutations Causes Factor X Deficiency in a Chinese Pedigree. Acta Haematol 2020; 144:176-181. [PMID: 32599596 DOI: 10.1159/000507689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/03/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Mutations in the F10-coding gene can cause factor X (FX) deficiency, leading to abnormal coagulation activity and severe tendency for hemorrhage. Therefore, identifying mutations in F10 is important for diagnosing congenital FX deficiency. METHODS We studied a 63-year-old male patient with FX deficiency and 10 of his family members. Clotting and immunological methods were used to determine activated partial thromboplastin time (aPTT), prothrombin time (PT), thrombin time (TT), fibrinogen levels, FX activity, and FX antigen levels. The platelet count was determined. A mixing study was performed to eliminate the presence of coagulation factor inhibitors and lupus anticoagulant. Mutations were searched using whole-exome sequencing and certified by Sanger sequencing. RESULTS Genetic analysis of the proband identified two single-base substitutions: c.1085G>A (p.Ser362Asn) and c.1152C>A (p.Tyr384Ter, termination codon, caused by the DNA sequence TAA). His FX activity and antigen levels were 1.7% and 408.53 pg/mL, respectively; aPTT and PT were 52.3 and 48.0 s, respectively. One brother had the same compound heterozygous mutations, and his FX activity and antigen levels were 1.3% and 465.47 pg/mL, respectively; his aPTT and PT were 65.2 and 54.5 s, respectively. His mother, another brother, and one sister were heterozygous for c.1085G>A (p.Ser362Asn), and his daughter and grandson (6 years old) were heterozygous for c.1152C>A (p.Tyr384Ter). CONCLUSION The heterozygous variants p.Ser362Asn or p.Tyr384Ter indicate mild FX deficiency, but the compound heterozygous mutation of the two causes severe congenital FX deficiency and bleeding. Genetic analysis of these two mutations may help characterize the bleeding tendency and confirm congenital FX deficiency.
Collapse
Affiliation(s)
- Songsong Lu
- Medical Laboratory Center, First Medical Center of Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Weicheng Lin
- Department of Orthopedic Trauma, Peking University People's Hospital, Beijing, China
| | - Huijuan Ji
- Intensive Care Unit, Peking University People's Hospital, Beijing, China
| | - Ming Su
- Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiaotao Zhao
- Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chengbin Wang
- Medical Laboratory Center, First Medical Center of Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China,
| |
Collapse
|
73
|
Trevisan BM, Porada CD, Atala A, Almeida-Porada G. Microfluidic devices for studying coagulation biology. Semin Cell Dev Biol 2020; 112:1-7. [PMID: 32563678 DOI: 10.1016/j.semcdb.2020.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
The ability to study the behavior of cells, proteins, and cell-cell or cell-protein interactions under dynamic forces such as shear stress under fluid flow, provides a more accurate understanding of the physiopathology of hemostasis. This review touches upon the traditional methods for studying blood coagulation and platelet aggregation and provides an overview on cellular and protein response to shear stress. We also elaborate on the biological aspects of how cells recognize mechanical forces and convert them into biochemical signals that can drive various signaling pathways. We give a detailed description of the various types of microfluidic devices that are employed to study the complex processes of platelet aggregation and blood coagulation under flow conditions as well as to investigate endothelial shear-response. We also highlight works mimicking artificial vessels as platforms to study the mechanisms of coagulation, and finish our review by describing anticipated clinical uses of microfluidics devices and their standardization.
Collapse
Affiliation(s)
- Brady M Trevisan
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
74
|
A Novel Frameshift Mutation in the FGA Gene (c.196 delT) Leading to Congenital Afibrinogenemia. J Pediatr Hematol Oncol 2020; 42:e98-e99. [PMID: 31725541 DOI: 10.1097/mph.0000000000001658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Congenital afibrinogenemia is characterized by the absence of fibrinogen. Congenital fibrinogen disorders result from several mutations in FGA, FGB, or FGG. Their epidemiology is not well known. OBSERVATION The present study reports on 2 children with congenital afibrinogenemia. The first child, a male who is now 9 years old, was diagnosed with afibrinogenemia after spontaneous intracranial bleeding at the age of 3 years. The second child is a 2-year-old female cousin of the first patient, who was diagnosed with afibrinogenemia after coagulation tests were carried out due to frequent epistaxis and mucocutaneous bleeding. At follow-up, blood samples of the patients and their parents were sent to the Department of Genetic Medicine and Development, University Medical Center, Switzerland, for polymerase chain reaction analysis. In both patients, the novel homozygous frameshift mutation in the FGA exon 3: c.196 delT was detected. The parents of the patients were both heterozygous for the same mutation. CONCLUSIONS Congenital afibrinogenemia is a rare coagulation disease. The molecular epidemiology of congenital fibrinogen disorders is complex, and the identification of new mutations will help shed light on this complex molecular structure. Therefore, a genetic analysis that includes more centers is needed.
Collapse
|
75
|
Bellon A, Fuseau E, Roumanie O, Lamazure J, Stevens W, Dahmane A, Barthez-Toullec M, Golly D, Henriet C, Bridey F. Population pharmacokinetics of a triple-secured fibrinogen concentrate administered to afibrinogenaemic patients: Observed age- and body weight-related differences and consequences for dose adjustment in children. Br J Clin Pharmacol 2020; 86:329-337. [PMID: 31658379 PMCID: PMC7015754 DOI: 10.1111/bcp.14147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/09/2019] [Accepted: 09/21/2019] [Indexed: 11/28/2022] Open
Abstract
Aims The pharmacokinetics (PK) of a triple‐secured fibrinogen concentrate (FC) was assessed in patients ≥40 kg by noncompartmental analysis over a period of 14 days with multiple blood samples. Limited PK time point assessments in children lead to consideration of using Bayesian estimation for paediatric data. The objectives were (i) to define the population PK of FC in patients with afibrinogenaemia; (ii) to detect age‐ and body weight‐related differences and consequences for dose adjustment. Methods A population PK model was built using plasma fibrinogen activity data collected in 31 patients aged 1 to 48 years who had participated in a single‐dose PK study with FC 0.06 g kg–1. Results A 1‐compartment model with allometric scaling accounting for body weight was found to best describe the kinetics of FC. Addition of age and sex as covariates did not improve the model. Incremental in vivo recovery assessed at the end of infusion with the predicted maximal concentrations was lower, weight‐adjusted clearance was higher, and fibrinogen elimination half‐life was shorter in patients <40 kg than patients ≥40 kg. Interpatient variability was similar in both groups. Conclusion Dosing in patients ≥40 kg based on the previous empirical finding using noncompartmental analysis where FC 1 g kg–1 raises the plasma fibrinogen activity by 23 g L–1 was confirmed. In patients <40 kg, (covering the age range from birth up to about 12 years old) FC 1 g kg–1 raises the plasma fibrinogen by 19 g L–1. Dosing should be adapted accordingly unless therapy is individualized.
Collapse
Affiliation(s)
- Anne Bellon
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | | | | | - Jennifer Lamazure
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Wil Stevens
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Amel Dahmane
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Malika Barthez-Toullec
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Dominique Golly
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Céline Henriet
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Françoise Bridey
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| |
Collapse
|
76
|
Prabhu S, Agarwal S, Prabhu S. Spontaneous Intracranial Extradural Hematoma Secondary to Hypofibrinogenemia: A Rare Case Report. Asian J Neurosurg 2020; 14:1288-1290. [PMID: 31903380 PMCID: PMC6896612 DOI: 10.4103/ajns.ajns_143_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Spontaneous extradural hematoma is a well-documented but fairly rare condition, which can be associated with conditions such as vascular malformations, sickle cell disease, metastasis to the skull, infectious diseases of the skull, coagulation disorders, and use of anticoagulants. Of these, very few cases are of life-threatening extradural hematomas in adults, where hypofibrinogenemia is the cause of bleed, without any history of trauma. In this report, we have discussed the management of such a patient.
Collapse
Affiliation(s)
- Santosh Prabhu
- Department of Neurosurgery, Western India Institute of Neurosciences, Kolhapur, Maharashtra, India
| | - Sidharth Agarwal
- Department of Neurosurgery, Western India Institute of Neurosciences, Kolhapur, Maharashtra, India
| | - Sujata Prabhu
- Department of Neuroanesthesia, Western India Institute of Neurosciences, Kolhapur, Maharashtra, India
| |
Collapse
|
77
|
Barbitoff YA, Skitchenko RK, Poleshchuk OI, Shikov AE, Serebryakova EA, Nasykhova YA, Polev DE, Shuvalova AR, Shcherbakova IV, Fedyakov MA, Glotov OS, Glotov AS, Predeus AV. Whole-exome sequencing provides insights into monogenic disease prevalence in Northwest Russia. Mol Genet Genomic Med 2019; 7:e964. [PMID: 31482689 PMCID: PMC6825859 DOI: 10.1002/mgg3.964] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Allele frequency data from large exome and genome aggregation projects such as the Genome Aggregation Database (gnomAD) are of ultimate importance to the interpretation of medical resequencing data. However, allele frequencies might significantly differ in poorly studied populations that are underrepresented in large-scale projects, such as the Russian population. METHODS In this work, we leveraged our access to a large dataset of 694 exome samples to analyze genetic variation in the Northwest Russia. We compared the spectrum of genetic variants to the dbSNP build 151, and made estimates of ClinVar-based autosomal recessive (AR) disease allele prevalence as compared to gnomAD r. 2.1. RESULTS An estimated 9.3% of discovered variants were not present in dbSNP. We report statistically significant overrepresentation of pathogenic variants for several Mendelian disorders, including phenylketonuria (PAH, rs5030858), Wilson's disease (ATP7B, rs76151636), factor VII deficiency (F7, rs36209567), kyphoscoliosis type of Ehlers-Danlos syndrome (FKBP14, rs542489955), and several other recessive pathologies. We also make primary estimates of monogenic disease incidence in the population, with retinal dystrophy, cystic fibrosis, and phenylketonuria being the most frequent AR pathologies. CONCLUSION Our observations demonstrate the utility of population-specific allele frequency data to the diagnosis of monogenic disorders using high-throughput technologies.
Collapse
Affiliation(s)
- Yury A. Barbitoff
- Bioinformatics InstituteSt. PetersburgRussia
- Department of Genetics and BiotechnologySt. Petersburg State UniversitySt. PetersburgRussia
| | | | | | - Anton E. Shikov
- Bioinformatics InstituteSt. PetersburgRussia
- City Hospital No. 40St. PetersburgRussia
| | - Elena A. Serebryakova
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology and ReproductionSt. PetersburgRussia
| | - Yulia A. Nasykhova
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology and ReproductionSt. PetersburgRussia
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
| | | | | | - Irina V. Shcherbakova
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
| | | | - Oleg S. Glotov
- City Hospital No. 40St. PetersburgRussia
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology and ReproductionSt. PetersburgRussia
| | - Andrey S. Glotov
- City Hospital No. 40St. PetersburgRussia
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology and ReproductionSt. PetersburgRussia
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
- Institute of Living SystemsImmanuel Kant Baltic Federal UniversityKaliningradRussia
| | | |
Collapse
|
78
|
Bilateral Cleft Lip and Palate Accompanied by 13q- Syndrome with Deficiencies of FVII and FX: A Case Report. J Clin Pediatr Dent 2019; 43:288-291. [PMID: 31094634 DOI: 10.17796/1053-4625-43.4.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 13q deletion syndrome is a rare genetic disorder caused by structural and functional monosomy of chromosome 13. On 13q34, which is the terminal of the long arm, causative genes of coagulation factors VII and X (FVII and FX) are mapped. Patients with a combination of FVII and FX deficiencies are extremely rare and there have been few articles about perioperative coagulation support for such patients. Herein, we report on a case of bilateral cleft lip and palate accompanied by 13q deletion syndrome with deficiencies of FVII and FX. The chromosomal investigation indicated 46, XX, del(13)(q33) by G-banding. Prothrombin time and activated partial thromboplastin time were found to be 21.0 seconds (sec) (prothrombin time-international normalized ratio 1.76) and 41.6 sec (normal range; 23.9 - 39.7 sec), respectively. The activities of coagulation FVII and FX were 22% and 36%, respectively. A two-stage cheiloplasty was performed at 4 and 7 months of age followed by a palatoplasty at 1 year and 6 months. Tranexamic acid was given intravenously three times a day for three days after each surgery. There were no adverse events such as bleeding from the oral or nasal cavities and healing of the surgical wound was good without dehiscence.
Collapse
|
79
|
Low factor V level ameliorates bleeding diathesis in patients with combined deficiency of factor V and factor VIII. Blood 2019; 134:1745-1754. [PMID: 31558466 DOI: 10.1182/blood.2018886069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/23/2019] [Indexed: 12/28/2022] Open
Abstract
Combined factor V and factor VIII deficiency is a rare disorder associated with relatively mild bleeding diathesis. Shao and colleagues elucidate the double role of factor V as both a pro- and anticoagulant protein, demonstrating that decreased factor V may ameliorate factor VIII deficiency through decreasing the level of tissue factor pathway inhibitor.
Collapse
|
80
|
Inherited Moderate Factor X Deficiency Presenting as Cardiac Tamponade. Case Rep Hematol 2019; 2019:9657516. [PMID: 31662920 PMCID: PMC6778902 DOI: 10.1155/2019/9657516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/29/2019] [Accepted: 09/11/2019] [Indexed: 11/20/2022] Open
Abstract
Factor X deficiency is a rare bleeding disorder that varies in the severity of its clinical manifestations. The symptoms of this disorder can occur at any age, although most severe cases appear in childhood. The rarity of this condition has not allowed for the establishment of evidence‐based management guidelines, and thus, individuals afflicted with factor X deficiency are treated based on limited literature and the opinions of clinicians with extensive experience. In this case report, we discuss a unique presentation of a 38-year-old male who was found to have cardiac tamponade as a result of his newly diagnosed inherited moderate factor X deficiency. This was discovered by obtaining a factor X activity assay and confirmed with genetic testing which demonstrated a missense variant on the factor X gene on chromosome 13. His management involved correction of his factor X deficiency with fresh frozen plasma, a pericardiocentesis, and placement of a pericardial window. He has been asymptomatic and without hemorrhagic episodes for the 10 months following his discharge.
Collapse
|
81
|
Clinical and laboratory diagnosis of rare coagulation disorders (RCDs). Thromb Res 2019; 196:603-608. [PMID: 31515069 DOI: 10.1016/j.thromres.2019.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/17/2022]
Abstract
Rare coagulation disorders (RCDs) are a group of diseases due to coagulation factors deficiency leading to life-long bleeding diathesis. The diagnosis of RCDs is challenging due to the limited knowledge of these disorders and the large heterogeneity of their bleeding patterns. The clinical symptoms of RCDs are extremely diverse in terms of bleeding type, site, severity, age at onset, and duration. The strength of the association between clotting factor activity level in plasma and clinical symptoms is also variable within each RCD. The clinical evaluation of RCDs starts with a detailed collection of clinical history and has been facilitated by bleeding assessment tools, however their effectiveness in diagnosing RCDs requires further investigation. The following laboratory diagnosis of RCDs involves coagulation screening tests, including activated partial thromboplastin time, prothrombin time, and thrombin time. After ruling out the presence of an inhibitor by mixing studies, in case of abnormal results, the specific deficiency is identified by performing one-stage clotting assays using the specific factor-depleted plasmas as substrate. In fibrinogen and FXIII deficiencies coagulation screening tests are not informative, therefore additional tests are needed. Global assays have been developed and are thought to aid in patient management, however, they are not well standardized yet. In addition to outlining the principles of clinical and laboratory diagnosis, this review explores molecular basis of RCDs and laboratory techniques for genetic analysis, and discusses the importance and effectiveness of quality control programs to ensure standardized laboratory results.
Collapse
|
82
|
Gupta S, Acharya S, Roberson C, Lail A, Soucie JM, Shapiro A. Potential of the Community Counts registry to characterize rare bleeding disorders. Haemophilia 2019; 25:1045-1050. [PMID: 31509320 DOI: 10.1111/hae.13847] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/13/2019] [Accepted: 08/23/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Rare bleeding disorders (RBDs) comprise of heterogeneous coagulation factor deficiencies and platelet disorders that are underreported worldwide. AIM First report on RBD data from United States haemophilia treatment center network (USHTCN). METHODS A national surveillance system for the federally recognized USHTCN developed in collaboration with the Centers for Disease Control and Prevention (CDC) and American Thrombosis and Haemostasis Network (ATHN) was queried for patients with RBDs. Patient counts were extracted from the HTC Population Profile (HTC PP) component including limited data on patients followed through the USHTCN, and from the Registry component, including patient authorized, detailed clinical data. The prevalence of RBDs in the United States was estimated based on the HTC PP data and compared to the expected national prevalence based on data extrapolated from Orphanet, an international registry. RESULTS Based on the estimated prevalence of RBD in the overall 2017 US population, the cases in the HTC network were lower than expected for FI, FII, FX, and FV + FVIII deficiencies by 36%, 61%, 75% and 94%, respectively, and higher than expected for FXIII, FV, FVII, and FXI deficiencies by 7%, 14%, 33% and 185%, respectively. The proportion of RBD patients reported in the HTC PP, enrolled in the Registry, was 10.8%. CONCLUSIONS There is a clear need to identify individuals with RBDs who could benefit from the comprehensive care provided in the USHTCN. In addition, increased enrolment of people with all RBDs in the Registry is needed to improve knowledge of treatment outcomes of patients with RBDs in the United States.
Collapse
Affiliation(s)
- Sweta Gupta
- Indiana Hemophilia and Thrombosis Center, Inc., Indianapolis, Indiana
| | - Suchitra Acharya
- Cohen Children's Medical Center, Northwell Health New Hyde Park, New York, New York
| | | | | | - J Michael Soucie
- National Center on Birth Defects and Developmental Disabilities, Division of Blood Disorders, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Amy Shapiro
- Indiana Hemophilia and Thrombosis Center, Inc., Indianapolis, Indiana
| |
Collapse
|
83
|
|
84
|
Andersen E, Chollet ME, Baroni M, Pinotti M, Bernardi F, Skarpen E, Sandset PM, Skretting G. The effect of the chemical chaperone 4-phenylbutyrate on secretion and activity of the p.Q160R missense variant of coagulation factor FVII. Cell Biosci 2019; 9:69. [PMID: 31467667 PMCID: PMC6712599 DOI: 10.1186/s13578-019-0333-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/17/2019] [Indexed: 01/09/2023] Open
Abstract
Background Congenital coagulation factor (F) VII deficiency is a rare bleeding disorder caused by mutations in the F7 gene. The missense factor FVII variant p.Q160R is the disease-causing mutation in all Norwegian FVII deficient patients and results in reduced biological activity and antigen levels of FVII in patient plasma. Previous in vitro studies on this variant demonstrated impaired intracellular trafficking and reduced secretion, possibly due to protein misfolding. The aim of the study was therefore to assess the impact of chemical chaperones on cellular processing and secretion of this variant using a cell model based on overexpression of the recombinant protein. Results Through screening of compounds, we identified 4-phenylbutyrate (4-PBA) to increase the secretion of recombinant (r) FVII-160R by ~ 2.5-fold. Additionally, treatment with 4-PBA resulted in a modest increase in specific biological activity. Intracellular localization studies revealed that upon treatment with 4-PBA, rFVII-160R was secreted through Golgi and Golgi reassembly-stacking protein (GRASP)-structures. Conclusions The present study demonstrates that the chemical chaperone 4-PBA, restores intracellular trafficking and increases the secretion of a missense FVII variant with functional properties in the extrinsic coagulation pathway.
Collapse
Affiliation(s)
- Elisabeth Andersen
- 1Department of Haematology, Oslo University Hospital, Oslo, Norway.,2Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,3Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Maria Eugenia Chollet
- 1Department of Haematology, Oslo University Hospital, Oslo, Norway.,2Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Marcello Baroni
- Department of Life Sciences and Biotechnology, and LTTA Centre, University of Ferrara, Oslo, Norway
| | - Mirko Pinotti
- Department of Life Sciences and Biotechnology, and LTTA Centre, University of Ferrara, Oslo, Norway
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, and LTTA Centre, University of Ferrara, Oslo, Norway
| | - Ellen Skarpen
- 5Core Facility for Advanced Light Microscopy, Institute for Cancer Research, Oslo University Hospital, Ferrara, Italy
| | - Per Morten Sandset
- 1Department of Haematology, Oslo University Hospital, Oslo, Norway.,2Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,3Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Grethe Skretting
- 1Department of Haematology, Oslo University Hospital, Oslo, Norway.,2Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
85
|
Abstract
Factor VII (FVII) deficiency is the most common of the Rare Inherited Coagulation Disorders. The inheritance is autosomal recessive but there is variable penetrance. Overall there is poor correlation between the FVII level and the bleeding phenotype. Heterozygotes may have significant bleeding and severe homozygotes, or compound heterozygotes can be asymptomatic. Typically, homozygotes have FVII levels <10% and heterozygotes have levels above that. In most cases bleeding is uncommon with FVII levels>10-20%. A personal and family history is essential to determine the bleeding risk and to plan for surgical and obstetrical prophylaxis. Severe bleeding complications including central nervous system bleeding, gastrointestinal system bleeding and bleeding into the joints occurs in 10-15% of FVII deficient patients. Mucocutaneous bleeding is a common symptom but 30% of patients are asymptomatic. Fifty to 69% of women have heavy menstrual bleeding. Due to the limited number of publications regarding this rare disorder there are no consensus guidelines. There is registry data which has led to the best recommendations for treatment of bleeding episodes, initiation of long-term prophylaxis in addition to surgical plus ante and peripartum prophylaxis. Recombinant FVII concentrate is the best replacement therapy and a review of treatment and prophylaxis dosing is discussed.
Collapse
Affiliation(s)
- K Sue Robinson
- Division of Hematology, Dalhousie University, Queen Elizabeth II Health Sciences Centre, Department of Medicine, Rm. 416, 4th Floor Bethune Bldg, 1276 South Park St., Halifax, NS, B3H 2Y9, Canada.
| |
Collapse
|
86
|
First description of the molecular and clinical characterization of hereditary factor V deficiency in Saudi Arabia. Blood Coagul Fibrinolysis 2019; 30:224-232. [DOI: 10.1097/mbc.0000000000000828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
87
|
Balestra D, Branchini A. Molecular Mechanisms and Determinants of Innovative Correction Approaches in Coagulation Factor Deficiencies. Int J Mol Sci 2019; 20:ijms20123036. [PMID: 31234407 PMCID: PMC6627357 DOI: 10.3390/ijms20123036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/07/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Molecular strategies tailored to promote/correct the expression and/or processing of defective coagulation factors would represent innovative therapeutic approaches beyond standard substitutive therapy. Here, we focus on the molecular mechanisms and determinants underlying innovative approaches acting at DNA, mRNA and protein levels in inherited coagulation factor deficiencies, and in particular on: (i) gene editing approaches, which have permitted intervention at the DNA level through the specific recognition, cleavage, repair/correction or activation of target sequences, even in mutated gene contexts; (ii) the rescue of altered pre-mRNA processing through the engineering of key spliceosome components able to promote correct exon recognition and, in turn, the synthesis and secretion of functional factors, as well as the effects on the splicing of missense changes affecting exonic splicing elements; this section includes antisense oligonucleotide- or siRNA-mediated approaches to down-regulate target genes; (iii) the rescue of protein synthesis/function through the induction of ribosome readthrough targeting nonsense variants or the correction of folding defects caused by amino acid substitutions. Overall, these approaches have shown the ability to rescue the expression and/or function of potentially therapeutic levels of coagulation factors in different disease models, thus supporting further studies in the future aimed at evaluating the clinical translatability of these new strategies.
Collapse
Affiliation(s)
- Dario Balestra
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy.
| | - Alessio Branchini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
88
|
Tsai-Nguyen G, Modrykamien AM, Bredeweg A. Hereditary afibrinogenemia and pulmonary-renal hydralazine-induced vasculitis. Proc (Bayl Univ Med Cent) 2019; 32:397-398. [PMID: 31384199 DOI: 10.1080/08998280.2019.1619397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 10/26/2022] Open
Abstract
Combined pulmonary-renal hydralazine-induced vasculitis is rare, and hereditary afibrinogenemia is also rare. We present a case of a 62-year-old man with a history of hereditary afibrinogenemia who presented with hemoptysis and hematuria. Although he had prior episodes of hemoptysis that resolved with repletion of fibrinogen levels, a hydralazine-induced vasculitis was the ultimate cause of his recurrent hemoptysis and hematuria. Hydralazine was held and after transfusion with cryoprecipitate, he was treated with prednisone and rituximab.
Collapse
Affiliation(s)
- Ginger Tsai-Nguyen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Baylor University Medical CenterDallasTexas
| | - Ariel M Modrykamien
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Baylor University Medical CenterDallasTexas
| | - Arthur Bredeweg
- Department of Pathology, Baylor University Medical CenterDallasTexas
| |
Collapse
|
89
|
Deng J, Li D, Mei H, Tang L, Wang HF, Hu Y. Combined deficiency of factors V and VIII in a Chinese family due to a novel nonsense mutation in lectin mannose binding protein 1. Int J Lab Hematol 2019; 42:e7-e9. [PMID: 31162802 DOI: 10.1111/ijlh.13054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/26/2019] [Accepted: 05/03/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Jun Deng
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Dan Li
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Mei
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Liang Tang
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Hua-Fang Wang
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Yu Hu
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| |
Collapse
|
90
|
Benzon HT, Park M, McCarthy RJ, Kendall MC, Lindholm PF. Mixing Studies in Patients With Prolonged Activated Partial Thromboplastin Time or Prothrombin Time. Anesth Analg 2019; 128:1089-1096. [PMID: 31094773 DOI: 10.1213/ane.0000000000003457] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Patients presenting for surgery may have isolated or combined prolonged activated partial thromboplastin time (aPTT) and/or prothrombin time (PT). In patients not receiving anticoagulants or with no identifiable cause for abnormal clot formation, a mixing study is performed. The index of circulating anticoagulant (ICA) has been used to predict the presence of an inhibitor, usually a lupus anticoagulant. METHODS We retrospectively reviewed the results of mixing studies performed at Northwestern Memorial Hospital, between January 1, 2010 and February 29, 2012. We determined the number of samples that normalized or remained prolonged, the clotting factors associated with prolonged test results, and the presence of coagulation inhibitors. We calculated the ICA in the samples with prolonged aPTT and PT to determine its ability to predict a lupus anticoagulant. The primary comparison of interest was the diagnostic utility of the ICA at cutoff values of 11% for predicting the presence of lupus anticoagulant. RESULTS There were 269 mixing studies performed: 131 samples with prolonged aPTT; 95 with prolonged PT; and 43 with both prolonged aPTT and prolonged PT. Of the samples with a prolonged aPTT, 55 of 131 (42%) normalized, 36 of 131 (27%) partially corrected, and 40 of 131 (31%) remained prolonged. Thirty-three of 95 samples (35%) with prolonged PT normalized, while 62 of 95 (65%) remained prolonged. Eight of 43 (19%) mixing studies of patients with prolonged PT and aPTT normalized; the aPTT normalized, but the PT remained prolonged in 17 of 43 (39%); the PT normalized, but the aPTT remained prolonged in 7 of 43 (16%); and both tests remained prolonged in 11 of 43 (26%) samples. Prolongations in the aPTT were primarily associated with low activities of CF XII, while the majority of the prolongations in PT were secondary to low activities in CF VII. Combined prolongations were secondary to deficiencies in both the intrinsic and extrinsic as well as the common pathways. An ICA >11% had 100% (95% CI, 59%-100%) sensitivity, 53% (95% CI, 35%-70%) specificity, and 77% (95% CI, 62%-92%) accuracy in predicting the presence of lupus anticoagulant in patients with prolonged aPTT. CONCLUSIONS Normalization of the aPTT and PT in a mixing study was associated with low clotting factor activity. The ICA may be helpful in predicting the presence of a lupus anticoagulant. As anesthesiologists take ownership of the perioperative surgical home, we need to understand the clinical implications of the results of mixing studies.
Collapse
Affiliation(s)
- Honorio T Benzon
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Meghan Park
- Department of Anesthesiology, University of Rochester, Rochester, New York
| | - Robert J McCarthy
- Department of Anesthesiology, Rush University Medical Center, Chicago, Illinois
| | - Mark C Kendall
- Department of Anesthesiology, Brown University Warren Alpert School of Medicine, Providence, Rhode Island
| | - Paul F Lindholm
- Department of Pathology (Coagulation Laboratory), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
91
|
Zaidi MH, Stanley A, Khan M. Acquired Factor VII deficiency - a rare but important consideration. Scott Med J 2019; 64:119-122. [PMID: 31142211 DOI: 10.1177/0036933019853167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Isolated acquired Factor VII deficiency is a rare coagulation disorder which is independent of vitamin K deficiency. The exact pathophysiological basis of this condition is unclear. We present a series of cases highlighting different clinical scenarios where this condition was encountered. CASE SERIES The first case presented with intra-abdominal sepsis. The second was a patient admitted with acute kidney injury and subsequently diagnosed with myeloma. The final case presented with microangiopathic haemolytic anaemia and was suspected of having atypical Haemolytic Uraemic Syndrome. In each case, there was no family or personal history of a bleeding disorder. Follow-up Factor VII levels after recovery from illness was normal in all three cases. CONCLUSION Acquired Factor VII deficiency is an uncommon but important finding which should be considered in the general medical setting when an isolated prolonged prothrombin time is detected.
Collapse
Affiliation(s)
| | - Arran Stanley
- 2 Foundation Doctor, Department of Haematology, Aberdeen Royal Infirmary, UK
| | - Mohammed Khan
- 3 Consultant Haematologist, Department of Haematology, Aberdeen Royal Infirmary, UK
| |
Collapse
|
92
|
Manderstedt E, Nilsson R, Lind-Halldén C, Ljung R, Astermark J, Halldén C. Targeted re-sequencing of F8, F9 and VWF: Characterization of Ion Torrent data and clinical implications for mutation screening. PLoS One 2019; 14:e0216179. [PMID: 31026269 PMCID: PMC6485758 DOI: 10.1371/journal.pone.0216179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Mutations are not identified in ~5% of hemophilia A and 10–35% of type 1 VWD patients. The bleeding tendency also varies among patients carrying the same causative mutation, potentially indicating variants in additional genes modifying the phenotype that cannot be identified by routine single-gene analysis. The F8, F9 and VWF genes were analyzed in parallel using an AmpliSeq strategy and Ion Torrent sequencing. Targeting all exonic positions showed an average read depth of >2000X and coverage close to 100% in 24 male patients with known disease-causing mutations. Discrimination between reference alleles and alternative/indel alleles was adequate at a 25% frequency threshold. In F8, F9 and VWF there was an absolute majority of all reference alleles at allele frequencies >95% and the average alternative allele and indel frequencies never reached above 10% and 15%, respectively. In VWF, 4–5 regions showed lower reference allele frequencies; in two regions covered by the pseudogene close to the 25% cut-off for reference alleles. All known mutations, including indels, gross deletions and substitutions, were identified. Additional VWF variants were identified in three hemophilia patients. The presence of additional mutations in 2 out of 16 (12%) randomly selected hemophilia patients indicates a potential mutational contribution that may affect the disease phenotype and counseling in these patients. Parallel identification of disease-causing mutations in all three genes not only confirms the deficiency, but differentiates phenotypic overlaps and allows for correct genetic counseling.
Collapse
Affiliation(s)
- Eric Manderstedt
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
| | - Rosanna Nilsson
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
- * E-mail:
| | - Christina Lind-Halldén
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
| | - Rolf Ljung
- Department of Clinical Sciences–Pediatrics and Malmö Center for Thrombosis and Hemostasis, Skåne University Hospital, Malmö, Sweden
| | - Jan Astermark
- Department for Hematology Oncology and Radiation Physics, Center for Thrombosis and Hemostasis, Skåne University Hospital, Malmö, Sweden
| | - Christer Halldén
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
93
|
Meidert AS, Kinzinger J, Möhnle P, Pekrul I, Spiekermann K, Thorsteinsdottir J, Briegel J, Huge V. Perioperative Management of a Patient with Severe Factor V Deficiency Presenting with Chronic Subdural Hematoma: A Clinical Report. World Neurosurg 2019; 127:409-413. [PMID: 30999086 DOI: 10.1016/j.wneu.2019.04.080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Severe factor V deficiency is an extremely rare coagulation disorder. Patients with factor V activity <5% usually become symptomatic in early childhood. CASE DESCRIPTION We report the case of an 82-year-old woman with incidentally diagnosed severe factor V deficiency, who developed a symptomatic chronic subdural hematoma, requiring burr hole craniostomy. Successful management was achieved by a multidisciplinary approach. Preoperatively, factor V activity was increased from 2% to 50% by administration of 25 mL/kg body weight of fresh frozen plasma over 30 minutes under close cardiopulmonary monitoring in the intensive care unit. Straight afterward, the patient was transferred to the operating room where surgery was performed under general anesthesia. Burr hole craniostomy could be performed without perioperative complications. In the postoperative days, there was no relevant recurrence of the subdural hematoma in the follow-up computed tomography scans under frequent control of coagulation parameters. However, despite further transfusion of fresh frozen plasma, factor V activity did not increase >16%. The patient was discharged without any neurologic deficits. In a hemostaseologic follow-up 2 months after surgery, factor V activity <1% was confirmed with evidence of a factor V inhibitor in the modified Bethesda assay. Most likely, the patient suffered from an acquired form of factor V deficiency with preformed antibodies that had been boosted by the initial treatment with fresh frozen plasma. CONCLUSIONS We conclude that in this rare bleeding disorder, intracranial surgery was successfully managed because of a thoroughly planned perioperative therapeutic strategy. However, if there is time prior to surgery, a full checkup of the bleeding disorder is advisable.
Collapse
Affiliation(s)
- Agnes S Meidert
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany.
| | - Johannes Kinzinger
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany
| | - Patrick Möhnle
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany; Department of Transfusion Medicine, Cellular Therapeutics and Hemostaseology, University Hospital of Munich (LMU), Munich, Germany
| | - Isabell Pekrul
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany; Department of Transfusion Medicine, Cellular Therapeutics and Hemostaseology, University Hospital of Munich (LMU), Munich, Germany
| | - Karsten Spiekermann
- Department of Internal Medicine III (Hematology and Oncology), University Hospital of Munich (LMU), Munich, Germany
| | - Jun Thorsteinsdottir
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| | - Josef Briegel
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany
| | - Volker Huge
- Department of Anaesthesiology, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
94
|
Grottke O, Moser O, Farrag A, Elbracht M, Orlikowsky T, Trepels-Kottek S. Plasma-derived Factor X therapy for treatment of intracranial bleeding in a patient with Factor X deficiency: a case report. Transfusion 2019; 59:2228-2233. [PMID: 30964547 DOI: 10.1111/trf.15308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/11/2019] [Accepted: 03/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Factor X (FX) deficiency (FXD) is an extremely rare autosomal recessive hereditary hematologic disorder, affecting approximately one in 1,000,000 of the general population. CASE REPORT This case report describes an infant with hereditary severe FXD who presented with a spontaneous, life-threatening intracranial hemorrhage and was treated with the first licensed plasma-derived FX (pdFX) concentrate. On admission, laboratory assays showed severe coagulopathy of unknown cause; the patient was empirically treated using a multimodal hemostatic approach with prothrombin complex concentrate, fresh-frozen plasma, and tranexamic acid. Subsequent single-factor coagulation and genetic analyses confirmed the hereditary FXD diagnosis, and the therapeutic regimen was changed to a targeted regimen of 250 IU pdFX daily. Based on careful monitoring of the coagulation profile, pdFX administration frequency was increased to twice daily, followed by a reduction to once every 18 hours. The patient was discharged after 7 weeks of hospitalization in good clinical condition and now receives prophylactic pdFX three times weekly.
Collapse
Affiliation(s)
- Oliver Grottke
- Department of Anaesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Olga Moser
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | - Ahmed Farrag
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany.,Pediatric Oncology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Miriam Elbracht
- Institute of Human Genetics, RWTH Aachen University Hospital, Aachen, Germany
| | - Thorsten Orlikowsky
- Section of Neonatology and Pediatric Intensive Care, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | - Sonja Trepels-Kottek
- Section of Neonatology and Pediatric Intensive Care, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
95
|
Djambas Khayat C, El Khorassani M, Lambert T, Gay V, Barthez-Toullec M, Lamazure J, Bellon A, Henriet C, Bridey F, Négrier C. Clinical pharmacology, efficacy and safety study of a triple-secured fibrinogen concentrate in adults and adolescent patients with congenital fibrinogen deficiency. J Thromb Haemost 2019; 17:635-644. [PMID: 30661302 DOI: 10.1111/jth.14392] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Indexed: 11/29/2022]
Abstract
Essentials A novel fibrinogen concentrate was evaluated in patients with congenital fibrinogen deficiency. An open-label, phase 2-3 trial studied pharmacology, efficacy, and safety in patients >6 years. The product offers safe and effective therapy in the treatment and prophylaxis of bleeding. Data in recovery show the need of adjusted treatment and further investigation in children. SUMMARY: Background Single-factor replacement therapy is considered the most suitable treatment option for hereditary fibrinogen deficiency. A triple-secured plasma-derived human fibrinogen product was developed to increase the safety of the former fibrinogen concentrate. Objectives This non-randomized, open-label, prospective study investigated pharmacokinetics, efficacy, and safety of a novel fibrinogen concentrate (FibCLOT® /CLOTTAFACT® LFB, France) in inherited deficiency. Patients/Methods Fourteen patients ≥40 kg received fibrinogen concentrate for pharmacology and 16 ≥ 23 kg received treatment for bleeding or surgery. Each treatment was followed by a 3-week safety observation period. Key outcomes included number of infusions, dose, bleeding control, daily assessment, hemoglobin, blood loss, transfusions, and physicians' global assessment of response. Results Incremental recovery was 2.35 mg mL-1 per mg kg-1 and maximal concentration 1.41 g L-1 (geometric mean) after 0.060 g kg-1 infusion in 14 afibrinogenemic patients. Terminal half-life was 69.3 h (non-compartmental analysis). The maximum clot firmness was increased by a mean of 10.3 mm from baseline to maximal effect. Sixteen patients participated to the efficacy phase: 32 bleeding episodes were treated in 9 patients, and 15 patients underwent 38 surgical/invasive procedures. All patients achieved appropriate hemostasis: response to treatment was successful in all bleeds (95% CI, 0.89-1.00) and procedures (95% CI, 0.91-1.00). Most (94%) bleeds were controlled with a single infusion (median 0.050 g kg-1 ). Two patients experienced asymptomatic distal venous thromboses identified by systematic ultrasound. Conclusion FibCLOT® /CLOTTAFACT® showed a pharmacokinetic profile comparable to that of other fibrinogen concentrates and provides safe and clinically effective substitution therapy for fibrinogen-deficient patients.
Collapse
Affiliation(s)
| | | | | | | | - Malika Barthez-Toullec
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Jennifer Lamazure
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Anne Bellon
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Céline Henriet
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Françoise Bridey
- Clinical Development, Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France
| | - Claude Négrier
- Hematology Division, Hemophilia Comprehensive Care Center, Louis Pradel Hospital, University Lyon1, Bron, France
| |
Collapse
|
96
|
Anzengruber J, Feichtinger M, Bärnthaler P, Haider N, Ilas J, Pruckner N, Benamara K, Scheiflinger F, Reipert BM, Malisauskas M. How Full-Length FVIII Benefits from Its Heterogeneity - Insights into the Role of the B-Domain. Pharm Res 2019; 36:77. [PMID: 30937539 PMCID: PMC6443606 DOI: 10.1007/s11095-019-2599-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/27/2019] [Indexed: 11/19/2022]
Abstract
Purpose To explore how the natural heterogeneity of human coagulation factor VIII (FVIII) and the processing of its B-domain specifically modulate protein aggregation. Methods Recombinant FVIII (rFVIII) molecular species containing 70% or 20% B-domain, and B-domain-deleted rFVIII (BDD-rFVIII), were separated from full-length recombinant FVIII (FL-rFVIII). Purified human plasma-derived FVIII (pdFVIII) was used as a comparator. Heterogeneity and aggregation of the various rFVIII molecular species, FL-rFVIII and pdFVIII were analysed by SDS-PAGE, dynamic light scattering, high-performance size-exclusion chromatography and flow cytometry-based particle analysis. Results FL-rFVIII and pdFVIII were heterogeneous in nature and demonstrated similar resistance to aggregation under physical stress. Differences were observed between these and among rFVIII molecular species. FVIII molecular species exhibited diverging aggregation pathways dependent on B-domain content. The propensity to form aggregates increased with decreasing proportions of B-domain, whereas the opposite was observed for oligomer formation. Development of cross-β sheet-containing aggregates in BDD-rFVIII induced effective homologous seeding and faster aggregation. Naturally heterogeneous FL-rFVIII and pdFVIII displayed the lowest propensity to aggregate in all experiments. Conclusions These results demonstrate that pdFVIII and FL-rFVIII have similar levels of molecular heterogeneity, and suggest that heterogeneity and the B-domain are involved in stabilising FVIII by modulating its aggregation pathway. Electronic supplementary material The online version of this article (10.1007/s11095-019-2599-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Anzengruber
- Research & Development, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria.
| | - Martin Feichtinger
- Technical Operations, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Philipp Bärnthaler
- Technical Operations, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Norbert Haider
- Technical Operations, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Josenato Ilas
- Research & Development, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Nina Pruckner
- Technical Operations, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Karima Benamara
- Research & Development, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | | | - Birgit M Reipert
- Research & Development, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Mantas Malisauskas
- Research & Development, Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| |
Collapse
|
97
|
Understanding the Impact of Aberrant Splicing in Coagulation Factor V Deficiency. Int J Mol Sci 2019; 20:ijms20040910. [PMID: 30791524 PMCID: PMC6412230 DOI: 10.3390/ijms20040910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 11/23/2022] Open
Abstract
Rare inherited coagulation disorders (RICDs) are congenital deficiencies of the plasma proteins that are involved in blood coagulation, which generally lead to lifelong bleeding manifestations. These diseases are generally qualitative and/or quantitative defects that are associated with monoallelic or biallelic mutations in the relevant gene. Among RICDs, factor V (FV) deficiency is one of the least characterized at the molecular level. Here, we investigated four unrelated patients with reduced plasma FV levels (three severe, one mild), which were associated with a moderately severe bleeding tendency. Sequence analysis of the FV gene identified seven different variants, five hitherto unknown (p.D1669G, c.5789-11C>A, c.5789-12C>A, c.5789-5T>G, and c.6528G>C), and two previously reported (c.158+1G>A and c.5789G>A). The possible pathogenic role of the newly identified missense variant was studied by in silico approaches. The remaining six genetic defects (all putative splicing mutations) were investigated for their possible effects on pre-mRNA splicing by transient transfection experiments in HeLa cells with plasmids expressing appropriate hybrid minigenes. The preparation of minigene constructs was instrumental to demonstrate that the two adjacent variants c.5789-11C>A and c.5789-12C>A are indeed present in cis in the analyzed FV-deficient patient (thus leading to the c.5789-11_12CC>AA mutation). Ex vivo experiments demonstrated that each variant causes either a skipping of the relevant exon or the activation of cryptic splice sites (exonic or intronic), eventually leading to the introduction of a premature termination codon.
Collapse
|
98
|
Tomaszewski M, Bienz M, Kherad O, Restellini S, Laflèche T, Barkun A, Warner M, Bessissow T. Low endoscopy bleeding risk in patients with congenital bleeding disorders. Haemophilia 2019; 25:289-295. [PMID: 30748066 DOI: 10.1111/hae.13691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/06/2018] [Accepted: 01/17/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Haemophilia A and haemophilia B, von Willebrand disease (VWD), factor VII deficiency and factor XI deficiency are congenital bleeding disorders predisposing to bleeding during invasive procedures. The ageing population of people with congenital bleeding disorders will likely increasingly require gastrointestinal endoscopy. The bleeding risk postgastrointestinal endoscopy and optimal prophylactic treatment regimens are not well described. METHODS We performed a retrospective chart review at the McGill University Health Centre. Adult patients with haemophilia A or B, VWD, FVII deficiency and FXI deficiency who underwent gastrointestinal endoscopic procedures were included. Bleeding prophylaxis included combinations of plasma-derived factor (VWD) or recombinant factor (haemophilia A and haemophilia B), desmopressin and/or tranexamic acid. Our primary outcome was the 72-hour postendoscopy bleeding rate. RESULTS One hundred and four endoscopies were performed in 48 patients. Haemophilia A (45.3% of endoscopies) was the most common bleeding disorder, followed by VWD (38.5%), FXI deficiency (8.7%), haemophilia B (4.8%) and FVII deficiency (2.9%). All patients were reviewed by the Haemophilia Treatment Center with peri-procedure treatment protocols put in place as required. The overall 72-hour bleeding rate was 0.96%, confidence interval (CI) 95% (0.17%-5.25%). The colonoscopic postpolypectomy bleeding rate was 1/21 (4.8%, CI 95% (0.9%-22.7%)) in comparison with the general population rate of 0.3%-10% for high-risk endoscopy (including colonoscopic polypectomy). CONCLUSION To the best of our knowledge, this is the largest study describing patients with inherited bleeding disorders undergoing gastrointestinal endoscopy. The bleeding risk is not significantly higher to the general population when haemostatically managed by a team experienced in bleeding disorders.
Collapse
Affiliation(s)
- Marcel Tomaszewski
- Internal Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marc Bienz
- Internal Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Omar Kherad
- Internal Medicine, Hôpital de la Tour and University of Geneva, Meyrin, Switzerland
| | - Sophie Restellini
- Service de gastroentérologie et d'hépatologie, Département des spécialités de Médecine, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Tania Laflèche
- Division of Hematology, McGill University Health Centre, Quebec, Canada
| | - Alan Barkun
- Division of Gastroenterology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Margaret Warner
- Division of Hematology, McGill University Health Centre, Quebec, Canada
| | - Talat Bessissow
- Division of Gastroenterology, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
99
|
Genotype analysis and identification of novel mutations in a multicentre cohort of patients with hereditary factor X deficiency. Blood Coagul Fibrinolysis 2019; 30:34-41. [DOI: 10.1097/mbc.0000000000000787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
100
|
Abdeltawab K, Yagan J, Megahed M, Zahab MA, Gheith OA, Rida S, Nair P, Mahmoud T, Maher AM, Al-Mohareb S, Mohamed M, Al-Otaibi T. Kidney Transplant in a Patient With Factor VII Deficiency: Case Report. EXP CLIN TRANSPLANT 2019; 17:142-144. [PMID: 30777540 DOI: 10.6002/ect.mesot2018.p9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Organ transplant in patients with congenital bleeding disorders is a challenge requiring an integrated approach of various specialists. Inherited factor VII deficiency is the most common of the rare bleeding disorders, with a wide set of hemorrhagic features. Although a kidney allograft is the most frequent type of solid-organ transplant, it is rarely performed in individuals with congenital hemorrhagic disorders. Here, we highlight the course of a patient with coagulation factor VII deficiency who underwent successful kidney transplant without significant coagulopathy. Our patient was a 19-year-old man with end-stage kidney disease and congenital coagulation factor VII deficiency. Perioperative bleeding was successfully prevented by administration of recombinant factor VII, confirming its safety in solid-organ transplants. Success requires evaluation of doses and therapeutic schedules, as well as a multidisciplinary approach.
Collapse
Affiliation(s)
- Khaled Abdeltawab
- From the Nephrology Department, Hamed Al-Essa Organ Transplant Center, Sabah Area, Kuwait
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|