Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Hand Surg Eur Vol ; 48(10): 1096-1102, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37728884

RESUMEN

Hand surgeons have the potential to improve patient care, both with their own research and by using evidenced-based practice. In this first part of a two-part article, we describe key steps for the analysis of clinical data using quantitative methodology. We aim to describe the principles of medical statistics and their relevance and use in hand surgery, with contemporaneous examples. Hand surgeons seek expertise and guidance in the clinical domain to improve their practice and patient care. Part of this process involves the critical analysis and appraisal of the research of others.


Asunto(s)
Especialidades Quirúrgicas , Cirujanos , Humanos , Mano/cirugía
2.
J Hand Surg Eur Vol ; 48(11): 1237-1243, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37747691

RESUMEN

In this second of a two-part article, we describe some of the common statistical pitfalls encountered in hand surgery research. These include dichotomania, the 'Table 2 fallacy', p-hacking, regression to the mean, overfitting and unaccounted data clustering. We explain the impact of these pitfalls on hand surgery research and describe techniques to avoid them. The aim of this two-part article was to provide a starting point for hand surgeons to refer to when conducting or analysing research and provide resources and references for interested readers to explore.


Asunto(s)
Especialidades Quirúrgicas , Cirujanos , Humanos , Mano/cirugía
4.
Hum Gene Ther ; 19(7): 690-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18576918

RESUMEN

Gliomas have a dismal prognosis, with the median survival of patients with the most common histology, glioblastoma multiforme, being only 12-15 months. Development of novel therapeutic agents is urgently needed. We have previously demonstrated that oncolytic measles virus strains derived from the Edmonston vaccine lineage have significant antitumor activity against gliomas [Phuong, L.K., Allen, C., Peng, K.W., Giannini, C., Greiner, S., Teneyck, C.J., Mishra, P.K., Macura, S.I., Russell, S.J., Galanis, E.C. (2003). Cancer. Res. 63, 2462-2469]. MV-CEA is an Edmonston vaccine lineage measles virus strain engineered to express the marker peptide carcinoembryonic antigen (CEA): CEA levels can serve as a correlate of viral gene expression. In support of a phase I clinical trial of intratumoral and resection cavity administration of MV-CEA to patients with recurrent gliomas, we assessed the neurotoxicity of MV-CEA in adult immune male rhesus macaques (Macaca mulatta). The animals ' immune status and administration schedule mimicked the trial population and proposed administration schema. Macaca mulatta represents the prototype animal species for assessment of measles neurotoxicity. The animals were stereotactically administered either vehicle (n = 1) or MV-CEA at 2 x 10(5)or 2 x 10(6) TCID(50) (each, n = 2) in the right frontal lobe in two injections on days 1 and 5. Macaques were closely monitored clinically for neurotoxicity. Body weight, temperature, complete blood count, CEA, clinical chemistries, coagulation, complement levels, immunoglobulin, measles antibody titers, viremia, and shedding (buccal swabs) were tested at multiple time points. Furthermore, cisterna magna spinal taps were performed on day 9 and 1 year after the first viral dose administration, and samples were analyzed for protein, glucose, cell differential, and presence of MV-CEA. Magnetic resonance imaging (MRI) was performed between 4 and 5 months after article administration to assess for subclinical neurotoxicity. To date, 36+ months from study initiation there has been no clinical or biochemical evidence of toxicity, including lack of neurological symptoms, fever, or other systemic symptoms and lack of immunosuppression. Quantitative RT-PCR analysis of blood, buccal swabs, and cerebrospinal fluid (CSF) was negative for MV-CEA at all time points, with the exception of viral genome deletion in the blood of one asymptomatic animal at the 2 x 10(6) TCID(50) dose level on day 85. Vero cell overlays of CSF cells and supernatant were negative for viral recovery. There was no detection of CEA in serum or CSF at any time point. MRI scans were negative for imaging abnormalities and showed no evidence of encephalitis. Our results support the safety of CNS administration of MV-CEA in glioma patients. A clinical trial of intratumoral and resection cavity administration of MV-CEA in patients with recurrent glioblastoma multiforme is currently ongoing.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer , Antígeno Carcinoembrionario/metabolismo , Vectores Genéticos , Glioma/terapia , Virus del Sarampión , Animales , Encéfalo/virología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/genética , Chlorocebus aethiops , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Efecto Citopatogénico Viral , Vías de Administración de Medicamentos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Glioma/genética , Glioma/patología , Glioma/prevención & control , Humanos , Macaca mulatta , Masculino , Virus del Sarampión/genética , Virus del Sarampión/patogenicidad , Recurrencia , Resultado del Tratamiento , Células Vero
5.
Mol Ther ; 15(3): 588-97, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17245355

RESUMEN

The innate antiviral responses of tumor cells are often impaired but may still be sufficient to impede the intratumoral spread of an oncolytic virus. Here, we establish that the oncolytic measles virus (MV-eGFP) induces interferon (IFN) production in human myeloma and ovarian cancer cells. In addition, MV gene expression and virus progeny production were inhibited by IFN treatment of these tumor cells. The P gene of wild-type measles virus encodes P/V/C proteins known to antagonize IFN induction and/or response. We therefore engineered MV-eGFP for IFN evasion and more efficient intratumoral spread by arming it with the P gene from wild-type IC-B strain MV, thus generating MV-eGFP-Pwt. The chimeric virus exhibited reduced IFN sensitivity and diminished capacity to induce IFN in BJAB lymphoma, ARH-77 myeloma cells, and activated peripheral blood mononuclear cells. Interestingly, unlike the wild-type MV, MV-eGFP-Pwt was unable to shut down IFN induction completely. In immunocompromised mice bearing human myeloma xenografts, intravenously administered MV-eGFP-Pwt showed significantly enhanced oncolytic potency compared to MV-eGFP. These results indicate that oncolytic viruses are subject to control by the innate immune defenses of human tumor cells and may therefore be more effective if their natural ability to combat innate immunity is maintained.


Asunto(s)
Inmunidad Innata/inmunología , Virus del Sarampión/genética , Virus Oncolíticos/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Genes Reporteros/genética , Ingeniería Genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inyecciones Intravenosas , Interferones/biosíntesis , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Sensibilidad y Especificidad , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Environ Sci Technol ; 40(21): 6709-14, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17144300

RESUMEN

We have measured the rate of scorodite (FeAsO4.2H2O) dissolution over an environmentally relevant range of pH and temperature conditions. Dissolution rates, calculated using arsenic (As) as the reaction progress variable, were slowest at pH 3 and increased with both decreasing and increasing pH. Comparison of the pH-dependence of the dissolution rates with a scorodite stability diagram suggests that our measurements of dissolution rate at pH 2 reflect congruent dissolution, and those at and above pH 3 reflect incongruent dissolution. Because As was used as the reaction progress variable, and recognizing that As may adsorb to iron hydroxides during incongruent dissolution of scorodite, the derived rates may be underestimated. The pH and temperature dependence of scorodite dissolution rates determined in these experiments have implications for the stability of scorodite at field sites and also for the potential use of scorodite to sequester As. Although scorodite dissolution is slow, it can be enhanced by up to a half order of magnitude by increases in pH and temperature.


Asunto(s)
Arsénico/toxicidad , Arsenicales/análisis , Arsénico/análisis , Arsénico/química , Arsenicales/química , Técnicas de Química Analítica/métodos , Compuestos Férricos/química , Concentración de Iones de Hidrógeno , Hierro/química , Cinética , Modelos Lineales , Modelos Químicos , Solubilidad , Temperatura , Termodinámica
7.
Clin Cancer Res ; 12(20 Pt 1): 6170-8, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17062694

RESUMEN

PURPOSE: Attenuated measles viruses are promising experimental anticancer agents currently being evaluated in a phase I dose escalation trial for ovarian cancer patients. Virus attachment, entry, and subsequent intercellular fusion between infected and uninfected neighboring cells are mediated via the two measles receptors (CD46 and SLAM). To minimize potential toxicity due to measles virus-associated immunosuppression and infection of nontarget tissues, we sought to develop an ovarian cancer exclusive fully retargeted measles virus. EXPERIMENTAL DESIGN AND RESULTS: Interactions of measles virus with its natural receptors were ablated, and a single-chain antibody (scFv) specific for alpha-folate receptor (FRalpha), a target overexpressed on 90% of nonmucinous ovarian cancer, was genetically engineered on the viral attachment protein (MV-alphaFR). Specificity of virus tropism was tested on tumor and normal cells. Biodistribution of measles virus infection was evaluated in measles-susceptible CD46 transgenic mice, whereas antitumor activity was monitored noninvasively by bioluminescence imaging in xenograft models. Tropism and fusogenic activity of MV-alphaFR was redirected exclusively to FRalpha without compromise to virus infectivity. In contrast to the parental virus, MV-alphaFR has no background infectivity on normal human cells. The antitumor activity of MV-alphaFR, as assessed by tumor volume reduction and overall survival increase, was equal to the parental virus in two models of human ovarian cancer (s.c. and i.p.). CONCLUSIONS: A FR-exclusive ovarian cancer targeted oncolytic virus was generated and shown to be therapeutically effective, thus introducing a new modality for FR targeting and a candidate measles virus for clinical testing.


Asunto(s)
Proteínas Portadoras/fisiología , Linfocitos/inmunología , Virus del Sarampión , Neoplasias Ováricas/inmunología , Receptores de Superficie Celular/fisiología , Vacunas contra el Cáncer , Femenino , Receptores de Folato Anclados a GPI , Vectores Genéticos , Humanos , Lentivirus/genética , Neoplasias Ováricas/virología , Valores de Referencia , Vacunas Atenuadas , Vacunas Virales
8.
J Virol ; 80(17): 8530-40, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16912303

RESUMEN

Antibodies to viral surface glycoproteins play a crucial role in immunity to measles by blocking both virus attachment and subsequent fusion with the host cell membrane. Here, we demonstrate that certain immunoglobulin G (IgG) antibodies can also enhance the entry of measles virus (MV) into monocytes and macrophages. Antibody-dependent enhancement of infectivity was observed in mouse and human macrophages using virions opsonized by a murine monoclonal antibody against the MV hemagglutinin (H) glycoprotein, polyclonal mouse anti-MV IgG, or diluted measles-immune human sera. Neither H-specific Fab fragments nor H-specific IgM could enhance MV entry in monocytes or macrophages, indicating involvement of a Fc gamma receptor (FcgammaR)-mediated mechanism. Preincubation with an anti-fusion protein (anti-F) monoclonal antibody or a fusion-inhibitory peptide blocked infection, indicating that a functional F protein was required for viral internalization. Classical complement pathway activation did not promote infection through complement receptors and inhibited anti-H IgG-mediated enhancement. In vivo, antibody-enhanced infection allowed MV to overcome a highly protective systemic immune response in preimmunized IfnarKo-Ge46 transgenic mice. These data demonstrate a previously unidentified mechanism that may contribute to morbillivirus pathogenesis where H-specific IgG antibodies promote the spread of MV infection among FcgammaR-expressing host cells. The findings point to a new model for the pathogenesis of atypical MV infection observed after immunization with formalin-inactivated MV vaccine and underscore the importance of the anti-F response after vaccination.


Asunto(s)
Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Inmunoglobulina G/inmunología , Virus del Sarampión/patogenicidad , Sarampión/inmunología , Animales , Línea Celular , Hemaglutininas Virales/inmunología , Humanos , Inmunización , Macrófagos/virología , Sarampión/fisiopatología , Sarampión/prevención & control , Sarampión/virología , Vacuna Antisarampión/administración & dosificación , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , Virus del Sarampión/inmunología , Ratones , Ratones Transgénicos , Monocitos/virología , Células U937/virología , Proteínas Virales de Fusión/inmunología
9.
Breast Cancer Res Treat ; 99(2): 177-84, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16642271

RESUMEN

Breast cancer is the most common malignancy and the second leading cause of female cancer mortality in the United States. There is an urgent need for development of novel therapeutic approaches. In this study, we investigated the antitumor potential of a novel viral agent, an attenuated strain of measles virus deriving from the Edmonston vaccine lineage, genetically engineered to produce carcinoembryonic antigen (CEA) against breast cancer. CEA production as the virus replicates can serve as a marker of viral gene expression. Infection of a variety of breast cancer cell lines including MDA-MB-231, MCF7 and SkBr3 at different multiplicities of infection (MOIs) from 0.1 to 10 resulted in significant cytopathic effect consisting of extensive syncytia formation and massive cell death at 72-96 h from infection. All breast cancer lines overexpressed the measles virus receptor CD46 and supported robust viral replication, which correlated with CEA production. TUNEL assays indicated an apoptotic mechanism of syncytial death. The efficacy of this approach in vivo was examined in a subcutaneous Balb C/nude mouse model of MDA-MB-231 cells. Intravenous administration of MV-CEA at a total dose of 1.2 x 10(7) TCID50 resulted in statistically significant tumor growth delay ( p=0.005) and prolongation of survival ( p=0.001). In summary, MV-CEA has potent antitumor activity against breast cancer lines and xenografts. Monitoring marker peptide levels in the serum could serve as a low-risk method of detecting viral gene expression during treatment and could allow dose optimization and individualization of treatment. Trackable measles virus derivatives merit further exploration in breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/terapia , Antígeno Carcinoembrionario/inmunología , Vacuna Antisarampión/farmacología , Virus del Sarampión/inmunología , Viroterapia Oncolítica , Animales , Apoptosis/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Antígeno Carcinoembrionario/genética , Chlorocebus aethiops , Efecto Citopatogénico Viral , Femenino , Humanos , Vacuna Antisarampión/genética , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , Proteína Cofactora de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas , Tasa de Supervivencia , Células Tumorales Cultivadas , Células Vero , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biochem Biophys Res Commun ; 337(1): 22-9, 2005 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-16171777

RESUMEN

A recombinant measles virus (MV) expressing the sodium iodide symporter (NIS) is being considered for therapy of advanced multiple myeloma. Auger electrons selectively damage cells in which the isotope decays. We hypothesized that the Auger electron emitting isotope 125I can be used to control viral proliferation. MV was engineered to express both carcinoembryonic antigen and NIS (MV-NICE). Cells were infected with MV-NICE and exposed to 125I with appropriate controls. MV-NICE replication in vitro is inhibited by the selective uptake of 125I by cells expressing NIS. Auger electron damage is partly mediated by free radicals and abrogated by glutathione. In myeloma xenografts, control of MV-NICE with 125I was not possible under the conditions of the experiment. MV-NICE does not replicate faster in the presence of radiation. Auger electron emitting isotopes effectively stop propagation of MV vectors expressing NIS in vitro. Additional work is necessary to translate these observations in vivo.


Asunto(s)
Electrones , Vectores Genéticos/efectos de la radiación , Radioisótopos de Yodo , Virus del Sarampión/genética , Mieloma Múltiple/terapia , Animales , Anticuerpos Antivirales/sangre , Antígeno Carcinoembrionario/genética , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Radicales Libres/metabolismo , Virus del Sarampión/inmunología , Virus del Sarampión/efectos de la radiación , Ratones , Mieloma Múltiple/inmunología , Simportadores/genética , Células Vero , Replicación Viral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Ther ; 12(5): 835-41, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16054438

RESUMEN

The sodium-iodide symporter (NIS) is primarily a thyroid protein, providing for the accumulation of iodide for biosynthesis of thyroid hormones. Native NIS expression has made possible the use of radioactive iodide to image and treat thyroid disease successfully. The current study, using adult male beagle dogs, was carried out in preparation for a Phase I clinical trial of adenovirus-mediated NIS gene (approved symbol SLC5A5) therapy for prostate cancer. Direct intraprostatic injection of virus (Ad5/CMV/NS) was followed by iv injection of 3 mCi 123I and serial image acquisition. The dogs were then given a therapeutic dose of 131I (116 mCi/m2) and observed for 7 days. SPECT/CT fusion imaging revealed clear images of the NIS-transduced prostates. Dosimetry calculations revealed an average absorbed dose to the prostate of 23 +/- 42 cGy/mCi 131I, with acceptably low radiation doses to other organs. This study demonstrated the successful introduction of localized NIS expression in the prostate gland of dogs, with no vector-related toxicity observed. None of the animals experienced any surgical complications, and serum chemistry panels showed no significant change following therapy. The results presented provide further evidence of the safety and efficacy of NIS as a therapeutic gene and support translation of this work into the clinical setting.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Neoplasias de la Próstata/terapia , Simportadores/uso terapéutico , Animales , Modelos Animales de Enfermedad , Perros , Radioisótopos de Yodo/uso terapéutico , Masculino , Neoplasias de la Próstata/diagnóstico por imagen , Radiometría , Cintigrafía , Radiofármacos/uso terapéutico , Simportadores/genética , Transducción Genética , Transfección
12.
Cancer Gene Ther ; 12(7): 593-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15746945

RESUMEN

Oncolytic viruses are promising cytoreductive agents for cancer treatment but extensive human testing will be required before they are made commercially available. Here, we investigated the oncolytic potential of two commercially available live attenuated vaccines, Moraten measles and Jeryl-Lynn mumps, in a murine model of intraperitoneal human ovarian cancer and compared their efficacies against a recombinant oncolytic measles virus (MV-CEA) that is being tested in a phase I clinical trial. The common feature of these viruses is that they express hemagglutinin and fusion therapeutic proteins that can induce extensive fusion of the infected cell with its neighbors, resulting in death of the cell monolayer. In vitro, the three viruses caused intercellular fusion in human ovarian cancer cells but with marked differences in fusion kinetics. MV-CEA was the fastest followed by Jeryl-Lynn mumps virus while Moraten measles virus was the slowest, although all viruses eventually caused comparable cell death 6 days postinfection. Tumor-bearing mice treated with 10(6) or 10(7) pfu (one thousand times the vaccine dose) of each of the three viruses responded favorably to therapy with significant prolongations in survival. All three viruses demonstrated equivalent antitumor potency. Commercially available Moraten measles and Jeryl-Lynn mumps vaccines warrant further investigation as potential anticancer agents.


Asunto(s)
Vacuna Antisarampión/uso terapéutico , Vacuna contra la Parotiditis/uso terapéutico , Neoplasias Ováricas/terapia , Animales , Antígeno Carcinoembrionario/sangre , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Efecto Citopatogénico Viral , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nat Biotechnol ; 23(2): 209-14, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15685166

RESUMEN

Live attenuated measles viruses of the Edmonston lineage (MV-Edm) have potent anti-tumor activity but are not entirely tumor-specific owing to widespread distribution of their native receptors, CD46 and SLAM. We have therefore developed a pseudoreceptor system that allows rescue and propagation of fully retargeted viruses displaying single-chain antibody fragments. Viruses retargeted to tumor-selective CD38, epidermal growth factor receptor (EGFR) or EGFR mutant vIII (EGFRvIII) efficiently entered cells through their respective targeted receptors in vitro and in vivo, but not through CD46 and SLAM. When administered intratumorally or intravenously to mice bearing human CD38 or EGFR-positive human tumor xenografts, the targeted viruses demonstrated specific receptor-mediated anti-tumor activity. These data provide an in vivo demonstration of antibody-directed tumor destruction by retargeted oncolytic viruses.


Asunto(s)
Terapia Biológica/métodos , Virus del Sarampión/fisiología , Neoplasias/virología , Virus/crecimiento & desarrollo , Virus/genética , Animales , Anticuerpos Antineoplásicos/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Mejoramiento Genético/métodos , Humanos , Ratones , Neoplasias/terapia , Receptores Virales/genética , Receptores Virales/metabolismo , Resultado del Tratamiento , Células Vero , Replicación Viral/genética
14.
Nat Biotechnol ; 22(3): 331-6, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14990955

RESUMEN

Membrane fusion has many potential applications in biotechnology. Here we show that antibody-targeted cell fusion can be achieved by engineering a fusogenic viral membrane glycoprotein complex. Three different single-chain antibodies were displayed at the extracellular C terminus of the measles hemagglutinin (H) protein, and combinations of point mutations were introduced to ablate its ability to trigger fusion through the native viral receptors CD46 and SLAM. When coexpressed with the measles fusion (F) protein, using plasmid cotransfection or bicistronic adenoviral vectors, the retargeted H proteins could mediate antibody-targeted cell fusion of receptor-negative or receptor-positive index cells with receptor-positive target cells. Adenoviral expression vectors mediating human epidermal growth factor receptor (EGFR)-targeted cell fusion were potently cytotoxic against EGFR-positive tumor cell lines and showed superior antitumor potency against EGFR-positive tumor xenografts as compared with control adenoviruses expressing native (untargeted) or CD38-targeted H proteins.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Membrana Celular/inmunología , Membrana Celular/patología , Sistemas de Liberación de Medicamentos/métodos , Fusión de Membrana/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Complejo Antígeno-Anticuerpo/genética , Antígenos CD/genética , Antígenos CD/inmunología , Línea Celular Tumoral , Membrana Celular/genética , Células Cultivadas , Chlorocebus aethiops , Femenino , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Proteína Cofactora de Membrana , Fusión de Membrana/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/terapia , Ingeniería de Proteínas/métodos , Receptores de Superficie Celular , Proteínas Recombinantes/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Resultado del Tratamiento , Células Vero
15.
Blood ; 103(5): 1641-6, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14604966

RESUMEN

The Edmonston vaccine strain of measles virus (MV-Edm) propagates efficiently in a broad range of human tumor cells, killing them selectively. However, the oncolytic potency of MV-Edm in different human tumor xenograft therapy models is highly variable and there is no convenient way to map the distribution of virus-infected tissues in vivo. To enhance the oncolytic potency of MV-Edm against radiosensitive malignancies and to facilitate noninvasive imaging of infected tissues, we generated a recombinant MV-Edm encoding the human thyroidal iodide symporter (NIS). MV-NIS replicated almost as efficiently as unmodified MV-Edm, and human tumor cells efficiently concentrated radioiodine when infected with MV-NIS. Intratumoral spread of MV-NIS was noninvasively demonstrated by serial gamma-camera imaging of iodine-123 (123I) uptake both in MV-sensitive KAS-6/1 myeloma xenografts, which regressed completely after a single intravenous dose of MV-NIS, and in MM1 myeloma xenografts, which were unresponsive to MVNIS therapy. However, MV-resistant MM1 tumors regressed completely when 131I was administered 9 days after a single intravenous injection of MV-NIS (radiovirotherapy). 131I alone had no effect on MM1 tumor growth. While the potential hematopoietic toxicity of this new therapy requires further evaluation, image-guided radiovirotherapy is a promising new approach to the treatment of multiple myeloma, an incurable but highly radiosensitive plasma cell malignancy. Testing in other radiosensitive cancers is warranted.


Asunto(s)
Terapia Combinada , Virus del Sarampión/genética , Mieloma Múltiple/terapia , Simportadores/genética , Glándula Tiroides/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , ADN Complementario/metabolismo , Femenino , Técnicas Genéticas , Humanos , Interleucina-6/metabolismo , Radioisótopos de Yodo/uso terapéutico , Ratones , Ratones SCID , Trasplante de Neoplasias , Factores de Tiempo , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Hum Gene Ther ; 14(16): 1565-77, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14577918

RESUMEN

In support of a proposed phase I clinical trial, we studied the biodistribution of virus-infected cells after intraperitoneal administration of oncolytic measles viruses to alpha/beta interferon-defective mice expressing human CD46 with human-like tissue specificity. Various marker genes were employed, and green fluorescent protein proved to be most informative. Mesothelium and ovarian surface epithelium were remarkably resistant to infection, but infected peritoneal macrophages were present in abundance both in peritoneal lavage fluid and in the greater omentum, where they were heavily concentrated in "milky spots". Infected macrophages were also identified outside the peritoneal cavity, along the peritoneal fluid drainage pathway and in the spleen. Thus, diaphragmatic stomata, thoracic lymphatic vessels, and parathymic lymph nodes contained numerous measles-infected cells, as did the marginal zones of the white pulp of the spleen. Splenic marginal zone macrophages were the predominant targets of infection after intravenous administration of oncolytic measles viruses. When measles-infected peritoneal macrophages were adoptively transferred, they did not migrate beyond the confines of the peritoneal cavity, suggesting that, after intraperitoneal virus administration, the positive cells in thoracic lymphatics, parathymic lymph nodes, and spleen are nonmigratory cells transduced in situ by viral particles that have exited from the peritoneal cavity.


Asunto(s)
Antígenos CD/fisiología , Inyecciones Intraperitoneales , Virus del Sarampión/fisiología , Glicoproteínas de Membrana/fisiología , Cavidad Peritoneal/citología , Proteínas Recombinantes/administración & dosificación , Animales , Proteínas Fluorescentes Verdes , Interferón Tipo I/metabolismo , Proteínas Luminiscentes/metabolismo , Macrófagos/virología , Proteína Cofactora de Membrana , Ratones , Ratones Transgénicos , Especificidad de Órganos , Cavidad Peritoneal/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA