Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 28(2): 548-560, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31870622

RESUMEN

The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor ß (TGF-ß) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.


Asunto(s)
Neoplasias de los Genitales Femeninos/inmunología , Inmunoterapia Adoptiva , Neoplasias Ováricas/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/genética , Epítopos/inmunología , Femenino , Neoplasias de los Genitales Femeninos/terapia , Humanos , Ratones , Neoplasias Ováricas/terapia , Receptores Quiméricos de Antígenos/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Molecules ; 21(12)2016 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-27999336

RESUMEN

Antibody drug conjugates (ADC), comprised of highly potent small molecule payloads chemically conjugated to a full-length antibody, represent a growing class of therapeutic agents. The targeting of cytotoxic payloads via the specificity and selectivity of the antibody has led to substantial clinical benefits. However, ADC potency can be altered by mechanisms of resistance such as overexpression of efflux pumps or anti-apoptotic proteins. DeBouganin is a de-immunized variant of bouganin, a ribosome-inactivating protein (RIP) that blocks protein synthesis, thereby leading to apoptosis. When conjugated to trastuzumab (T-deB), deBouganin was more potent than ado-trastuzumab-emtansine (T-DM1) and unaffected by resistance mechanisms to which DM1 is susceptible. To further highlight the differentiating mechanism of action of deBouganin, HCC1419 and BT-474 tumor cells that survived T-DM1 or trastuzumab-MMAE (T-MMAE) treatment were treated with an anti-HER2 C6.5 diabody-deBouganin fusion protein or T-deB. C6.5 diabody-deBouganin and T-deB were potent against HCC1419 and BT-474 cells that were resistant to T-DM1 or T-MMAE killing. The resistant phenotype involved MDR pumps, Bcl-2 family members, and the presence of additional unknown pathways. Overall, the data suggest that deBouganin is effective against tumor cell resistance mechanisms selected in response to ADCs composed of anti-microtubule payloads.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inmunoconjugados/farmacología , Microtúbulos/efectos de los fármacos , Antineoplásicos/química , Antineoplásicos/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Portadoras/química , Línea Celular Tumoral , Proliferación Celular , Cromatografía Líquida de Alta Presión , Escherichia coli , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/toxicidad , Péptidos y Proteínas de Señalización Intracelular , Células MCF-7 , Proteínas de la Membrana/química , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/toxicidad , Trastuzumab
3.
FASEB J ; 27(2): 581-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104982

RESUMEN

Fibroblast activation protein (FAP) is a serine protease selectively expressed on tumor stromal fibroblasts in epithelial carcinomas and is important in cancer growth, adhesion, and metastases. As FAP enzymatic activity is a potent therapeutic target, we aimed to identify inhibitory antibodies. Using a competitive inhibition strategy, we used phage display techniques to identify 53 single-chain variable fragments (scFvs) after three rounds of panning against FAP. These scFvs were expressed and characterized for binding to FAP by surface plasmon resonance and flow cytometry. Functional assessment of these antibodies yielded an inhibitory scFv antibody, named E3, which could attenuate 35% of FAP cleavage of the fluorescent substrate Ala-Pro-7-amido-4-trifluoromethylcoumarin compared with nonfunctional scFv control. Furthermore, a mutant E3 scFv was identified by yeast affinity maturation. It had higher affinity (4-fold) and enhanced inhibitory effect on FAP enzyme activity (3-fold) than E3. The application of both inhibitory anti-FAP scFvs significantly affected the formation of 3-dimensional FAP-positive cell matrix, as demonstrated by reducing the fibronectin fiber orientation from 41.18% (negative antibody control) to 34.06% (E3) and 36.15% (mutant E3), respectively. Thus, we have identified and affinity-maturated the first scFv antibody capable of inhibiting FAP function. This scFv antibody has the potential to disrupt the role of FAP in tumor invasion and metastasis.


Asunto(s)
Gelatinasas/antagonistas & inhibidores , Gelatinasas/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Serina Endopeptidasas/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Afinidad de Anticuerpos , Endopeptidasas , Citometría de Flujo , Gelatinasas/genética , Gelatinasas/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Terapia Molecular Dirigida , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Biblioteca de Péptidos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Resonancia por Plasmón de Superficie
4.
ACS Nano ; 16(12): 20021-20033, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36264003

RESUMEN

To address the key challenges in the development of next-generation drug delivery systems (DDS) with desired physicochemical properties to overcome limitations regarding safety, in vivo efficacy, and solid tumor penetration, an ultrasmall folate receptor alpha (FRα) targeted silica nanoparticle (C'Dot) drug conjugate (CDC; or folic acid CDC) was developed. A broad array of methods was employed to screen a panel of CDCs and identify a lead folic acid CDC for clinical development. These included comparing the performance against antibody-drug conjugates (ADCs) in three-dimensional tumor spheroid penetration ability, assessing in vitro/ex vivo cytotoxic efficacy, as well as in vivo therapeutic outcome in multiple cell-line-derived and patient-derived xenograft models. An ultrasmall folic acid CDC, EC112002, was identified as the lead candidate out of >500 folic acid CDC formulations evaluated. Systematic studies demonstrated that the lead formulation, EC112002, exhibited highly specific FRα targeting, multivalent binding properties that would mediate the ability to outcompete endogenous folate in vivo, enzymatic responsive payload cleavage, stability in human plasma, rapid in vivo clearance, and minimal normal organ retention organ distribution in non-tumor-bearing mice. When compared with an anti-FRα-DM4 ADC, EC112002 demonstrated deeper penetration into 3D cell-line-derived tumor spheroids and superior specific cytotoxicity in a panel of 3D patient-derived tumor spheroids, as well as enhanced efficacy in cell-line-derived and patient-derived in vivo tumor xenograft models expressing a range of low to high levels of FRα. With the growing interest in developing clinically translatable, safe, and efficacious DDSs, EC112002 has the potential to address some of the critical limitations of the current systemic drug delivery for cancer management.


Asunto(s)
Receptor 1 de Folato , Sistema de Administración de Fármacos con Nanopartículas , Neoplasias , Animales , Humanos , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptor 1 de Folato/metabolismo , Receptor 1 de Folato/uso terapéutico , Ácido Fólico/química , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Dióxido de Silicio/uso terapéutico
5.
Anal Chem ; 83(9): 3392-7, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21449604

RESUMEN

Rapid and sensitive detection of serum tumor biomarkers are needed to monitor cancer patients for disease progression. Highly sensitive piezoelectric microcantilever sensors (PEMS) offer an attractive tool for biomarker detection; however, their utility in the complex environment encountered in serum has yet to be determined. As a proof of concept, we have functionalized PEMS with antibodies that specifically bind to HER2, a biomarker (antigen) that is commonly overexpressed in the blood of breast cancer patients. The function and sensitivity of these anti-HER2 PEMS biosensors was initially assessed using recombinant HER2 spiked into human serum. Their ability to detect native HER2 present in the serum of breast cancer patients was then determined. We have found that the anti-HER2 PEMS were able to accurately detect both recombinant and naturally occurring HER2 at clinically relevant levels (>2 ng/mL). This indicates that PEMS-based biosensors provide a potentially effective tool for biomarker detection.


Asunto(s)
Análisis Químico de la Sangre/métodos , Neoplasias de la Mama/sangre , Electricidad , Espacio Extracelular , Receptor ErbB-2/sangre , Receptor ErbB-2/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/química , Estudios de Casos y Controles , Humanos , Masculino , Estructura Terciaria de Proteína , Receptor ErbB-2/inmunología , Trastuzumab
6.
Sens Actuators B Chem ; 160(1): 349-356, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22888196

RESUMEN

We have investigated real-time, label-free, in-situ detection of human epidermal growth factor receptor 2 (Her2) in diluted serum using the first longitudinal extension mode of a lead zirconate-lead titanate (PZT)/glass piezoelectric microcantilever sensor (PEMS) with H3 single-chain variable fragment (scFv) immobilized on the 3-mercaptopropyltrimethoxysilane (MPS) insulation layer of the PEMS surface. We showed that with the longitudinal extension mode, the PZT/glass PEMS consisting of a 1 mm long and 127 µm thick PZT layer bonded with a 75 µm thick glass layer with a 1.8 mm long glass tip could detect Her2 at a concentration of 6-60 ng/ml (or 0.06-0.6 nM) in diluted human serum, about 100 times lower than the concentration limit obtained using the lower-frequency flexural mode of a similar PZT/glass PEMS. We further showed that with the longitudinal mode, the PZT/glass PEMS determined the equilibrium H3-Her2 dissociation constant K(d) to be 3.3±0.3 × 10(-8) M consistent with the value, 3.2±0.28 ×10(-8) M deduced by the surface plasmon resonance method (BIAcore).

7.
Sensors (Basel) ; 11(5): 5520-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22413149

RESUMEN

Piezoelectric microcantilever sensors (PEMS) can be sensitive tools for the detection of proteins and cells in biological fluids. However, currently available PEMS can only be used a single time or must be completely stripped and refunctionalized prior to subsequent uses. Here we report the successful use of an alternative regeneration protocol employing high salt concentrations to remove the target, leaving the functional probe immobilized on the microcantilever surface. Our model system employed the extracellular domain (ECD) of recombinant human Epidermal Growth Factor Receptor (EGFR) as the probe and anti-human EGFR polyclonal antibodies as the target. We report that high concentrations of MgCl2 dissociated polyclonal antibodies specifically bound to EGFR ECD immobilized on the sensor surface without affecting its bioactivity. This simple regeneration protocol both minimized the time required to re-conjugate the probe and preserved the density of probe immobilized on PEMS surface, yielding identical biosensor sensitivity over a series of assays.


Asunto(s)
Técnicas Biosensibles/instrumentación , Anticuerpos/química , Antígenos/química , Biomarcadores/análisis , Técnicas Biosensibles/métodos , Diseño de Equipo , Humanos
8.
PLoS Pathog ; 4(11): e1000197, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18989460

RESUMEN

Phylogenetic analyses have provided strong evidence that amino acid changes in spike (S) protein of animal and human SARS coronaviruses (SARS-CoVs) during and between two zoonotic transfers (2002/03 and 2003/04) are the result of positive selection. While several studies support that some amino acid changes between animal and human viruses are the result of inter-species adaptation, the role of neutralizing antibodies (nAbs) in driving SARS-CoV evolution, particularly during intra-species transmission, is unknown. A detailed examination of SARS-CoV infected animal and human convalescent sera could provide evidence of nAb pressure which, if found, may lead to strategies to effectively block virus evolution pathways by broadening the activity of nAbs. Here we show, by focusing on a dominant neutralization epitope, that contemporaneous- and cross-strain nAb responses against SARS-CoV spike protein exist during natural infection. In vitro immune pressure on this epitope using 2002/03 strain-specific nAb 80R recapitulated a dominant escape mutation that was present in all 2003/04 animal and human viruses. Strategies to block this nAb escape/naturally occurring evolution pathway by generating broad nAbs (BnAbs) with activity against 80R escape mutants and both 2002/03 and 2003/04 strains were explored. Structure-based amino acid changes in an activation-induced cytidine deaminase (AID) "hot spot" in a light chain CDR (complementarity determining region) alone, introduced through shuffling of naturally occurring non-immune human VL chain repertoire or by targeted mutagenesis, were successful in generating these BnAbs. These results demonstrate that nAb-mediated immune pressure is likely a driving force for positive selection during intra-species transmission of SARS-CoV. Somatic hypermutation (SHM) of a single VL CDR can markedly broaden the activity of a strain-specific nAb. The strategies investigated in this study, in particular the use of structural information in combination of chain-shuffling as well as hot-spot CDR mutagenesis, can be exploited to broaden neutralization activity, to improve anti-viral nAb therapies, and directly manipulate virus evolution.


Asunto(s)
Anticuerpos Antivirales/genética , Evolución Biológica , Selección Genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Regiones Determinantes de Complementariedad/genética , Citidina Desaminasa/genética , Epítopos , Humanos , Sueros Inmunes/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología
9.
Clin Cancer Res ; 14(3): 875-82, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18245551

RESUMEN

PURPOSE: Successful radioimmunotherapy strategies depend on selecting radioisotopes with physical properties complementary to the biological properties of the targeting vehicle. Small, engineered antitumor antibody fragments are capable of rapid, highly specific tumor targeting in immunodeficient mouse models. We hypothesized that the C6.5 diabody, a noncovalent anti-HER2 single-chain Fv dimer, would be an ideal radioisotope carrier for the radioimmunotherapy of established tumors using the short-lived alpha-emitting radioisotope (211)At. EXPERIMENTAL DESIGN: Immunodeficient nude mice bearing established HER2/neu-positive MDA-MB-361/DYT2 tumors treated with N-succinimidyl N-(4-[(211)At]astatophenethyl)succinamate ((211)At-SAPS)-C6.5 diabody. Additional cohorts of mice were treated with (211)At-SAPS T84.66 diabody targeting the carcinoembryonic antigen or (211)At-SAPS on a diabody specific for the Müllerian inhibiting substance type II receptor, which is minimally expressed on this tumor cell line. RESULTS: A single i.v. injection of (211)At-SAPS C6.5 diabody led to a 30-day delay in tumor growth when a 20 muCi dose was administered and a 57-day delay in tumor growth (60% tumor-free after 1 year) when a 45 muCi dose was used. Treatment of mice bearing the same tumors with (211)At-SAPS T84.66 diabody at the same doses led to a delay in tumor growth, but no complete responses, likely due to substantially lower expression of this antigen on the MDA-MB-361/DYT2 tumors. In contrast, a dose of 20 muCi of (211)At-SAPS on the anti-Müllerian-inhibiting substance type II receptor diabody did not affect tumor growth rate, demonstrating specificity of the therapeutic effect. CONCLUSIONS: These findings indicate that diabody molecules can be effective agents for targeted radioimmunotherapy of solid tumors using powerful, short-lived alpha-emitting radioisotopes.


Asunto(s)
Astato/uso terapéutico , Neoplasias de la Mama/radioterapia , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Ratones SCID , Fragmentos de Péptidos , Radioinmunoterapia , Receptor ErbB-2/inmunología , Distribución Tisular , Trasplante Heterólogo
10.
J Mol Biol ; 371(4): 934-47, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17602702

RESUMEN

To determine the importance of single-chain Fv (scFv) affinity on binding, uptake, and cytotoxicity of tumor-targeting nanoparticles, the affinity of the epidermal growth factor receptor (EGFR) scFv antibody C10 was increased using molecular evolution and yeast display. A library containing scFv mutants was created by error-prone PCR, displayed on the surface of yeast, and higher affinity clones selected by fluorescence activated cell sorting. Ten mutant scFv were identified that had a 3-18-fold improvement in affinity (KD=15-88 nM) for EGFR-expressing A431 tumor cells compared to C10 scFv (KD=264 nM). By combining mutations, higher affinity scFv were generated with KD ranging from 0.9 nM to 10 nM. The highest affinity scFv had a 280-fold higher affinity compared to that of the parental C10 scFv. Immunoliposome nanoparticles (ILs) were prepared using EGFR scFv with a 280-fold range of affinities, and their binding and uptake into EGFR-expressing tumor cells was quantified. At scFv densities greater than 148 scFv/IL, there was no effect of scFv affinity on IL binding and uptake into tumor cells, or on cytotoxicity. At lower scFv densities, there was less uptake and binding for ILs constructed from the very low affinity C10 scFv. The results show the importance of antibody fragment density on nanoparticle uptake, and suggest that engineering ultrahigh affinity scFv may be unnecessary for optimal nanoparticle targeting.


Asunto(s)
Anticuerpos/inmunología , Afinidad de Anticuerpos/inmunología , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Nanopartículas , Neoplasias/inmunología , Neoplasias/metabolismo , Secuencia de Aminoácidos , Anticuerpos/química , Anticuerpos/genética , Anticuerpos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Regulación de la Expresión Génica , Humanos , Liposomas/inmunología , Datos de Secuencia Molecular , Mutación/genética , Neoplasias/genética , Neoplasias/patología , Alineación de Secuencia , Solubilidad , Topotecan/toxicidad
11.
Nat Biotechnol ; 23(9): 1147-57, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16151408

RESUMEN

The most significant recent advances in the application of monoclonal antibodies (mAbs) to oncology have been the introduction and approval of bevacizumab (Avastin), an anti-vascular endothelial growth factor antibody, and of cetuximab (Erbitux), an anti-epidermal growth factor antibody. In combination with standard chemotherapy regimens, bevacizumab significantly prolongs the survival of patients with metastatic cancers of the colorectum, breast and lung. Cetuximab, used alone or with salvage chemotherapy, produces clinically meaningful anti-tumor responses in patients with chemotherapy-refractory cancers of the colon and rectum. In addition, the anti-HER2/neu antibody trastuzumab (Herceptin), in combination with standard adjuvant chemotherapy, has been shown to reduce relapses and prolong disease-free and overall survival in high-risk patients after definitive local therapy for breast cancer. These exciting recent results provide optimism for the development of mAbs that bind novel targets, exploit novel mechanisms of action or possess improved tumor targeting. Progress in the clinical use of radioimmunoconjugates remains hindered by complexity of administration, toxicity concerns and insufficiently selective tumor targeting.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoterapia/métodos , Neoplasias/terapia , Radioinmunoterapia/métodos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Sitios de Unión , Neoplasias de la Mama/terapia , Cetuximab , Proteínas del Sistema Complemento , Supervivencia sin Enfermedad , Fibronectinas/química , Sustancias de Crecimiento/química , Sustancias de Crecimiento/metabolismo , Humanos , Ligandos , Neoplasias Pulmonares/terapia , Modelos Biológicos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Transducción de Señal
12.
Rev Sci Instrum ; 79(7): 076101, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18681738

RESUMEN

Using 3-mercaptopropyltrimethoxysilane (MPS)-coated (PbMg 1/3 Nb 2/3 O3)0.63-(PbTiO3)0.37 (PMN-PT)/tin and lead zirconate titanate/glass piezoelectric microcantilever sensors (PEMSs) with single-chain variable fragment (scFv) immobilized on the MPS surface, we have demonstrated real-time, label-free detection of human epidermal growth factor receptor 2 (Her2) in a background of 1 mg/ml bovine serum albumin. Coupled with a scFv with a KD of 3.4 x 10(-8)M, the MPS-insulated PMN-PT/tin PEMS 560 microm long and 720 microm wide exhibited a Her2 concentration sensitivity of 5 ng/ml in a background of 1 mg/ml BSA.


Asunto(s)
Plomo/química , Receptor ErbB-2/análisis , Titanio/química , Animales , Bovinos , Línea Celular , Diseño de Equipo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Vidrio , Humanos , Cinética , Compuestos de Organosilicio , Receptor ErbB-2/biosíntesis , Reproducibilidad de los Resultados , Albúmina Sérica Bovina/química , Silanos/química
13.
Nucl Med Biol ; 34(6): 609-18, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17707800

RESUMEN

INTRODUCTION: The cellular binding and processing of an epidermal growth factor receptor (EGFR) targeting affibody molecule, (Z(EGFR:955))(2), was studied. This new and small molecule is aimed for applications in nuclear medicine. The natural ligand epidermal growth factor (EGF) and the antibody cetuximab were studied for comparison. METHODS: All experiments were made with cultured A431 squamous carcinoma cells. Receptor specificity, binding time patterns, retention and preliminary receptor binding site localization studies were all made after (125)I labeling. Internalization was studied using Oregon Green 488, Alexa Fluor 488 and CypHer5E markers. RESULTS: [(125)I](Z(EGFR:955))(2) and [(125)I]cetuximab gave a maximum cellular uptake of (125)I within 4 to 8 h of incubation, while [(125)I]EGF gave a maximum uptake already after 2 h. The retention studies showed that the cell-associated fraction of (125)I after 48 h of incubation was approximately 20% when delivered as [(125)I](Z(EGFR:955))(2) and approximately 25% when delivered as [(125)I]cetuximab. [(125)I]EGF-mediated delivery gave a faster (125)I release, where almost all cell-associated radioactivity had disappeared within 24 h. All three substances were internalized as demonstrated with confocal microscopy. Competitive binding studies showed that both EGF and cetuximab inhibited binding of (Z(EGFR:955))(2) and indicated that the three substances competed for an overlapping binding site. CONCLUSION: The results gave information on cellular processing of radionuclides when delivered with (Z(EGFR:955))(2) in comparison to delivery with EGF and cetuximab. Competition assays suggested that [(125)I](Z(EGFR:955))(2) bind to Domain III of EGFR. The affibody molecule (Z(EGFR:955))(2) can be a candidate for EGFR imaging applications in nuclear medicine.


Asunto(s)
Receptores ErbB/metabolismo , Radiofármacos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Ácidos Carboxílicos , Línea Celular Tumoral , Cetuximab , Factor de Crecimiento Epidérmico/farmacocinética , Técnica del Anticuerpo Fluorescente , Fluorobencenos , Humanos , Radioisótopos de Yodo/farmacocinética , Marcaje Isotópico , Microscopía Confocal
14.
Clin Cancer Res ; 12(5): 1599-605, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16533787

RESUMEN

Radiolabeled single-chain Fv (sFv) molecules display highly specific tumor retention in the severe combined immunodeficient (SCID) mouse model; however, the absolute quantity of sFv retained in the tumors is diminished by the rapid renal elimination resulting from the small size of the sFv molecules (Mr 27,000) and by dissociation of the monovalent sFv from tumor-associated antigen. We previously reported significant improvement in tumor retention without a loss of targeting specificity on converting monovalent sFv into divalent [(sFv')2] dimers, linked by a disulfide bond between COOH-terminal cysteinyl peptides engineered into the sFv'. However, our data for enhanced dimer localization in tumors could not distinguish between the contributions of enhanced avidity and increased systemic retention associated with the larger size of 54 kDa [(sFv')2] dimers relative to 27-kDa sFv. In this investigation, we have compared tumor targeting of divalent anti-c-erbB-2/HER2/neu 741F8-1 (sFv')2 homodimers with monovalent 741F8/26-10 (sFv')2 heterodimers (Mr 54,000) and 741F8 sFv monomers (741F8 sFv has binding specificity for erbB-2/HER2/neu and 26-10 sFv specificity for digoxin and related cardiac glycosides). These studies allowed us to distinguish the dominant effect of valency over molecular weight in accounting for the superior tumor retention of 741F8-1 (sFv')2 homodimers. Each of the radioiodinated species was administered i.v. to SCID mice bearing SK-OV-3 human tumor xenografts and tumor localization at 24 hours post i.v. injection was determined for 125I-741F8-1 (sFv')2 (3.57 %ID/g), 125I-741F8/26-10 (sFv')2 (1.13 %ID/g), and 125I-741F8-1 sFv (1.25 %ID/g). These findings substantiate that the improved tumor retention of (sFv')2 homodimers over sFv monomers results from the availability of dual binding sites rather than from the slower systemic clearance of homodimers.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Fragmentos de Inmunoglobulinas/metabolismo , Neoplasias Ováricas/metabolismo , Receptor ErbB-2/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Dimerización , Femenino , Humanos , Fragmentos de Inmunoglobulinas/administración & dosificación , Fragmentos de Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos ICR , Ratones SCID , Neoplasias Ováricas/diagnóstico por imagen , Cintigrafía , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Distribución Tisular , Trasplante Heterólogo , Trastuzumab , Células Tumorales Cultivadas
15.
Cancer Res ; 65(4): 1471-8, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735035

RESUMEN

Positron emission tomography (PET) provides an effective means of both diagnosing/staging several types of cancer and evaluating efficacy of treatment. To date, the only U.S. Food and Drug Administration-approved radiotracer for oncologic PET is (18)F-fluoro-deoxyglucose, which measures glucose accumulation as a surrogate for malignant activity. Engineered antibody fragments have been developed with the appropriate targeting specificity and systemic elimination properties predicted to allow for effective imaging of cancer based on expression of tumor associated antigens. We evaluated a small engineered antibody fragment specific for the HER2 receptor tyrosine kinase (C6.5 diabody) for its ability to function as a PET radiotracer when labeled with iodine-124. Our studies revealed HER2-dependent imaging of mouse tumor xenografts with a time-dependent increase in tumor-to-background signal over the course of the experiments. Radioiodination via an indirect method attenuated uptake of radioiodine in tissues that express the Na/I symporter without affecting the ability to image the tumor xenografts. In addition, we validated a method for using a clinical PET/computed tomography scanner to quantify tumor uptake in small-animal model systems; quantitation of the tumor targeting by PET correlated with traditional necropsy-based analysis at all time points analyzed. Thus, diabodies may represent an effective molecular structure for development of novel PET radiotracers.


Asunto(s)
Inmunoconjugados , Fragmentos de Inmunoglobulinas , Neoplasias Ováricas/diagnóstico por imagen , Radiofármacos , Receptor ErbB-2/inmunología , Animales , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Radioisótopos de Yodo/química , Marcaje Isotópico/métodos , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Distribución Tisular , Trasplante Heterólogo
16.
Mol Cancer Ther ; 5(8): 2096-105, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16928831

RESUMEN

The Müllerian inhibiting substance type II receptor (MISIIR) is involved in Müllerian duct regression as part of the development of the male reproductive system. In adult females, MISIIR is present on ovarian surface epithelium and is frequently expressed on human epithelial ovarian cancer cells. Müllerian inhibiting substance has been found to be capable of inhibiting the growth of primary human ovarian cancer cells derived from ascites and ovarian cancer cell lines. This suggested to us that MISIIR could be an attractive target for antibody-based tumor targeting and growth inhibition strategies. Here, we describe the production of recombinant human MISIIR extracellular domain-human immunoglobulin Fc domain fusion proteins and their use as targets for the selection of MISIIR-specific human single-chain variable fragments (scFv) molecules from a human nonimmune scFv phage display library. The binding kinetics of the resulting anti-MISIIR scFv clones were characterized and two were employed as the basis for the construction of bivalent scFv:Fc antibody-based molecules. Both bound specifically to human ovarian carcinoma cells in flow cytometry assays and cross-reacted with mouse MISIIR. These results indicate that antibody-based constructs may provide a highly specific means of targeting MISIIR on human ovarian carcinoma cells for the purpose of diagnosing and treating this disease.


Asunto(s)
Anticuerpos/genética , Ingeniería de Proteínas/métodos , Receptores de Péptidos/inmunología , Anticuerpos/metabolismo , Afinidad de Anticuerpos , Femenino , Citometría de Flujo/métodos , Humanos , Región Variable de Inmunoglobulina/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Biblioteca de Péptidos , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas
17.
Mol Imaging Biol ; 19(5): 656-664, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28213834

RESUMEN

PURPOSE: Competitive radiolabeled antibody imaging can determine the unlabeled intact antibody dose that fully blocks target binding but may be confounded by heterogeneous tumor penetration. We evaluated the hypothesis that smaller radiolabeled constructs can be used to more accurately evaluate tumor expressed receptors. PROCEDURES: The Krogh cylinder distributed model, including bivalent binding and variable intervessel distances, simulated distribution of smaller constructs in the presence of increasing doses of labeled antibody forms. RESULTS: Smaller constructs <25 kDa accessed binding sites more uniformly at large distances from blood vessels compared with larger constructs and intact antibody. These observations were consistent for different affinity and internalization characteristics of constructs. As predicted, a higher dose of unlabeled intact antibody was required to block binding to these distant receptor sites. CONCLUSIONS: Small radiolabeled constructs provide more accurate information on total receptor expression in tumors and reveal the need for higher antibody doses for target receptor blockade.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Simulación por Computador , Imagen Molecular/métodos , Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Humanos , Peso Molecular
18.
Cancer Res ; 64(17): 6200-6, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342405

RESUMEN

Antitumor diabody molecules are noncovalent single-chain Fv dimers that recapitulate the divalent binding properties of native IgG antibodies. Diabodies are capable of substantial accumulation in tumor xenografts expressing relevant antigens in immunodeficient mouse models. With a Mr of approximately 55,000, diabodies are rapidly cleared from the circulation, resulting in tumor-to-blood ratios that significantly exceed those achieved early after the administration of monoclonal antibodies. We have evaluated the therapeutic potential of the beta-emitting isotope yttrium-90 (t1/2, 64 hours) conjugated to the C6.5K-A diabody that specifically targets the HER2/neu human tumor-associated antigen. We have found that a single intravenous dose of 150 microCi (200 microg) 90Y-CHX-A"-C6.5K-A diabody substantially inhibits the growth rates of established MDA-361/DYT2 human breast tumor xenografts in athymic nude mice. In contrast, 300 microCi (300 microg) 90Y-CHX-A"-C6.5K-A diabody resulted in only a minor delay in the growth of SK-OV-3 human ovarian cancer xenografts. The maximum tolerated dose was also dependent on the tumor xenograft model used. These studies indicate that genetically engineered antitumor diabody molecules can be used as effective vehicles for radioimmunotherapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Inmunotoxinas/farmacología , Isotiocianatos/farmacología , Neoplasias Ováricas/radioterapia , Ácido Pentético/análogos & derivados , Ácido Pentético/farmacología , Radioisótopos de Itrio/farmacología , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Región Variable de Inmunoglobulina/administración & dosificación , Región Variable de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/farmacología , Inmunotoxinas/farmacocinética , Isotiocianatos/administración & dosificación , Isotiocianatos/farmacocinética , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Ácido Pentético/administración & dosificación , Ácido Pentético/farmacocinética , Radioinmunoterapia , Cintigrafía , Receptor ErbB-2/biosíntesis , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto , Radioisótopos de Itrio/administración & dosificación , Radioisótopos de Itrio/farmacocinética
19.
J Immunother ; 39(3): 117-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26938945

RESUMEN

The development of antibody drug conjugates has provided enhanced potency to tumor-targeting antibodies by the addition of highly potent payloads. In the case of trastuzumab-DM1 (T-DM1), approved for the treatment of metastatic breast cancer, the addition of mertansine (DM1) to trastuzumab substantially increased progression-free survival. Despite these improvements, most patients eventually relapse due to complex mechanisms of resistance often associated with small molecule chemotherapeutics. Therefore, identifying payloads with different mechanisms of action (MOA) is critical for increasing the efficacy of targeted therapeutics and ultimately improving patient outcomes. To evaluate payloads with different MOA, deBouganin, a deimmunized plant toxin that inhibits protein synthesis, was conjugated to trastuzumab and compared with T-DM1 both in vitro and in vivo. The trastuzumab-deBouganin conjugate (T-deB) demonstrated greater potency in vitro against most cells lines with high levels of Her2 expression. In addition, T-deB, unlike T-DM1, was unaffected by inhibitors of multidrug resistance, Bcl-2-mediated resistance, or Her2-Her3 dimerization. Contrary to T-DM1 that showed only minimal cytotoxicity, T-deB was highly potent in vitro against tumor cells with cancer stem cell properties. Overall, the results demonstrate the potency and efficacy of deBouganin and emphasize the importance of using payloads with different MOAs. The data suggest that deBouganin could be a highly effective against tumor cell phenotypes not being addressed by current antibody drug conjugate formats and thereby provide prolonged clinical benefit.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Inmunoconjugados/farmacología , Maitansina/análogos & derivados , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Ado-Trastuzumab Emtansina , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Genes bcl-2 , Genes erbB-2 , Humanos , Maitansina/farmacología , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Trastuzumab , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Mol Cancer Ther ; 15(2): 313-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26719579

RESUMEN

T cells recognize cancer cells via HLA/peptide complexes, and when disease overtakes these immune mechanisms, immunotherapy can exogenously target these same HLA/peptide surface markers. We previously identified an HLA-A2-presented peptide derived from macrophage migration inhibitory factor (MIF) and generated antibody RL21A against this HLA-A2/MIF complex. The objective of the current study was to assess the potential for targeting the HLA-A2/MIF complex in ovarian cancer. First, MIF peptide FLSELTQQL was eluted from the HLA-A2 of the human cancerous ovarian cell lines SKOV3, A2780, OV90, and FHIOSE118hi and detected by mass spectrometry. By flow cytometry, RL21A was shown to specifically stain these four cell lines in the context of HLA-A2. Next, partially matched HLA-A*02:01+ ovarian cancer (n = 27) and normal fallopian tube (n = 24) tissues were stained with RL21A by immunohistochemistry to assess differential HLA-A2/MIF complex expression. Ovarian tumor tissues revealed significantly increased RL21A staining compared with normal fallopian tube epithelium (P < 0.0001), with minimal staining of normal stroma and blood vessels (P < 0.0001 and P < 0.001 compared with tumor cells) suggesting a therapeutic window. We then demonstrated the anticancer activity of toxin-bound RL21A via the dose-dependent killing of ovarian cancer cells. In summary, MIF-derived peptide FLSELTQQL is HLA-A2-presented and recognized by RL21A on ovarian cancer cell lines and patient tumor tissues, and targeting of this HLA-A2/MIF complex with toxin-bound RL21A can induce ovarian cancer cell death. These results suggest that the HLA-A2/MIF complex should be further explored as a cell-surface target for ovarian cancer immunotherapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Antígeno HLA-A2/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Neoplasias Ováricas/metabolismo , Péptidos/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Línea Celular Tumoral , Femenino , Antígeno HLA-A2/inmunología , Humanos , Neoplasias Ováricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA