Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
ACS Synth Biol ; 10(5): 1176-1183, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33856201

RESUMEN

Various antibody-redirected immunotherapeutic approaches, including antibody-drug conjugates (ADCs), bispecific antibodies (bsAbs), and chimeric antigen receptor-T (CAR-T) cells, have been devised to produce specific activity against various cancer types. Using genetically encoded unnatural amino acids, we generated a homogeneous Her2-targeted ADC, a T cell-redirected bsAb, and a FITC-modified antibody capable of redirecting anti-FITC CAR-T (switchable CAR-T; sCAR-T) cells to target different Her2-expressing breast cancers. sCAR-T cells showed activity against Her2-expressing tumor cells comparable to that of conventional anti-Her2 CAR-T cells and superior to that of ADC- and bsAb-based approaches. To prevent antigen escape, we designed bispecific sCAR-T cells targeting both the Her2 receptor and IGF1R, which showed an overall improved activity against cancer cells with low Her2 expression. This study increases our understanding of various explored cancer therapeutics and underscores the efficient application of sCAR-T cells as a promising therapeutic option for breast cancer patients with low or heterogeneous antigen expression.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Neoplasias de la Mama/metabolismo , Inmunoconjugados/inmunología , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/inmunología , Receptor IGF Tipo 1/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Aminoácidos/genética , Deriva y Cambio Antigénico/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Femenino , Fluoresceína-5-Isotiocianato , Humanos , Inmunoterapia Adoptiva/métodos , Terapia Molecular Dirigida/métodos
2.
Immunotherapy ; 13(2): 125-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33172323

RESUMEN

Aim: We report results of a first-in-human study of pasotuxizumab, a PSMA bispecific T-cell engager (BiTE®) immune therapy mediating T-cell killing of tumor cells in patients with advanced castration-resistant prostate cancer. Patients & methods: We assessed once-daily subcutaneous (SC) pasotuxizumab. All SC patients developed antidrug antibodies; therefore, continuous intravenous (cIV) infusion was assessed. Results: A total of 47 patients received pasotuxizumab (SC: n = 31, 0.5-172 µg/d; cIV: n = 16, 5-80 µg/d). The SC maximum tolerated dose was 172.0 µg/d. A sponsor change stopped the cIV cohort early; maximum tolerated dose was not determined. PSA responders occurred (>50% PSA decline: SC, n = 9; cIV, n = 3), including two long-term responders. Conclusion: Data support pasotuxizumab safety in advanced castration-resistant prostate cancer and represent evidence of BiTE monotherapy efficacy in solid tumors. Clinical trial registration: NCT01723475 (ClinicalTrials.gov).


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos Inmunológicos , Neoplasias de la Próstata Resistentes a la Castración , Anciano , Anciano de 80 o más Años , Humanos , Masculino , Persona de Mediana Edad , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/uso terapéutico , Antígenos de Superficie/inmunología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor/sangre , Complejo CD3/inmunología , Glutamato Carboxipeptidasa II/inmunología , Inmunoterapia , Infusiones Intravenosas , Inyecciones Subcutáneas , Dosis Máxima Tolerada , Neoplasias de la Próstata Resistentes a la Castración/sangre , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Resultado del Tratamiento
3.
JCI Insight ; 5(7)2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32271166

RESUMEN

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Afinidad de Anticuerpos , Antineoplásicos Inmunológicos/inmunología , Receptor ErbB-2/inmunología , Animales , Anticuerpos Biespecíficos/química , Antineoplásicos Inmunológicos/química , Complejo CD3/química , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Macaca fascicularis , Receptor ErbB-2/química
4.
Clin Cancer Res ; 26(9): 2203-2215, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969333

RESUMEN

PURPOSE: Multiple myeloma (MM) patients with disease refractory to all available drugs have a poor outcome, indicating the need for new agents with novel mechanisms of action. EXPERIMENTAL DESIGN: We evaluated the anti-MM activity of the fully human BCMA×CD3 bispecific antibody JNJ-7957 in cell lines and bone marrow (BM) samples. The impact of several tumor- and host-related factors on sensitivity to JNJ-7957 therapy was also evaluated. RESULTS: We show that JNJ-7957 has potent activity against 4 MM cell lines, against tumor cells in 48 of 49 BM samples obtained from MM patients, and in 5 of 6 BM samples obtained from primary plasma cell leukemia patients. JNJ-7957 activity was significantly enhanced in patients with prior daratumumab treatment, which was partially due to enhanced killing capacity of daratumumab-exposed effector cells. BCMA expression did not affect activity of JNJ-7957. High T-cell frequencies and high effector:target ratios were associated with improved JNJ-7957-mediated lysis of MM cells. The PD-1/PD-L1 axis had a modest negative impact on JNJ-7957 activity against tumor cells from daratumumab-naïve MM patients. Soluble BCMA impaired the ability of JNJ-7957 to kill MM cells, although higher concentrations were able to overcome this negative effect. CONCLUSIONS: JNJ-7957 effectively kills MM cells ex vivo, including those from heavily pretreated MM patients, whereby several components of the immunosuppressive BM microenvironment had only modest effects on its killing capacity. Our findings support the ongoing trial with JNJ-7957 as single agent and provide the preclinical rationale for evaluating JNJ-7957 in combination with daratumumab in MM.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Antígeno de Maduración de Linfocitos B/inmunología , Complejo CD3/inmunología , Mieloma Múltiple/tratamiento farmacológico , Anticuerpos Biespecíficos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos/farmacología , Médula Ósea/patología , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Inmunoterapia/métodos , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Tumorales Cultivadas
5.
BioDrugs ; 34(2): 111-119, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31916225

RESUMEN

In autoimmune diseases, a highly complex network comprising diverse cytokines and their receptors on immune cells drives the inflammatory response. A number of therapeutic antibodies targeting these disease-related molecules have been approved for the treatment of autoimmune diseases. Bispecific antibodies (bsAbs), with binding specificity for two different target molecules, have recently been developed for a range of autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis, and tested in clinical trials. This review briefly describes the three main categories of bsAb structures developed for autoimmune diseases, including immunoglobulin G (IgG)-like, natural IgG, and tandem antibody fragment formats. The bsAbs developed and evaluated to date mainly target the depletion of T or B cells, the inhibition of T cell differentiation or activation, or the neutralization of proinflammatory cytokines. The clinical evaluation of bsAbs in autoimmune diseases is ongoing, with both successes (phase II trials of obexelimab in systemic lupus erythematosus) and failures (phase II trials of lutikizumab in osteoarthritis and romilkimab in idiopathic pulmonary fibrosis), and this review aims to provide a comprehensive, up-to-date summary of the clinical progress of bsAbs in this therapeutic area. Although many challenges remain, bsAbs offer new therapeutic options in the future direction of autoimmune disease treatments.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Ensayos Clínicos como Asunto , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Terapia Molecular Dirigida , Resultado del Tratamiento
6.
J Immunol ; 203(3): 585-592, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31332079

RESUMEN

Effector lymphocytes are multifunctional cells of the immune system that promote cytolysis of pathogen-infected cells and nascent tumors. Tumors must learn to evade effectors and employ a wide variety of mechanisms to do so. Bispecific Abs (BsAbs) are an emerging cancer immunotherapy approach seeking to re-engage either T effectors or NK cells with malignant cells. Possessing specificity for effector cells on one end and a tumor Ag on the other, these molecules work by attracting effectors to the target cell to build an immunologic synapse and induce tumor cell killing. The BsAb blinatumomab, for example, has specificity for the T cell-activating cell surface protein CD3 and the B cell Ag CD19. The only BsAb with regulatory approval currently, blinatumomab is used in the treatment of relapsed or refractory B cell acute lymphoblastic leukemia. Many additional BsAbs are in preclinical development, however, targeting many different tumor types. The variety of potential effector cells and cancer Ags, along with potential combination therapies, make BsAbs an active area of drug development. In this review, we discuss cancer recognition by the immune system and structural and mechanistic aspects of BsAbs. We summarize key steps in preclinical development and subsequent translation to medical practice. Future directions for BsAbs include combinations with a wide variety of both immunologic and nonimmunologic therapies. Defining their optimum clinical use is at early stages.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfocitos T/inmunología , Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Complejo CD3/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología
7.
Nat Rev Drug Discov ; 18(8): 585-608, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31175342

RESUMEN

The term bispecific antibody (bsAb) is used to describe a large family of molecules designed to recognize two different epitopes or antigens. BsAbs come in many formats, ranging from relatively small proteins, merely consisting of two linked antigen-binding fragments, to large immunoglobulin G (IgG)-like molecules with additional domains attached. An attractive bsAb feature is their potential for novel functionalities - that is, activities that do not exist in mixtures of the parental or reference antibodies. In these so-called obligate bsAbs, the physical linkage of the two binding specificities creates a dependency that can be temporal, with binding events occurring sequentially, or spatial, with binding events occurring simultaneously, such as in linking an effector to a target cell. To date, more than 20 different commercialized technology platforms are available for bsAb creation and development, 2 bsAbs are marketed and over 85 are in clinical development. Here, we review the current bsAb landscape from a mechanistic perspective, including a comprehensive overview of the pipeline.


Asunto(s)
Anticuerpos Biespecíficos , Diseño de Fármacos , Neoplasias , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Sitios de Unión de Anticuerpos , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
8.
MAbs ; 11(5): 861-869, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31099718

RESUMEN

Despite recent advances in the development of tools to predict immunogenicity risk of biotherapeutic molecules, the ability of a protein to elicit the formation of anti-drug antibodies (ADA) remains one of the most common causes for termination of clinical development programs. In this study, we use ADA assays to detect and measure pre-existing reactivity or the ability of a molecule to produce an ADA-like response in serum from treatment-naïve, healthy donors. We report herein that the magnitude of pre-existing reactivity evaluated pre-clinically and expressed as the 90th percentile of Tier 2 inhibition correlates with the subsequent rate of ADA emergence in the clinic. Furthermore, a multi-domain biotherapeutic (IgG-scFv bispecific antibody) showed the highest pre-existing reactivity and incidence of treatment-emergent ADA (TE-ADA) (57% and 93%, respectively). Using the components of the multidomain molecule in the Tier 2 step of the ADA assay, we were able to identify the scFv as the target of the serum pre-existing reactivity. Most importantly, the domain specificity of pre-existing ADA was the same as that of the TE-ADA from patients treated with the molecule. Based on these data, we propose the evaluation of the magnitude and of the domain specificity of pre-existing reactivity as a powerful tool to understand the immunogenic potential of novel biotherapeutics.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos de Cadena Única/inmunología , Adulto , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/sangre , Formación de Anticuerpos , Terapia Biológica/efectos adversos , Epítopos/inmunología , Femenino , Humanos , Sueros Inmunes/inmunología , Masculino , Persona de Mediana Edad , Riesgo , Anticuerpos de Cadena Única/efectos adversos , Anticuerpos de Cadena Única/sangre , Adulto Joven
9.
Clin Cancer Res ; 24(11): 2631-2641, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29463552

RESUMEN

Purpose: Flotetuzumab (MGD006 or S80880) is a bispecific molecule that recognizes CD3 and CD123 membrane proteins, redirecting T cells to kill CD123-expressing cells for the treatment of acute myeloid leukemia. In this study, we developed a mathematical model to characterize MGD006 exposure-response relationships and to assess the impact of its immunogenicity in cynomolgus monkeys.Experimental Design: Thirty-two animals received multiple escalating doses (100-300-600-1,000 ng/kg/day) via intravenous infusion continuously 4 days a week. The model reflects sequential binding of MGD006 to CD3 and CD123 receptors. Formation of the MGD006/CD3 complex was connected to total T cells undergoing trafficking, whereas the formation of the trimolecular complex results in T-cell activation and clonal expansion. Activated T cells were used to drive the peripheral depletion of CD123-positive cells. Anti-drug antibody development was linked to MGD006 disposition as an elimination pathway. Model validation was tested by predicting the activity of MGD006 in eight monkeys receiving continuous 7-day infusions.Results: MGD006 disposition and total T-cell and CD123-positive cell profiles were well characterized. Anti-drug antibody development led to the suppression of T-cell trafficking but did not systematically abolish CD123-positive cell depletion. Target cell depletion could persist after drug elimination owing to the self-proliferation of activated T cells generated during the first cycles. The model was externally validated with the 7-day infusion dosing schedule.Conclusions: A translational model was developed for MGD006 that features T-cell activation and expansion as a key driver of pharmacologic activity and provides a mechanistic quantitative platform to inform dosing strategies in ongoing clinical studies. Clin Cancer Res; 24(11); 2631-41. ©2018 AACR.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Algoritmos , Animales , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/inmunología , Complejo CD3/antagonistas & inhibidores , Complejo CD3/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Subunidad alfa del Receptor de Interleucina-3/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-3/inmunología , Isoanticuerpos/sangre , Isoanticuerpos/inmunología , Macaca fascicularis , Masculino , Modelos Teóricos , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Mol Immunol ; 67(2 Pt A): 95-106, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25637431

RESUMEN

Bispecific antibodies are on the cusp of coming of age as therapeutics more than half a century after they were first described. Two bispecific antibodies, catumaxomab (Removab(®), anti-EpCAM×anti-CD3) and blinatumomab (Blincyto(®), anti-CD19×anti-CD3) are approved for therapy, and >30 additional bispecific antibodies are currently in clinical development. Many of these investigational bispecific antibody drugs are designed to retarget T cells to kill tumor cells, whereas most others are intended to interact with two different disease mediators such as cell surface receptors, soluble ligands and other proteins. The modular architecture of antibodies has been exploited to create more than 60 different bispecific antibody formats. These formats vary in many ways including their molecular weight, number of antigen-binding sites, spatial relationship between different binding sites, valency for each antigen, ability to support secondary immune functions and pharmacokinetic half-life. These diverse formats provide great opportunity to tailor the design of bispecific antibodies to match the proposed mechanisms of action and the intended clinical application.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Complejo CD3/inmunología , Complejo CD3/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Molécula de Adhesión Celular Epitelial , Humanos , Modelos Inmunológicos , Neoplasias/inmunología , Neoplasias/metabolismo
11.
Anal Chem ; 87(5): 2952-8, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25622025

RESUMEN

The multianalyte immunoassay (MIA) has attracted increasing attention due to its high sample throughput, short assay time, low sample consumption, and reduced overall cost. However, up to now, the reported MIA methods commonly require multiple antibodies since each antibody can recognize only one antigen. Herein, a novel bispecific monoclonal antibody (BsMcAb) that could bind methyl parathion and imidacloprid simultaneously was produced by a hybrid hybridomas strategy. A chemiluminescence (CL) reaction kinetics-resolved strategy was designed for MIA of methyl parathion and imidacloprid using the BsMcAb as the unique recognition reagent. Horseradish peroxidase (HRP) and alkaline phosphatase (ALP) were adopted as the signal probes to tag the haptens of the two pesticides due to their very different CL kinetic characteristics. After competitive immunoreactions, the HRP-tagged methyl parathion hapten and the ALP-tagged imidacloprid hapten were simultaneously bound to the BsMcAb since there were two different antigen-binding sites in it. Then, two CL reactions were simultaneously triggered by adding the CL coreactants, and the signals for methyl parathion and imidacloprid detections were collected at 0.6 and 1000 s, respectively. The linear ranges for methyl parathion and imidacloprid were both 1.0-500 ng/mL, with detection limits of 0.33 ng/mL (S/N = 3). The proposed method was successfully used to detect pesticides spiked in ginseng and American ginseng with acceptable recoveries of 80-118%. This proof-of-principle work demonstrated the feasibility of MIA using only one antibody.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Imidazoles/análisis , Inmunoensayo/métodos , Mediciones Luminiscentes/métodos , Metil Paratión/análisis , Nitrocompuestos/análisis , Panax/química , Plaguicidas/análisis , Fosfatasa Alcalina/metabolismo , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/metabolismo , Femenino , Peroxidasa de Rábano Silvestre/metabolismo , Hibridomas , Imidazoles/inmunología , Imidazoles/metabolismo , Inmunización , Insecticidas/análisis , Insecticidas/inmunología , Insecticidas/metabolismo , Límite de Detección , Metil Paratión/inmunología , Metil Paratión/metabolismo , Ratones , Ratones Endogámicos BALB C , Neonicotinoides , Nitrocompuestos/inmunología , Nitrocompuestos/metabolismo
12.
Mol Cancer Ther ; 13(7): 1803-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24944121

RESUMEN

Bispecific antibodies (BsAb) have proven to be useful targeting vectors for pretargeted radioimmunotherapy (PRIT). We sought to overcome key PRIT limitations such as high renal radiation exposure and immunogenicity (e.g., of streptavidin-antibody fusions), to advance clinical translation of this PRIT strategy for diasialoganglioside GD2-positive [GD2(+)] tumors. For this purpose, an IgG-scFv BsAb was engineered using the sequences for the anti-GD2 humanized monoclonal antibody hu3F8 and C825, a murine scFv antibody with high affinity for the chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) complexed with ß-particle-emitting radiometals such as (177)Lu and (90)Y. A three-step regimen, including hu3F8-C825, a dextran-based clearing agent, and p-aminobenzyl-DOTA radiolabeled with (177)Lu (as (177)Lu-DOTA-Bn; t1/2 = 6.71 days), was optimized in immunocompromised mice carrying subcutaneous human GD2(+) neuroblastoma (NB) xenografts. Absorbed doses for tumor and normal tissues were approximately 85 cGy/MBq and ≤3.7 cGy/MBq, respectively, with therapeutic indices (TI) of 142 for blood and 23 for kidney. A therapy study (n = 5/group; tumor volume, 240 ± 160 mm(3)) with three successive PRIT cycles (total (177)Lu: ∼33 MBq; tumor dose ∼3,400 cGy), revealed complete tumor response in 5 of 5 animals, with no recurrence up to 28 days after treatment. Tumor ablation was confirmed histologically in 4 of 5 mice, and normal organs showed minimal overall toxicities. All nontreated mice required sacrifice within 12 days (>1.0-cm(3) tumor volume). We conclude that this novel anti-GD2 PRIT approach has sufficient TI to successfully ablate subcutaneous GD2(+)-NB in mice while sparing kidney and bone marrow.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Gangliósidos/inmunología , Inmunoglobulina G/inmunología , Radiofármacos/farmacología , Animales , Anticuerpos Biespecíficos/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/farmacología , Lutecio/administración & dosificación , Lutecio/química , Ratones , Ratones Desnudos , Octreótido/análogos & derivados , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/farmacología , Radioinmunoterapia , Radiofármacos/química , Radiofármacos/inmunología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto , Radioisótopos de Itrio/administración & dosificación , Radioisótopos de Itrio/química
13.
Cancer Immunol Immunother ; 62(8): 1369-80, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23715927

RESUMEN

Combinatory strategies are becoming of increasing interest in cancer immunotherapy. Costimulation by individual members of the immunoglobulin-like (Ig)- and TNF superfamily have already shown promising antitumor potential, thus prompting the exploration of their synergistic abilities in combinatorial approaches. Here, we pursued a targeted strategy with antibody-fusion proteins composed of a tumor-directed antibody and the extracellular domain of the costimulatory ligand B7.1, 4-1BBL, OX40L, GITRL or LIGHT, respectively. Costimulatory activity was assessed in an experimental setting where initial T cell activation was induced by a bispecific antibody (tumor-related antigen × CD3). Advantage of combined targeted costimulation was shown for either B7.1 or 4-1BBL with OX40L, GITRL, LIGHT and 4-1BBL in terms of T cell proliferation and IFN-γ release. Since encouraging results were obtained by the combination of B7.1 and 4-1BBL, we adapted the model system for a time-shift setting. Here, enhanced proliferation and granzyme B expression as well as reduced PD-1 expression on the T cell population demonstrated the benefit of costimulation-assisted restimulation. Finally, the antitumor potential of this combinatorial setting was confirmed in vivo in a lung metastasis mouse model. Thus, combinatorial approaches with costimulatory antibody-ligand fusion proteins seem a promising strategy to be further investigated for cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Experimentales/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Ligando 4-1BB/genética , Ligando 4-1BB/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Granzimas/inmunología , Granzimas/metabolismo , Células HEK293 , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ligandos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
14.
Arthritis Rheum ; 62(7): 1933-43, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506263

RESUMEN

OBJECTIVE: To exploit the physiologic Fcgamma receptor IIb (CD32B) inhibitory coupling mechanism to control B cell activation by constructing a novel bispecific diabody scaffold, termed a dual-affinity retargeting (DART) molecule, for therapeutic applications. METHODS: DART molecules were constructed by pairing an Fv region from a monoclonal antibody (mAb) directed against CD32B with an Fv region from a mAb directed against CD79B, the beta-chain of the invariant signal-transducing dimer of the B cell receptor complex. DART molecules were characterized physicochemically and for their ability to simultaneously bind the target receptors in vitro and in intact cells. The ability of the DART molecules to negatively control B cell activation was determined by calcium mobilization, by tyrosine phosphorylation of signaling molecules, and by proliferation and Ig secretion assays. A DART molecule specific for the mouse ortholog of CD32B and CD79B was also constructed and tested for its ability to inhibit B cell proliferation in vitro and to control disease severity in a collagen-induced arthritis (CIA) model. RESULTS: DART molecules were able to specifically bind and coligate their target molecules on the surface of B cells and demonstrated a preferential simultaneous binding to both receptors on the same cell. DART molecules triggered the CD32B-mediated inhibitory signaling pathway in activated B cells, which translated into inhibition of B cell proliferation and Ig secretion. A DART molecule directed against the mouse orthologs was effective in inhibiting the development of CIA in DBA/1 mice. CONCLUSION: This innovative bispecific antibody scaffold that simultaneously engages activating and inhibitory receptors enables novel therapeutic approaches for the treatment of rheumatoid arthritis and potentially other autoimmune and inflammatory diseases in humans.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Linfocitos B/efectos de los fármacos , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Receptores de IgG , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Antígenos CD79/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dimerización , Femenino , Humanos , Inmunoglobulinas/metabolismo , Inmunosupresores/inmunología , Inmunosupresores/farmacocinética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Receptores de IgG/inmunología , Transducción de Señal , Bazo/citología , Andamios del Tejido
15.
MAbs ; 1(4): 348-56, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20068406

RESUMEN

The administration of anti-CD3 antibodies, either unmodified or in bispecific formats, has been shown to kill tumors. However, their activity needs to be carefully controlled. We have approached this problem by inhibiting their anti-CD3 activity until it is required. Folated anti-human CD3 antibody bispecific conjugates were therefore synthesised in which the folate portion of the conjugates remained free to bind to folate receptor (FR) expressing cancer cells, whilst their anti-CD3 activity was reversibly inhibited. On irradiation with UV-A light, the T-cell binding activity of the anti-CD3 antibody can be restored only when and where it is required, i.e., adjacent to a tumor. Conjugate bound to FR expressed on normal tissues in other parts of the body remains inactive. This report describes the preclinical in vivo testing of these conjugates in transgenic mice whose T-cells express human CD3 molecules. When the 'cloaked' conjugates were reactivated in the region of the primary tumor, both primary tumor growth and liver metastasis were markedly reduced. That the deliberate targeting of T-cell activity locally to the primary tumor also resulted in reduced distant metastatic growth was a key finding. Light-activatable bispecific antibody conjugates similar to those described here offer a means to control T-cell targeting with a much higher degree of specificity to tumors because they minimize potentially dangerous and unwanted side effects in non-illuminated areas. The addition of light-specific targeting to the inherent tumor specific targeting of therapeutic antibody conjugates could result in the development of safer treatments for patients.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3 , Luz , Neoplasias Ováricas/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Complejo CD3/inmunología , Complejo CD3/uso terapéutico , Proteínas Portadoras , Evaluación Preclínica de Medicamentos , Femenino , Receptores de Folato Anclados a GPI , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias Ováricas/inmunología , Receptores de Superficie Celular , Linfocitos T
16.
J Immunol Methods ; 306(1-2): 80-92, 2005 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16202424

RESUMEN

We have developed a universal DC targeting vehicle that could be a convenient method to deliver any type of antigen to DC. P125, a quadroma (hybrid-hybridoma) secreting bispecific monoclonal antibodies (bsmAb), with one paratope specific for mouse DC DEC-205 and another paratope specific for biotin, was developed by PEG-fusion of the two parental hybridomas and selected by a fluorescence activated cell sorter. The bsmAb were purified using a biotin-Agarose column and the bsmAb activity was demonstrated using ELISA method employing mouse bone marrow DC and biotinylated BSA. Both confocal microscopy and ELISA studies have shown enhanced binding and internalization of biotinylated and FITC-labelled M13 to DC cell in the presence of bsmAb. In vivo studies in mice with biotinylated OVA has shown that in the presence of bsmAb and anti-CD40 mAb, both humoral and cell-mediated responses can be augmented. In addition, only a low concentration of antigen (500 fold less) is required using bsmAb to achieve a similar immune response in mice that were immunized using complete Freund's adjuvant. In the absence of traditional adjuvants, bsmAb targeting of biotinylated antigens to DC could be an alternative, convenient method to deliver antigens to DC. Moreover, this method could be an alternative method to ex vivo stimulation of DC to overcome DC defects and for treatment of cancer.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Presentación de Antígeno , Antígenos CD/inmunología , Biotina/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/química , Antígenos/química , Antígenos/inmunología , Biotina/química , Biotinilación , Femenino , Hibridomas , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Polietilenglicoles/química
17.
J Immunol ; 158(8): 3965-70, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9103467

RESUMEN

Anti-CD3 x anti-tumor-bispecific Abs have been used to redirect cytotoxic T cells to tumor cells in an MHC-unrestricted fashion and to induce their rejection in vivo. We have recently described a recombinant bispecific single-chain Ab that combines four different V regions of two Abs, anti-17-1A and anti-CD3, on one polypeptide chain. It folds correctly to a 60-kDa globular protein and is secreted in fully functional form by a high producer Chinese hamster ovary cell line. In this work, we report that its remarkable cytotoxicity against 17-1A+ tumor cells is exerted via T cells without an apparent engagement of a detectable costimulatory pathway. T cells are activated only by the bispecific Ab when coincubated with 17-1A+ target cells. In a chromium release assay, CD8+ T cells reach maximal tumor cell cytotoxicity within 4 h, while CD4+ T cells need about 20 h to reach similar levels of cytotoxicity. Addition of costimulatory CD28 Abs did not lead to a further increase in cytotoxicity. Its remarkable stability at 37 degrees in serum, the ease of production, and purification by affinity chromatography via polyhistidine tail make this smaller version of a bispecific Ab a promising candidate for a therapeutic trial in patients with solid tumor. Because adjuvant therapy with an intact, much less cytotoxic IgG2a Ab against the 17-1A target had already increased the 7-yr survival of colorectal cancer patients by 30%, the presented small bispecific construct lacking the immunogenic murine Fc region as well as autochthonous T lymphocyte stimulatory activity warrants a therapeutic trial in patients with minimal residual 17-1A+ cancer.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígenos de Neoplasias/inmunología , Complejo CD3/inmunología , Citotoxicidad Inmunológica , Linfocitos T/inmunología , Animales , Células CHO , Cricetinae , Humanos , Ratones , Células Tumorales Cultivadas
18.
Surg Oncol ; 3(5): 279-85, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7889221

RESUMEN

Bispecific antibodies (BAb) direct T-lymphocytes to lyse selected tumour targets, both in vitro and in vivo. Significant tumour cell lysis with BAb requires pre-expansion of T-lymphocytes, which may be achieved in vitro by the addition of anti-CD3 monoclonal antibody plus interleukin-2 (IL-2), but anti-CD3 may cause immunosuppression. We investigated an alternative agent for in vivo immunostimulation, staphyloccal enterotoxin B (SEB), which selectively activates certain T-cell subsets and may result in less immunosuppression than with anti-CD3. We activated T-lymphocytes in vivo with SEB, expanded them in vitro with IL-2, and directed them against a tumour target with the BAb 500A2 x 96.5, specific for the murine CD3 antigen and the melanoma p97 antigen expressed by the CL62 tumour. C3H mice received SEB 50 micrograms intraperitoneally (i.p.). After 18 h mice were sacrificed and splenocytes extracted and either passed over a nylon wool column to isolate T-lymphocytes, or cultured in vitro for 3 to 7 days with 100 U ml-1 of IL-2. A 4-h chromium-release assay was used to assess the ability of T-lymphocytes to lyse the tumour target CL 62 in the presence or absence of the bispecific antibody 500A2 x 96.5. The addition of BAb significantly enhanced tumour lysis by SEB activated cells after a period of in vitro culture with IL-2. In vivo SEB results in the activation of T-lymphocytes which may be directed by bispecific antibodies to increase the lysis of selected tumour targets in vitro.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Enterotoxinas/farmacología , Melanoma Experimental/inmunología , Linfocitos T/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Citotoxicidad Inmunológica , Femenino , Citometría de Flujo , Técnicas In Vitro , Interleucina-2/farmacología , Activación de Linfocitos , Ratones , Ratones Endogámicos C3H , Staphylococcus aureus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA