Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Intervalo de año de publicación
1.
Mol Oncol ; 13(11): 2441-2459, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31545551

RESUMEN

Advanced colorectal carcinoma is currently incurable, and new therapies are urgently needed. We report that phosphotyrosine-dependent Eph receptor signaling sustains colorectal carcinoma cell survival, thereby uncovering a survival pathway active in colorectal carcinoma cells. We find that genetic and biochemical inhibition of Eph tyrosine kinase activity or depletion of the Eph ligand EphrinB2 reproducibly induces colorectal carcinoma cell death by autophagy. Spautin and 3-methyladenine, inhibitors of early steps in the autophagic pathway, significantly reduce autophagy-mediated cell death that follows inhibition of phosphotyrosine-dependent Eph signaling in colorectal cancer cells. A small-molecule inhibitor of the Eph kinase, NVP-BHG712 or its regioisomer NVP-Iso, reduces human colorectal cancer cell growth in vitro and tumor growth in mice. Colorectal cancers express the EphrinB ligand and its Eph receptors at significantly higher levels than numerous other cancer types, supporting Eph signaling inhibition as a potential new strategy for the broad treatment of colorectal carcinoma.


Asunto(s)
Autofagia , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Terapia Molecular Dirigida , Receptores de la Familia Eph/metabolismo , Transducción de Señal , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Efrina-B2/metabolismo , Femenino , Silenciador del Gen/efectos de los fármacos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
2.
J Natl Cancer Inst ; 110(4): 390-399, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29202196

RESUMEN

Background: Deleted in Liver Cancer 1 (DLC1) is a tumor suppressor gene frequently deleted in cancer. However, DLC1 is not known to be deleted in angiosarcoma, an aggressive malignancy of endothelial cell derivation. Additionally, the physiologic functions of DLC1 protein in endothelial cells are poorly defined. Methods: We investigated the effects of shRNA-induced DLC1 depletion in endothelial cells. Cell growth was measured by 3H thymidine incorporation, IncuCyte imaging, and population doublings; cell death by cell cycle analysis; gene expression by Affimetrix arrays and quantitative polymerase chain reaction; NF-κB activity by reporter assays; and protein levels by immunoblotting and immunofluorescence staining. We tested Tanespimycin/17-AAG and Fasudil treatment in groups of nine to 10 mice bearing ISOS-1 angiosarcoma. All statistical tests were two-sided. Results: We discovered that DLC1 is a critical regulator of cell contact inhibition of proliferation in endothelial cells, promoting statistically significant (P < .001) cell death when cells are confluent (mean [SD] % viability: control DLC1 = 15.6 [19.3]; shDLC1 = 73.4 [13.1]). This prosurvival phenotype of DLC1-depleted confluent endothelial cells is attributable to a statistically significant and sustained increase of NF-κB activity (day 5, P = .001; day 8, P = .03) associated with increased tumor necrosis factor alpha-induced protein 3 (TNFAIP3/A20) signaling. Consistently, we found that DLC1 is statistically significantly reduced (P < .001 in 5 of 6) and TNFAIP3/A20 is statistically significantly increased (P < .001 in 2 of 3 and P = 0.02 in 1 of 3) in human angiosarcoma compared with normal adjacent endothelium. Treatment with the NF-κB inhibitor Tanespimycin/17-AAG statistically significantly reduced angiosarcoma tumor growth in mice (treatment tumor weight vs control, 0.50 [0.19] g vs 0.91 [0.21] g, P = .001 experiment 1; 0.66 [0.26] g vs 1.10 [0.31] g, P = .01 experiment 2). Conclusions: These results identify DLC1 as a previously unrecognized regulator of endothelial cell contact inhibition of proliferation that is depleted in angiosarcoma and support NF-κB targeting for the treatment of angiosarcoma where DLC1 is lost.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Clusterina/metabolismo , Inhibición de Contacto , Proteínas Activadoras de GTPasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Hemangiosarcoma/patología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Biomarcadores de Tumor/genética , Ciclo Celular , Movimiento Celular , Proliferación Celular , Clusterina/genética , Progresión de la Enfermedad , Femenino , Proteínas Activadoras de GTPasa/genética , Hemangiosarcoma/genética , Hemangiosarcoma/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Pronóstico , Transducción de Señal , Células Tumorales Cultivadas , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteínas Supresoras de Tumor/genética
3.
J Natl Cancer Inst ; 109(9)2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28376153

RESUMEN

Background: Primary effusion lymphoma (PEL) is a Kaposi's sarcoma herpes virus (KSHV)-induced lymphoma that typically arises in body cavities of HIV-infected patients. PEL cells are often co-infected with Epstein-Barr virus (EBV). "PEL-like" lymphoma is a KSHV-unrelated lymphoma that arises in body cavities of HIV-negative patients. "PEL-like" lymphoma is sometimes EBV positive. The derivation of PEL/"PEL-like" cells is unclear. Methods: Mesothelial cells were cultured from body cavity effusions of 23 patients. Cell proliferation, cytokine secretion, marker phenotypes, KSHV/EBV infection, and clonality were evaluated by standard methods. Gene expression was measured by quantitative polymerase chain reaction and immunoblotting. A mouse model of PEL (3 mice/group) was used to evaluate tumorigenicity. Results: We found that the mesothelia derived from six effusions of HIV-infected patients with PEL or other KSHV-associated diseases contained rare KSHV + or EBV + mesothelial cells. After extended culture (16-17 weeks), some mesothelial cells underwent a trans-differentiation process, generating lymphoid-type CD45 + /B220 + , CD5 + , CD27 + , CD43 + , CD11c + , and CD3 - cells resembling "B1-cells," most commonly found in mouse body cavities. These "B1-like" cells were short lived. However, long-term KSHV + EBV - and EBV + KSHV - clonal cell lines emerged from mesothelial cultures from two patients that were clonally distinct from the monoclonal or polyclonal B-cell populations found in the patients' original effusions. Conclusions: Mesothelial-to-lymphoid transformation is a newly identified in vitro process that generates "B1-like" cells and is associated with the emergence of long-lived KSHV or EBV-infected cell lines in KSHV-infected patients. These results identify mesothelial cultures as a source of PEL cells and lymphoid cells in humans.


Asunto(s)
Epitelio/patología , Linfoma de Efusión Primaria/patología , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 8/aislamiento & purificación , Humanos , Linfoma de Efusión Primaria/virología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Adulto Joven
4.
Trends Biotechnol ; 33(5): 292-301, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25819764

RESUMEN

Antibody-based drugs represent one of the most successful and promising therapeutic approaches in oncology. Large combinatorial phage antibody libraries are available for the identification of therapeutic antibodies and various technologies exist for their further conversion into multivalent and multispecific formats optimized for the desired pharmacokinetics and the pathological context. However, there is no technology for antigen profiling of intact tumors to identify tumor markers targetable with antibodies. Such constraints have led to a relative paucity of tumor-associated antigens for antibody targeting in oncology. Here we review novel approaches aimed at the identification of antibody-targetable, accessible antigens in intact tumors. We hope that such advanced selection approaches will be useful in the development of next-generation antibody therapies for cancer.


Asunto(s)
Anticuerpos Antineoplásicos/química , Antineoplásicos/química , Oncología Médica/tendencias , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Neoplasias/química , Antineoplásicos/uso terapéutico , Técnicas de Visualización de Superficie Celular , Descubrimiento de Drogas , Humanos , Neoplasias/inmunología , Microambiente Tumoral
5.
Cancer Res ; 74(7): 2038-49, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24520074

RESUMEN

Myeloid cells that orchestrate malignant progression in the tumor microenvironment offer targets for a generalized strategy to attack solid tumors. Through an analysis of tumor microenvironments, we explored an experimental model of lung cancer that uncovered a network of Dll4/Notch/TGF-ß1 signals that links myeloid cells to cancer progression. Myeloid cells attracted to the tumor microenvironment by the tumor-derived cytokines CCL2 and M-CSF expressed increased levels of the Notch ligand Dll4, thereby activating Notch signaling in the tumor cells and amplifying tumor-intrinsic Notch activation. Heightened Dll4/Notch signaling in tumor cells magnified TGF-ß-induced pSMAD2/3 signaling and was required to sustain TGF-ß-induced tumor cell growth. Conversely, Notch blockade reduced TGF-ß signaling and limited lung carcinoma tumor progression. Corroborating these findings, by interrogating RNAseq results from tumor and adjacent normal tissue in clinical specimens of human head and neck squamous carcinoma, we found evidence that TGF-ß/Notch crosstalk contributed to progression. In summary, the myeloid cell-carcinoma signaling network we describe uncovers novel mechanistic links between the tumor microenvironment and tumor growth, highlighting new opportunities to target tumors where this network is active.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Células Mieloides/patología , Neoplasias Experimentales/patología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Microambiente Tumoral , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Proliferación Celular , Progresión de la Enfermedad , Femenino , Masculino , Ratones
7.
PLoS One ; 8(8): e72957, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23951338

RESUMEN

Metastatic breast cancer is the leading cause of death by malignancy in women worldwide. Tumor metastasis is a multistep process encompassing local invasion of cancer cells at primary tumor site, intravasation into the blood vessel, survival in systemic circulation, and extravasation across the endothelium to metastasize at a secondary site. However, only a small percentage of circulating cancer cells initiate metastatic colonies. This fact, together with the inaccessibility and structural complexity of target tissues has hampered the study of the later steps in cancer metastasis. In addition, most data are derived from in vivo models where critical steps such as intravasation/extravasation of human cancer cells are mediated by murine endothelial cells. Here, we developed a new mouse model to study the molecular and cellular mechanisms underlying late steps of the metastatic cascade. We have shown that a network of functional human blood vessels can be formed by co-implantation of human endothelial cells and mesenchymal cells, embedded within a reconstituted basement membrane-like matrix and inoculated subcutaneously into immunodeficient mice. The ability of circulating cancer cells to colonize these human vascularized organoids was next assessed in an orthotopic model of human breast cancer by bioluminescent imaging, molecular techniques and immunohistological analysis. We demonstrate that disseminated human breast cancer cells efficiently colonize organoids containing a functional microvessel network composed of human endothelial cells, connected to the mouse circulatory system. Human breast cancer cells could be clearly detected at different stages of the metastatic process: initial arrest in the human microvasculature, extravasation, and growth into avascular micrometastases. This new mouse model may help us to map the extravasation process with unprecedented detail, opening the way for the identification of relevant targets for therapeutic intervention.


Asunto(s)
Neoplasias de la Mama/patología , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Organoides/irrigación sanguínea , Organoides/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Desnudos
8.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23918357

RESUMEN

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Biomarcadores de Tumor/inmunología , Inmunoterapia/métodos , Proteínas Musculares/metabolismo , Neoplasias de la Próstata/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Anticuerpos Antineoplásicos/metabolismo , Especificidad de Anticuerpos , Western Blotting , Técnicas de Visualización de Superficie Celular , Cromatografía de Afinidad , Cromatografía Liquida , Sistemas de Liberación de Medicamentos/métodos , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/terapia , Estadísticas no Paramétricas , Espectrometría de Masas en Tándem
9.
Mol Ther Nucleic Acids ; 2: e93, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23695536

RESUMEN

A human single-chain variable fragment (scFv) antibody library was expressed on the surface of human T cells after transduction with lentiviral vectors (LVs). The repertoire was fused to a first-generation T cell receptor ζ (TCRζ)-based chimeric antigen receptor (CAR). We used this library to isolate antibodies termed CARbodies that recognize antigens expressed on the tumor cell surface in a proof-of-principle system. After three rounds of activation-selection there was a clear repertoire restriction, with the emergence dominant clones. The CARbodies were purified from bacterial cultures as soluble and active proteins. Furthermore, to validate its potential application for adoptive cell therapy, human T cells were transduced with a LV encoding a second-generation costimulatory CAR (CAR(v2)) bearing the selected CARbodies. Transduced human primary T cells expressed significant levels of the CARbodies-based CAR(v2) fusion protein on the cell surface, and importantly could be specifically activated, after stimulation with tumor cells. This approach is a promising tool for the generation of antibodies fully adapted to the display format (CAR) and the selection context (cell synapse), which could extend the scope of current adoptive cell therapy strategies with CAR-redirected T cells.Molecular Therapy-Nucleic Acids (2013) 2, e93; doi:10.1038/mtna.2013.19; published online 21 May 2013.

10.
MAbs ; 5(1): 70-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23221741

RESUMEN

Here, we describe a new class of multivalent and multispecific antibody-based reagents for therapy. The molecules, termed "trimerbodies," use a modified version of the N-terminal trimerization region of human collagen XVIII noncollagenous 1 domain flanked by two flexible linkers as trimerizing scaffold. By fusing single-chain variable fragments (scFv) with the same or different specificity to both N- and C-terminus of the trimerizing scaffold domain, we produced monospecific or bispecific hexavalent molecules that were efficiently secreted as soluble proteins by transfected mammalian cells. A bispecific anti-laminin x anti-CD3 N-/C-trimerbody was found to be trimeric in solution, very efficient at recognizing purified plastic-immobilized laminin and CD3 expressed at the surface of T cells, and remarkably stable in human serum. The bispecificity was further demonstrated in T cell activation studies. In the presence of laminin-rich substrate, the bispecific anti-laminin x anti-CD3 N-/C-trimerbody stimulated a high percentage of human T cells to express surface activation markers. These results suggest that the trimerbody platform offers promising opportunities for the development of the next-generation therapeutic antibodies, i.e., multivalent and bispecific molecules with a format optimized for the desired pharmacokinetics and adapted to the pathological context.


Asunto(s)
Anticuerpos Biespecíficos , Colágeno Tipo XVIII , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Complejo CD3/inmunología , Complejo CD3/metabolismo , Colágeno Tipo XVIII/química , Colágeno Tipo XVIII/genética , Colágeno Tipo XVIII/inmunología , Colágeno Tipo XVIII/metabolismo , Células HEK293 , Humanos , Células Jurkat , Laminina/inmunología , Laminina/metabolismo , Activación de Linfocitos , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/inmunología , Transfección
11.
MAbs ; 4(2): 226-32, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22453098

RESUMEN

We recently described the in vitro and in vivo properties of an engineered homotrimeric antibody made by fusing the N-terminal trimerization region of collagen XVIII NC1 domain to the C-terminus of a scFv fragment [trimerbody (scFv-NC1) 3; 110 kDa]. Here, we demonstrated the utility of the N-terminal trimerization region of collagen XV NC1 domain in the engineering of trivalent antibodies. We constructed several scFv-based trimerbodies containing the human type XV trimerization domain and demonstrated that all the purified trimerbodies were trimeric in solution and exhibited excellent antigen binding capacity. Importantly, type XV trimerbodies demonstrated substantially greater thermal and serum stability and resistance to protease digestion than type XVIII trimerbodies. In summary, the small size, high expression level, solubility and stability of the trimerization domain of type XV collagen make it the ideal choice for engineering homotrimeric antibodies for cancer detection and therapy.


Asunto(s)
Anticuerpos Antineoplásicos , Colágeno , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única , Animales , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/genética , Anticuerpos Antineoplásicos/inmunología , Colágeno/biosíntesis , Colágeno/genética , Colágeno/inmunología , Células HEK293 , Humanos , Ratones , Neoplasias/inmunología , Neoplasias/patología , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
12.
Curr Opin Biotechnol ; 22(6): 924-30, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21435857

RESUMEN

Purified proteins such as antibodies are widely used as therapeutic agents in clinical medicine. However, clinical-grade proteins for therapeutic use require sophisticated technologies and are extremely expensive to produce. In vivo secretion of therapeutic proteins by genetically engineered human cells may advantageously replace injection of highly purified proteins. The use of gene transfer methods circumvents problems related to large-scale production and purification and offers additional benefits by achieving sustained concentrations of therapeutic protein with a syngenic glycosylation pattern that make the protein potentially less immunogenic. The feasibility of the in vivo production of therapeutic proteins by diverse cells/tissues has now been demonstrated using different techniques, such as ex vivo genetically modified cells and in vivo gene transfer mediated by viral vectors.


Asunto(s)
Anticuerpos/metabolismo , Ingeniería Genética , Terapia Genética , Proteínas/metabolismo , Formación de Anticuerpos , Ingeniería Genética/economía , Terapia Genética/economía , Terapia Genética/tendencias , Humanos , Proteínas/genética
13.
J Biol Chem ; 286(7): 5197-203, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21156793

RESUMEN

Tumor-associated cell surface antigens and tumor-associated vascular markers have been used as a target for cancer intervention strategies. However, both types of targets have limitations due to accessibility, low and/or heterogeneous expression, and presence of tumor-associated serum antigen. It has been previously reported that a mitochondrial/cell surface protein, p32/gC1qR, is the receptor for a tumor-homing peptide, LyP-1, which specifically recognizes an epitope in tumor cells, tumor lymphatics, and tumor-associated macrophages/myeloid cells. Using antibody phage technology, we have generated an anti-p32 human monoclonal antibody (2.15). The 2.15 antibody, expressed in single-chain fragment variable and in trimerbody format, was then characterized in vivo using mice grafted subcutaneously with MDA-MB-231 human breast cancers cells, revealing a highly selective tumor uptake. The intratumoral distribution of the antibody was consistent with the expression pattern of p32 in the surface of some clusters of cells. These results demonstrate the potential of p32 for antibody-based tumor targeting strategies and the utility of the 2.15 antibody as targeting moiety for the selective delivery of imaging and therapeutic agents to tumors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Portadoras/inmunología , Portadores de Fármacos/farmacología , Proteínas Mitocondriales/inmunología , Proteínas de Neoplasias/inmunología , Anticuerpos de Cadena Única/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Péptidos Cíclicos/inmunología , Péptidos Cíclicos/metabolismo , Anticuerpos de Cadena Única/inmunología
14.
PLoS One ; 4(9): e7174, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19777065

RESUMEN

Since their onset, display technologies have proven useful for the selection of antibodies against a variety of targets; however, most of the antibodies selected with the currently available platforms need to be further modified for their use in humans, and are restricted to accessible antigens. Furthermore, these platforms are not well suited for in vivo selections. We present here a novel cell based antibody display platform, which takes advantage of the functional capabilities of T lymphocytes. The display of antibodies on the surface of T lymphocytes, as a part of a chimeric-immune receptor (CIR) mediating signaling, may ideally link the antigen-antibody interaction to a demonstrable change in T cell phenotype, due to subsequent expression of the early T cell activation marker CD69. In this proof-of-concept, an in vitro selection was carried out using a human T cell line lentiviral-transduced to express a tumor-specific CIR on the surface, against a human tumor cell line expressing the carcinoembryonic antigen. Based on an effective interaction between the CIR and the tumor antigen, we demonstrated that combining CIR-mediated activation with FACS sorting of CD69(+) T cells, it is possible to isolate binders to tumor specific cell surface antigen, with an enrichment factor of at least 10(3)-fold after two rounds, resulting in a homogeneous population of T cells expressing tumor-specific CIRs.


Asunto(s)
Técnicas Inmunológicas , Activación de Linfocitos/fisiología , Linfocitos T/citología , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Separación Celular , Citometría de Flujo , Haptenos/química , Células HeLa , Humanos , Células Jurkat , Lectinas Tipo C , Lentivirus/metabolismo , Fenotipo , Transducción de Señal
15.
PLoS One ; 4(4): e5381, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19401768

RESUMEN

There is an urgent need to develop new and effective agents for cancer targeting. In this work, a multivalent antibody is characterized in vivo in living animals. The antibody, termed "trimerbody", comprises a single-chain antibody (scFv) fragment connected to the N-terminal trimerization subdomain of collagen XVIII NC1 by a flexible linker. As indicated by computer graphic modeling, the trimerbody has a tripod-shaped structure with three highly flexible scFv heads radially outward oriented. Trimerbodies are trimeric in solution and exhibited multivalent binding, which provides them with at least a 100-fold increase in functional affinity than the monovalent scFv. Our results also demonstrate the feasibility of producing functional bispecific trimerbodies, which concurrently bind two different ligands. A trimerbody specific for the carcinoembryonic antigen (CEA), a classic tumor-associated antigen, showed efficient tumor targeting after systemic administration in mice bearing CEA-positive tumors. Importantly, a trimerbody that recognizes an angiogenesis-associated laminin epitope, showed excellent tumor localization in several cancer types, including fibrosarcomas and carcinomas. These results illustrate the potential of this new antibody format for imaging and therapeutic applications, and suggest that some laminin epitopes might be universal targets for cancer targeting.


Asunto(s)
Anticuerpos Antineoplásicos , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/inmunología , Animales , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/genética , Antígeno Carcinoembrionario/inmunología , Línea Celular , Línea Celular Tumoral , Colágeno Tipo XVIII/química , Colágeno Tipo XVIII/genética , Simulación por Computador , Epítopos , Femenino , Colorantes Fluorescentes , Células HeLa , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/genética , Laminina/inmunología , Ratones , Ratones Desnudos , Modelos Moleculares , Trasplante de Neoplasias , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Ingeniería de Proteínas , Estructura Cuaternaria de Proteína , Trasplante Heterólogo
16.
Stem Cells ; 27(3): 753-60, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19096041

RESUMEN

Mesenchymal stem cells (MSCs) are appealing as gene therapy cell vehicles given their ease of expansion and transduction. However, MSCs exhibit immunomodulatory and proangiogenic properties that may pose a risk in their use in anticancer therapy. For this reason, we looked for a strategy to confine MSCs to a determined location, compatible with a clinical application. Human MSCs genetically modified to express luciferase (MSC(luc)), seeded in a synthetic extracellular matrix (sECM) scaffold (sentinel scaffold) and injected subcutaneously in immunodeficient mice, persisted for more than 40 days, as assessed by bioluminescence imaging in vivo. MSCs modified to express a bispecific alpha-carcinoembryonic antigen (alphaCEA)/alphaCD3 diabody (MSC(dAb)) and seeded in an sECM scaffold (therapeutic scaffolds) supported the release of functional diabody into the bloodstream at detectable levels for at least 6 weeks after implantation. Furthermore, when therapeutic scaffolds were implanted into CEA-positive human colon cancer xenograft-bearing mice and human T lymphocytes were subsequently transferred, circulating alphaCEA/alphaCD3 diabody activated T cells and promoted tumor cell lysis. Reduction of tumor growth in MSC(dAb)-treated mice was statistically significant compared with animals that only received MSC(luc). In summary, we report here for the first time that human MSCs genetically engineered to secrete a bispecific diabody, seeded in an sECM scaffold and implanted in a location distant from the primary tumor, induce an effective antitumor response and tumor regression.


Asunto(s)
Matriz Extracelular , Terapia Genética/métodos , Inmunoterapia/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Andamios del Tejido , Animales , Línea Celular , Proliferación Celular , Células Cultivadas , Neoplasias del Colon/terapia , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Lentivirus/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Microvasc Res ; 75(3): 308-14, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18252255

RESUMEN

Angiogenesis is a multistep process that encompasses complex molecular and cellular interactions that can not be recapitulated in vitro. Here, we demonstrate that vasculature generated from lentivirally transduced human primary endothelial cells expressing firefly luciferase and co-implanted with human bone marrow mesenchymal stem cells in immunodeficient mice can be assessed quantitatively by in vivo whole body bioluminescence imaging for more than 120 days. Luciferase activity correlated with the formation of a network of functional, mature blood vessels of human nature inside the implant that critically depend on the presence of mesenchymal stem cells. In summary, our study offers an unprecedented opportunity to perform long-term serial analysis of the molecular events involved in the angiogenic process and monitoring responses to anti-angiogenic agents.


Asunto(s)
Vasos Sanguíneos/citología , Células de la Médula Ósea/citología , Endotelio Vascular/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/fisiología , Pared Abdominal/irrigación sanguínea , Anciano , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Endotelio Vascular/metabolismo , Endotelio Vascular/trasplante , Femenino , Humanos , Lentivirus/genética , Luciferasas/genética , Luciferasas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Modelos Animales , Transducción Genética/métodos , Venas Umbilicales/citología
19.
Rev. mex. anestesiol ; 15(2): 62-8, abr.-jun. 1992. tab
Artículo en Español | LILACS | ID: lil-118486

RESUMEN

Se estudiaron 40 pacientes de ambos sexos, los cuales fueron divididos aleatoriamente en 2 grupos de 20 cada uno. Todos los pacientes fueron sometidos a diferentes procedimientos de cirugía mayor bajo anestesia general o regional. Se calculó el volumen sanguíneo de los pacientes, y se cuantificó la hemorragia y el porciento del volumen sanguíneo perdido en el transoperatorio. En un grupo se usó dextrán 40 y en otro pentalmidón como parte del régimen de líquidos para restituir la volemia. Se midieron los TP, TPT y proteínas totales antes de iniciar la administración de los expansores del plasma y a las 12, 24 y 36 horas posteriores al uso de dichos coloides, así como el 5o. día o el día de alta de los pacientes. Se tipificó la sangre, se calculó la presión oncótica, y se cuantificaron las plaquetas en los mismos tiempos. La prueba de t para muestras independientes mostró que el pentalmidón mantiene una presión oncótica significativamente mayor que la que produce el dextrán 40 (P<0.02 a P<0.001). No se encontraron diferencias significativas entre ambos grupos en los TP, TPT y cuenta de plaquetas (P>0.02). No se obserbaron cambios en la tipificació de la sangre.


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Recuento de Plaquetas , Sangre , Sustitutos del Plasma , Anestesia General , Pérdida de Sangre Quirúrgica , Presión Sanguínea , Pruebas de Coagulación Sanguínea , Volumen Sanguíneo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...