Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Virol J ; 8: 529, 2011 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-22152060

RESUMEN

Modified vaccinia virus Ankara (MVA) has become a promising vaccine vector due to its immunogenicity and its proven safety in humans. As a general approach for stringent and rapid selection of recombinant MVA, we assessed marker rescue of the essential viral D4R gene in an engineered deletion mutant that is fully replication defective in wild-type cells. Recombinant, replicating virus was obtained by re-introduction of the deleted viral gene as a dominant selection marker into the deletion mutant.


Asunto(s)
Genes Esenciales , Genes Virales , Vectores Genéticos , Recombinación Genética , Uracil-ADN Glicosidasa/genética , Virus Vaccinia/genética , Animales , Línea Celular , Chlorocebus aethiops , Replicación del ADN , Humanos , Eliminación de Secuencia , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/metabolismo , Virus Vaccinia/patogenicidad , Células Vero , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
2.
PLoS One ; 6(9): e24505, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931732

RESUMEN

BACKGROUND: Currently existing yellow fever (YF) vaccines are based on the live attenuated yellow fever virus 17D strain (YFV-17D). Although, a good safety profile was historically attributed to the 17D vaccine, serious adverse events have been reported, making the development of a safer, more modern vaccine desirable. METHODOLOGY/PRINCIPAL FINDINGS: A gene encoding the precursor of the membrane and envelope (prME) protein of the YFV-17D strain was inserted into the non-replicating modified vaccinia virus Ankara and into the D4R-defective vaccinia virus. Candidate vaccines based on the recombinant vaccinia viruses were assessed for immunogenicity and protection in a mouse model and compared to the commercial YFV-17D vaccine. The recombinant live vaccines induced γ-interferon-secreting CD4- and functionally active CD8-T cells, and conferred full protection against lethal challenge already after a single low immunization dose of 10(5) TCID(50). Surprisingly, pre-existing immunity against wild-type vaccinia virus did not negatively influence protection. Unlike the classical 17D vaccine, the vaccinia virus-based vaccines did not cause mortality following intracerebral administration in mice, demonstrating better safety profiles. CONCLUSIONS/SIGNIFICANCE: The non-replicating recombinant YF candidate live vaccines induced a broad immune response after single dose administration, were effective even in the presence of a pre-existing immunity against vaccinia virus and demonstrated an excellent safety profile in mice.


Asunto(s)
Virus Vaccinia/metabolismo , Vacunas Virales/uso terapéutico , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Animales , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Chlorocebus aethiops , Células HeLa , Humanos , Sistema Inmunológico , Ratones , Ratones Endogámicos BALB C , Plásmidos/metabolismo , Vacunas Atenuadas/uso terapéutico , Células Vero , Proteínas del Envoltorio Viral/química
3.
J Infect Dis ; 203(11): 1556-64, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21592984

RESUMEN

After vaccination of humans with tick-borne encephalitis virus (TBEV) vaccine, the extent of cross-neutralization between viruses of the European, Far Eastern, and Siberian subtypes of TBEV and Omsk hemorrhagic fever virus (OHFV) was analyzed. Hybrid viruses that encode the TBEV surface proteins for representative viruses within all subtypes, and OHFV, were constructed using the West Nile virus (WNV) backbone as vector. These viruses allow for unbiased head-to-head comparison in neutralization assays because they exhibit the antigenic characteristics of the TBEV strains from which the surface proteins were derived and showed equivalent biologic properties in cell culture. Human serum samples derived from a TBEV vaccine trial were analyzed and revealed comparable neutralizing antibody titers against European, Far Eastern, and Siberian subtype viruses, indicating equally potent cross-protection against these TBEV strains and a somewhat reduced but still protective neutralization capacity against more distantly related viruses, such as OHFV.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Vacunas Virales/inmunología , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Análisis de Varianza , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Línea Celular Tumoral , Chlorocebus aethiops , Clonación Molecular , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/crecimiento & desarrollo , Encefalitis Transmitida por Garrapatas/sangre , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/prevención & control , Humanos , Cinética , Persona de Mediana Edad , Datos de Secuencia Molecular , Pruebas de Neutralización , Fenotipo , Alineación de Secuencia , Células Vero , Vacunas Virales/genética , Cultivo de Virus , Virus del Nilo Occidental/genética , Adulto Joven
4.
Vaccine ; 29(24): 4132-41, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21477673

RESUMEN

Ross River Virus has caused reported outbreaks of epidemic polyarthritis, a chronic debilitating disease associated with significant long-term morbidity in Australia and the Pacific region since the 1920s. To address this public health concern, a formalin- and UV-inactivated whole virus vaccine grown in animal protein-free cell culture was developed and tested in preclinical studies to evaluate immunogenicity and efficacy in animal models. After active immunizations, the vaccine dose-dependently induced antibodies and protected adult mice from viremia and interferon α/ß receptor knock-out (IFN-α/ßR(-/-)) mice from death and disease. In passive transfer studies, administration of human vaccinee sera followed by RRV challenge protected adult mice from viremia and young mice from development of arthritic signs similar to human RRV-induced disease. Based on the good correlation between antibody titers in human sera and protection of animals, a correlate of protection was defined. This is of particular importance for the evaluation of the vaccine because of the comparatively low annual incidence of RRV disease, which renders a classical efficacy trial impractical. Antibody-dependent enhancement of infection, did not occur in mice even at low to undetectable concentrations of vaccine-induced antibodies. Also, RRV vaccine-induced antibodies were partially cross-protective against infection with a related alphavirus, Chikungunya virus, and did not enhance infection. Based on these findings, the inactivated RRV vaccine is expected to be efficacious and protect humans from RRV disease.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Inmunización/métodos , Virus del Río Ross/inmunología , Vacunas Virales/inmunología , Adolescente , Adulto , Infecciones por Alphavirus/mortalidad , Infecciones por Alphavirus/patología , Animales , Biomarcadores , Virus Chikungunya/inmunología , Protección Cruzada , Femenino , Humanos , Masculino , Ratones , Análisis de Supervivencia , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas Virales/administración & dosificación , Viremia/prevención & control , Adulto Joven
5.
PLoS One ; 6(1): e16247, 2011 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-21283631

RESUMEN

BACKGROUND: New highly pathogenic H5N1 influenza viruses are continuing to evolve with a potential threat for an influenza pandemic. So far, the H5N1 influenza viruses have not widely circulated in humans and therefore constitute a high risk for the non immune population. The aim of this study was to evaluate the cross-protective potential of the hemagglutinins of five H5N1 strains of divergent clades using a live attenuated modified vaccinia Ankara (MVA) vector vaccine. METHODOLOGY/PRINCIPAL FINDINGS: The replication-deficient MVA virus was used to express influenza hemagglutinin (HA) proteins. Specifically, recombinant MVA viruses expressing the HA genes of the clade 1 virus A/Vietnam/1203/2004 (VN/1203), the clade 2.1.3 virus A/Indonesia/5/2005 (IN5/05), the clade 2.2 viruses A/turkey/Turkey/1/2005 (TT01/05) and A/chicken/Egypt/3/2006 (CE/06), and the clade 2.3.4 virus A/Anhui/1/2005 (AH1/05) were constructed. These experimental live vaccines were assessed in a lethal mouse model. Mice vaccinated with the VN/1203 hemagglutinin-expressing MVA induced excellent protection against all the above mentioned clades. Also mice vaccinated with the IN5/05 HA expressing MVA induced substantial protection against homologous and heterologous AH1/05 challenge. After vaccination with the CE/06 HA expressing MVA, mice were fully protected against clade 2.2 challenge and partially protected against challenge of other clades. Mice vaccinated with AH1/05 HA expressing MVA vectors were only partially protected against homologous and heterologous challenge. The live vaccines induced substantial amounts of neutralizing antibodies, mainly directed against the homologous challenge virus, and high levels of HA-specific IFN-γ secreting CD4 and CD8 T-cells against epitopes conserved among the H5 clades and subclades. CONCLUSIONS/SIGNIFICANCE: The highest level of cross-protection was induced by the HA derived from the VN/1203 strain, suggesting that pandemic H5 vaccines utilizing MVA vector technology, should be based on the VN/1203 hemagglutinin. Furthermore, the recombinant MVA-HA-VN, as characterized in the present study, would be a promising candidate for such a vaccine.


Asunto(s)
Protección Cruzada/genética , Vectores Genéticos , Hemaglutininas/biosíntesis , Subtipo H5N1 del Virus de la Influenza A/química , Vacunas/inmunología , Virus Vaccinia/genética , Animales , Humanos , Ratones , Especificidad de la Especie , Vacunación
6.
J Virol ; 85(4): 1896-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21123389

RESUMEN

West Nile virus (WNV)-neutralizing intravenous immune globulins (IVIG) were fractionated into IgG subclasses, and the contribution of each subclass to in vitro neutralization of and in vivo protection against WNV was evaluated. The results indicate that IgG1 (i) is the main subclass induced following WNV infection of humans, (ii) contained nearly all the in vitro WNV neutralization capacity, and (iii) mediates effector functions in vivo that render it superior to other subclasses in protection against WNV. The importance of human IgG1 indicates that a candidate WNV vaccine should induce an immune response that includes WNV-specific IgG1.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Inmunoglobulina G/clasificación , Inmunoglobulina G/uso terapéutico , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/mortalidad , Vacunas contra el Virus del Nilo Occidental
7.
PLoS One ; 5(8): e12217, 2010 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-20808939

RESUMEN

BACKGROUND: The development of novel influenza vaccines inducing a broad immune response is an important objective. The aim of this study was to evaluate live vaccines which induce both strong humoral and cell-mediated immune responses against the novel human pandemic H1N1 influenza virus, and to show protection in a lethal animal challenge model. METHODOLOGY/PRINCIPAL FINDINGS: For this purpose, the hemagglutinin (HA) and neuraminidase (NA) genes of the influenza A/California/07/2009 (H1N1) strain (CA/07) were inserted into the replication-deficient modified vaccinia Ankara (MVA) virus--a safe poxviral live vector--resulting in MVA-H1-Ca and MVA-N1-Ca vectors. These live vaccines, together with an inactivated whole virus vaccine, were assessed in a lung infection model using immune competent Balb/c mice, and in a lethal challenge model using severe combined immunodeficient (SCID) mice after passive serum transfer from immunized mice. Balb/c mice vaccinated with the MVA-H1-Ca virus or the inactivated vaccine were fully protected from lung infection after challenge with the influenza H1N1 wild-type strain, while the neuraminidase virus MVA-N1-Ca induced only partial protection. The live vaccines were already protective after a single dose and induced substantial amounts of neutralizing antibodies and of interferon-gamma-secreting (IFN-gamma) CD4- and CD8 T-cells in lungs and spleens. In the lungs, a rapid increase of HA-specific CD4- and CD8 T cells was observed in vaccinated mice shortly after challenge with influenza swine flu virus, which probably contributes to the strong inhibition of pulmonary viral replication observed. In addition, passive transfer of antisera raised in MVA-H1-Ca vaccinated immune-competent mice protected SCID mice from lethal challenge with the CA/07 wild-type virus. CONCLUSIONS/SIGNIFICANCE: The non-replicating MVA-based H1N1 live vaccines induce a broad protective immune response and are promising vaccine candidates for pandemic influenza.


Asunto(s)
Brotes de Enfermedades , Inmunización Pasiva/métodos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Vacunación/métodos , Animales , Formación de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Reacciones Cruzadas/inmunología , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunocompetencia/inmunología , Pulmón/inmunología , Ratones , Neuraminidasa/inmunología , Bazo/inmunología , Vacunas Atenuadas/inmunología
8.
Vaccine ; 28(19): 3318-24, 2010 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-20211218

RESUMEN

A cDNA comprising the complete genome of West Nile Virus (WNV) was generated by chemical synthesis using published sequence data, independent of any preformed viral components. The synthetic WNV, produced by transfection of in vitro transcribed RNA into cell culture, exhibited undistinguishable biological properties compared to the corresponding animal-derived wild-type virus. No differences were found concerning viral growth in mammalian and insect cell lines and concerning expression of viral proteins in cells. There were also no significant differences in virulence in mice following intranasal challenge. After immunizations of mice with experimental vaccines derived from the synthetic and wild-type viruses, protection from lethal challenge was achieved with similar amounts of antigen. Both vaccine preparations also induced comparable levels of neutralizing antibodies in mice. In addition, the synthetic approach turned out to be very accurate, since the rescued WNV genome contained no undesired mutations. Thus, the first flavivirus based on chemical gene synthesis was indistinguishable from the parent virus. This demonstrates that virus isolates from animal sources are dispensable to derive seed viruses for vaccine production or research.


Asunto(s)
ADN Complementario/genética , Vacunas Virales/inmunología , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Genoma Viral , Insectos , Ratones , Ratones Endogámicos BALB C , ARN Viral/genética , Análisis de Supervivencia , Transfección , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología , Vacunas Virales/genética , Virulencia , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/patogenicidad
9.
J Virol ; 83(10): 5192-203, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19279103

RESUMEN

The timely development of safe and effective vaccines against avian influenza virus of the H5N1 subtype will be of the utmost importance in the event of a pandemic. Our aim was first to develop a safe live vaccine which induces both humoral and cell-mediated immune responses against human H5N1 influenza viruses and second, since the supply of embryonated eggs for traditional influenza vaccine production may be endangered in a pandemic, an egg-independent production procedure based on a permanent cell line. In the present article, the generation of a complementing Vero cell line suitable for the production of safe poxviral vaccines is described. This cell line was used to produce a replication-deficient vaccinia virus vector H5N1 live vaccine, dVV-HA5, expressing the hemagglutinin of a virulent clade 1 H5N1 strain. This experimental vaccine was compared with a formalin-inactivated whole-virus vaccine based on the same clade and with different replicating poxvirus-vectored vaccines. Mice were immunized to assess protective immunity after high-dose challenge with the highly virulent A/Vietnam/1203/2004(H5N1) strain. A single dose of the defective live vaccine induced complete protection from lethal homologous virus challenge and also full cross-protection against clade 0 and 2 challenge viruses. Neutralizing antibody levels were comparable to those induced by the inactivated vaccine. Unlike the whole-virus vaccine, the dVV-HA5 vaccine induced substantial amounts of gamma interferon-secreting CD8 T cells. Thus, the nonreplicating recombinant vaccinia virus vectors are promising vaccine candidates that induce a broad immune response and can be produced in an egg-independent and adjuvant-independent manner in a proven vector system.


Asunto(s)
Vectores Genéticos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Chlorocebus aethiops , Virus Defectuosos/genética , Femenino , Subtipo H5N1 del Virus de la Influenza A/genética , Interferón gamma/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Infecciones por Orthomyxoviridae/inmunología , Virus Vaccinia/genética , Células Vero , Cultivo de Virus
10.
Virology ; 337(2): 235-41, 2005 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15882885

RESUMEN

The Escherichia coli thymidine kinase/thymidylate kinase (tk/tmk) fusion gene encodes an enzyme that efficiently converts the prodrug 3'-azido-2',3'-dideoxythymidine (AZT) into its toxic triphosphate derivative, a substance which stops DNA chain elongation. Integration of this marker gene into vaccinia virus that normally is not inhibited by AZT allowed the establishment of a powerful selection procedure for recombinant viruses. In contrast to the conventional vaccinia thymidine kinase (tk) selection that is performed in tk-negative cell lines, AZT selection can be performed in normal (tk-positive) cell lines. The technique is especially useful for the generation of replication-deficient vaccinia viruses and may also be used for gene knock-out studies of essential vaccinia genes.


Asunto(s)
Fusión Artificial Génica , Nucleósido-Fosfato Quinasa/genética , Profármacos , Timidina Quinasa/genética , Virus Vaccinia/enzimología , Zidovudina/metabolismo , Animales , Fármacos Anti-VIH/metabolismo , Biotransformación , Línea Celular , Chlorocebus aethiops , Escherichia coli/enzimología , Escherichia coli/genética
11.
J Virol ; 77(12): 7017-25, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12768020

RESUMEN

Recombinant vaccinia viruses that express defective retroviral vectors upon a single infection event in normal host cells were constructed. The gag-pol and envelope genes and a retroviral vector unit were inserted as vaccinia virus promoter-controlled transcription units at three separate loci. The triple recombinant virus was used to infect such diverse cell types as monkey and rabbit kidney, human lung, and primary chicken cells, resulting in the production of transduction-competent defective retroviral vectors. Infection of Chinese hamster ovary cells, which are nonpermissive for vaccinia virus replication, also resulted in production of retroviral vectors and secondary permanent transduction of the host cells. Since vaccinia virus supports the expression of cytotoxic proteins, the vesicular stomatitis virus G glycoprotein could be chosen as the envelope allowing a broad host range of transduction. Functionality of particles was monitored by expression of the green fluorescent protein in transduced 3T3 cell clones. This is the first description of a single chimeric virus encoding and releasing functional retroviral vectors, providing proof of principle of the new concept. No replication-competent retrovirus was detectable by sensitive reverse transcriptase assays. Since vaccinia virus has a broad host range, is extremely robust, and can be obtained at high titers and safe nonreplicating vaccinia virus strains are available, the hybrid system may open new perspectives for gene delivery.


Asunto(s)
Vectores Genéticos , Recombinación Genética , Retroviridae/genética , Transducción Genética , Virus Vaccinia/genética , Células 3T3 , Animales , Células CHO , Línea Celular , Chlorocebus aethiops , Cricetinae , Humanos , Glicoproteínas de Membrana/genética , Ratones , Conejos , Retroviridae/fisiología , Virus Vaccinia/fisiología , Células Vero , Proteínas del Envoltorio Viral/genética , Replicación Viral
12.
Protein Expr Purif ; 29(1): 58-69, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12729726

RESUMEN

Current hepatitis B virus (HBV) vaccines consist of preparations of recombinant HBV major surface antigen (sAg) and are protective in about 90-95% of vaccinated subjects. In improved vaccines, the frequency of nonresponders to the classical vaccine could be reduced by including additional epitopes from the preS-domains of the middle and large surface antigens. In this report, the development and characterization of a CHO cell line for HBsAg, expressing major, middle, and large antigens are described. Despite the previously reported retention of secreted proteins by the preS1 domain, cell lines could be amplified that secreted large amounts of the complete set of antigens. A producer line was established that expressed 1mg HBsAg per 100ml suspension culture per week during exponential growth. The productivity per cell increased further by at least threefold when the culture reached the stationary phase at high cell densities. In the production cell line, several hundred copies of the HBV vector were integrated at two adjacent sites into chromosome 2. The cell line was adapted to growth in a defined protein-free medium minimizing the risk of adventitious agents introduced by animal derived supplements. The cell line stably produced antigen over several months. In the candidate vaccine, both preS2 and preS1 domains were present at ratios similar to HBsAg from human sera. In summary, a production cell line for an improved HBV vaccine is presented with properties such as high productivity, long term stability of expression, and growth in protein-free media.


Asunto(s)
Antígenos de Superficie/química , Antígenos de Superficie de la Hepatitis B/inmunología , Virus de la Hepatitis B/metabolismo , Animales , Southern Blotting , Western Blotting , Células CHO , Técnicas de Cultivo de Célula/métodos , Clonación Molecular , Cricetinae , Medio de Cultivo Libre de Suero/farmacología , Bases de Datos como Asunto , Epítopos , Vectores Genéticos , Antígenos de Superficie de la Hepatitis B/metabolismo , Hibridación Fluorescente in Situ , Cinética , Microscopía Fluorescente , Modelos Genéticos , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Factores de Tiempo , Transfección , Proteínas del Envoltorio Viral/química
14.
Cancer Gene Ther ; 10(1): 30-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12489026

RESUMEN

Poor efficiency of gene transfer into cancer cells constitutes the major bottleneck of current cancer gene therapy. We reasoned that because tumors are masses of rapidly dividing cells, they would be most efficiently transduced with vector systems allowing transgene propagation. We thus designed two replicative retrovirus-derived vector systems: one inherently replicative vector, and one defective vector propagated by a helper retrovirus. In vitro, both systems achieved very efficient transgene propagation. In immunocompetent mice, replicative vectors transduced >85% tumor cells, whereas defective vectors transduced <1% under similar conditions. It is noteworthy that viral propagation could be efficiently blocked by azido-thymidine, in vitro and in vivo. In a model of established brain tumors treated with suicide genes, replicative retroviral vectors (RRVs) were approximately 1000 times more efficient than defective adenoviral vectors. These results demonstrate the advantage and potential of RRVs and strongly support their development for cancer gene therapy.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos , Virus de la Leucemia Murina de Moloney/genética , Neoplasias/terapia , Células 3T3 , Animales , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...