Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33627393

RESUMEN

Herpes simplex virus (HSV) is a promising tool for developing oncolytic virotherapy. We recently reported a platform for receptor-retargeted oncolytic HSVs that incorporates single-chain antibodies (scFvs) into envelope glycoprotein D (gD) to mediate virus entry via tumor-associated antigens. Therefore, it would be useful to develop an efficient system that can screen antibodies that might mediate HSV entry when they are incorporated as scFvs into gD. We created an HSV-based screening probe by the genetic fusion of a gD mutant with ablated binding capability to the authentic HSV entry receptors and the antibody-binding C domain of streptococcal protein G. This engineered virus failed to enter cells through authentic receptors. In contrast, when this virus was conjugated with an antibody specific to an antigen on the cell membrane, it specifically entered cells expressing the cognate antigen. This virus was used as a probe to identify antibodies that mediate virus entry via recognition of certain molecules on the cell membrane other than authentic receptors. Using this method, we identified an antibody specific to epiregulin (EREG), which has been investigated mainly as a secreted growth factor and not necessarily for its precursor that is expressed in a transmembrane form. We constructed an scFv from the anti-EREG antibody for insertion into the retargeted HSV platform and found that the recombinant virus entered cells specifically through EREG expressed by the cells. This novel antibody-screening system may contribute to the discovery of unique and unexpected molecules that might be used for the entry of receptor-retargeted oncolytic HSVs.IMPORTANCE The tropism of the cellular entry of HSV is dependent on the binding of the envelope gD to one of its authentic receptors. This can be fully retargeted to other receptors by inserting scFvs into gD with appropriate modifications. In theory, upon binding to the engineered gD, receptors other than authentic receptors should induce a conformational change in the gD, which activates downstream mechanisms required for viral entry. However, prerequisite factors for receptors to be used as targets of a retargeted virus remain poorly understood, and it is difficult to predict which molecules might be suitable for our retargeted HSV construct. Our HSV-based probe will allow unbiased screening of antibody-antigen pairs that mediate virus entry and might be a useful tool to identify suitable pairs for our construct and to enhance our understanding of virus-cell interactions during infection by HSV and possibly other viruses.


Asunto(s)
Epirregulina/metabolismo , Herpesvirus Humano 1/metabolismo , Virus Oncolíticos/fisiología , Anticuerpos de Cadena Única/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Células CHO , Línea Celular Tumoral , Chlorocebus aethiops , Cricetulus , Humanos , Neoplasias/terapia , Viroterapia Oncolítica , Células Vero , Tropismo Viral
2.
J Virol ; 90(24): 11096-11105, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27707922

RESUMEN

Membrane fusion, which is the key process for both initial cell entry and subsequent lateral spread of herpes simplex virus (HSV), requires the four envelope glycoproteins gB, gD, gH, and gL. Syncytial mutations, predominantly mapped to the gB and gK genes, confer hyperfusogenicity on HSV and cause multinucleated giant cells, termed syncytia. Here we asked whether interaction of gD with a cognate entry receptor remains indispensable for initiating membrane fusion of syncytial strains. To address this question, we took advantage of mutant viruses whose viral entry into cells relies on the uniquely specific interaction of an engineered gD with epidermal growth factor receptor (EGFR). We introduced selected syncytial mutations into gB and/or gK of the EGFR-retargeted HSV and found that these mutations, especially when combined, enabled formation of extensive syncytia by human cancer cell lines that express the target receptor; these syncytia were substantially larger than the plaques formed by the parental retargeted HSV strain. We assessed the EGFR dependence of entry and spread separately by using direct entry and infectious center assays, respectively, and we found that the syncytial mutations did not override the receptor specificity of the retargeted viruses at either stage. We discuss the implications of these results for the development of more effective targeted oncolytic HSV vectors. IMPORTANCE: Herpes simplex virus (HSV) is investigated not only as a human pathogen but also as a promising agent for oncolytic virotherapy. We previously showed that both the initial entry and subsequent lateral spread of HSV can be retargeted to cells expressing tumor-associated antigens by single-chain antibodies fused to a receptor-binding-deficient envelope glycoprotein D (gD). Here we introduced syncytial mutations into the gB and/or gK gene of gD-retargeted HSVs to determine whether viral tropism remained dependent on the interaction of gD with the target receptor. Entry and spread profiles of the recombinant viruses indicated that gD retargeting does not abolish the hyperfusogenic activity of syncytial mutations and that these mutations do not eliminate the dependence of HSV entry and spread on a specific gD-receptor interaction. These observations suggest that syncytial mutations may be valuable for increasing the tumor-specific spreading of retargeted oncolytic HSV vectors.


Asunto(s)
Receptores ErbB/metabolismo , Herpesvirus Humano 1/genética , Mutación , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/genética , Animales , Células CHO , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Cricetulus , Receptores ErbB/genética , Expresión Génica , Células Gigantes/metabolismo , Células Gigantes/ultraestructura , Células Gigantes/virología , Herpesvirus Humano 1/metabolismo , Interacciones Huésped-Patógeno , Humanos , Fusión de Membrana , Mutagénesis Sitio-Dirigida , Viroterapia Oncolítica , Receptores Virales/genética , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
3.
Biochem Biophys Res Commun ; 458(4): 877-82, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25701778

RESUMEN

TROP-2 is a type Ⅰ transmembrane glycoprotein that is highly expressed in various epithelial cancer cells, and its increased expression correlates with poor prognosis. Although several anti-TROP-2 antibodies have been described, they were found unsuitable for antitumor therapy use in vivo as naked antibodies. In this study, we established a novel anti-TROP-2 antibody, designated Pr1E11, from mice immunized with primary prostate cancer cells. Antibody screening was based on the infection activity of Adv-LacZ-FZ33, which displays an immunoglobulin G binding domain in the adenoviral fiber protein. We found that Pr1E11 specifically binds to TROP-2 with high affinity and recognizes diverse epithelial cancer cell lines and primary pancreatic cancer tissues. Epitope analysis using TROP-2 deletion mutants revealed that binding site of Pr1E11 is a cysteine-rich domain, a unique epitope compared with other available anti-TROP-2 antibodies. In addition, Pr1E11 exhibited low internalization activity, which may make it suitable for naked antibody therapeutics. Our results suggest that Pr1E11 may stimulate different biological activities from other anti-TROP-2 antibodies based on its unique binding epitope, and is a potential candidate for naked antibody therapeutics for various epithelial cancer treatments.


Asunto(s)
Adenoviridae/inmunología , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Epítopos/inmunología , Neoplasias de la Próstata/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Humanos , Masculino , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Próstata/inmunología , Eliminación de Secuencia , Células Tumorales Cultivadas
4.
Biochem Biophys Res Commun ; 454(4): 600-3, 2014 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-25450699

RESUMEN

Antibody-drug conjugates (ADCs), drugs developed by conjugation of an anticancer agent to a monoclonal antibody (mAb), have lately attracted attention in cancer therapy because ADCs can directly bind cancer cells and kill them. Although mAbs for ADCs must be internalized by the target cells, few methods are available for screening mAbs for their ability to be internalized by cells. We have developed a recombinant protein, termed DT3C, which consists of diphtheria toxin (DT) lacking the receptor-binding domain but containing the C1, C2, and C3 domains of Streptococcus protein G (3C). When a mAb-DT3C conjugate, which functions in vitro like an ADC, reduces the viability of cancer cells, the mAb being tested must have been internalized by the target cells. DT3C can thus be a tool to identify efficiently and easily mAbs that can be internalized by cells, thereby enhancing the development of promising ADCs.


Asunto(s)
Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Supervivencia Celular/efectos de los fármacos , Toxina Diftérica/química , Toxina Diftérica/inmunología , Toxina Diftérica/farmacología , Toxina Diftérica/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Recombinantes/análisis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
5.
Int J Cancer ; 133(6): 1479-88, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23444104

RESUMEN

Oncolytic viruses (OVs) are novel cancer therapeutics with great promise, but host antiviral immunity represents the hurdle for their efficacy. Immunosuppression by cyclophosphamide (CP) has thus been shown to enhance the oncolytic efficacy of many OVs, but its effects on OVs armed with therapeutic genes remain unknown. We have previously reported on the efficacy of AxE1CAUP, an oncolytic adenovirus (OAd) expressing uracil phosphoribosyltransferase (UPRT), an enzyme that markedly enhanced the toxicity of 5-fluorouracil (5-FU), in immunodeficient, Ad-nonpermissive nude mice. Here we explored the efficacy and safety of intratumoral (i.t.) AxE1CAUP/5-FU therapy and of its combination with CP for syngenic HaP-T1 pancreatic cancers in immunocompetent, Ad-permissive Syrian hamsters. AxE1CAUP infected, replicated, expressed UPRT, and increased the sensitivity to 5-FU in HaP-T1 cells in vitro. I.t. AxE1CAUP/5-FU treatment inhibited the growth of subcutaneous HaP-T1 allografts. The combination with high-dose CP inhibited serum Ad-neutralizing antibody formation, increased intratumoral AxE1CAUP replication and UPRT expression, and resulted in further enhanced therapeutic effects with 5-FU. Neither body weight nor histology of the liver and lung changed during these treatments. A clinically-approved, intermediate-dose CP also enhanced the efficacy of i.t. AxE1CAUP/5-FU treatment in these hamsters, which was not affected by preexisting immunity to the vector. These data demonstrate the excellent antitumor efficacy and safety of an OAd armed with a suicide gene in combination with CP for treating syngenic tumors in immunocompetent, Ad-permissive animals, indicating the efficacy of CP in overcoming the hurdle of antiviral immunity for effective OV-mediated gene therapy.


Asunto(s)
Ciclofosfamida/uso terapéutico , Virus Oncolíticos/genética , Neoplasias Pancreáticas/terapia , Pentosiltransferasa/genética , Animales , Línea Celular Tumoral , Cricetinae , Femenino , Fluorouracilo/uso terapéutico , Inmunocompetencia , Mesocricetus , Transducción Genética
6.
Nature ; 446(7134): 444-8, 2007 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-17334357

RESUMEN

Cardiac hypertrophy occurs as an adaptive response to increased workload to maintain cardiac function. However, prolonged cardiac hypertrophy causes heart failure, and its mechanisms are largely unknown. Here we show that cardiac angiogenesis is crucially involved in the adaptive mechanism of cardiac hypertrophy and that p53 accumulation is essential for the transition from cardiac hypertrophy to heart failure. Pressure overload initially promoted vascular growth in the heart by hypoxia-inducible factor-1 (Hif-1)-dependent induction of angiogenic factors, and inhibition of angiogenesis prevented the development of cardiac hypertrophy and induced systolic dysfunction. Sustained pressure overload induced an accumulation of p53 that inhibited Hif-1 activity and thereby impaired cardiac angiogenesis and systolic function. Conversely, promoting cardiac angiogenesis by introducing angiogenic factors or by inhibiting p53 accumulation developed hypertrophy further and restored cardiac dysfunction under chronic pressure overload. These results indicate that the anti-angiogenic property of p53 may have a crucial function in the transition from cardiac hypertrophy to heart failure.


Asunto(s)
Gasto Cardíaco Bajo/fisiopatología , Cardiomegalia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Aorta/patología , Aorta/fisiopatología , Presión Sanguínea , Cardiomegalia/patología , Circulación Coronaria , Progresión de la Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Neovascularización Patológica , Proteína p53 Supresora de Tumor/genética
7.
Br J Haematol ; 157(6): 674-86, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22463758

RESUMEN

The regulation of human early lymphopoiesis remains unclear. B- and T-lineage cells cannot develop simultaneously with conventional stromal cultures. Here we show that telomerized human bone marrow stromal cells supported simultaneous generation of CD19(+) CD34(lo/-) CD10(+) cyCD79a(+) CD20(+/-) VpreB(-) pro-B cells and CD7(+) CD34(+) CD45RA(+) CD56(-) cyCD3(-) early T/Natural Killer (NK) cell precursors from human haematopoietic progenitors, and the generation of both lymphoid precursors was promoted by flt3 ligand (flt3L). On the other hand, stem cell factor or thrombopoietin had little or no effect when used alone. However, both acted synergistically with flt3L to augment the generation of both lymphoid precursors. Characteristics of these lymphoid precursors were evaluated by gene expression profiles, rearrangements of IgH genes, or replating assays. Similar findings were observed with primary human bone marrow stromal cells. Notably, these two lymphoid-lineage precursors were generated without direct contact with stromal cells, indicating that early B and T/NK development can occur, at least in part, by stromal cell-derived humoral factors. In serum-free cultures, flt3L elicited similar effects and appeared particularly important for B cell development. The findings of this study identified the potential of human bone marrow stromal cells to support human early B and T lymphopoiesis and a principal role for flt3L during early lymphopoiesis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Linfocitos B/metabolismo , Células Asesinas Naturales/metabolismo , Células Progenitoras Linfoides/metabolismo , Linfopoyesis/efectos de los fármacos , Proteínas de la Membrana/farmacología , Linfocitos T/metabolismo , Antígenos CD/biosíntesis , Linfocitos B/citología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Reordenamiento Génico de Cadena Pesada de Linfocito B/efectos de los fármacos , Reordenamiento Génico de Cadena Pesada de Linfocito B/fisiología , Humanos , Células Asesinas Naturales/citología , Células Progenitoras Linfoides/citología , Linfopoyesis/fisiología , Masculino , Células del Estroma/citología , Células del Estroma/metabolismo , Linfocitos T/citología
8.
Biochem Biophys Res Commun ; 421(2): 349-54, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22510406

RESUMEN

The interaction between pancreatic cancer cells and pancreatic stellate cells (PSCs), a major profibrogenic cell type in the pancreas, is receiving increasing attention. There is accumulating evidence that PSCs promote the progression of pancreatic cancer by increasing cancer cell proliferation and invasion as well as by protecting them from radiation- and gemcitabine-induced apoptosis. Recent studies have identified that a portion of cancer cells, called "cancer stem cells", within the entire cancer tissue harbor highly tumorigenic and chemo-resistant phenotypes, which lead to the recurrence after surgery or re-growth of the tumor. The mechanisms that maintain the "stemness" of these cells remain largely unknown. We hypothesized that PSCs might enhance the cancer stem cell-like phenotypes in pancreatic cancer cells. Indirect co-culture of pancreatic cancer cells with PSCs enhanced the spheroid-forming ability of cancer cells and induced the expression of cancer stem cell-related genes ABCG2, Nestin and LIN28. In addition, co-injection of PSCs enhanced tumorigenicity of pancreatic cancer cells in vivo. These results suggested a novel role of PSCs as a part of the cancer stem cell niche.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Fibroblastos/patología , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Humanos , Ratones , Trasplante de Neoplasias , Esferoides Celulares/patología
9.
Int J Cancer ; 129(5): 1244-53, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21710497

RESUMEN

A critical issue in adenovirus (Ad)-based cancer gene therapy is to improve the specificity of gene delivery to cancer cells for better efficacy and safety. We explored methods of retargeting Ad vectors for selective gene therapy of human biliary cancers using the Ad incorporating an IgG Fc-binding motif (Z33) from the Staphylococcus protein A (Ad-FZ33) combined with tumor-specific antibodies. Flow cytometry analysis revealed high-expression levels of epithelial cell adhesion molecule (EpCAM) and epidermal growth factor receptor (EGFR) on human biliary cancer cells. Ad-FZ33 expressing LacZ combined with antibodies against EpCAM or EGFR, followed by ß-gal assay, demonstrated highly efficient gene transduction in these biliary cancer cells, compared to the treatment with control antibody or without antibody. Ad-FZ33 expressing uracil phosphoribosyl transferase (UPRT), an enzyme which greatly enhances the toxicity of 5-fluorouracil (FU), combined with antibodies against EpCAM or EGFR, remarkably enhanced the sensitivity of biliary cancer cells to 5-FU. By contrast, the treatment did not affect the 5-FU sensitivity of the cells not expressing EpCAM or EGFR including normal hepatocytes. Finally, treatments with the UPRT-expressing Ad-FZ33 with antibodies against EpCAM or EGFR, followed by 5-FU administration, significantly suppressed the growth of biliary cancer xenografts in nude mice. These results indicate that the gene therapy mediated by the Z33 fiber modified Ad with anti-EpCAM or anti-EGFR antibodies offers a potentially effective therapeutic modality against biliary cancers.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Antígenos de Neoplasias/genética , Neoplasias del Sistema Biliar/terapia , Moléculas de Adhesión Celular/genética , Receptores ErbB/genética , Terapia Genética , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/inmunología , Western Blotting , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/inmunología , Terapia Combinada , Molécula de Adhesión Celular Epitelial , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Femenino , Citometría de Flujo , Fluorouracilo/uso terapéutico , Vectores Genéticos/uso terapéutico , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Estafilocócica A/genética , Células Tumorales Cultivadas
10.
Cancer Sci ; 102(4): 808-14, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21214674

RESUMEN

In adenovirus-derived gene therapy, one of the problems is the difficulty in specific targeting. We have recently demonstrated that monoclonal antibody (mAb) libraries screened by fiber-modified adenovirus vector (Adv-FZ33), which is capable of binding to immunoglobulin-G (IgG), provide a powerful approach for the identification of suitable target antigens for prostate cancer therapy. Hybridoma libraries from mice immunized with androgen-dependent prostate cancer cell line LNCaP were screened and mAb were selected. Through this screening, we obtained one mAb, designated LNI-29, that recognizes a glycoprotein with an apparent molecular mass of 100 kD. It was identified as neural cell adhesion molecule 2 (NCAM2). Some prostate and breast cancer cell lines highly expressed NCAM2 whereas normal prostate cell lines expressed NCAM2 at low levels. In contrast to the low efficiency of gene transduction by Adv-FZ33 with a control antibody, LNI-29-mediated Adv-FZ33 infection induces high rates of gene delivery in NCAM2-positive cancers. NCAM2-mediated therapeutic gene transduction of uracil phosphoribosyltransferase (UPRT) had a highly effective cytotoxic effect on NCAM2-positive cancer cells, whereas it had less of an effect in cases with a control antibody. In conclusion, NCAM2 should be a novel gene therapy target for the treatment of prostate and breast cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos/genética , Terapia Genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neoplasias de la Próstata/terapia , Adenoviridae/genética , Adenoviridae/inmunología , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Antimetabolitos Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Células Cultivadas , Femenino , Citometría de Flujo , Fluorouracilo/farmacología , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Hibridomas , Inmunización , Inmunoglobulina G/inmunología , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/inmunología , Moléculas de Adhesión de Célula Nerviosa , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , ARN Interferente Pequeño/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Células Tumorales Cultivadas
11.
Artículo en Inglés | MEDLINE | ID: mdl-20626230

RESUMEN

A higher production of functional mast cells (MCs) can be generated by co-culturing cord blood-derived CD34+ cells with a human bone marrow stromal cell line under serum-free conditions supplemented with stem cell factor and IL-6. We addressed the question of whether the higher proliferation of MCs in this co-culture system might be due to the higher proliferation of MC progenitors. The stromal cell line increased the cell numbers of MC progenitors derived from cord blood-derived CD34+ cells, in a combination of cell-cell interactions between stromal cells and CD34+ cells, and as yet unidentified soluble factors derived from stromal cells.


Asunto(s)
Proliferación Celular , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Interleucina-6/metabolismo , Mastocitos/citología , Factor de Células Madre/metabolismo , Antígenos CD34/metabolismo , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo/métodos , Medios de Cultivo Condicionados , Medio de Cultivo Libre de Suero , Sangre Fetal/inmunología , Humanos , Mastocitos/metabolismo , Células Madre Mesenquimatosas/citología , Células del Estroma/citología , Células del Estroma/metabolismo
12.
Mol Cancer Ther ; 20(12): 2329-2340, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34413126

RESUMEN

Trophoblast cell surface antigen 2 (TROP2) is highly expressed on various epithelial tumors and correlates with poor prognosis. We developed the novel TROP2-directed antibody-drug conjugate (ADC), datopotamab deruxtecan (Dato-DXd, DS-1062a), with a potent DNA topoisomerase I inhibitor (DXd), and evaluated its antitumor activity and safety profiles in preclinical models.The pharmacologic activity and mechanism of action of Dato-DXd were investigated in several human cancer cell lines and xenograft mouse models including patient-derived xenograft (PDX) models. Safety profiles were also assessed in rats and cynomolgus monkeys.Dato-DXd bound specifically to TROP2 and was internalized into tumor cells followed by intracellular trafficking to lysosome and DXd release, which induced DNA damage and apoptosis in TROP2-expressing tumor cells in vitro. Dato-DXd exhibited in vivo antitumor activity with DNA damage induced by the accumulated DXd in TROP2-expressing xenograft tumors, but neither isotype control IgG-ADC nor anti-TROP2 antibody had this effect. Dato-DXd also showed potent antitumor activity with tumor regression in several TROP2-expressing xenograft tumors including NSCLC PDX models. Safety profiles of Dato-DXd in rats and cynomolgus monkeys were acceptable.Dato-DXd demonstrated potent antitumor activity against TROP2-expressing tumors by efficient payload delivery into tumors and acceptable safety profiles in preclinical models. These results suggest Dato-DXd could be a valuable treatment option for patients with TROP2-expressing tumors in the clinical setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Inmunoconjugados/uso terapéutico , Animales , Antineoplásicos/farmacología , Humanos , Inmunoconjugados/farmacología , Macaca fascicularis , Masculino , Ratones , Ratones Desnudos , Ratas
13.
J Neurosci ; 29(47): 14932-41, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19940189

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) derived from bone marrow has been shown to improve functional outcome in spinal cord injury (SCI). We transplanted MSCs derived from human bone marrow (hMSCs) to study their potential therapeutic effect in SCI in the rat. In addition to hMSCs, we used gene-modified hMSCs to secrete brain-derived neurotrophic factor (BDNF-hMSCs). After a dorsal transection lesion was induced at T9, cells were microinjected on each side of the transection site. Fluorogold (FG) was injected into the epicenter of the lesion cavity to identify transected corticospinal tract (CST) neurons. At 5 weeks after transplantation, the animals were perfused. Locomotor recovery improvement was observed for the BDNF-hMSC group, but not in the hMSC group. Structurally there was increased sprouting of injured corticospinal tract and serotonergic projections after hMSC and BDNF-hMSC transplantation. Moreover, an increased number of serotonergic fibers was observed in spinal gray matter including the ventral horn at and below the level of the lesion, indicating increased innervation in the terminal regions of a descending projection important for locomotion. Stereological quantification was performed on the brains to determine neuronal density in primary motor (M1) cortex. The number of FG backfilled cells demonstrated an increased cell survival of CST neurons in M1 cortex in both the hMSC and BDNF-hMSC groups at 5 weeks, but the increase for the BDNF-hMSC group was greater. These results indicate that transplantation of hMSCs hypersecreting BDNF results in structural changes in brain and spinal cord, which are associated with improved functional outcome in acute SCI.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Regeneración Nerviosa/fisiología , Tractos Piramidales/cirugía , Traumatismos de la Médula Espinal/cirugía , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Citoprotección/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica/fisiología , Vectores Genéticos/farmacología , Vectores Genéticos/uso terapéutico , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Humanos , Plasticidad Neuronal/fisiología , Tractos Piramidales/citología , Tractos Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Transfección/métodos , Trasplante Heterólogo/métodos , Resultado del Tratamiento
14.
Angiogenesis ; 13(3): 203-10, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20458615

RESUMEN

Sendai viral vector (SeV) is emerging as a promising vector for gene therapy. However, little information is available regarding the combination of SeV-mediated gene and mesenchymal stem cell (MSC) therapy in dealing with ischemic diseases. In this study, we infected SeV to the MSCs in vitro; and injected MSCs modified with SeV harboring human angiopoietin-1 gene (SeVhAng-1) into the ischemic limb of rats in vivo. We found SeV had high transductive efficiency to the MSCs. Both MSCs and SeVhAng-1-modified MSCs improved the blood flow recovery and increased the capillary density of the ischemic limb, compared with the control. However, in contrast to MSCs, SeVhAng-1-modified MSCs had a better improvement of blood flow recovery in the ischemic limb. We further found the ischemic limb injected with SeVhAng-1-modified MSCs had strong expression of p-Akt, which improved survival of MSCs injected into the ischemic limb. This indicated SeVhAng-1 modification enhanced angiogenetic effect of MSCs by both angiogenesis and cell protection. We conclude that SeVhAng-1-modified MSCs may serve as a more effective tool in dealing with ischemic limb disease.


Asunto(s)
Angiopoyetina 1/genética , Angiopoyetina 1/uso terapéutico , Extremidades/irrigación sanguínea , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Virus Sendai/genética , Animales , Capilares/metabolismo , Capilares/patología , Extremidades/patología , Terapia Genética , Vectores Genéticos/genética , Humanos , Inyecciones , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Lew , Flujo Sanguíneo Regional , Análisis de Supervivencia , Transducción Genética
15.
J Virol ; 83(11): 5918-27, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297502

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs) have been reported to migrate to brain lesions in experimental models of ischemia, tumors, and neurodegenerative diseases and to ameliorate functional deficits. In this study, we attempted to evaluate the therapeutic potential of MSCs for treating prion diseases. Immortalized human MSCs (hMSCs) that express the LacZ gene were transplanted into the unilateral hippocampi or thalami of mice, and their distributions were monitored by the expression of beta-galactosidase. In mice infected with prions, hMSCs transplanted at 120 days postinoculation (dpi) were detected on the contralateral side at 2 days after transplantation and existed there even at 3 weeks after transplantation. In contrast, few hMSCs were detected on the contralateral side for mock-infected mice. Interestingly, the migration of hMSCs appeared to correlate with the severity of neuropathological lesions, including disease-specific prion protein deposition. The hMSCs also migrated to a prion-specific lesion in the brain, even when intravenously injected. Although the effects were modest, intrahippocampal and intravenous transplantation of hMSCs prolonged the survival of mice infected with prions. A subpopulation of hMSCs in the brains of prion-infected mice produced various trophic factors and differentiated into cells of neuronal and glial lineages. These results suggest that MSCs have promise as a cellular vehicle for the delivery of therapeutic genes to brain lesions associated with prion diseases and, furthermore, that they may help to regenerate neuronal tissues damaged by prion propagation.


Asunto(s)
Diferenciación Celular/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Enfermedades por Prión/inmunología , Animales , Movimiento Celular/inmunología , Proliferación Celular , Femenino , Humanos , Ratones , Enfermedades por Prión/patología , Enfermedades por Prión/cirugía , Tasa de Supervivencia
16.
Angiogenesis ; 12(3): 243-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19322669

RESUMEN

Sendai virus vector is emerging as a promising vector for gene therapy, and angiopoietin-1 (Ang-1) has been reported to improve the blood flow recovery in the ischemic limb or heart. In this study, we constructed a human Ang-1-expressing Sendai viral vector (SeVhAng-1) and injected it into the ischemic limb of rats. We found that SeVhAng-1 improved the blood flow recovery and increased the capillary density of the ischemic limb, compared with the controls. We also found that SeVhAng-1 increased p-Akt during the early period of limb ischemia, and decreased apoptosis in ischemic limb. It suggests that SeVhAng-1 may serve as a potential therapeutic tool in ischemic limb disease.


Asunto(s)
Angiopoyetina 1/administración & dosificación , Vectores Genéticos , Isquemia/terapia , Extremidad Inferior/irrigación sanguínea , Enfermedades Vasculares Periféricas/terapia , Virus Sendai/genética , Angiopoyetina 1/genética , Animales , Constricción Patológica/genética , Constricción Patológica/patología , Constricción Patológica/terapia , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Arteria Ilíaca/patología , Inyecciones Intramusculares , Isquemia/genética , Isquemia/patología , Extremidad Inferior/patología , Masculino , Enfermedades Vasculares Periféricas/genética , Enfermedades Vasculares Periféricas/patología , Ratas , Ratas Endogámicas Lew , Virus Sendai/fisiología
17.
Hepatology ; 48(3): 828-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18756484

RESUMEN

UNLABELLED: Although conditionally replicable adenovirus (CRAd) has been used in the clinical treatment of hepatocellular carcinoma (HCC), it suffers from the inherent drawback of having relatively low antitumor activity. Here, we have sought to overcome this drawback. First, we combined CRAd (AdAFPep/Rep) driven by alpha-fetoprotein enhancer/promoter (AFPep) with a replication-incompetent adenovirus carrying a p53 transgene that is also driven by AFPep. The synergism of this combination produced a significantly improved tumoricidal effect on the human HCC cell line Hep3B, which has a relatively short doubling time in comparison with other human HCC cell lines, through the transactivation of p53 by early region 1A transcribed by AdAFPep/Rep. This synergistic interaction was augmented by the addition of a subtumoricidal dose (0.5 microg/mL) of 5-fluorouracil (5-FU), which enhanced p53 expression and facilitated the release of virions from tumor cells. When relatively large (10-mm-diameter) Hep3B tumors grown in nude mice were injected with the two viruses in combination, they showed significantly impaired growth in comparison with those treated with each virus separately. The growth suppression effect of the virus combination was enhanced by a low dose (600 microg) of 5-FU. Survival of the tumor-bearing mice treated with these three agents was significantly longer than that of control mice. Moreover, the tumor completely disappeared with the repeated injection of these agents. CONCLUSION: This combination strategy holds promise for the treatment of relatively large and rapidly growing HCCs that may be encountered clinically.


Asunto(s)
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Fluorouracilo/uso terapéutico , Neoplasias Hepáticas/terapia , Virus Oncolíticos/genética , Proteína p53 Supresora de Tumor/genética , alfa-Fetoproteínas/genética , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Células Cultivadas , Terapia Combinada , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica/métodos , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-Fetoproteínas/metabolismo
18.
J Surg Res ; 157(1): e63-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19345377

RESUMEN

BACKGROUND: The prognosis of bile duct cancer is quite poor because of the low resection rate and the tolerance of the cancer to chemotherapy and radiotherapy. We investigated the feasibility of an oncolytic adenovector with two suicide genes for the treatment of bile duct cancer. MATERIALS AND METHODS: We developed a new conditionally replicating adenovirus (AxE1CAUT) with the uracil phosphoribosyltransferase (UPRT) gene and the herpes simplex virus thymidine kinase (HSV-tk) gene, and compared its antitumor effects with a replication defective adenovector (AxCAUT) that has both the UPRT and HSV-tk genes. We evaluated the effects of these adenoviruses with 5-fluorouracil (5-FU) and/or ganciclovir (GCV) on human cholangiocarcinoma cells (HuCCT1, with mutant p53) in vitro and in vivo. RESULTS: The drug sensitivity of HuCCT1 cells to 5-FU and/or GCV was increased with an increase in the multiplicity of infection (MOI). The antitumor effect increased when 5-FU and GCV were given at the same time. Subcutaneous tumors of nude mice directly injected with AxCAUT showed a higher response to 5-FU/GCV than 5-FU or GCV alone, but there was no difference between AxCAUT and AxE1CAUT. However, AxE1CAUT with 5-FU/GCV produced a decrease in tumor weight and better survival than AxCAUT in a peritoneal dissemination model infected by intraperitoneal administration of the adenovectors. CONCLUSION: Oncolytic double suicide gene therapy is effective against human cholangiocarcinoma cells in nude mouse models.


Asunto(s)
Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos , Colangiocarcinoma/terapia , Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Adenoviridae/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Antivirales/farmacología , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Línea Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Terapia Combinada , Femenino , Fibroblastos/citología , Fluorouracilo/farmacología , Ganciclovir/farmacología , Herpes Simple/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Wound Repair Regen ; 17(3): 436-46, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19660053

RESUMEN

In this study, we regenerated skin and its appendages by transplanting cultured normal dermal fibroblasts, into which morphogen genes had been introduced. We cultured normal dermal fibroblasts obtained from Fisher 344 rats on the surface of hydroxyapatite beads, and then adsorbed them onto the surface of a collagen sponge, which was transplanted into a full-thickness skin defect prepared on the backs of rats. Before transplantation, genes were introduced into the dermal fibroblasts via adenovirus vector (ad)-bone morphogenetic protein 2 and ad-wingless int 3 genes in addition to fibroblast growth factor-2 protein. By Week 4, the appearance of follicle germs or primitive hair germs was observed only in the ad-bone morphogenetic protein 2+ad-wingless int 3 combined with the fibroblast growth factor-2 protein group. By Week 16, in that same group, hair follicles having mature pilosebaceous systems with equally spaced localization had formed in the ulcer wound.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/trasplante , Folículo Piloso/fisiología , Regeneración/fisiología , Proteínas Wnt/genética , Heridas y Lesiones/cirugía , Adenoviridae/genética , Animales , Proteína Morfogenética Ósea 2/metabolismo , Dermis/citología , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos , Ratas , Ratas Endogámicas F344 , Trasplante de Piel/métodos , Ingeniería de Tejidos , Proteínas Wnt/metabolismo , Proteína Wnt3 , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología , Proteínas de Xenopus
20.
Sci Rep ; 9(1): 1281, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718742

RESUMEN

Malignant melanoma is one of the untreatable cancers in which conventional therapeutic strategies, including chemotherapy, are hardly effective. Therefore, identification of novel therapeutic targets involved in melanoma progression is urgently needed for developing effective therapeutic methods. Overexpression of interleukin-13 receptor α2 (IL13Rα2) is observed in several cancer types including glioma and pancreatic cancer. Although IL13Rα2 is implicated in the progression of various types of cancer, its expression and roles in the malignant melanoma have not yet been elucidated. In the present study, we showed that IL13Rα2 was expressed in approximately 7.5% melanoma patients. While IL13Rα2 expression in human melanoma cells decreased their proliferation in vitro, it promoted in vivo tumour growth and angiogenesis in melanoma xenograft mouse model. We also found that the expression of amphiregulin, a member of the epidermal growth factor (EGF) family, was correlated with IL13Rα2 expression in cultured melanoma cells, xenograft tumour tissues and melanoma clinical samples. Furthermore, expression of amphiregulin promoted tumour growth, implicating causal relationship between the expression of IL13Rα2 and amphiregulin. These results suggest that IL13Rα2 enhances tumorigenicity by inducing angiogenesis in malignant melanoma, and serves as a potential therapeutic target of malignant melanoma.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Subunidad alfa2 del Receptor de Interleucina-13/biosíntesis , Melanoma/metabolismo , Proteínas de Neoplasias/biosíntesis , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Humanos , Subunidad alfa2 del Receptor de Interleucina-13/genética , Melanoma/genética , Melanoma/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA