Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 389(7): 620-631, 2023 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-37585628

RESUMEN

BACKGROUND: Patients with the Crigler-Najjar syndrome lack the enzyme uridine diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1), the absence of which leads to severe unconjugated hyperbilirubinemia that can cause irreversible neurologic injury and death. Prolonged, daily phototherapy partially controls the jaundice, but the only definitive cure is liver transplantation. METHODS: We report the results of the dose-escalation portion of a phase 1-2 study evaluating the safety and efficacy of a single intravenous infusion of an adeno-associated virus serotype 8 vector encoding UGT1A1 in patients with the Crigler-Najjar syndrome that was being treated with phototherapy. Five patients received a single infusion of the gene construct (GNT0003): two received 2×1012 vector genomes (vg) per kilogram of body weight, and three received 5×1012 vg per kilogram. The primary end points were measures of safety and efficacy; efficacy was defined as a serum bilirubin level of 300 µmol per liter or lower measured at 17 weeks, 1 week after discontinuation of phototherapy. RESULTS: No serious adverse events were reported. The most common adverse events were headache and alterations in liver-enzyme levels. Alanine aminotransferase increased to levels above the upper limit of the normal range in four patients, a finding potentially related to an immune response against the infused vector; these patients were treated with a course of glucocorticoids. By week 16, serum bilirubin levels in patients who received the lower dose of GNT0003 exceeded 300 µmol per liter. The patients who received the higher dose had bilirubin levels below 300 µmol per liter in the absence of phototherapy at the end of follow-up (mean [±SD] baseline bilirubin level, 351±56 µmol per liter; mean level at the final follow-up visit [week 78 in two patients and week 80 in the other], 149±33 µmol per liter). CONCLUSIONS: No serious adverse events were reported in patients treated with the gene-therapy vector GNT0003 in this small study. Patients who received the higher dose had a decrease in bilirubin levels and were not receiving phototherapy at least 78 weeks after vector administration. (Funded by Genethon and others; ClinicalTrials.gov number, NCT03466463.).


Asunto(s)
Síndrome de Crigler-Najjar , Terapia Genética , Glucuronosiltransferasa , Humanos , Administración Intravenosa , Bilirrubina/sangre , Síndrome de Crigler-Najjar/sangre , Síndrome de Crigler-Najjar/complicaciones , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/terapia , Dependovirus , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glucuronosiltransferasa/administración & dosificación , Glucuronosiltransferasa/genética , Hiperbilirrubinemia/sangre , Hiperbilirrubinemia/etiología , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/terapia , Trasplante de Hígado , Fototerapia
2.
J Thromb Haemost ; 21(9): 2354-2361, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353081

RESUMEN

The introduction of adeno-associated virus-mediated, liver-directed gene therapy into the hemophilia treatment landscape brings not only great promise but also considerable uncertainty to a community that has a history punctuated by the devastating effects of HIV and hepatitis C virus. These infections were introduced into people with hemophilia through the innovation of factor concentrates in the 1970s and 1980s. Concentrates, heralded as a major advance in treatment at the time, brought devastation and death to the community already challenged by the complications of bleeding into joints, vital organs, and the brain. Over the past 5 decades, considerable advances in hemophilia treatment have improved the survival, quality of life, and participation of people with hemophilia, although challenges remain and health equity with their unaffected peers has not yet been achieved. The decision to take a gene therapy product is one in which an informed, holistic, and shared decision-making approach must be employed. Bias on the part of health care professionals and people with hemophilia must be addressed and minimized. Here, we review data leading to the regulatory authorization of valoctocogene roxaparvovec, an adeno-associated virus 5 gene therapy, in Europe to treat hemophilia A and etranacogene dezaparvovec-drlb in the United States and Europe to treat hemophilia B. We also provide an overview of the decision-making process and recommend steps that should be taken by the hemophilia community to ensure the safety of and optimal outcomes for people with hemophilia who choose to receive a gene therapy product.


Asunto(s)
Hemofilia A , Hemofilia B , Humanos , Hemofilia A/genética , Hemofilia A/terapia , Calidad de Vida , Hemofilia B/genética , Hemofilia B/terapia , Terapia Genética/efectos adversos
3.
Viruses ; 13(6)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208264

RESUMEN

Oncolytic virotherapy (OV) is an emerging class of immunotherapeutic drugs. Their mechanism of action is two-fold: direct cell lysis and unmasking of the cancer through immunogenic cell death, which allows the immune system to recognize and eradicate tumours. Breast cancer is the most common cancer in women and is challenging to treat with immunotherapy modalities because it is classically an immunogenically "cold" tumour type. This provides an attractive niche for OV, given viruses have been shown to turn "cold" tumours "hot," thereby opening a plethora of treatment opportunities. There has been a number of pre-clinical attempts to explore the use of OV in breast cancer; however, these have not led to any meaningful clinical trials. This review considers both the potential and the barriers to OV in breast cancer, namely, the limitations of monotherapy and the scope for combination therapy, improving viral delivery and challenges specific to the breast cancer population (e.g., tumour subtype, menopausal status, age).


Asunto(s)
Neoplasias de la Mama/terapia , Terapia Genética , Viroterapia Oncolítica , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/etiología , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Resultado del Tratamiento
4.
Viruses ; 13(6)2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34198859

RESUMEN

Oncolytic viruses have emerged as a promising strategy for cancer therapy due to their dual ability to selectively infect and lyse tumor cells and to induce systemic anti-tumor immunity. Among various candidate viruses, coxsackievirus group B (CVBs) have attracted increasing attention in recent years. CVBs are a group of small, non-enveloped, single-stranded, positive-sense RNA viruses, belonging to species human Enterovirus B in the genus Enterovirus of the family Picornaviridae. Preclinical studies have demonstrated potent anti-tumor activities for CVBs, particularly type 3, against multiple cancer types, including lung, breast, and colorectal cancer. Various approaches have been proposed or applied to enhance the safety and specificity of CVBs towards tumor cells and to further increase their anti-tumor efficacy. This review summarizes current knowledge and strategies for developing CVBs as oncolytic viruses for cancer virotherapy. The challenges arising from these studies and future prospects are also discussed in this review.


Asunto(s)
Enterovirus Humano B/genética , Ingeniería Genética , Vectores Genéticos/genética , Virus Oncolíticos/genética , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Enterovirus Humano B/fisiología , Ingeniería Genética/métodos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Neoplasias/terapia , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Resultado del Tratamiento , Replicación Viral
5.
Value Health ; 24(6): 839-845, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34119082

RESUMEN

OBJECTIVES: To evaluate alternative methods to calculate and/or attribute economic surplus in the cost-effectiveness analysis of single or short-term therapies. METHODS: We performed a systematic literature review of articles describing alternative methods for cost-effectiveness analysis of potentially curative therapies whose assessment using traditional methods may suggest unaffordable valuations owing to the magnitude of estimated long-term quality-adjusted life-year (QALY) gains or cost offsets. Through internal deliberation and discussion with staff at the Health Technology Assessment bodies in England and Canada, we developed the following 3 alternative methods for further evaluation: (1) capping annual costs in the comparator arm at $150 000 per year; (2) "sharing" the economic surplus with the health sector by apportioning only 50% of cost offsets or 50% of cost offsets and QALY gains to the value of the therapy; and (3) crediting the therapy with only 12 years of the average annual cost offsets or cost offsets and QALY gains over the lifetime horizon. The impact of each alternative method was evaluated by applying it in an economic model of 3 hypothetical condition-treatment scenarios meant to reflect a diversity of chronicity and background healthcare costs. RESULTS: The alternative with greatest impact on threshold price for the fatal pediatric condition spinal muscular atrophy type 1 was the 12-year cutoff scenario. For a hypothetical one-time treatment for hemophilia A, capping cost offsets at $150 000 per year had the greatest impact. For chimeric antigen receptor T-cell treatment of non-Hodgkin's lymphoma, capping cost offsets or using 12-year threshold had little impact, whereas 50% sharing of surplus including QALY gains and cost offsets greatly reduced threshold pricing. CONCLUSIONS: Health Technology Assessment bodies and policy makers will wrestle with how to evaluate single or short-term potentially curative therapies and establish pricing and payment mechanisms to ensure sustainability. Scenario analyses using alternative methods for calculating and apportioning economic surplus can provide starkly different assessment results. These methods may stimulate important societal dialogue on fair pricing for these novel treatments.


Asunto(s)
Quimioterapia/economía , Terapia Genética/economía , Costos de la Atención en Salud , Inmunoterapia Adoptiva/economía , Evaluación de la Tecnología Biomédica/economía , Anticuerpos Biespecíficos/economía , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/economía , Anticuerpos Monoclonales Humanizados/uso terapéutico , Productos Biológicos/economía , Productos Biológicos/uso terapéutico , Ahorro de Costo , Análisis Costo-Beneficio , Costos de los Medicamentos , Terapia Genética/efectos adversos , Hemofilia A/tratamiento farmacológico , Hemofilia A/economía , Humanos , Inmunoterapia Adoptiva/efectos adversos , Linfoma no Hodgkin/economía , Linfoma no Hodgkin/terapia , Modelos Económicos , Años de Vida Ajustados por Calidad de Vida , Proteínas Recombinantes de Fusión/economía , Proteínas Recombinantes de Fusión/uso terapéutico , Inducción de Remisión , Atrofias Musculares Espinales de la Infancia/economía , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/terapia , Factores de Tiempo , Resultado del Tratamiento
6.
Mol Ther ; 29(2): 597-610, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309883

RESUMEN

Evaluation of immune responses to adeno-associated virus (AAV)-mediated gene therapies prior to and following dose administration plays a key role in determining therapeutic safety and efficacy. This report describes up to 3 years of immunogenicity data following administration of valoctocogene roxaparvovec (BMN 270), an AAV5-mediated gene therapy encoding human B domain-deleted FVIII (hFVIII-SQ) in a phase 1/2 clinical study of adult males with severe hemophilia A. Patients with pre-existing humoral immunity to AAV5 or with a history of FVIII inhibitors were excluded from the trial. Blood plasma and peripheral blood mononuclear cell (PBMC) samples were collected at regular intervals following dose administration for assessment of humoral and cellular immune responses to both the AAV5 vector and transgene-expressed hFVIII-SQ. The predominant immune response elicited by BMN 270 administration was largely limited to the development of antibodies against the AAV5 capsid that were cross-reactive with other common AAV serotypes. No FVIII inhibitor responses were observed within 3 years following dose administration. In a context of prophylactic or on-demand corticosteroid immunosuppression given after vector infusion, AAV5 and hFVIII-SQ peptide-specific cellular immune responses were intermittently detected by an interferon (IFN)-γ and tumor necrosis factor (TNF)-α FluoroSpot assay, but they were not clearly associated with detrimental safety events or changes in efficacy measures.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Hemofilia A/genética , Hemofilia A/terapia , Adulto , Reacciones Cruzadas/inmunología , Dependovirus/inmunología , Factor VIII/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Humoral , Masculino , Transgenes , Resultado del Tratamiento
7.
Mol Ther ; 28(8): 1759-1771, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32592692

RESUMEN

Targeted delivery of oligonucleotides to liver hepatocytes using N-acetylgalactosamine (GalNAc) conjugates that bind to the asialoglycoprotein receptor has become a breakthrough approach in the therapeutic oligonucleotide field. This technology has led to the approval of givosiran for the treatment of acute hepatic porphyria, and there are another seven conjugates in registrational review or phase 3 trials and at least another 21 conjugates at earlier stages of clinical development. This review highlights some of the recent chemical and preclinical advances in this space, leading to a large number of clinical candidates against a diverse range of targets in liver hepatocytes. The review focuses on the use of this delivery system for small interfering RNAs (siRNAs) and antisense molecules that cause downregulation of target mRNA and protein. A number of other approaches such as anti-microRNAs and small activating RNAs are starting to exploit the technology, broadening the potential of this approach for therapeutic oligonucleotide intervention.


Asunto(s)
Acetilgalactosamina , Técnicas de Transferencia de Gen , Terapia Genética , Hígado/metabolismo , Oligonucleótidos/administración & dosificación , Acetilgalactosamina/química , Animales , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Oligonucleótidos/química , Oligonucleótidos/genética , ARN Mensajero/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Investigación , Investigación Biomédica Traslacional
8.
Mol Ther ; 28(3): 723-746, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31972133

RESUMEN

Gene therapy with adeno-associated virus (AAV) vectors has demonstrated safety and long-term efficacy in a number of trials across target organs, including eye, liver, skeletal muscle, and the central nervous system. Since the initial evidence that AAV vectors can elicit capsid T cell responses in humans, which can affect the duration of transgene expression, much progress has been made in understanding and modulating AAV vector immunogenicity. It is now well established that exposure to wild-type AAV results in priming of the immune system against the virus, with development of both humoral and T cell immunity. Aside from the neutralizing effect of antibodies, the impact of pre-existing immunity to AAV on gene transfer is still poorly understood. Herein, we review data emerging from clinical trials across a broad range of gene therapy applications. Common features of immune responses to AAV can be found, suggesting, for example, that vector immunogenicity is dose-dependent, and that innate immunity plays an important role in the outcome of gene transfer. A range of host-specific factors are also likely to be important, and a comprehensive understanding of the mechanisms driving AAV vector immunogenicity in humans will be key to unlocking the full potential of in vivo gene therapy.


Asunto(s)
Dependovirus/inmunología , Vectores Genéticos/efectos adversos , Inmunidad , Animales , Ensayos Clínicos como Asunto , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunidad Innata , Especificidad de Órganos , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
JACC Cardiovasc Interv ; 13(1): 1-19, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31918927

RESUMEN

Despite optimal combination of guideline-directed anti-ischemic therapies and myocardial revascularization, a substantial proportion of patients with stable coronary artery disease continues to experience disabling symptoms and is often referred as "no-option." The appraisal of the pathways linking ischemia to symptom perception indicates a complex model of heart-brain interactions in the generation of the subjective anginal experience and inspired novel approaches that may be clinically effective in alleviating the angina burden of this population. Conversely, the prevailing ischemia-centered view of angina, with the focus on traditional myocardial revascularization as the sole option to address ischemia on top of medical therapy, hinders the experimental characterization and broad-scale clinical implementation of strongly needed therapeutic options. The interventionist, often the first physician to establish the diagnosis of refractory angina pectoris (RAP) following coronary angiography, should be aware of the numerous emerging technologies with the potential to improve quality of life in the growing population of RAP patients. This review describes the current landscape and the future perspectives on nonpharmacological treatment technologies for patients with RAP, with a view on the underlying physiopathological rationale and current clinical evidence.


Asunto(s)
Angina de Pecho/terapia , Enfermedad de la Arteria Coronaria/terapia , Contrapulsación , Terapia por Estimulación Eléctrica , Tratamiento con Ondas de Choque Extracorpóreas , Terapia Genética , Corazón/inervación , Terapia por Láser , Trasplante de Células Madre , Angina de Pecho/genética , Angina de Pecho/metabolismo , Angina de Pecho/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/fisiopatología , Circulación Coronaria , Contrapulsación/efectos adversos , Terapia por Estimulación Eléctrica/efectos adversos , Metabolismo Energético , Tratamiento con Ondas de Choque Extracorpóreas/efectos adversos , Terapia Genética/efectos adversos , Humanos , Terapia por Láser/efectos adversos , Miocardio/metabolismo , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento
10.
Hum Gene Ther ; 31(1-2): 20-46, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31802714

RESUMEN

Gene therapy and gene editing technologies are complex and it can be difficult for the public to understand their possible benefits or side effects. However, patient and public support is critical for the successful adoption of any new technology. Given the recent advances in gene therapy and gene editing, their potential clinical benefits, and the significant attention that has been given to the first-known successful attempt at permanent and heritable changes to the human genome, a systematic review was performed to assess beliefs and attitudes toward gene therapy and gene editing for human use, and to highlight the factors that influence acceptability. A systematic search following Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines was undertaken in April 2018 to identify articles examining opinions and attitudes regarding the acceptability of gene therapy and gene editing. Overall, 1,561 records were retrieved from 4 databases (Ovid Medline, PsycINFO, Scopus, and Web of Science). Duplicates were removed, and titles and abstracts independently screened, leaving 86 full-text articles assessed for eligibility. Following full-text review, 33 were included, with 5 articles added after forward/backward searching. An additional three articles were added following an updated search in March 2019 (total n = 41). Findings from the studies were integrated according to common themes: the impact of demographics; risks versus benefits of success; treatment specifics (e.g., medical vs. other reasons; disease severity and status; somatic vs. germ line; and mode of delivery); moral or ethical issues; and changes with time. In general, perceptions were positive, particularly for medical reasons and fatal diseases, but were also influenced by perceived risk. Somatic therapies had higher levels of acceptability than germ line therapies. While available in various forms, limitations exist in the measurement of perceptions of gene therapy and gene editing. Treatment acceptability is essential for future clinical trials, so it is important for scientists and clinicians to be clear about the risks and benefits of these technologies, and how these are communicated to the public, while encouraging education about genetic therapies to a broad range of individuals.


Asunto(s)
Edición Génica , Terapia Genética , Aceptación de la Atención de Salud , Opinión Pública , Terapias Complementarias , Análisis Factorial , Edición Génica/ética , Edición Génica/métodos , Técnicas de Transferencia de Gen , Enfermedades Genéticas Congénitas/epidemiología , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/terapia , Terapia Genética/efectos adversos , Terapia Genética/ética , Terapia Genética/métodos , Terapia Genética/psicología , Conocimientos, Actitudes y Práctica en Salud , Política de Salud , Humanos , Aceptación de la Atención de Salud/psicología , Aceptación de la Atención de Salud/estadística & datos numéricos , Mejoramiento de la Calidad , Medición de Riesgo , Índice de Severidad de la Enfermedad
11.
BMC Cancer ; 19(1): 1126, 2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31747895

RESUMEN

BACKGROUND: TAE-gene therapy for hepatoma, incorporating the tumor-targeted therapeutic efficacy of trans-arterial embolization, hydroxyapatite nanoparticles (nHAP) and anti-cancer wild-type p53 gene (wt-p53), was presented in our former studies (Int J Nanomedicine 8:3757-68, 2013, Liver Int 32:998-1007, 2012). However, the incompletely antitumoral effect entails defined guidelines on searching properer materials for this novel therapy. METHODS: Unmodified nHAP, Ca(2+) modified nHAP, poly-lysine modified nHAP and liposome were separately used to form U-nanoplex, Ca-nanoplex, Pll-nanoplex, L-nanoplex respectively with wt-p53 expressing plasmid. The four nanoplexs were then applied in vitro for human normal hepacyte L02 and hepatoma HePG2 cell line, and in vivo for rabbits with hepatic VX2 tumor by injection of nanoplexs/lipiodol emulsion into the hepatic artery in a tumor target manner. The distribution, superficial potential, physical structure, morphology and chemical compositions of nanoplexs were evaluated by TEM, SEM, EDS etc., with the objective of understanding their roles in hepatoma TAE-gene therapy. RESULTS: In vitro, L-nanoplex managed the highest gene transferring efficiency. Though with the second highest transfection activity, Pll-nanoplex showed the strongest tumor inhibition activity while maintaining safe to the normal hepacyte L02. In fact, only Pll-nanoplex can combine both the antitumoral effect to HePG2 and safe procedure to L02 among the four systems above. In vivo, being the only one with successful gene transference to hepatic VX2 tumor, Pll-nanoplex/lipiodol emulsion can target the tumor more specifically, which may explain its best therapeutic effect and hepatic biologic response. Further physical characterizations of the four nanoplexs suggested particle size and proper electronic organic surface may be crucial for nano-TAE gene therapy. CONCLUSION: Pll-nanoplex is the most proper system for the combined therapy due to its selectively retention in liver cancer cells, secondary to its morphological and physico-chemical properties of nanometric particle size, steady emulsion, proper organic and electronic surface.


Asunto(s)
Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Terapia Genética , Neoplasias Hepáticas/terapia , Proteína p53 Supresora de Tumor/genética , Animales , Carcinoma Hepatocelular/diagnóstico , Quimioembolización Terapéutica/efectos adversos , Quimioembolización Terapéutica/métodos , Emulsiones , Aceite Etiodizado/administración & dosificación , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Neoplasias Hepáticas/diagnóstico , Masculino , Nanopartículas , Conejos , Nanomedicina Teranóstica
12.
Biomed Pharmacother ; 106: 610-617, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29990850

RESUMEN

Radiation countermeasures are radioprotective agents that reduce the harmful effects of ionizing radiation. They have wide range of applications extending from protection of normal tissues of cancer patients during radiotherapy to safeguard people aftermath of radiologic or nuclear accidents. Despite the screening of thousands of natural and synthetic compounds, only few found place in clinic with limited tolerance. Therefore, mechanistic understanding is essential in the development of more suitable and customized radiation countermeasure agents. This review focuses on the mechanisms of radioprotection imparted by these agents. Radioprotectors are diverse and act through widely varying mechanisms that can be classified in 10 categories: 1) scavenging of free radicals; 2) enhancing DNA repair; 3) synchronizing of cells; 4) modulating redox sensitive genes; 5) modulating growth factors and cytokines; 6) inhibiting apoptosis; 7) repurposing of drug; 8) interacting and chelating of radionuclides; and therapeutic methods of tissue regeneration such as 9) gene therapy; and 10) stem cell therapy. The most common mechanism of radioprotection is the scavenging of free radicals whereas, modulation of growth factors, cytokines and redox genes emerge as effective strategies. Gene and stem cell therapies as therapeutic radiation countermeasures are being developed and can be applied in the near future to minimize the side effects of radiation exposure through tissues regenerations. Thus, the management of radiation exposure may require a holistic multi-mechanistic approaches to achieve optimal radiation protection during radiotherapy of cancer patients and in cases of nuclear eventualities.


Asunto(s)
Terapia Genética/métodos , Exposición a la Radiación/efectos adversos , Traumatismos por Radiación/prevención & control , Protección Radiológica/métodos , Protectores contra Radiación/uso terapéutico , Trasplante de Células Madre/métodos , Animales , Terapia Genética/efectos adversos , Humanos , Factores Protectores , Dosis de Radiación , Traumatismos por Radiación/genética , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Protectores contra Radiación/efectos adversos , Medición de Riesgo , Factores de Riesgo , Trasplante de Células Madre/efectos adversos
13.
Biotechnol Bioeng ; 115(7): 1866-1877, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29578571

RESUMEN

Further quantitative understanding of the biological effects and mechanisms involved in cellular and intracellular delivery of nucleic acid materials is required to produce clinical applications of gene therapy. Several modeling approaches have been used in this field; however, a comprehensive approach that integrates all the key pharmacological issues into a holistic framework that is applicable for in vivo conditions is still lacking. This contribution presents a pharmacokinetic/pharmacodynamic model-based control study of non-viral siRNA delivery describing the dynamics of the delivery process and takes into account the main multi-objective optimization issues such as efficacy and toxicity, as well as the effect of uncertainty in cell doubling time. The methodology developed in this work is used to predict the optimal dosage injection rate and optimal intracellular exposure of siRNAs in order to improve the pharmacological effects before cell division occurs. The present analysis successfully provides quantitative predictions of non-viral siRNA activity paving the path for further experimental work to probe more efficient delivery systems.


Asunto(s)
Productos Biológicos/farmacología , Productos Biológicos/farmacocinética , Terapia Genética/métodos , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/farmacocinética , Productos Biológicos/administración & dosificación , Productos Biológicos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Silenciador del Gen , Terapia Genética/efectos adversos , Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Humanos , Modelos Estadísticos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/toxicidad
14.
Hum Gene Ther ; 28(10): 782-799, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28854824

RESUMEN

Hematopoietic stem cell-directed gene therapy (HSC-GT) provides an innovative treatment option for hematological disorders. Gene therapy promises to cure the disease "at the root" and is therefore exceptional in its potential, but also formidable in its challenges, as long-term side effects are hard to predict and clinical experience remains limited. Many excellent reviews on the topic by designated experts in the field of HSC-GT have come forth, elucidating the successes and pitfalls in the various clinical studies. This review attempts to discuss what we understand from those studies to represent current state of the art with respect to vectors, stem cell transduction, and pretransplant preparatory regimes, what limitations may remain, and which types of diseases may be more suited for HSC-GT than others (targets). We thus discuss the available vector platforms (tools) and preclinical/clinical and basic research (tricks) that contribute to our current understanding of HSC-GT, as well as some overarching principles we can conclude from these. The field has also learned from previous shortcomings, although some of the major concerns of the past, specifically insertional mutagenesis, may not be of relevance for future trials. This very positive development in HSC-GT, however, has to compete with the improvements in hematopoietic stem cell transplantation or enzyme-replacement therapy, leaving a narrow margin for gene therapy.


Asunto(s)
Terapia Genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/terapia , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Pruebas de Mutagenicidad , Transgenes
15.
Hematol Oncol Clin North Am ; 31(5): 823-834, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28895850

RESUMEN

Transfer of gene-corrected autologous hematopoietic stem cells in patients with primary immunodeficiencies has emerged as a new therapeutic approach. Patients with various conditions lacking a suitable donor have been treated with retroviral vectors and a gene-addition strategy. Initial promising results were shadowed by the occurrence of malignancies in some of these patients. Current trials, developed in the last decade, use safer viral vectors to overcome the risk of genotoxicity and have led to improved clinical outcomes. This review reflects the progresses made in specific disorders, including adenosine deaminase deficiency, X-linked severe combined immunodeficiency, chronic granulomatous disease, and Wiskott-Aldrich syndrome.


Asunto(s)
Terapia Genética , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Expresión Génica , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/clasificación , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Transducción Genética , Transgenes , Acondicionamiento Pretrasplante/métodos
16.
Hematol Oncol Clin North Am ; 31(5): 835-852, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28895851

RESUMEN

Gene therapy for hemoglobinopathies is currently based on transplantation of autologous hematopoietic stem cells genetically modified with a lentiviral vector expressing a globin gene under the control of globin transcriptional regulatory elements. Preclinical and early clinical studies showed the safety and potential efficacy of this therapeutic approach as well as the hurdles still limiting its general application. In addition, for both beta-thalassemia and sickle cell disease, an altered bone marrow microenvironment reduces the efficiency of stem cell harvesting as well as engraftment. These hurdles need be addressed for gene therapy for hemoglobinopathies to become a clinical reality.


Asunto(s)
Terapia Genética , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinas/genética , Humanos , Transducción Genética , Talasemia beta/genética , Talasemia beta/terapia
17.
EBioMedicine ; 12: 34-42, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27720213

RESUMEN

MicroRNAs (miRNAs) are an evolutionarily conserved class of small, regulatory non-coding RNAs that negatively regulate protein coding gene and other non-coding transcripts expression. miRNAs have been established as master regulators of cellular processes, and they play a vital role in tumor initiation, progression and metastasis. Further, widespread deregulation of microRNAs have been reported in several cancers, with several microRNAs playing oncogenic and tumor suppressive roles. Based on these, miRNAs have emerged as promising therapeutic tools for cancer management. In this review, we have focused on the roles of miRNAs in tumorigenesis, the miRNA-based therapeutic strategies currently being evaluated for use in cancer, and the advantages and current challenges to their use in the clinic.


Asunto(s)
Terapia Genética , MicroARNs/genética , MicroARNs/uso terapéutico , Neoplasias/genética , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Humanos , Inmunomodulación/genética , Neoplasias/inmunología , Neoplasias/patología , Interferencia de ARN , Resultado del Tratamiento , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
18.
Reg Anesth Pain Med ; 41(4): 511-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27224659

RESUMEN

Chronic neuropathic pain is a widespread problem with negative personal and societal consequences. Despite considerable clinical neuroscience research, the goal of developing effective, reliable, and durable treatments has remained elusive. The critical role played by the dorsal root ganglion (DRG) in the induction and maintenance of chronic pain has been largely overlooked in these efforts, however. It may be that, by targeting this site, robust new options for pain management will be revealed. This review summarizes recent advances in the knowledge base for DRG-targeted treatments for neuropathic pain:• Pharmacological options including the chemical targeting of voltage-dependent calcium channels, transient receptor potential channels, neurotrophin production, potentiation of opioid transduction pathways, and excitatory glutamate receptors.• Ablation or modulation of the DRG via continuous thermal radiofrequency and pulsed radiofrequency treatments.• Implanted electrical neurostimulator technologies.• Interventions involving the modification of DRG cellular function at the genetic level by using viral vectors and gene silencing methods.


Asunto(s)
Analgésicos/uso terapéutico , Ablación por Catéter , Dolor Crónico/terapia , Desnervación/métodos , Terapia por Estimulación Eléctrica , Ganglios Espinales , Terapia Genética/métodos , Neuralgia/terapia , Analgésicos/efectos adversos , Animales , Ablación por Catéter/efectos adversos , Dolor Crónico/diagnóstico , Dolor Crónico/genética , Dolor Crónico/fisiopatología , Desnervación/efectos adversos , Terapia por Estimulación Eléctrica/instrumentación , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Ganglios Espinales/cirugía , Terapia Genética/efectos adversos , Humanos , Neuroestimuladores Implantables , Neuralgia/diagnóstico , Neuralgia/genética , Neuralgia/fisiopatología , Dimensión del Dolor , Percepción del Dolor , Umbral del Dolor , Resultado del Tratamiento
19.
J Clin Oncol ; 34(12): 1330-8, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26926680

RESUMEN

PURPOSE: Autologous monocyte-derived dendritic cells (DCs) electroporated with synthetic mRNA (TriMixDC-MEL) are immunogenic and have antitumor activity as a monotherapy in patients with pretreated advanced melanoma. Ipilimumab, an immunoglobulin G1 monoclonal antibody directed against the cytotoxic T-lymphocyte-associated protein 4 receptor that counteracts physiologic suppression of T-cell function, improves the overall survival of patients with advanced melanoma. This phase II study investigated the combination of TriMixDC-MEL and ipilimumab in patients with pretreated advanced melanoma. PATIENTS AND METHODS: Thirty-nine patients were treated with TriMixDC-MEL (4 × 10(6) cells administered intradermally and 20 × 10(6) cells administered intravenously) plus ipilimumab (10 mg/kg every 3 weeks for a total of four administrations, followed by maintenance therapy every 12 weeks in patients who remained progression free). Six-month disease control rate according to the immune-related response criteria served as the primary end point. RESULTS: The 6-month disease control rate was 51% (95% CI, 36% to 67%), and the overall tumor response rate was 38% (including eight complete and seven partial responses). Seven complete responses and one partial tumor response are ongoing after a median follow-up time of 36 months (range, 22 to 43 months). The most common treatment-related adverse events (all grades) consisted of local DC injection site skin reactions (100%), transient post-DC infusion chills (38%) and flu-like symptoms (84%), dermatitis (64%), hepatitis (13%), hypophysitis (15%), and diarrhea/colitis (15%). Grade 3 or 4 immune-related adverse events occurred in 36% of patients. There was no grade 5 adverse event. CONCLUSION: The combination of TriMixDC-MEL and ipilimumab is tolerable and results in an encouraging rate of highly durable tumor responses in patients with pretreated advanced melanoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Células Dendríticas/trasplante , Electroporación , Terapia Genética/métodos , Melanoma/terapia , ARN Mensajero/genética , Neoplasias Cutáneas/terapia , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Células Cultivadas , Quimioterapia Adyuvante , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Esquema de Medicación , Femenino , Terapia Genética/efectos adversos , Terapia Genética/mortalidad , Humanos , Ipilimumab , Estimación de Kaplan-Meier , Masculino , Melanoma/genética , Melanoma/inmunología , Melanoma/mortalidad , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , ARN Mensajero/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA