Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Infection ; 49(6): 1277-1287, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34642875

RESUMO

PURPOSE: Over the course of COVID-19 pandemic, evidence has accumulated that SARS-CoV-2 infections may affect multiple organs and have serious clinical sequelae, but on-site clinical examinations with non-hospitalized samples are rare. We, therefore, aimed to systematically assess the long-term health status of samples of hospitalized and non-hospitalized SARS-CoV-2 infected individuals from three regions in Germany. METHODS: The present paper describes the COVIDOM-study within the population-based cohort platform (POP) which has been established under the auspices of the NAPKON infrastructure (German National Pandemic Cohort Network) of the national Network University Medicine (NUM). Comprehensive health assessments among SARS-CoV-2 infected individuals are conducted at least 6 months after the acute infection at the study sites Kiel, Würzburg and Berlin. Potential participants were identified and contacted via the local public health authorities, irrespective of the severity of the initial infection. A harmonized examination protocol has been implemented, consisting of detailed assessments of medical history, physical examinations, and the collection of multiple biosamples (e.g., serum, plasma, saliva, urine) for future analyses. In addition, patient-reported perception of the impact of local pandemic-related measures and infection on quality-of-life are obtained. RESULTS: As of July 2021, in total 6813 individuals infected in 2020 have been invited into the COVIDOM-study. Of these, about 36% wished to participate and 1295 have already been examined at least once. CONCLUSION: NAPKON-POP COVIDOM-study complements other Long COVID studies assessing the long-term consequences of an infection with SARS-CoV-2 by providing detailed health data of population-based samples, including individuals with various degrees of disease severity. TRIAL REGISTRATION: Registered at the German registry for clinical studies (DRKS00023742).


Assuntos
COVID-19 , Qualidade de Vida , COVID-19/complicações , Humanos , Pandemias , SARS-CoV-2 , Resultado do Tratamento , Síndrome de COVID-19 Pós-Aguda
2.
Sci Rep ; 8(1): 8214, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844458

RESUMO

Genome editing is the introduction of directed modifications in the genome, a process boosted to therapeutic levels by designer nucleases. Building on the experience of ex vivo gene therapy for severe combined immunodeficiencies, it is likely that genome editing of haematopoietic stem/progenitor cells (HSPC) for correction of inherited blood diseases will be an early clinical application. We show molecular evidence of gene correction in a mouse model of primary immunodeficiency. In vitro experiments in DNA-dependent protein kinase catalytic subunit severe combined immunodeficiency (Prkdc scid) fibroblasts using designed zinc finger nucleases (ZFN) and a repair template demonstrated molecular and functional correction of the defect. Following transplantation of ex vivo gene-edited Prkdc scid HSPC, some of the recipient animals carried the expected genomic signature of ZFN-driven gene correction. In some primary and secondary transplant recipients we detected double-positive CD4/CD8 T-cells in thymus and single-positive T-cells in blood, but no other evidence of immune reconstitution. However, the leakiness of this model is a confounding factor for the interpretation of the possible T-cell reconstitution. Our results provide support for the feasibility of rescuing inherited blood disease by ex vivo genome editing followed by transplantation, and highlight some of the challenges.


Assuntos
Edição de Genes , Imunodeficiência Combinada Severa/genética , Animais , Proteína Quinase Ativada por DNA/genética , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos SCID , Proteínas Nucleares/genética
3.
Ann Oncol ; 28(1): 142-148, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27687311

RESUMO

Background: Sinonasal carcinomas (SNCs) comprise various rare tumor types that are characterized by marked histologic diversity and largely unknown molecular profiles, yet share an overall poor prognosis owing to an aggressive clinical course and frequent late-stage diagnosis. The lack of effective systemic therapies for locally advanced or metastatic SNC poses a major challenge to therapeutic decision making for individual patients. We here aimed to identify actionable genetic alterations in a patient with metastatic SNC whose tumor, despite all diagnostic efforts, could not be assigned to any known SNC category and was refractory to multimodal therapy. Patients and methods: We used whole-exome and transcriptome sequencing to identify a KIT exon 11 mutation (c.1733_1735del, p.D579del) as potentially druggable target in this patient and carried out cancer hotspot panel sequencing to detect secondary resistance-conferring mutations in KIT. Furthermore, as a step towards clinical exploitation of the recently described signatures of mutational processes in cancer genomes, we established and applied a novel bioinformatics algorithm that enables supervised analysis of the mutational catalogs of individual tumors. Results: Molecularly guided treatment with imatinib in analogy to the management of gastrointestinal stromal tumor (GIST) resulted in a dramatic and durable response with remission of nearly all tumor manifestations, indicating a dominant driver function of mutant KIT in this tumor. KIT dependency was further validated by a secondary KIT exon 17 mutation (c.2459_2462delATTCinsG, p.D820_S821delinsG) that was detected upon tumor progression after 10 months of imatinib treatment and provided a rationale for salvage therapy with regorafenib, which has activity against KIT exon 11/17 mutant GIST. Conclusions: These observations highlight the potential of unbiased genomic profiling for uncovering the vulnerabilities of individual malignancies, particularly in rare and unclassifiable tumors, and underscore that KIT exon 11 mutations represent tractable therapeutic targets across different histologies.


Assuntos
Carcinoma/diagnóstico , Carcinoma/genética , Neoplasias dos Seios Paranasais/diagnóstico , Neoplasias dos Seios Paranasais/genética , Proteínas Proto-Oncogênicas c-kit/genética , Adulto , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/análise , Carcinoma/tratamento farmacológico , Análise Mutacional de DNA , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mesilato de Imatinib/uso terapêutico , Imuno-Histoquímica , Masculino , Mutação , Neoplasias dos Seios Paranasais/tratamento farmacológico
4.
Leukemia ; 30(4): 937-46, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26582644

RESUMO

Activating BRAF mutations, in particular V600E/K, drive many cancers and are considered mutually exclusive with mutant RAS, whereas inactivating BRAF mutations in the D(594)F(595)G(596) motif cooperate with RAS via paradoxical MEK/ERK activation. Due to the increasing use of comprehensive tumor genomic profiling, many non-V600 BRAF mutations are being detected whose functional consequences and therapeutic actionability are often unknown. We investigated an atypical BRAF mutation, F595L, which was identified along with mutant HRAS in histiocytic sarcoma and also occurs in epithelial cancers, melanoma and neuroblastoma, and determined its interaction with mutant RAS. Unlike other DFG motif mutants, BRAF(F595L) is a gain-of-function variant with intermediate activity that does not act paradoxically, but nevertheless cooperates with mutant RAS to promote oncogenic signaling, which is efficiently blocked by pan-RAF and MEK inhibitors. Mutation data from patients and cell lines show that BRAF(F595L), as well as other intermediate-activity BRAF mutations, frequently coincide with mutant RAS in various cancers. These data define a distinct class of activating BRAF mutations, extend the spectrum of patients with systemic histiocytoses and other malignancies who are candidates for therapeutic blockade of the RAF-MEK-ERK pathway and underscore the value of comprehensive genomic testing for uncovering the vulnerabilities of individual tumors.


Assuntos
Regulação Neoplásica da Expressão Gênica , Sarcoma Histiocítico/genética , Sarcoma Histiocítico/patologia , Mutação/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Adulto , Animais , Biomarcadores Tumorais/genética , Western Blotting , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Exoma/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Sarcoma Histiocítico/metabolismo , Humanos , Masculino , Camundongos , Estadiamento de Neoplasias , Prognóstico , Transdução de Sinais
5.
Gene Ther ; 22(9): 707-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25965393

RESUMO

Monocyte-derived conventional dendritic cells (ConvDCs) loaded with melanoma antigens showed modest responses in clinical trials. Efficacy studies were hampered by difficulties in ConvDC manufacturing and low potency. Overcoming these issues, we demonstrated higher potency of lentiviral vector (LV)-programmed DCs. Monocytes were directly induced to self-differentiate into DCs (SmartDC-TRP2) upon transduction with a tricistronic LV encoding for cytokines (granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4)) and a melanoma antigen (tyrosinase-related protein 2 (TRP2)). Here, SmartDC-TRP2 generated with monocytes from five advanced melanoma patients were tested in autologous DC:T cell stimulation assays, validating the activation of functional TRP2-specific cytotoxic T lymphocytes (CTLs) for all patients. We described methods compliant to good manufacturing practices (GMP) to produce LV and SmartDC-TRP2. Feasibility of monocyte transduction in a bag system and cryopreservation following a 24-h standard operating procedure were achieved. After thawing, 50% of the initial monocyte input was recovered and SmartDC-TRP2 self-differentiated in vitro, showing uniform expression of DC markers, detectable LV copies and a polyclonal LV integration pattern not biased to oncogenic loci. GMP-grade SmartDC-TRP2 expanded TRP2-specific autologous CTLs in vitro. These results demonstrated a simpler GMP-compliant method of manufacturing an effective individualized DC vaccine. Such DC vaccine, when in combination with checkpoint inhibition therapies, might provide higher specificity against melanoma.


Assuntos
Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Lentivirus/metabolismo , Melanoma/terapia , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Vetores Genéticos , Células HEK293 , Humanos , Imunoterapia/métodos , Lentivirus/genética , Melanoma/imunologia , Linfócitos T Citotóxicos/imunologia
6.
Cancer Gene Ther ; 21(9): 373-80, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25145311

RESUMO

Precise oncotropism is required for successful systemic administration of next-generation oncolytic measles viruses (MVs). We have previously established a system for efficient post-entry targeting by insertion of synthetic microRNA target sites (miRTS) into the MV genome, thereby repressing replication in the presence of cognate microRNAs. Thus, differential expression of microRNAs, as frequently observed in normal compared with malignant tissues, can be exploited to increase vector specificity and safety. Here we report the combination of miRTS for different microRNAs in a single vector to detarget pivotal organs at risk during systemic administration (liver, brain, gastrointestinal tract). Accordingly, miRTS for miR-122, miR-7 and miR-148a that are enriched in these tissues were inserted to create multi-tissue-detargeted MV (MV-EGFP(mtd)). Replication of MV-EGFP(mtd) is repressed in cell lines as well as in non-transformed primary human hepatocytes and liver slices expressing cognate microRNAs. Oncolytic potency of MV-EGFP(mtd) is retained in a model of pancreatic cancer in vitro and in vivo. This work is a proof-of-concept that favorable expression profiles of multiple microRNAs can be exploited concomitantly to reshape the tropism of MV without compromising oncolytic efficacy. This strategy can be adapted to different vectors and cancer entities for safe and efficient high-dose systemic administration in clinical trials.


Assuntos
Vetores Genéticos/genética , Vírus do Sarampo/genética , MicroRNAs/genética , Vírus Oncolíticos/genética , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral , Modelos Animais de Doenças , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Ordem dos Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , MicroRNAs/química , Dados de Sequência Molecular , Neoplasias/genética , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Interferência de RNA , Transdução Genética , Células Vero , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Urologe A ; 52(9): 1283-9, 2013 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-23942742

RESUMO

BACKGROUND: Urologic cancers comprise one quarter of all newly diagnosed cancers per year in Germany. In addition to the increasing incidence treatment of solid and hematological tumors has become more differentiated, complex and potentially more effective as well as more expensive. Following the example of the USA multidisciplinary translational comprehensive cancer centers (CCCs) have been established in Germany. The financial support from the government and nonprofit organizations, such as the German Cancer Aid aims to ensure and to optimize treatment of tumor patients now and in the future. Coupled with this development new funding opportunities for translational research are opening up for the participating clinical and scientific partners. DISCUSSION: Just as attractive and coherent integration of urology into the structures of a CCC where available appears to be, just as controversial is the professional modus operandi. Using the example of the National Center for Tumor Diseases in Heidelberg (NCT), the current manuscript discusses the risks and opportunities of this new centralized form of oncological care in urology. Detailed knowledge of organizational structures, clinical operations and funding is a prerequisite for any partner of a CCC to succeed in such a highly demanding environment as a specialty instead of becoming mere surgical proceduralists.


Assuntos
Modelos Organizacionais , Serviço Hospitalar de Oncologia/organização & administração , Pesquisa Translacional Biomédica/organização & administração , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/terapia , Urologia/organização & administração , Alemanha , Humanos , Objetivos Organizacionais
10.
Virchows Arch ; 461(1): 79-86, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22653529

RESUMO

Tissue banks constitute decisive and rate-limiting resource and technology platforms for basic and translational biomedical research, notably in the area of cancer. Thus, it is essential to plan and structure tissue banking and allocate resources according to research needs, but essential requirements are still incompletely defined. The tissue bank of the National Center of Tumor Diseases Heidelberg (NCT) was founded with the intention to provide tissues of optimal quality and to prioritize the realization of research projects. We analysed its structure and prospective project management registration as well as tracking records for all projects of the NCT tissue bank as of its start in 2005 in order to obtain information that may be relevant for tissue bank planning. All project proposals submitted to the NCT tissue bank (n = 681) were included in the study. For a detailed evaluation of provided services, only projects that were completed until July 2011 (n = 605) were analysed. For these 605 projects, NCT tissue bank provided 769 specific services. In all projects/services, we recorded project leader, type and amount of material provided, type of research (basic/translational), work load of project and project completion. Furthermore, all completed projects were tracked after 90 days according to a standard protocol to determine principal investigators' (PI) satisfaction and quality of the provided material. Until July 2011, 605 projects had been successfully completed as documented by material transfer agreement. Of the projects, 72.7 % addressed basic research, 22.3 % were translational research projects and 3 % concerned epidemiological research; 91 % (n = 546) concerned a single PI and the NTC tissue bank. For these projects, 769 specific services were provided. Of these services, 288 concerned providing formalin-fixed and paraffin-embedded (FFPE) tissue (extracts, full size sections), 126 providing fresh frozen materials (including fresh frozen sections), 137 providing tissue micro-array (TMA)-based sections and 199 providing immunohistochemical services. Project tracking demonstrated that all projects had started within 90 days after reception of the material by the PIs, and PI satisfaction with provided material exceeded 97 %. Standardized registration and tracking provides valuable structural information for planning and financing of tissue banks and allocation of resources. The high number of completed projects as well as high user satisfaction demonstrates that structuring of tissue banks should be preferably research-oriented and highly efficient. The comparable number of requests for FFPE and fresh frozen tissue as well as TMA-based services underpins the need for a broad approach in terms of methods and material types in order to fulfil research needs.


Assuntos
Bancos de Tecidos/organização & administração , Bancos de Tecidos/estatística & dados numéricos , Alemanha , Humanos
11.
Cancer Gene Ther ; 19(3): 181-91, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22076043

RESUMO

First-line treatment of recurrent and/or refractory head and neck squamous cell carcinoma (HNSCC) is based on platinum, 5-fluorouracil (5-FU) and the monoclonal antiEGFR antibody cetuximab. However, in most cases this chemoimmunotherapy does not cure the disease, and more than 50% of HNSCC patients are dying because of local recurrence of the tumors. In the majority of cases, HNSCC overexpress the epidermal growth factor receptor (EGFR), and its presence is associated with a poor outcome. In this study, we engineered an EGFR-targeted oncolytic measles virus (MV), armed with the bifunctional enzyme cytosine deaminase/uracil phosphoribosyltransferase (CD/UPRT). CD/UPRT converts 5-fluorocytosine (5-FC) into the chemotherapeutic 5-FU, a mainstay of HNSCC chemotherapy. This virus efficiently replicates in and lyses primary HNSCC cells in vitro. Arming with CD/UPRT mediates efficient prodrug activation with high bystander killing of non-infected tumor cells. In mice bearing primary HNSCC xenografts, intratumoral administration of MV-antiEGFR resulted in statistically significant tumor growth delay and prolongation of survival. Importantly, combination with 5-FC is superior to virus-only treatment leading to significant tumor growth inhibition. Thus, chemovirotherapy with EGFR-targeted and CD/UPRT-armed MV is highly efficacious in preclinical settings with direct translational implications for a planned Phase I clinical trial of MV for locoregional treatment of HNSCC.


Assuntos
Carcinoma de Células Escamosas/terapia , Citosina Desaminase/genética , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/terapia , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/métodos , Pentosiltransferases/genética , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Citosina Desaminase/biossíntese , Citosina Desaminase/metabolismo , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Flucitosina/farmacocinética , Flucitosina/farmacologia , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Vírus do Sarampo/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pentosiltransferases/biossíntese , Pentosiltransferases/metabolismo , Pró-Fármacos/farmacocinética , Carcinoma de Células Escamosas de Cabeça e Pescoço , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Gene Ther ; 18(8): 598-608, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21701532

RESUMO

No curative therapy is currently available for locally advanced or metastatic pancreatic cancer. Therefore, new therapeutic approaches must be considered. Measles virus (MV) vaccine strains have shown promising oncolytic activity against a variety of tumor entities. For specific therapy of pancreatic cancer, we generated a fully retargeted MV that enters cells exclusively through the prostate stem cell antigen (PSCA). Besides a high-membrane frequency on prostate cancer cells, this antigen is expressed on pancreatic adenocarcinoma, but not on non-neoplastic tissue. PSCA expression levels differ within heterogeneous tumor bulks and between human pancreatic cell lines, and we could show specific infection of pancreatic adenocarcinoma cell lines with both high- and low-level PSCA expression. Furthermore, we generated a fully retargeted and armed MV-PNP-anti-PSCA to express the prodrug convertase purine nucleoside phosphorylase (PNP). PNP, which activates the prodrug fludarabine effectively, enhanced the oncolytic efficacy of the virus on infected and bystander cells. Beneficial therapeutic effects were shown in a pancreatic cancer xenograft model. Moreover, in the treatment of gemcitabine-resistant pancreatic adenocarcinoma cells, no cross-resistance to both MV oncolysis and activated prodrug was detected.


Assuntos
Adenocarcinoma/terapia , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/imunologia , Adenocarcinoma/virologia , Animais , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Terapia Combinada , Feminino , Proteínas Ligadas por GPI/metabolismo , Humanos , Masculino , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Camundongos , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/virologia , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/metabolismo , Células Vero , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados , Vidarabina/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Gene Ther ; 18(2): 210-2, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21068779

RESUMO

More than 10 years ago, we developed an efficient protocol for serum-free retroviral transduction of human hematopoietic stem cells derived from mobilized peripheral blood. After upscaling of the methodology, serum-free retroviral gibbon-ape leukemia virus (GALV) pseudotype PG13/LN vector supernatant produced under strict good manufacturing practice (GMP) conditions was used in the first clinical gene-marking trial in Germany. In this study, we analyzed the titer and transduction efficiency of this serum-free clinical-grade retroviral supernatant 10 years after production to evaluate the long-term stability. Long-term storage and transport on dry ice resulted in modestly decreased titers and levels of transduction efficiency in CD34+ cells ranging from 38.4 to 49.1%. We conclude that the stability of retroviral vectors in serum-free medium allows extended storage and distribution of approved clinical-grade retroviral vector stocks to distant sites in multicenter clinical trials.


Assuntos
Meios de Cultura Livres de Soro , Vetores Genéticos , Células-Tronco Hematopoéticas , Vírus da Leucemia do Macaco Gibão/genética , Preservação Biológica , Transdução Genética , Fatores de Tempo
14.
Curr Gene Ther ; 10(6): 414-22, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21054249

RESUMO

In the past 5 years, European investigators have played a major role in the development of clinical gene therapy. The provision of substantial funds by some individual member states to construct GMP facilities makes it an opportune time to network available gene therapy GMP facilities at an EU level. The integrated coordination of GMP production facilities and human skills for advanced gene and genetically-modified (GM) cell therapy, can dramatically enhance academic-led "First-in-man" gene therapy trials. Once proof of efficacy is gathered, technology can be transferred to the private sector which will take over further development taking advantage of knowledge and know-how. Complex technical challenges require existing production facilities to adapt to emerging technologies in a coordinated manner. These include a mandatory requirement for the highest quality of production translating gene-transfer technologies with pharmaceutical-grade GMP processes to the clinic. A consensus has emerged on the directions and priorities to adopt, applying to advanced technologies with improved efficacy and safety profiles, in particular AAV, lentivirus-based and oncolytic vectors. Translating cutting-edge research into "First-in-man" trials require that pre-normative research is conducted which aims to develop standard assays, processes and candidate reference materials. This research will help harmonise practices and quality in the production of GMP vector lots and GM-cells. In gathering critical expertise in Europe and establish conditions for interoperability, the PEVI infrastructure will contribute to the demands of the advanced therapy medicinal products* regulation and to both health and quality of life of EU-citizens.


Assuntos
Terapia Genética/tendências , Vetores Genéticos , Academias e Institutos , Transplante de Células/tendências , Ensaios Clínicos como Assunto , Desenho de Fármacos , Indústria Farmacêutica/normas , Europa (Continente) , Humanos
15.
BMC Cancer ; 8: 349, 2008 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19036146

RESUMO

BACKGROUND: Disease progression of hepatocellular cancer (HCC) in patients eligible for liver transplantation (LTx) occurs in up to 50% of patients, resulting in withdrawal from the LTx waiting list. Transarterial chemoembolization (TACE) is used as bridging therapy with highly variable response rates. The oral multikinase inhibitor sorafenib significantly increases overall survival and time-to-progression in patients with advanced hepatocellular cancer. DESIGN: The HeiLivCa study is a double-blinded, controlled, prospective, randomized multi-centre phase III trial. Patients in study arm A will be treated with transarterial chemoembolization plus sorafenib 400 mg bid. Patients in study arm B will be treated with transarterial chemoembolization plus placebo. A total of 208 patients with histologically confirmed hepatocellular carcinoma or HCC diagnosed according to EASL criteria will be enrolled. An interim patients' analysis will be performed after 60 events. Evaluation of time-to-progression as primary endpoint (TTP) will be performed at 120 events. Secondary endpoints are number of patients reaching LTx, disease control rates, OS, progression free survival, quality of live, toxicity and safety. DISCUSSION: As TACE is the most widely used primary treatment of HCC before LTx and sorafenib is the only proven effective systemic treatment for advanced HCC there is a strong rational to combine both treatment modalities. This study is designed to reveal potential superiority of the combined TACE plus sorafenib treatment over TACE alone and explore a new neo-adjuvant treatment concept in HCC before LTx.


Assuntos
Antineoplásicos/uso terapêutico , Benzenossulfonatos/uso terapêutico , Carcinoma Hepatocelular/terapia , Quimioembolização Terapêutica , Neoplasias Hepáticas/terapia , Piridinas/uso terapêutico , Adulto , Carcinoma Hepatocelular/tratamento farmacológico , Terapia Combinada , Método Duplo-Cego , Feminino , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Transplante de Fígado , Masculino , Niacinamida/análogos & derivados , Compostos de Fenilureia , Projetos de Pesquisa , Sorafenibe
16.
Gene Ther ; 13(13): 1057-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16511518

RESUMO

Following gene therapy of SCID-X1 using murine leukemia virus (MLV) derived vector, two patients developed leukemia owing to an activating vector integration near the LMO2 gene. We found that these integrations reside within FRA11E, a common fragile site known to correlate with chromosomal breakpoints in tumors. Further analysis showed that fragile sites attract a nonrandom number of MLV integrations, shedding light on its integration mechanism and risk-to-benefit ratio in gene therapy.


Assuntos
Sítios Frágeis do Cromossomo , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Vírus da Leucemia Murina/genética , Imunodeficiência Combinada Severa/terapia , Integração Viral/genética , Células Cultivadas , Fragilidade Cromossômica , Terapia Genética/métodos , Vetores Genéticos/genética , Células HeLa/virologia , Humanos , Leucemia/imunologia , Leucemia/virologia , Mutagênese Insercional , Medição de Risco , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/virologia
18.
Semin Hematol ; 41(4): 303-18, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15508116

RESUMO

Two serious adverse events involving activation of the LMO2 oncogene through retrovirus vector insertion in the otherwise extremely successful first gene therapy trial for X-linked severe combined immunodeficieny type 1 (SCID-X1) had initially caused widespread concern in the patient and research communities. Careful consideration 1 year after diagnosis of the second case still finds 12 of the treated patients clearly benefiting from gene therapy (freedom from treatment failure, 80%; survival 100%), a situation that should not portend the end of gene therapy for this disease, and is, in fact encouraging. While current approaches are justified to treat patients with otherwise life-threatening disorders, a broad consensus has developed that systematic basic research is required to further understand the pathophysiology of these serious adverse events and to provide new insights, enabling safer and more effective gene therapy strategies. With the continued success of SCID-X1 gene therapy in the majority of patients treated, it is of even greater importance to understand exactly which vector element or combination of elements predispose to toxicity. An in-depth study of the mechanisms behind the activation of the LMO2 and gammac genes will be highly instructive for the development of safer procedures and vectors. We summarize the central observations, ongoing experimental approaches, new concepts, and developments relevant to understanding, interpreting, and eventually overcoming the real and perceived obstacles posed by insertional mutagenesis due to gene transfer vectors.


Assuntos
Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Imunodeficiência Combinada Severa/terapia , Proteínas Adaptadoras de Transdução de Sinal , Dano ao DNA , Proteínas de Ligação a DNA/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Proteínas com Domínio LIM , Metaloproteínas/genética , Proteínas Proto-Oncogênicas , Imunodeficiência Combinada Severa/complicações
19.
Science ; 302(5644): 415-9, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-14564000

RESUMO

We have previously shown correction of X-linked severe combined immunodeficiency [SCID-X1, also known as gamma chain (gamma(c)) deficiency] in 9 out of 10 patients by retrovirus-mediated gamma(c) gene transfer into autologous CD34 bone marrow cells. However, almost 3 years after gene therapy, uncontrolled exponential clonal proliferation of mature T cells (with gammadelta+ or alphabeta+ T cell receptors) has occurred in the two youngest patients. Both patients' clones showed retrovirus vector integration in proximity to the LMO2 proto-oncogene promoter, leading to aberrant transcription and expression of LMO2. Thus, retrovirus vector insertion can trigger deregulated premalignant cell proliferation with unexpected frequency, most likely driven by retrovirus enhancer activity on the LMO2 gene promoter.


Assuntos
Proteínas de Ligação a DNA/genética , Terapia Genética/efeitos adversos , Vetores Genéticos , Leucemia-Linfoma de Células T do Adulto/etiologia , Metaloproteínas/genética , Retroviridae/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos T/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Ensaios Clínicos como Assunto , Células Clonais/fisiologia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lactente , Proteínas com Domínio LIM , Mutagênese Insercional , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Proto-Oncogenes , Receptores de Interleucina-2/genética , Retroviridae/fisiologia , Transcrição Gênica , Integração Viral , Replicação Viral
20.
Ann N Y Acad Sci ; 938: 146-55; discussion 155-6, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11458502

RESUMO

The semirandom location of retroviral integration in the target cell genome introduces a marker in the form of a fusion sequence composed of a genomic and a proviral part that is unique for each transduced cell and its clonal progeny. High-sensitivity detection of these fusion sequences would allow the tracking of clonal contributions of individual, marked hematopoietic progenitor, and stem cells in vivo. Clone detection by Southern blot has helped to analyze models of oligoclonal repopulation but is limited in sensitivity and specificity. Inverse PCR (Nolta et al., Proc. Natl. Acad. Sci. USA 93: 2414-2419) can demonstrate the clonal identity by sequencing but does not permit simultaneous detection of multiple clones. In an efficiently transduced rhesus macaque model (Tisdale et al., Blood 92: 2681-2687; Wu et al., Mol. Ther. 1: 285-293) Kim et al. (Blood 96: 1-8) have identified more than 40 insertion sequences from marrow CFU by inverse PCR. However, no previous study has been able to directly analyze the number of clones active in vivo. Here we demonstrate that the application of a recently developed PCR technology allows the simultaneous visualization of multiple integration sites from small clonal contributions to hematopoietic cells. By combining solid-phase primer extension with ligation-mediated PCR, direct genomic sequencing of retroviral integration sites was obtained in murine bone marrow samples. Further development of this technology will allow analysis of the clonal composition of marked hematopoiesis in small and large animals as well as in human gene transfer.


Assuntos
DNA Viral/análise , Células-Tronco Hematopoéticas/citologia , Vírus da Leucemia Murina/genética , Reação em Cadeia da Polimerase/métodos , Provírus/genética , Integração Viral/genética , Animais , Células Sanguíneas/citologia , Southern Blotting , Células da Medula Óssea/química , Células da Medula Óssea/citologia , Células Clonais/química , Células Clonais/citologia , Primers do DNA/genética , Marcadores Genéticos , Sobrevivência de Enxerto , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/química , Vírus da Leucemia Murina/isolamento & purificação , Camundongos , Modelos Biológicos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...