Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Biol Chem ; 285(27): 20844-9, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20444691

RESUMEN

We previously reported the utility for cancer immunotherapy of a humanized bispecific diabody (hEx3) that targets epidermal growth factor receptor and CD3. Here, we used dynamic and static light scattering measurements to show that the multimer fraction observed in hEx3 in solution is a monodisperse tetramer. The multimerization into tetramers increased the inhibition of cancer cell growth by the hEx3 diabody. Furthermore, 1:2 stoichiometric binding for both antigens was observed in a thermodynamic analysis, indicating that the tetramer has bivalent binding activity for each target, and the structure may be in a circular configuration, as is the case for the single-chain Fv tetrabody. In addition to enhanced cytotoxicity, the functional affinity and stability of the hEx3 tetrabody were superior to those of the hEx3 diabody. The increase in molecular weight is also expected to improve the pharmacokinetics of the bispecific diabody, making the hEx3 tetrabody attractive as a therapeutic antibody fragment for cancer immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos/toxicidad , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/uso terapéutico , Antígenos/inmunología , Complejo CD3/inmunología , División Celular/efectos de los fármacos , Cromatografía en Gel , Dimerización , Receptores ErbB/inmunología , Humanos , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/inmunología , Cinética , Luz , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Pliegue de Proteína , Proteínas Recombinantes/uso terapéutico , Dispersión de Radiación , Programas Informáticos , Resonancia por Plasmón de Superficie , Termodinámica
2.
Clin Cancer Res ; 12(13): 4036-42, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818703

RESUMEN

PURPOSE: Bispecific antibodies (BsAb) have been exploited as both cancer immunodiagnostics and cancer therapeutics and show promise in clinical trials of cancer imaging and therapy. For development of BsAbs as clinical reagents, we have focused on construction of small recombinant BsAbs, called bispecific diabodies. Here, we constructed and characterized a humanized bispecific diabody. EXPERIMENTAL DESIGN: We have reported significant antitumor activity of an anti-epidermal growth factor receptor (EGFR) x anti-CD3 bispecific diabody (Ex3) in in vitro cytotoxicity assays and in vivo. We humanized the Ex3 diabody (hEx3) by grafting the complementarity-determining region and compared its biological properties with those of Ex3. We also tested its physiologic stability and ability to alter survival in xenografted mice. RESULTS: The final yield of hEx3 was 10 times that of Ex3, and refolded hEx3 and Ex3 showed identical binding profiles in EGFR-positive cell lines and EGFR-transfected Chinese hamster ovary cells. hEx3 showed dose-dependent cytotoxicity to EGFR-positive cell lines, which could be specifically inhibited by parental monoclonal antibody IgGs against EGFR or CD3 antigens. The heterodimeric structure was retained in PBS for 6 months, and growth inhibition was maintained after incubation under physiologic conditions. Coadministration of hEx3 with T-LAK cells and interleukin-2 prolonged the survival of nude mice with human colon carcinoma. CONCLUSIONS: The humanized diabody hEx3 is an attractive molecule for cancer therapy and may provide important insights into the development of EGFR-based cancer-targeting reagents.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Reacciones Antígeno-Anticuerpo , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Complejo CD3/inmunología , Células CHO , Carcinoma/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Clonación Molecular , Neoplasias del Colon/inmunología , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/inmunología , Citometría de Flujo , Humanos , Ratones , Ratones Desnudos , Unión Proteica , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
FEBS Lett ; 528(1-3): 70-6, 2002 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-12297282

RESUMEN

Interleukin-21 (IL-21) has recently been identified as a novel 4-helix-bundle type I cytokine possessing a cytokine receptor gamma chain essential for the immune response. We report the preparation and functional characterization of Escherichia coli-expressed recombinant human IL-21 (rIL-21). The rIL-21, expressed as insoluble inclusion bodies in E. coli, was solubilized and then refolded by using a modified dialysis method. The introduction of redox reagents during refolding led to a dramatic increase in the refolding efficiency. Circular dichroism spectrum analysis showed that the refolded rIL-21 had an alpha-helix as a secondary structure, which is a characteristic of type I cytokines. Flow cytometry confirmed previous reports that rIL-21 binds to CD3-activated T cells (T-LAK) and to cell lines Raji, HL60, and Jurkat. rIL-21 stimulated the proliferation of T-LAK but not peripheral blood mononuclear cells, and this effect seems to be identical to that of co-stimulation with anti-CD3 antibody. Growth inhibition assay indicated that enhancement of the cytotoxicity of T-LAK to the human bile duct carcinoma TFK-1 depended on the concentration of rIL-21. Thus, refolded rIL-21 had activity identical to that of authentic IL-21 and enhanced the anti-tumor activity of T-LAK. These conclusions suggest the potential use of the refolded cytokine in adoptive immunotherapy using T-LAK cells and in the discovery of other functions of the cytokine.


Asunto(s)
Antineoplásicos/farmacología , Interleucinas/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Secuencia de Bases , Citotoxicidad Inmunológica , ADN Recombinante/genética , Escherichia coli/química , Escherichia coli/genética , Humanos , Inmunoterapia Adoptiva , Técnicas In Vitro , Cuerpos de Inclusión/química , Interleucinas/genética , Interleucinas/aislamiento & purificación , Células Asesinas Activadas por Linfocinas/inmunología , Datos de Secuencia Molecular , Neoplasias/inmunología , Neoplasias/terapia , Ingeniería de Proteínas , Pliegue de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Subgrupos de Linfocitos T/inmunología , Células Tumorales Cultivadas
4.
Cancer Gene Ther ; 9(4): 330-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960283

RESUMEN

In order to induce specific antitumor immunity in mice, we attempted to immunize C57BL/6 mice with DNA vaccine encoding MUC1 polypeptide. When the mice immunized with MUC1 DNA were challenged with EL4-muc, MUC1-transfected syngeneic lymphoma cells, they completely rejected tumors. When DNA vaccine was given to the EL4-muc tumor-bearing mice, this vaccination was insufficient to suppress tumor growth in the mice. However, activated, but nonprimed dendritic cells (DCs) obtained from syngeneic mice and MUC1 DNA vaccine were given simultaneously to the same site of EL4-muc tumor-bearing mice, tumor growth was markedly suppressed accompanying prolongation of survival time. MUC1 antigen was detected on the DCs at the vaccination site and in regional nodes in the mice which received MUC1 DNA vaccine and DCs. These mice showed markedly enhanced cellular immune responses specific for MUC1 compared to those in mice vaccinated with MUC1 DNA alone. No significant difference in titers of antibodies to MUC1 between the two groups was observed. These results suggest that nonprimed DCs inoculated at the DNA vaccine site are essential for eliciting strong antitumor cellular immunity to suppress tumor growth efficiently in DNA-vaccinated mice. This animal model is useful for developing DNA vaccine for anti-cancer immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia/métodos , Mucina-1/genética , Mucina-1/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Vacunas de ADN/administración & dosificación , Animales , Formación de Anticuerpos , Antígenos CD/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunidad Celular , Inyecciones Subcutáneas , Linfoma/prevención & control , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Transfección , Células Tumorales Cultivadas , Vacunación/métodos
5.
Immunol Lett ; 81(2): 99-106, 2002 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-11852114

RESUMEN

For the purpose of establishing a new adoptive immunotherapy for bile duct carcinoma (BDC), we previously constructed two kinds of bispecific antibodies (bsAbs), anti-MUC1 x anti-CD3 (M x 3) and anti-MUC1 x anti-CD28 (M x 28), which activate T cells and form bridges between them and MUC1-expressing tumor cells. In our previous studies [Cancer Res. 56 (1996) 4205] specific targeting therapy (STT) consisting of i.v. administration of lymphokine activated killer cells with a T cell phenotype (T-LAK) sensitized with two kinds of bsAbs to human BDC-grafted severe combined immunodeficient (SCID) mice demonstrated remarkable inhibition of tumor growth. However, complete cures could not be obtained. In order to improve antitumor efficacy, we have paid attention to anti-CD2 monoclonal antibodies (mAbs), thought to play an important roles in signal transduction in T cell activation or control of T cell receptor (TCR)-driven activation. Therefore, we developed another bsAb, anti-MUC1 x anti-CD2 (M x 2), in order to examine if this would show synergism with the two previously described bsAbs. The combination of the three bsAbs (M x 3, M x 28 and M x 2 bsAbs) showed highest cytotoxicity against MUC1-expressing BDC cells when given simultaneously with peripheral blood mononuclear cells (PBMCs) or T-LAK cells in vitro. When 2 x 10(7) T-LAK cells sensitized with different combinations of bsAbs were administered four times i.v. to BDC-grafted SCID mice, the best therapeutic result was obtained with a combination of all three bsAbs. These results indicate usefulness of combination of three bsAbs for targeting cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de los Conductos Biliares/terapia , Antígenos CD2/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Inmunoterapia Adoptiva/métodos , Mucina-1/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Femenino , Humanos , Interferón gamma/biosíntesis , Células Asesinas Activadas por Linfocinas/inmunología , Leucocitos/citología , Leucocitos/inmunología , Subgrupos Linfocitarios/inmunología , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/terapia , Células Tumorales Cultivadas
6.
J Biochem ; 132(6): 903-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12473192

RESUMEN

Recombinant fragments of the variable region of antibodies are useful in many experimental and clinical applications. However, it can be difficult to obtain these materials in soluble form after their expression in bacteria. Here, we report an efficient procedure for preparing several variable-domain fragments (Fv), single-chain Fv (scFv), and a diabody (the smallest functional bispecific antibody) of anti-carcinoembryonic antigen (CEA) antibody by overexpression in Escherichia coli in inclusion bodies, using a refolding system to obtain renatured proteins. Two types of refolded Fv were prepared: (i) Heavy and light chains of the immunoglobulin variable regions (VH and VL, respectively) were coexpressed with a dicistronic expression vector (designated Fv(co)); (ii) VH and VL were expressed separately, mixed stoichiometrically, and refolded (designated Fv(mix)). All samples refolded with high efficiency; Fv(co), Fv(mix), scFv, and the bispecific diabody bound to several CEA-positive cell lines, exactly as did soluble Fv fragments secreted by E. coli (Fv(sol)) and the parent IgG. The refolded fragments inhibited binding of the parent IgG to CEA-positive cell lines, indicating that their epitope is identical to that of IgG. The bispecific diabody, which combined variable-region fragments of anti-CEA antibody with variable-region fragments of anti-CD3 antibody, was also prepared using the refolding system. This refolded diabody could bind to lymphokine-activated killer cells. In addition, its cytotoxicity toward human bile duct carcinoma TFK-1 and other several other CEA-positive cell lines was concentration-dependent. Taken together, our results suggest that a refolding procedure can be used to prepare various functional antibody fragments (Fv, scFv, and diabody).


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Antígeno Carcinoembrionario/inmunología , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Pliegue de Proteína , Proteínas Recombinantes/metabolismo , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Antineoplásicos/química , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antígeno Carcinoembrionario/metabolismo , Línea Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Fragmentos de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina , Cuerpos de Inclusión/química , Ratones , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
7.
Anticancer Res ; 22(2A): 769-76, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12014649

RESUMEN

BACKGROUND: The bacterial superantigen staphylococcal enterotoxin A (SEA) is an extremely potent activator of T lymphocytes when presented on major histocompatibility complex (MHC) class II molecules. To develop a tumor-specific superantigen for cancer therapy, we constructed a recombinant fusion protein of SEA and the single-chain variable fragment (scFv) of the FU-MK-1 antibody, which recognizes a glycoprotein antigen (termed MK-1 antigen) present on most carcinomas. MATERIALS AND METHODS: We employed recombinant DNA techniques to fuse recombinant mutant SEA to an scFv antibody derived from FU-MK-1 and the resulting fusion protein (SEA/FUscFv) was produced by a bacterial expression system, purified with a metal-affinity column, and characterized for its MK-1-binding specificity and its antitumor activity. RESULTS: The SEA/FUscFv fusion protein retained the reactivity with MK-1-expressing tumor cells, introduced a specific cytotoxicity of lymphokine-activated killer T-cells to the tumor cells, and consequently suppressed the tumor growth in a SCID mouse xenograft model. CONCLUSION: This genetically engineered SEA/FUscFv fusion protein may serve as a potentially useful immunotherapeutic reagent for human MK-1-expressing tumors.


Asunto(s)
Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Enterotoxinas/inmunología , Neoplasias Gástricas/inmunología , Superantígenos/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/farmacología , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/farmacología , Citotoxicidad Inmunológica , Enterotoxinas/genética , Enterotoxinas/farmacología , Molécula de Adhesión Celular Epitelial , Humanos , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/inmunología , Inmunoterapia/métodos , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Células Asesinas Activadas por Linfocinas/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones SCID , Pliegue de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/terapia , Superantígenos/genética , Superantígenos/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
MAbs ; 6(5): 1243-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25517309

RESUMEN

One approach to creating more beneficial therapeutic antibodies is to develop bispecific antibodies (bsAbs), particularly IgG-like formats with tetravalency, which may provide several advantages such as multivalent binding to each target antigen. Although the effects of configuration and antibody-fragment type on the function of IgG-like bsAbs have been studied, there have been only a few detailed studies of the influence of the variable fragment domain order. Here, we prepared four types of hEx3-scDb-Fc, IgG-like bsAbs, built from a single-chain hEx3-Db (humanized bispecific diabody [bsDb] that targets epidermal growth factor receptor and CD3), to investigate the influence of domain order and fusion manner on the function of a bsDb with an Fc fusion format. Higher cytotoxicities were observed with hEx3-scDb-Fcs with a variable light domain (VL)-variable heavy domain (VH) order (hEx3-scDb-Fc-LHs) compared with a VH-VL order, indicating that differences in the Fc fusion manner do not affect bsDb activity. In addition, flow cytometry suggested that the higher cytotoxicities of hEx3-scDb-Fc-LH may be attributable to structural superiority in cross-linking. Interestingly, enhanced degradation resistance and prolonged in vivo half-life were also observed with hEx3-scDb-Fc-LH. hEx3-scDb-Fc-LH and its IgG2 variant exhibited intense in vivo antitumor effects, suggesting that Fc-mediated effector functions are dispensable for effective anti-tumor activities, which may cause fewer side effects. Our results show that merely rearranging the domain order of IgG-like bsAbs can enhance not only their antitumor activity, but also their degradation resistance and in vivo half-life, and that hEx3-scDb-Fc-LHs are potent candidates for next-generation therapeutic antibodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antineoplásicos/inmunología , Inmunoglobulina G/inmunología , Neoplasias/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/farmacocinética , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Área Bajo la Curva , Sitios de Unión/inmunología , Complejo CD3/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Relación Dosis-Respuesta a Droga , Receptores ErbB/inmunología , Femenino , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/farmacología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células MCF-7 , Ratones SCID , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Unión Proteica/inmunología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Protein Eng Des Sel ; 26(5): 359-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23468569

RESUMEN

The domains of bispecific diabodies (BsDbs) can be ordered in four different ways; however, the influence of domain order on the cytotoxicity of BsDbs that retarget immune cells against tumor cells had not been addressed. We previously reported the marked antitumor effects of a humanized BsDb that targets epidermal growth factor receptor and CD3 (hEx3-Db). Here, we rearranged the domains of hEx3-Db to examine the influence of domain order on the function of BsDbs. We successfully prepared homogenous dimers of hEx3-Db in all four domain configurations. Interestingly, all three rearranged hEx3s inhibited cancer growth more effectively than did the original hEx3-Db, in which both components were in variable heavy domain (VH)-variable light domain (VL) order (redesignated as hEx3-HL), and the highest effects were observed with hEx3-LH (hEx3-Db with both components in VL-VH order). In addition, hEx3-LH had comparable in vitro growth inhibitory effects to those of the tandem single-chain variable fragment (scFv) format of hEx3-Db (hEx3-tandem scFv (taFv)), which we previously showed to have greater cytotoxicity than does hEx3-HL. Flow cytometry suggested that the enhanced cytotoxicity of hEx3-LH is attributable to structural superiority for cross-linking, similar to that of hEx3-taFv. Furthermore, hEx3-LH inhibited cancer growth in mice more effectively than did hEx3-taFv; this difference may be due to differences in antibody stability. Our results show that merely rearranging the domain order of BsDbs can enhance their effects beyond those with structural format conversion.


Asunto(s)
Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Complejo CD3/inmunología , Receptores ErbB/inmunología , Neoplasias/terapia , Animales , Anticuerpos Biespecíficos/inmunología , Antineoplásicos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Neoplasias/inmunología , Multimerización de Proteína , Estructura Terciaria de Proteína
10.
FEBS J ; 279(2): 223-33, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22074399

RESUMEN

We previously reported the construction and activity of a humanized, bispecific diabody (hEx3) that recruited T cells towards an epidermal growth factor receptor (EGFR) positive tumor. Herein, we describe the construction of a second functional, fully humanized, anti-EGFR bispecific diabody that recruits another subset of lymphocyte effectors, the natural killer cells, to EGFR-expressing tumor cells. After we confirmed that an anti-EGFR × anti-CD16 bispecific diabody (Ex16) consisting of a previously humanized anti-EGFR variable fragment (Fv) and a mouse anti-CD16 Fv had growth inhibitory activity, we designed a humanized anti-CD16 Fv to construct the fully humanized Ex16 (hEx16). However, the humanized form had lower activity for inhibition of cancer growth. To restore its growth inhibitory activity, we introduced mutations into the Vernier zone, which is located near the complementarity-determining regions and is involved in their binding activity. We efficiently prepared 15 different hEx16 mutants by expressing each chimeric single-chain component for hEx16 separately. We then used our in vitro refolding system to select the most functional mutant, which had a growth inhibitory effect comparable with that of the commercially available chimeric anti-EGFR antibody, cetuximab. Our refolding system could aid in the efficient optimization of other proteins with heterodimeric structure.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Monoclonales Humanizados/biosíntesis , Diseño de Fármacos , Receptores ErbB/antagonistas & inhibidores , Factores Inmunológicos/metabolismo , Receptores de IgG/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/farmacología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Receptores ErbB/metabolismo , Proteínas Ligadas a GPI/antagonistas & inhibidores , Humanos , Hibridomas , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/genética , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/biosíntesis , Proteínas Mutantes Quiméricas/química , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Replegamiento Proteico , Alineación de Secuencia
11.
Oncol Rep ; 26(4): 949-55, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21743971

RESUMEN

We performed in vitro and in vivo experiments of the anti-epidermal growth factor receptor (EGFR) x anti-CD3 bispecific diabody (hEx3-Db) with the IgG-like bispecific antibodies (BsAbs) (hEx3-scFv-Fc and hEx3-scDb-Fc) and the anti-EGFR therapeutic antibody cetuximab to assess the effect of BsAbs on cancer growth inhibition. In vitro, efficacy of the BsAbs and cetuximab were compared by growth inhibition assays of human cell lines of bile duct (TFK-1, HuCC-T1, OCUCh-LM1), epidermoid (A431), gastric (Kato-III), colon (DLD-1, SW480), and breast (SK-BR-3, MCF-7) cancer. In vivo, in three mouse models, we evaluated the anti-tumor activity of hEx3-Db and cetuximab, assessed the effect of hEx3-Db alone, and compared the antitumor activity of hEx3-Db with the IgG-like BsAbs. In vitro, hEx3-scFv-Fc showed nearly 100% killing activity for all cell lines. Both in vitro and in vivo, hEx3-Db needed CD3-positive phenotypes to induce a growth inhibitory effect. In contrast, IgG-like BsAbs showed monotherapeutic effects in vivo by inducing antibody-dependent cellular cytotoxicity (ADCC) similar to cetuximab. However, enhancement was not observed when lymphokine-activated killer cells with the T-cell phenotype were co-injected. Results suggest that IgG-like BsAbs could not efficiently direct T lymphocytes toward tumor cells to induce ADCC due to steric hindrance on binding to CD3- and Fc-receptor-positive phenotypes. Although hEx3-scFv-Fc showed high cytotoxicity in vitro, its high molecular weight limits its usefulness. With an in vivo effect comparable to hEx3-scFv-Fc and its realistic molecular weight, hEx3-scDb-Fc shows promise as a novel recombinant therapeutic antibody and may be modified to enhance its potency by prevention of steric hindrance.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Receptores ErbB/inmunología , Neoplasias/terapia , Animales , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Línea Celular Tumoral , Cetuximab , Femenino , Humanos , Inmunización Pasiva , Ratones , Ratones SCID , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
FEBS J ; 277(2): 477-87, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20015073

RESUMEN

We previously reported the use of a humanized bi-specific diabody that targets epidermal growth factor receptor and CD3 (hEx3-Db) for cancer immunotherapy. Bacterial expression can be used to express small recombinant antibodies on a large scale; however, their overexpression often results in the formation of insoluble aggregates, and in most cases artificial affinity peptide tags need to be fused to the antibodies for purification by affinity chromatography. Here, we propose a novel method for preparing refined, functional, tag-free bi-specific diabodies from IgG-like bi-specific antibodies (BsAbs) in a mammalian expression system. We created an IgG-like BsAb in which bi-specific diabodies were fused to the human Fc region via a designed human rhinovirus 3C (HRV3C) protease recognition site. The BsAb was purified by protein A affinity chromatography, and the refined tag-free hEx3-Db was efficiently produced from the Fc fusion format by protease digestion. The tag-free hEx3-Db from the Fc fusion format showed a greater inhibition of cancer growth than affinity-tagged hEx3-Db prepared directly from Chinese hamster ovary cells. We also applied our novel method to another small recombinant antibody fragment, hEx3 single-chain diabody (hEx3-scDb), and demonstrated the versatility and advantages of our proposed method compared with papain digestion of hEx3-scDb. This approach may be used for industrial-scale production of functional tag-free small therapeutic antibodies.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/genética , Ingeniería de Proteínas/métodos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/farmacología , Células CHO , Línea Celular Tumoral , Cromatografía de Afinidad , Cricetinae , Cricetulus , Expresión Génica , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoterapia , Neoplasias/terapia , Péptido Hidrolasas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Rhinovirus/enzimología , Rhinovirus/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Protein Eng Des Sel ; 21(10): 597-603, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18621782

RESUMEN

We report the utility of in vitro refolding in the preparation of monomorphous hEx3 bispecific diabodies with epidermal growth factor receptor and CD3 retargeting from insoluble aggregates in Escherichia coli. Appropriate interaction between cognate variable heavy and light chains led to the formation of functional hEx3 heterodimers in a diabody format rather than inactive homodimers. The refolded hEx3 was found to exhibit almost the equivalent activity to the hEx3 and single-chain hEx3 (hEx3-scDb) prepared in a mammalian secretion system. We suggest that the preparation of hEx3 from bacterial insoluble material by means of in vitro refolding would be useful for industrial-scale production of the diabody for its potential use in clinical studies.


Asunto(s)
Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Receptores ErbB/inmunología , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Biespecíficos/farmacología , Antineoplásicos/química , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía en Gel , Cricetinae , Cricetulus , Dimerización , Expresión Génica , Humanos , Cadenas Pesadas de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/metabolismo , Renaturación de Proteína , Estructura Cuaternaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Especificidad por Sustrato
14.
J Immunother ; 31(8): 752-61, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18779744

RESUMEN

Recently, recombinant antibodies have been dissected into antigen-binding regions and rebuilt into multivalent high-avidity formats. These new structural designs are expected to improve in vivo pharmacokinetics and efficacy in clinical use. Here, we designed effective recombinant bispecific antibody (BsAb) formats based on hEx3, a humanized bispecific diabody with epidermal growth factor receptor and CD3 retargeting. The bispecific and bivalent IgG-like antibodies engineered from hEx3 (or its single-chain form, hEx3-scDb) and the human Fc region showed stronger binding to each target cell than did monovalent diabody formats, and their affinity was identical to that of the corresponding parent IgG. The bivalent effect of the constructed IgG-like BsAbs resulted in cell cytotoxicity 10 times that of monovalent diabodies, and further, the fusion of Fc portion contributed intense cytotoxicity in peripheral blood mononuclear cells by the induction of the antibody-dependent cellular cytotoxicity. The growth-inhibition effects of IgG-like BsAbs were superior to those of the approved therapeutic antibody cetuximab, which recognizes the same epidermal growth factor receptor antigen, even when peripheral blood mononuclear cells were used as effector cells. We thus demonstrated a critical improvement in the effect of hEx3 by the bottom-up construction of IgG-like BsAbs; in adoptive immunotherapy, monotherapy without supplemental molecules may be able to induce antibody-dependent cellular cytotoxicity.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Receptores ErbB/inmunología , Inmunoglobulina G/inmunología , Células Asesinas Activadas por Linfocinas/inmunología , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Citotóxicos , Animales , Anticuerpos Biespecíficos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Complejo CD3/metabolismo , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Humanos , Inmunoglobulina G/genética , Proteínas Recombinantes de Fusión/genética , Transfección
15.
J Biol Chem ; 282(38): 27659-65, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17644522

RESUMEN

We previously reported the marked in vitro and in vivo antitumor activity of hEx3, a humanized diabody (small recombinant bispecific antibody) with epidermal growth factor receptor (EGFR) and CD3 retargeting. Here, we fabricated a tetravalent IgG-like bispecific antibody with two kinds of single-chain Fv (scFv), i.e. humanized anti-EGFR scFv and anti-CD3 scFv, that contains the same four variable domains as hEx3, on the platform of human IgG1 (hEx3-scFv-Fc). hEx3-scFv-Fc prepared from mammalian cells showed specific binding to both EGFR and CD3 target antigens. At one-thousandth (0.1-100 fmol/ml) of the dose of normal hEx3, hEx3-scFv-Fc showed intense cytotoxicity to an EGFR-positive cell line in a growth-inhibition assay using lymphokine-activated killer cells with the T-cell phenotype (T-LAK cells). The enhanced antitumor effect was more clearly observed when peripheral blood mononuclear cells (PBMCs) were used as effector cells, indicating the utility of IgG-like fabrication. These results suggested that the intense antitumor activity is attributable to the multivalency and the presence of the fused human Fc, a hypothesis that was supported by the results of flow cytometry, PBMC proliferation assay, and protein kinase inhibition assay. Furthermore, the growth inhibition effects of hEx3-scFv-Fc were considerably superior to those of the approved therapeutic antibody, cetuximab, which recognizes the same EGFR antigen even when using PBMCs as effector cells. The high potency of hEx3-scFv-Fc may translate into improved antitumor therapy and lower costs of production because of the smaller doses needed.


Asunto(s)
Anticuerpos Biespecíficos/química , Inmunoglobulina G/química , Inmunoterapia/métodos , Linfocitos/metabolismo , Neoplasias/patología , Animales , Complejo CD3/química , Células CHO , Proliferación Celular , Cricetinae , Cricetulus , Receptores ErbB/metabolismo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Modelos Biológicos , Neoplasias/inmunología
16.
Int Immunol ; 19(10): 1191-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17698559

RESUMEN

IL-21 regulates certain functions of T cells, B cells, NK cells and dendritic cells. Although activated CD4(+) T cells produce IL-21, data identifying the specific CD4(+) T cell subsets that produce IL-21 are conflicting. In a previous study, mouse IL-21 message was detected in T(H)2, whereas human IL-21 (hIL-21) message was found in both T(H)1 and follicular helper T cells. To identify the IL-21-secreting cell populations in human, we established a hybridoma cell line producing an anti-hIL-21 mAb. Intracellular hIL-21-staining experiments showed that hIL-21 was mainly expressed in activated CD4(+) central memory T cells and in activated CD4(+) effector memory T cells, but not in activated CD4(+) naive T cells. Moreover, IL-21 was produced upon activation by some IFN-gamma-producing T(H)1-polarized cells and some IL-17-producing T(H)17-polarized cells, but not by IL-4-producing T(H)2-polarized cells. These results suggest that specific CD4(+) T cell populations produce IL-21. In the functional analysis, we found that IL-21 significantly enhanced the cytokine-driven proliferation of CD4(+) helper T cells synergistically with IL-7 and IL-15 without T cell activation stimuli. Taken together, IL-21 produced from CD4(+) memory T cells may have a supportive role in the maintenance of CD4(+) T cell subsets.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Interleucinas/metabolismo , Activación de Linfocitos , Subgrupos de Linfocitos T/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos CD4/análisis , Linfocitos T CD4-Positivos/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Citocinas/farmacología , Factores de Transcripción Forkhead/análisis , Humanos , Hibridomas , Interleucinas/análisis , Interleucinas/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Células TH1/inmunología
17.
Biochem Biophys Res Commun ; 328(1): 98-105, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15670756

RESUMEN

Integration of lymphocyte-activating cytokines (e.g., interleukin-12: IL-12) to tumor cells offers promise for cancer immunotherapy, but the preparation of such heterodimeric proteins by refolding is difficult because of subunit instability. We achieved the refolding of Escherichia coli-expressed human IL-12 by a stepwise dialysis method, preventing the formation of insoluble aggregates by adding a redox reagent and an aggregation suppressor. We also constructed a tumor-specific IL-12 protein, each subunit of which was fused with one chain of variable domain fragment (Fv) of anticarcinoembryonic antigen (CEA) antibody T84.66 (aCEA-IL12). Fusion of IL-12 with Fv greatly increased the yield of functional heterodimer. Several assays have indicated that the Fv domain and IL-12 domain of the fused protein had cognate biological activities, and it enhanced the cytotoxicity of T-LAK cells for the cancer cell line.


Asunto(s)
Anticuerpos/administración & dosificación , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Antígeno Carcinoembrionario/administración & dosificación , Interleucina-12/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Neoplasias de los Conductos Biliares/inmunología , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/administración & dosificación , Citocinas/química , Citocinas/genética , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Humanos , Interleucina-12/química , Interleucina-12/genética , Interleucina-12/inmunología , Microdiálisis/métodos , Pliegue de Proteína , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química
18.
Blood ; 101(7): 2620-7, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12411307

RESUMEN

Interleukin-17 (IL-17) is a CD4 T-cell-derived proinflammatory cytokine. We investigated the effects of locally produced IL-17 by tumors as a means to evaluate its biologic function. Although recombinant IL-17 protein or retroviral transduction of IL-17 gene into tumors did not affect in vitro proliferation, IL-17 transfectants grew more rapidly in vivo when compared with controls. Immunostaining for Factor VIII revealed that tumors transduced with IL-17 had significantly higher vascular density when compared with controls. IL-17 indeed elicited neovascularization in rat cornea. In addition, angiogenic activity present in the conditioned media of CD4 T cells was markedly suppressed by neutralizing monoclonal antibody to IL-17. IL-17 had no direct effect on the growth of vascular endothelial cells, whereas IL-17 significantly stimulated migration. IL-17 also markedly promoted the cord formation of vascular endothelial cells. In addition, IL-17 up-regulated elaboration of a variety of proangiogenic factors by fibroblasts as well as tumor cells. These findings reveal a novel role for IL-17 as a CD4 T-cell-derived mediator of angiogenesis that stimulates vascular endothelial cell migration and cord formation and regulates production of a variety of proangiogenic factors. Furthermore, they suggest that inhibition of biologic action of IL-17 may have therapeutic benefits when applied to angiogenesis-related disorders.


Asunto(s)
Interleucina-17/farmacología , Neoplasias Experimentales/patología , Neovascularización Patológica/inducido químicamente , Animales , División Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Córnea/irrigación sanguínea , Córnea/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Sustancias de Crecimiento/biosíntesis , Humanos , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/etiología , Ratas , Transducción Genética , Venas Umbilicales/citología , Regulación hacia Arriba/efectos de los fármacos
19.
Cancer Immunol Immunother ; 52(2): 97-106, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12594573

RESUMEN

4-1BB ligand (4-1BBL), a member of the tumor necrosis factor (TNF) superfamily, interacts with 4-1BB (CDw137) expressed on activated T cells and delivers a costimulatory signal for T cell activation and growth. Various studies have demonstrated a role for murine 4-1BB in immune function, but relatively few investigations of human 4-1BB have been conducted. Here we report on the construction of a recombinant E1/E3-deleted adenovirus encoding human 4-1BBL (Ad4-1BBL) and its stimulation of antitumor immunity. Ad4-1BBL was able to efficiently infect several human adenocarcinoma cell lines and induce 4-1BBL expression on the cell surface within 24 h, this enhancing the antitumor activity not only of lymphokine-activated killer cells with a T cell phenotype (T-LAK) but also naive peripheral blood mononuclear cells (PBMC). This antitumor activity with T-LAK cells was further enhanced by addition of bispecific antibody (BsAb; anti-MUC1xanti-CD3). Cocultivation of Ad4-1BBL-infected tumor cells with either T-LAK cells or PBMC resulted in significant elevation of interferon-gamma (IFN-gamma), interleukin-2 (IL-2), and granulocyte-macrophage colony-stimulating factor (GM-CSF) production. Furthermore, remarkable tumor growth inhibition was observed in cholangiocarcinoma-grafted severe combined immunodeficient (SCID) mice to which Ad4-1BBL and T-LAK cells were administered when tumor size exceeded 5 mm in diameter. These results provide strong evidence in support of the efficacy of adenovirally delivered 4-1BBL for genetic immunotherapy of cancer.


Asunto(s)
Adenoviridae/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/terapia , Terapia Genética , Células Asesinas Activadas por Linfocinas/inmunología , Factor de Necrosis Tumoral alfa/genética , Ligando 4-1BB , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD , Neoplasias de los Conductos Biliares/terapia , Complejo CD3/inmunología , Colangiocarcinoma/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Ratones , Ratones SCID , Mucina-1/inmunología , Trasplante de Neoplasias , Neoplasias Experimentales/terapia , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Células Tumorales Cultivadas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/fisiología
20.
J Hum Genet ; 48(3): 130-7, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12624724

RESUMEN

Screening for protein-truncating mutations of the BRCA1 and BRCA2 genes is useful in genetic testing for familial breast cancer because, first, the methods are usually simple and not expensive, and second, the detected mutations indicate pathogenic mutations in general. We evaluated the diagnostic accuracy of the stop codon (SC) assay for detecting protein-truncating mutations in the BRCA1 and BRCA2 genes by comparing the results with DNA sequencing in samples from 29 patients with breast cancer from 24 Japanese families with a history of breast cancer. Protein-truncating mutations were detected in 5 of the 24 families (20.8%; two in the BRCA1 gene and three in the BRCA2 gene). Among the 176 DNA fragments examined using the SC assay, the existence of three protein-truncating mutations (one in the BRCA1 gene and two in the BRCA2gene) was predicted correctly by the assay. Only one reverse transcriptase-polymerase chain reaction fragment was positive for the SC assay but was negative using DNA sequencing. Our study showed clearly that the SC assay is sensitive (3 of 3, 100%) and specific (172 of 173, 99%) for detecting pathogenic protein-truncating mutations in the BRCA1 and BRCA2 genes, and that it could be useful for screening larger populations.


Asunto(s)
Bioensayo , Neoplasias de la Mama/genética , Codón de Terminación , Genes BRCA1 , Genes BRCA2 , Neoplasias de la Mama/etiología , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Mutación , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA