Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Thromb Res ; 231: 99-111, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37839151

RESUMEN

Congenital coagulopathies have, throughout the history of medicine, been a focus of scientific study and of great interest as they constitute an alteration of one of the most important and conserved pathways of evolution. The first therapeutic strategies developed to address them were aimed at restoring the blood components lost during hemorrhage by administering whole blood or plasma. Later on, the use of cryoprecipitates was a significant breakthrough as it made it possible to decrease the volumes of blood infused. In the 1970' and 80', clotting factor concentrates became the treatment and, from the 1990's to the present day, recombinant factors -with increasingly longer half-lives- have taken over as the treatment of choice for certain coagulopathies in a seamless yet momentous transition from biological to biotechnological drugs. The beginning of this century, however, saw the emergence of new advanced (gene and cell) treatments, which are currently transforming the therapeutic landscape. The possibility to use cells and viruses as well as specific or bispecific antibodies as medicines is likely to spark a revolution in the world of pharmacology where therapies will be individualized and have long-term effects. Specifically, attention is nowadays focused on the development of gene editing strategies, chiefly those based on CRISPR/Cas technology. Rare coagulopathies such as hemophilia A and B, or even ultra-rare ones such as factor V deficiency, could be among those deriving the greatest benefit from these new developments.


Asunto(s)
Productos Biológicos , Hemofilia A , Hemofilia B , Humanos , Hemofilia B/genética , Edición Génica , Sistemas CRISPR-Cas , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Productos Biológicos/uso terapéutico
2.
Int J Mol Sci ; 24(7)2023 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-37047215

RESUMEN

Proteostasis, i.e., the homeostasis of proteins, responsible for ensuring protein turnover, is regulated by proteases, which also participate in the etiopathogenesis of multiple conditions. The magic of proteases is such that, in blood coagulation, one same molecule, such as coagulation factor V, for example, can perform both a procoagulant and an anticoagulant function as a result of the activity of proteases. However, this magic has an insidious side to it, as it may also prevent the completion of the clinical value chain of factor V deficiency. This value chain encompasses the discovery of knowledge, the transfer of this knowledge, and its translation to clinical practice. In the case of rare and ultra-rare diseases like factor V deficiency, this value chain has not been completed as the knowledge acquisition phase has dragged out over time, holding up the transfer of knowledge to clinical practice. The reason for this is related to the small number of patients afflicted with these conditions. As a result, new indications must be found to make the therapies cost-effective. In the case of factor V, significant research efforts have been directed at developing a recombinant factor V capable of resisting the action of the proteases capable of inactivating this factor. This is where bioethics and health equity considerations come into the equation.


Asunto(s)
Deficiencia del Factor V , Factor V , Humanos , Factor V/genética , Factor V/metabolismo , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Péptido Hidrolasas/farmacología , Coagulación Sanguínea , Endopeptidasas/farmacología
3.
Haematologica ; 108(6): 1544-1554, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36700401

RESUMEN

Hemophilia A (HA) cell therapy approaches in pediatric individuals require suitable factor (F)VIII-producing cells for stable engraftment. Liver sinusoidal endothelial cells (LSEC) and hematopoietic stem cells (HSC) have been demonstrated to be suitable for the treatment of adult HA mice. However, after transplantation in busulfan (BU)-conditioned newborn mice, adult LSEC/HSC cannot efficiently engraft, while murine fetal liver (FL) hemato/vascular cells from embryonic day 11-13 of gestation (E11-E13), strongly engraft the hematopoietic and endothelial compartments while also secreting FVIII. Our aim was to investigate the engraftment of FL cells in newborn HA mice to obtain a suitable "proof of concept" for the development of a new HA treatment in neonates. Hence, we transplanted FL E11 or E13 cells and adult bone marrow (BM) cells into newborn HA mice with or without BU preconditioning. Engraftment levels and FVIII activity were assessed starting from 6 weeks after transplantation. FL E11-E13+ BU transplanted newborns reached up to 95% engraftment with stable FVIII activity levels observed for 16 months. FL E13 cells showed engraftment ability even in the absence of BU preconditioning, while FL E11 cells did not. BM BU transplanted newborn HA mice showed high levels of engraftment; nevertheless, in contrast to FL cells, BM cells cannot engraft HA newborns in BU non-conditioning regimen. Finally, none of the transplanted mice developed anti-FVIII antibodies. Overall, this study sheds some light on the therapeutic potential of healthy FL cells in the cure of HA neonatal/pediatric patients.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Hemofilia A , Ratones , Animales , Hemofilia A/terapia , Células Endoteliales , Hígado , Células Madre Hematopoyéticas , Trasplante de Células Madre , Busulfano , Ratones Endogámicos C57BL
4.
Int J Mol Sci ; 23(15)2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-35955419

RESUMEN

The vascular endothelium has several important functions, including hemostasis. The homeostasis of hemostasis is based on a fine balance between procoagulant and anticoagulant proteins and between fibrinolytic and antifibrinolytic ones. Coagulopathies are characterized by a mutation-induced alteration of the function of certain coagulation factors or by a disturbed balance between the mechanisms responsible for regulating coagulation. Homeostatic therapies consist in replacement and nonreplacement treatments or in the administration of antifibrinolytic agents. Rebalancing products reestablish hemostasis by inhibiting natural anticoagulant pathways. These agents include monoclonal antibodies, such as concizumab and marstacimab, which target the tissue factor pathway inhibitor; interfering RNA therapies, such as fitusiran, which targets antithrombin III; and protease inhibitors, such as serpinPC, which targets active protein C. In cases of thrombophilia (deficiency of protein C, protein S, or factor V Leiden), treatment may consist in direct oral anticoagulants, replacement therapy (plasma or recombinant ADAMTS13) in cases of a congenital deficiency of ADAMTS13, or immunomodulators (prednisone) if the thrombophilia is autoimmune. Monoclonal-antibody-based anti-vWF immunotherapy (caplacizumab) is used in the context of severe thrombophilia, regardless of the cause of the disorder. In cases of disseminated intravascular coagulation, the treatment of choice consists in administration of antifibrinolytics, all-trans-retinoic acid, and recombinant soluble human thrombomodulin.


Asunto(s)
Factor V/metabolismo , Trombofilia , Factor de von Willebrand , Anticoagulantes , Endotelio Vascular/metabolismo , Factor VIII/genética , Factor VIII/uso terapéutico , Homeostasis , Humanos , Proteína C/uso terapéutico , Trombofilia/genética , Factor de von Willebrand/metabolismo
5.
Int J Mol Sci ; 23(10)2022 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-35628611

RESUMEN

Factor V deficiency, an ultra-rare congenital coagulopathy, is characterized by bleeding episodes that may be more or less intense as a function of the levels of coagulation factor activity present in plasma. Fresh-frozen plasma, often used to treat patients with factor V deficiency, is a scarcely effective palliative therapy with no specificity to the disease. CRISPR/Cas9-mediated gene editing, following precise deletion by non-homologous end-joining, has proven to be highly effective for modeling on a HepG2 cell line a mutation similar to the one detected in the factor V-deficient patient analyzed in this study, thus simulating the pathological phenotype. Additional CRISPR/Cas9-driven non-homologous end-joining precision deletion steps allowed correction of 41% of the factor V gene mutated cells, giving rise to a newly developed functional protein. Taking into account the plasma concentrations corresponding to the different levels of severity of factor V deficiency, it may be argued that the correction achieved in this study could, in ideal conditions, be sufficient to turn a severe phenotype into a mild or asymptomatic one.


Asunto(s)
Deficiencia del Factor V , Factor V , Sistemas CRISPR-Cas/genética , Factor V/genética , Deficiencia del Factor V/genética , Edición Génica , Humanos , Mutación
6.
Front Vet Sci ; 9: 846216, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35419447

RESUMEN

Factor V together with activated factor X forms the prothrombinase complex, which transforms prothrombin into thrombin. The Mus musculus species is characterized by very high levels of this factor and short clotting times, which hinders accurate measurements. For that reason, a detailed characterization of such parameters is indispensable. A method was designed as part of this study to provide an accurate determination and standardization of factor V levels, prothrombin time and activated partial thromboplastin time in Mus musculus. Those parameters were evaluated in a sample of 66 healthy animals using a semi-automated coagulometer and human diagnostic reagents in an attempt to determine the most appropriate time of day for the extractions. A mouse-based protocol was designed, capable of making corrections to the samples at dilutions of 1:100 for factor V and at of 1:3 for prothrombin time. The goal was to smoothen the calibration curves, which often present with steep slopes and narrow measurement ranges between one calibration point and another. It was found that the most stable period for blood sample extraction was that comprised between the first 6 h of light. No clinical differences were observed between the sexes and reference intervals were established for factor V (95.80% ± 18.14; 25.21 s ± 1.34), prothrombin time (104.31% ± 14.52; 16.85 s ± 1.32) and activated partial thromboplastin time (32.86 s ± 3.01). The results obtained are applicable to human or veterinary biomedical research, to transfusional medicine or to pathological models for diseases such as factor V deficiency.

7.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34575869

RESUMEN

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.


Asunto(s)
Análisis Mutacional de ADN , Deficiencia del Factor V/genética , Deficiencia del Factor V/terapia , Factor V/genética , Adolescente , Coagulación Sanguínea , Trastornos de la Coagulación Sanguínea Heredados/genética , Pruebas de Coagulación Sanguínea , Plaquetas/metabolismo , Preescolar , Codón sin Sentido , ADN Complementario/metabolismo , Salud de la Familia , Femenino , Mutación del Sistema de Lectura , Humanos , Masculino , Pakistán , Proteínas Recombinantes/química , Análisis de Secuencia de ADN , España
8.
Biomed Pharmacother ; 142: 112059, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34467894

RESUMEN

Deficiency of factor V is a congenital autosomal recessive coagulopathy associated with mutations in the F5 gene that results in mild-to-severe bleeding episodes. Factor V is a component of the prothrombinase complex responsible for accelerating conversion of prothrombin to thrombin. At the present time there are no therapeutic factor V concentrates available. This study was designed to lay the preliminary foundations for future cell-based therapy for patients with severe factor V deficiency. The study showed that hepatospheres, which produce coagulation factors VIII, IX, and V, synthetize and store intracellular glycogen and express albumin levels up to 8 times higher than those of undifferentiated cells. Factor IX and factor V gene expression increased significantly in hepatospheres as compared to undifferentiated cells, whereas factor VIII gene expression remained constant. The factor V protein was detected in the hepatospheres´ secretome. Considering the enormous potential of mesenchymal stem cells as therapeutic agents, this study proposes a highly reproducible method to induce differentiation of mesenchymal stem cells from human placenta to factor V-producing hepatospheres. This strategy constitutes a preliminary step towards a curative treatment of factor V deficiency through advanced therapies such as cell therapy.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Decidua/citología , Deficiencia del Factor V/terapia , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Albúminas/genética , Albúminas/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Factor IX/genética , Factor IX/metabolismo , Factor V/genética , Factor V/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Secretoma/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
9.
Int J Mol Sci ; 22(14)2021 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-34299267

RESUMEN

Hemophilia is a monogenic mutational disease affecting coagulation factor VIII or factor IX genes. The palliative treatment of choice is based on the use of safe and effective recombinant clotting factors. Advanced therapies will be curative, ensuring stable and durable concentrations of the defective circulating factor. Results have so far been encouraging in terms of levels and times of expression using mainly adeno-associated vectors. However, these therapies are associated with immunogenicity and hepatotoxicity. Optimizing the vector serotypes and the transgene (variants) will boost clotting efficacy, thus increasing the viability of these protocols. It is essential that both physicians and patients be informed about the potential benefits and risks of the new therapies, and a register of gene therapy patients be kept with information of the efficacy and long-term adverse events associated with the treatments administered. In the context of hemophilia, gene therapy may result in (particularly indirect) cost savings and in a more equitable allocation of treatments. In the case of hemophilia A, further research is needed into how to effectively package the large factor VIII gene into the vector; and in the case of hemophilia B, the priority should be to optimize both the vector serotype, reducing its immunogenicity and hepatotoxicity, and the transgene, boosting its clotting efficacy so as to minimize the amount of vector administered and decrease the incidence of adverse events without compromising the efficacy of the protein expressed.


Asunto(s)
Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Animales , Factor IX/genética , Factor IX/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patología , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patología , Humanos
10.
BMJ Case Rep ; 12(4)2019 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-30962210

RESUMEN

Haemophilia is a hereditary X-linked recessive disorder caused by a deficiency of either clotting factor VIII (haemophilia A) or IX (haemophilia B). Conventional treatment is currently based on the use of either plasma derived or recombinant coagulation factors. This paper reports on the case of a patient with severe haemophilia who presented with mesial decay and interproximal tartar build-up, for which extraction and scaling to remove tartar deposits were indicated. Following extraction, the usual haemostasis techniques were applied, and postoperative prophylactic antihaemophilic treatment was indicated for 2 or 3 days. The patient presented with moderate bleeding for a few minutes immediately after the procedure. Administration of factor VIII before surgery as well as the patient's favourable pharmacokinetic response allowed for an optimal result. This treatment has afforded patients with haemophilia a better quality of life, and safe and efficient access to invasive surgical procedures.


Asunto(s)
Factor VIII/administración & dosificación , Hemofilia A/tratamiento farmacológico , Cuidados Preoperatorios/métodos , Extracción Dental/métodos , Factor IX , Factor VIII/farmacocinética , Hemofilia A/complicaciones , Hemorragia/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Cuidados Posoperatorios/métodos , Proteínas Recombinantes/uso terapéutico , Extracción Dental/efectos adversos
11.
Thromb Haemost ; 118(8): 1370-1381, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29991091

RESUMEN

The development of new strategies based on cell therapy approaches to correct haemophilia A (HA) requires further insights into new cell populations capable of producing coagulation factor VIII (FVIII) and presenting stable engraftment potential. The major producers of FVIII in the adult are liver sinusoidal endothelial cells (LSECs) and in a lesser degree bone marrow-derived cells, both of which have been shown to ameliorate the bleeding phenotype in adult HA mice after transplantation. We have previously shown that cells from the foetal liver (FL) and the aorta-gonads-mesonephros (AGM) haematopoietic locations possess higher LSEC engraftment potential in newborn mice compared with adult-derived LSECs, constituting likely therapeutic targets for the treatment of HA in neonates. However, less is known about the production of FVIII in embryonic locations. Quantitative polymerase chain reaction and Western blot analysis were performed to assess the relative level of FVIII production in different embryonic tissues and at various developmental stages, identifying the FL and AGM region from day 12 (E12) as prominent sources of FVIII. Furthermore, FL-derived VE-cad+CD45-Lyve1+/- endothelial/endothelial progenitor cells, presenting vascular engraftment potential, produced high levels of F8 ribonucleic acid compared with CD45+ blood progenitors or Dlk1+ hepatoblasts. In addition, we show that the E11 AGM explant cultures expanded cells with LSEC repopulation activity, instrumental to further understand signals for in vitro generation of LSECs. Taking into account the capacity for FVIII expression, culture expansion and newborn engraftment potential, these results support the use of cells with foetal characteristics for correction of FVIII deficiency in young individuals.


Asunto(s)
Aorta/metabolismo , Células Progenitoras Endoteliales/metabolismo , Factor VIII/metabolismo , Gónadas/metabolismo , Hemofilia A/metabolismo , Hígado/metabolismo , Mesonefro/metabolismo , Animales , Aorta/embriología , Aorta/trasplante , Diferenciación Celular , Células Progenitoras Endoteliales/trasplante , Factor VIII/genética , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Gónadas/embriología , Gónadas/trasplante , Hemofilia A/genética , Hemofilia A/cirugía , Hígado/embriología , Mesonefro/embriología , Mesonefro/trasplante , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trasplante de Células Madre/métodos , Técnicas de Cultivo de Tejidos
12.
PeerJ ; 4: e1907, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27114871

RESUMEN

Background. Hemophilia is a rare recessive X-linked disease characterized by a deficiency of coagulation factor VIII or factor IX. Its current treatment is merely palliative. Advanced therapies are likely to become the treatment of choice for the disease as they could provide a curative treatment. Methods. The present study looks into the use of a safe non-viral transfection method based on nucleofection to express and secrete human clotting factor IX (hFIX) where human adipose tissue derived mesenchymal stem cells were used as target cells in vitro studies and NOD. Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice were used to analyze factor IX expression in vivo studies. Previously, acute liver injury was induced by an injected intraperitoneal dose of 500 mg/kg body weight of acetaminophen. Results. Nucleofection showed a percentage of positive cells ranging between 30.7% and 41.9% and a cell viability rate of 29.8%, and cells were shown to secrete amounts of hFIX between 36.8 and 71.9 ng/mL. hFIX levels in the blood of NSG mice injected with ASCs transfected with this vector, were 2.7 ng/mL 48 h after injection. Expression and secretion of hFIX were achieved both in vitro cell culture media and in vivo in the plasma of mice treated with the transfected ASCs. Such cells are capable of eventually migrating to a previously damaged target tissue (the liver) where they secrete hFIX, releasing it to the bloodstream over a period of at least five days from administration. Conclusions. The results obtained in the present study may form a preliminary basis for the establishment of a future ex vivo non-viral gene/cellular safe therapy protocol that may eventually contribute to advancing the treatment of hemophilia.

13.
Expert Opin Biol Ther ; 15(5): 713-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25826280

RESUMEN

INTRODUCTION: In hemophilia, advanced therapies are warranted from a conceptual and methodological standpoint. Current advanced therapy strategies are centered on the use of adeno-associated viral vectors, although problems related to immunogenicity and hepatotoxicity still remain. AREAS COVERED: Future clinical trials will have to scrupulously observe international bioethical standards in terms of patient selection, particularly children. Patient recruitment rates are likely to remain low due to the stringent exclusion criteria usually imposed on the trial population regarding their hepatic and immunological markers and the presence of viral coinfection; and to the existence of an optimal palliative treatment. EXPERT OPINION: Accordingly, the results obtained are likely to be of low statistical significance, which could hinder their application to clinical practice. Another important issue is the degree to which society embraces these new emerging therapies. The unfamiliarity of society with these new methods, together with the many unresolved questions about them that remain, may threaten their acceptance not only by society at large but also by health-care professionals, which would limit their translational application to clinical practice.


Asunto(s)
Terapia Biológica/ética , Hemofilia A/genética , Hemofilia A/terapia , Selección de Paciente/ética , Distancia Psicológica , Terapia Biológica/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/ética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/ética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Cuidados Paliativos/ética , Cuidados Paliativos/métodos
15.
Orphanet J Rare Dis ; 7: 97, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23237078

RESUMEN

Monogenic diseases are ideal candidates for treatment by the emerging advanced therapies, which are capable of correcting alterations in protein expression that result from genetic mutation. In hemophilia A and B such alterations affect the activity of coagulation factors VIII and IX, respectively, and are responsible for the development of the disease. Advanced therapies may involve the replacement of a deficient gene by a healthy gene so that it generates a certain functional, structural or transport protein (gene therapy); the incorporation of a full array of healthy genes and proteins through perfusion or transplantation of healthy cells (cell therapy); or tissue transplantation and formation of healthy organs (tissue engineering). For their part, induced pluripotent stem cells have recently been shown to also play a significant role in the fields of cell therapy and tissue engineering. Hemophilia is optimally suited for advanced therapies owing to the fact that, as a monogenic condition, it does not require very high expression levels of a coagulation factor to reach moderate disease status. As a result, significant progress has been possible with respect to these kinds of strategies, especially in the fields of gene therapy (by using viral and non-viral vectors) and cell therapy (by means of several types of target cells). Thus, although still considered a rare disorder, hemophilia is now recognized as a condition amenable to gene therapy, which can be administered in the form of lentiviral and adeno-associated vectors applied to adult stem cells, autologous fibroblasts, platelets and hematopoietic stem cells; by means of non-viral vectors; or through the repair of mutations by chimeric oligonucleotides. In hemophilia, cell therapy approaches have been based mainly on transplantation of healthy cells (adult stem cells or induced pluripotent cell-derived progenitor cells) in order to restore alterations in coagulation factor expression.


Asunto(s)
Hemofilia A/terapia , Terapia Genética , Hemofilia B/terapia , Humanos
17.
J Transl Med ; 8: 131, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21143967

RESUMEN

There is much to be investigated about the specific characteristics of stem cells and about the efficacy and safety of the new drugs based on this type of cells, both embryonic as adult stem cells, for several therapeutic indications (cardiovascular and ischemic diseases, diabetes, hematopoietic diseases, liver diseases). Along with recent progress in transference of nuclei from human somatic cells, as well as iPSC technology, has allowed availability of lineages of all three germ layers genetically identical to those of the donor patient, which permits safe transplantation of organ-tissue-specific adult stem cells with no immune rejection. The main objective is the need for expansion of stem cell characteristics to maximize stem cell efficacy (i.e. the proper selection of a stem cell) and the efficacy (maximum effect) and safety of stem cell derived drugs. Other considerations to take into account in cell therapy will be the suitability of infrastructure and technical staff, biomaterials, production costs, biobanks, biosecurity, and the biotechnological industry. The general objectives in the area of stem cell research in the next few years, are related to identification of therapeutic targets and potential therapeutic tests, studies of cell differentiation and physiological mechanisms, culture conditions of pluripotent stem cells and efficacy and safety tests for stem cell-based drugs or procedures to be performed in both animal and human models in the corresponding clinical trials. A regulatory framework will be required to ensure patient accessibility to products and governmental assistance for their regulation and control. Bioethical aspects will be required related to the scientific and therapeutic relevance and cost of cryopreservation over time, but specially with respect to embryos which may ultimately be used for scientific uses of research as source of embryonic stem cells, in which case the bioethical conflict may be further aggravated.


Asunto(s)
Discusiones Bioéticas , Regulación Gubernamental , Investigación con Células Madre/ética , Investigación con Células Madre/legislación & jurisprudencia , Trasplante de Células Madre/ética , Trasplante de Células Madre/legislación & jurisprudencia , Células Madre/citología , Biotecnología , Unión Europea , Prueba de Histocompatibilidad , Humanos , Investigación con Células Madre/economía , Trasplante de Células Madre/tendencias , Bancos de Tejidos , Estados Unidos , United States Food and Drug Administration
18.
Int Arch Med ; 3: 10, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20540744

RESUMEN

The University is tasked with drawing together, transmitting and maintaining knowledge, while creating an area where the ethical "sense" required for working in the field of Biology and Biomedicine can be provided. Although scientific knowledge is present on an overwhelming scale in nature and, therefore, its discovery is unceasing, this does not mean that, as a human being, the researcher has no limitations. It is Bioethics that sets this limit. The successful spreading of knowledge, therefore, which is proclaimed with the creation of a Global Higher Education Area, should also pursue the establishment of the bioethical principles necessary for the credibility of science and its progress so that the society that it promotes and sustains becomes a reality.

19.
Blood Coagul Fibrinolysis ; 19(5): 333-40, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18600079

RESUMEN

The period between isolation of HIV in the early 1980s and the development of effective viral inactivation procedures able to eradicate the virus from the blood supply was long and unfortunately many recipients of blood-derived products became infected; this translated into a devastating impact on their quality of life, quality of care as well as on their life expectancy. Some years later, hepatitis C virus infection was identified as another known blood-borne disease complicating the treatment of haemophilia. Nowadays, the potential threat of emerging new pathogens has stressed the need to provide effective but primarily safe products with regard to infectious agents, as well as to regularly update therapeutic guidelines for haemophilia. The aim of the present publication was to review some of the crucial aspects related to the choice of haemostatic concentrates for the treatment of haemophilia and other inherited bleeding disorders, to analyse the current situation in the United States, Canada and European Union countries and to report the most relevant aspects of the Spanish consensus opinion of haemophilia-treating doctors for the use of therapeutic products for haemophilia recently issued. Essentially, it suggests that a gradual switch to recombinant concentrates may be a beneficial decision for patients with haemophilia and for the National Health Service.


Asunto(s)
Transfusión de Componentes Sanguíneos , Infecciones por VIH/prevención & control , Hemofilia A/terapia , Hepatitis C/prevención & control , Inactivación de Virus , Patógenos Transmitidos por la Sangre , VIH , Infecciones por VIH/transmisión , Hepatitis C/transmisión , Guías de Práctica Clínica como Asunto , Calidad de la Atención de Salud , Calidad de Vida , España
20.
Int Arch Med ; 1(1): 9, 2008 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-18573217

RESUMEN

It has been long since French physician Jean-Baptiste Denys carried out the first successful blood transfusion to a human being. Using bird feathers as canules, sheep blood was transfused to a young man. The patient died soon after Denys' treatment and Denys was accused of murder. In the XXI century, known as the biotechnology century, we face new challenges in Medicine. New emerging and reemerging diseases, such as Creutzfeldt-Jakob disease (CJD) or "mad cow disease" and its human variant (vCJD), challenge the biosafety aspects of a widely extended and extremely useful technique, that is, the perfusion of blood, of its derived components and of other pharmacological products obtained from plasma. To face these new challenges we need innovative prevention strategies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA