Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 21(4): 895-903, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23459515

RESUMEN

Natural killer (NK) cells represent a key component of the innate immune system against cancer. Nevertheless, malignant diseases arise in immunocompetent individuals despite tumor immunosurveillance. Hodgkin lymphoma (HL) is characterized by CD30(+) tumor cells and a massive infiltration of immune effector cells in affected lymph nodes. The latter obviously fail to eliminate the malignant cell population. Here, we tested for functional NK cell defects in HL and suggest an improvement of NK function by therapeutic means. We demonstrate that peripheral NK cells (pNK) from patients with HL fail to eliminate HL cell lines in ex vivo killing assays. Impaired NK cell function correlated with elevated serum levels of soluble ligands for NK cell receptors NKp30 (BAG6/BAT3) and NKG2D (MICA), factors known to constrict NK cell function. In vitro, NK cell cytotoxicity could be restored by an NKG2D/NKp30-independent bispecific antibody construct (CD30xCD16A). It artificially links the tumor receptor CD30 with the cytotoxicity NK cell receptor CD16A. Moreover, we observed that NK cells from patients treated with this construct were generally activated and displayed a restored cytotoxicity against HL target cells. These data suggest that reversible suppression of NK cell activity contributes to immune evasion in HL and can be antagonized therapeutically.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Enfermedad de Hodgkin/terapia , Células Asesinas Naturales/inmunología , Anticuerpos Biespecíficos/farmacología , Línea Celular Tumoral , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica
2.
Cancer Immunol Immunother ; 61(10): 1869-75, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22976535

RESUMEN

Bispecific tetravalent antibodies (TandAb) directed against the B cell surface marker CD19 and activating receptors on T or NK cells (CD19 × CD3 or CD19 × CD16) have shown promising effects in vitro and in preclinical studies. Here, we examine the cytotoxic efficacy of T and NK cells from patients with B cell Non-Hodgkin's Lymphoma (NHL) against B-lymphoma cells following the binding of the matching TandAb. The addition of CD19 × CD16 TandAb led to a threefold increase in NK cell activation in the presence of B-lymphoma cells. Similarly, T cells displayed a sevenfold increase in cytotoxic activity after the addition of CD19 × CD3 TandAb. Comparison of T and NK cell effector function of patients and healthy controls showed comparable levels of cytotoxic activity in response to lymphoma cells and no reduction in functional activity due to age, disease stage or the type and amount of previous therapy. Thus, T and NK cells of patients with B cell NHL are fully capable of being activated by therapeutic crosslinking antibodies. These results provide a rationale for the use of TandAbs for patients with B cell NHL, particularly in cases where remission with minimal residual disease could be achieved by cytotoxic chemotherapy.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Células Asesinas Naturales/inmunología , Linfoma de Células B/terapia , Linfocitos T Citotóxicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Biespecíficos/inmunología , Antígenos CD19/inmunología , Antineoplásicos/inmunología , Complejo CD3/inmunología , Células Cultivadas , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfoma de Células B/inmunología , Masculino , Persona de Mediana Edad , Receptores de IgG/inmunología
3.
Antibodies (Basel) ; 11(1)2022 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-35225870

RESUMEN

Innate cell engager (ICE®) constructs are bispecific tetravalent antibodies targeting specific tumor antigens and simultaneously engaging natural killer (NK) cell and macrophage receptors for the destruction of tumor cells. Pre-complexing of ICE® constructs with adoptive NK cells is a novel approach to enhance NK cell activity. The suitability of such complexes for cryopreservation, whilst retaining the biological activity and specificity, may enable the development of off-the-shelf NK cell products. This study investigates the binding affinity of ICE® constructs targeting EpCAM and NK cell receptors CD16A, NKG2D, or NKp46 to the corresponding antigens, the ICE® antitumor activity, and feasibility of cryopreservation. Cell surface retention assays using primary NK cells confirmed a substantially slower ICE® construct dissociation kinetics compared with control molecules, suggesting the formation of durable complexes independently of the CD16A polymorphism. The high-affinity NK cell and EpCAM/CD16A ICE® complexes were superior to those engaging NKG2D or NKp46 receptors when tested for the NK-cell-mediated elimination of EpCAM-expressing tumor cells. Moreover, the potency and efficacy of these complexes were unaffected after a single freeze-thaw cycle. CD16A-selective ICE® drug candidates complexed with NK cells hold promise as novel cryopreserved off-the-shelf NK cell products with chimeric antigen receptor-like NK cell properties, capable of effective depletion of tumor cells.

4.
Leukemia ; 36(4): 1006-1014, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35001074

RESUMEN

Despite the recent progress, multiple myeloma (MM) is still essentially incurable and there is a need for additional effective treatments with good tolerability. RO7297089 is a novel bispecific BCMA/CD16A-directed innate cell engager (ICE®) designed to induce BCMA+ MM cell lysis through high affinity binding of CD16A and retargeting of NK cell cytotoxicity and macrophage phagocytosis. Unlike conventional antibodies approved in MM, RO7297089 selectively targets CD16A with no binding of other Fcγ receptors, including CD16B on neutrophils, and irrespective of 158V/F polymorphism, and its activity is less affected by competing IgG suggesting activity in the presence of M-protein. Structural analysis revealed this is due to selective interaction with a single residue (Y140) uniquely present in CD16A opposite the Fc binding site. RO7297089 induced tumor cell killing more potently than conventional antibodies (wild-type and Fc-enhanced) and induced lysis of BCMA+ cells at very low effector-to-target ratios. Preclinical toxicology data suggested a favorable safety profile as in vitro cytokine release was minimal and no RO7297089-related mortalities or adverse events were observed in cynomolgus monkeys. These data suggest good tolerability and the potential of RO7297089 to be a novel effective treatment of MM patients.


Asunto(s)
Anticuerpos Biespecíficos , Mieloma Múltiple , Antígeno de Maduración de Linfocitos B , Humanos , Mieloma Múltiple/tratamiento farmacológico , Fagocitosis , Receptores de IgG
5.
MAbs ; 13(1): 1950264, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34325617

RESUMEN

Epidermal growth factor receptor (EGFR)-targeted cancer therapy such as anti-EGFR monoclonal antibodies and tyrosine kinase inhibitors have demonstrated clinical efficacy. However, there remains a medical need addressing limitations of these therapies, which include a narrow therapeutic window mainly due to skin and organ toxicity, and primary and secondary resistance mechanisms of the EGFR-signaling cascade (e.g., RAS-mutated colorectal cancer). Using the redirected optimized cell killing (ROCK®) antibody platform, we have developed AFM24, a novel bispecific, IgG1-scFv fusion antibody targeting CD16A on innate immune cells, and EGFR on tumor cells. We herein demonstrate binding of AFM24 to CD16A on natural killer (NK) cells and macrophages with KD values in the low nanomolar range and to various EGFR-expressing tumor cells. AFM24 was highly potent and effective for antibody-dependent cell-mediated cytotoxicity via NK cells, and also mediated antibody-dependent cellular phagocytosis via macrophages in vitro. Importantly, AFM24 was effective toward a variety of EGFR-expressing tumor cells, regardless of EGFR expression level and KRAS/BRAF mutational status. In vivo, AFM24 was well tolerated up to the highest dose (75 mg/kg) when administered to cynomolgus monkeys once weekly for 28 days. Notably, skin and other toxicities were not observed. A transient elevation of interleukin-6 levels was detected at all dose levels, 2-4 hours post-dose, which returned to baseline levels after 24 hours. These results emphasize the promise of bispecific innate cell engagers as an alternative cancer therapy and demonstrate the potential for AFM24 to effectively target tumors expressing varying levels of EGFR, regardless of their mutational status.Abbreviations: ADA: antidrug antibody; ADCC: antibody-dependent cell-mediated cytotoxicity; ADCP: antibody-dependent cellular phagocytosis; AUC: area under the curve; CAR: chimeric-antigen receptor; CD: Cluster of differentiation; CRC :colorectal cancer; ECD: extracellular domain; EGF: epidermal growth factorEGFR epidermal growth factor receptor; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; Fc: fragment, crystallizableFv variable fragment; HNSCC: head and neck squamous carcinomaIL interleukinm; Ab monoclonal antibody; MOA: mechanism of action; NK :natural killer; NSCLC: non-small cell lung cancer; PBMC: peripheral blood mononuclear cell; PBS: phosphate-buffered saline; PD: pharmacodynamic; ROCK: redirected optimized cell killing; RSV: respiratory syncytial virus; SABC: specific antibody binding capacity; SD: standard deviation; TAM: tumor-associated macrophage; TKI: tyrosine kinase inhibitor; WT: wildtype.


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos Inmunológicos , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Proteínas de Neoplasias , Neoplasias/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Receptores de IgG , Células A549 , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Células HCT116 , Células HT29 , Humanos , Células Asesinas Naturales/patología , Células MCF-7 , Macaca fascicularis , Macrófagos/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/inmunología
6.
Eur J Nucl Med Mol Imaging ; 37(7): 1397-407, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20157706

RESUMEN

PURPOSE: Recombinant antibodies isolated from human antibody libraries have excellent affinities and high target specificity. As full-length IgGs are cleared inadequately slowly from the circulation, the aim of this work was to figure out which kind of recombinant antibody fragment proves to be appropriate for imaging epithelial cell adhesion molecule (EpCAM)-expressing tumours with the short-living radioisotope (68)Ga. METHODS: In order to combine the promising tumour targeting properties of antibodies with (68)Ga, four antibody variants with the same specificity and origin only differing in molecular weight were constructed for comparison. Therefore, the binding domains of a single-chain fragment variable (scFv) isolated from a human naïve antibody library were modified genetically to construct the respective full-length IgG, the tria- and diabody variants. These molecules were conjugated with the bifunctional chelating agent N,N'-bis[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC) to enable (68)Ga labelling at ambient temperature and compared in biodistribution and immuno-PET imaging experiments. RESULTS: The antibody variants with identical specificity proved to have the correct molecular weight, high binding affinity and specificity to their antigen, EpCAM. Radiometal complexation was efficiently performed at room temperature leading to (68)Ga-labelled antibodies with unchanged binding properties compared to the original antibody variants. The best targeting properties were obtained with the scFv and especially with the diabody. The triabody showed higher absolute tumour uptake but only moderate clearance from circulation. CONCLUSION: The antibody variants differed considerably in normal organ uptake, clearance from circulation and tumour accumulation. The data demonstrate the feasibility of imaging solid tumours with the (68)Ga-labelled diabody format. This type of recombinant protein might be a promising carrier even for the short-lived radiometal (68)Ga to support e.g. the management of immunotherapy which may provide important information regarding receptor expression of solid tumours.


Asunto(s)
Fragmentos de Inmunoglobulinas , Neoplasias/diagnóstico por imagen , Neoplasias/inmunología , Tomografía de Emisión de Positrones/métodos , Proteínas Recombinantes , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Femenino , Radioisótopos de Galio , Regulación Neoplásica de la Expresión Génica , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética
7.
Mol Immunol ; 45(1): 144-51, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17576014

RESUMEN

Transmissible spongiform encephalopathies are a group of neurological disorders associated with the deposition of PrP(Sc), an abnormal form of the cellular prion protein PrP(c). The 37 kDa/67 kDa laminin receptor (LRP/LR) has been identified as a prion receptor and several lines of evidence strongly suggest that this protein plays a role during prion pathogenesis. Here we report the selection of recombinant single chain antibodies (scFvs) directed against LRP from naïve and synthetic phage scFv libraries for therapeutic application. Western blotting and FACS analysis confirmed a specific LRP/LR recognition pattern of the two selected scFvs S18 and N3. Both scFvs specifically interfered with the PrP/LRP interaction in vitro. High yield production of the scFvs of approx. 1mg/l of culture medium was achieved in E. coli. Passive immunotransfer of the scFv S18 antibody reduced PrP(Sc) levels by approx. 40% in the spleen of scrapie infected C57BL/6 mice 90 days post scFv injection, suggesting that scFv S18 interferes with peripheral PrP(Sc) propagation, without a significant prolongation of incubation and survival times.


Asunto(s)
Anticuerpos/farmacología , Región Variable de Inmunoglobulina/farmacología , Enfermedades por Prión/terapia , Receptores de Laminina/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Especificidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos/inmunología , Modelos Animales de Enfermedad , Mapeo Epitopo , Femenino , Humanos , Inmunización Pasiva , Cinética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Peso Molecular , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Unión Proteica/efectos de los fármacos , Scrapie/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología
8.
J Immunother ; 42(5): 180-188, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31090657

RESUMEN

Immunotherapy of B-cell malignancies with bispecific antibodies is an emerging treatment option. However, not all patients benefit from these therapies, presumably due to pretreatment regimens. Therefore, we determined the effect of different treatment lines on the activity of T cells and their responsiveness to AFM11. AFM11 is a tetravalent, bispecific CD19/CD3 immunoengager based on Affimed's ROCK platform, currently being investigated in phase I clinical trials for non-Hodgkin lymphoma and acute lymphoblastic leukemia. T cells from B-cell lymphoma patients treated with either rituximab+bendamustine (R-Benda), rituximab+CHOP (R-CHOP), or with high-dose BEAM chemotherapy (HD-BEAM) and autologous HSCT were compared with T cells from healthy donors. Overall, in these patients, T-cell numbers were significantly reduced. To determine whether distinct chemotherapy affects AFM11 efficacy, functional T-cell assays were performed. It is interesting to note that, only in assays that combine target cell lysis, cytokine production and proliferation over 4 days at an effector to target ratio of up to 1:25 significant differences could be detected between the different treatment groups: T cells after R-CHOP showed only modest decrease in their functionality when compared with healthy controls, whereas R-Benda and HD-BEAM had a profound effect on AFM11-induced T-cell cytotoxicity. In conclusion, T cells from lymphoma patients are reduced in number and have functional defects following treatment with certain chemotherapy regimens, also reducing AFM11 efficacy. Importantly, AFM11 was still able to trigger B-cell-directed T-cell immunity in all treatment groups.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Complejo CD3/antagonistas & inhibidores , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Anciano , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD19/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular , Citocinas/metabolismo , Femenino , Humanos , Leucemia de Células B/tratamiento farmacológico , Leucemia de Células B/inmunología , Leucemia de Células B/metabolismo , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Masculino , Persona de Mediana Edad , Linfocitos T/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
9.
MAbs ; 11(5): 899-918, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31172847

RESUMEN

Redirection of immune cells to efficiently eliminate tumor cells holds great promise. Natural killer cells (NK), macrophages, or T cells are specifically engaged with target cells expressing markers after infection or neoplastic transformation, resulting in their activation and subsequent killing of those targets. Multiple strategies to redirect immunity have been developed in the past two decades, but they have technical hurdles or cause undesirable side-effects, as exemplified by the T cell-based chimeric antigen receptor approaches (CAR-T therapies) or bispecific T cell engager platforms. Our first-in-class bispecific antibody redirecting innate immune cells to tumors (AFM13, a CD30/CD16A-specific innate immune cell engager) has shown signs of clinical efficacy in CD30-positive lymphomas and the potential to be safely administered, indicating a wider therapeutic window compared to T cell engaging therapies. AFM13 is the most advanced candidate from our fit-for-purpose redirected optimized cell killing (ROCK®) antibody platform, which comprises a plethora of CD16A-binding innate immune cell engagers with unique properties. Here, we discuss aspects of this modular platform, including the advantages of innate immune cell engagement over classical monoclonal antibodies and other engager concepts. We also present details on its potential to engineer a fit-for-purpose innate immune cell engager format that can be equipped with unique CD16A domains, modules that influence pharmacokinetic properties and molecular architectures that influence the activation of immune effectors, as well as tumor targeting. The ROCK® platform is aimed at the activation of innate immunity for the effective lysis of tumor cells and holds the promise of overcoming limitations of other approaches that redirect immune cells by widening the therapeutic window.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Animales , Anticuerpos Biespecíficos/farmacocinética , Línea Celular Tumoral , Femenino , Inmunoglobulina G/sangre , Inmunoterapia/métodos , Antígeno Ki-1/inmunología , Ratones , Receptores de IgG/inmunología
10.
Cancer Immunol Res ; 6(5): 517-527, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29514797

RESUMEN

CD16A is a potent cytotoxicity receptor on human natural killer (NK) cells, which can be exploited by therapeutic bispecific antibodies. So far, the effects of CD16A-mediated activation on NK cell effector functions beyond classical antibody-dependent cytotoxicity have remained poorly elucidated. Here, we investigated NK cell responses after exposure to therapeutic antibodies such as the tetravalent bispecific antibody AFM13 (CD30/CD16A), designed for the treatment of Hodgkin lymphoma and other CD30+ lymphomas. Our results reveal that CD16A engagement enhanced subsequent IL2- and IL15-driven NK cell proliferation and expansion. This effect involved the upregulation of CD25 (IL2Rα) and CD132 (γc) on NK cells, resulting in increased sensitivity to low-dose IL2 or to IL15. CD16A engagement initially induced NK cell cytotoxicity. The lower NK cell reactivity observed 1 day after CD16A engagement could be recovered by reculture in IL2 or IL15. After reculture in IL2 or IL15, these CD16A-experienced NK cells exerted more vigorous IFNγ production upon restimulation with tumor cells or cytokines. Importantly, after reculture, CD16A-experienced NK cells also exerted increased cytotoxicity toward different tumor targets, mainly through the activating NK cell receptor NKG2D. Our findings uncover a role for CD16A engagement in priming NK cell responses to restimulation by cytokines and tumor cells, indicative of a memory-like functionality. Our study suggests that combination of AFM13 with IL2 or IL15 may boost NK cell antitumor activity in patients by expanding tumor-reactive NK cells and enhancing NK cell reactivity, even upon repeated tumor encounters. Cancer Immunol Res; 6(5); 517-27. ©2018 AACR.


Asunto(s)
Proliferación Celular , Citotoxicidad Inmunológica/fisiología , Memoria Inmunológica/fisiología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/fisiología , Neoplasias/inmunología , Receptores de IgG/inmunología , Adulto , Animales , Anticuerpos Biespecíficos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Memoria Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Células Jurkat , Células K562 , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Neoplasias/terapia , Receptores de IgG/metabolismo
11.
Protein Eng Des Sel ; 30(9): 673-684, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28981915

RESUMEN

Bispecific antibodies that redirect the lytic activity of cytotoxic immune effector cells, such as T- and NK cells, onto tumor cells have emerged as a highly attractive and clinically validated treatment modality for hematological malignancies. Advancement of this therapeutic concept into solid tumor indications, however, is hampered by the scarcity of targetable antigens that are surface-expressed on tumor cells but demonstrate only limited expression on healthy tissues. To overcome this limitation, the concept of dual-targeting, i.e. the simultaneous targeting of two tumor-expressed surface antigens with limited co-expression on non-malignant cells, with multispecific antibodies has been proposed to increase tumor selectivity of antibody-induced effector cell cytotoxicity. Here, a novel CD16A (FcγRIIIa)-directed trispecific, tetravalent antibody format, termed aTriFlex, is described, that is capable of redirecting NK cell cytotoxicity to two surface-expressed antigens. Using a BCMA/CD200-based in vitro model system, the potential use of aTriFlex antibodies for dual-targeting and selective induction of NK cell-mediated target cell lysis was investigated. Bivalent bispecific target cell binding was found to result in significant avidity gains and up to 17-fold increased in vitro potency. These data suggest trispecific aTriFlex antibodies may support dual-targeting strategies to redirect NK cell cytotoxicity with increased selectivity to enable targeting of solid tumor antigens.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Antineoplásicos/biosíntesis , Citotoxicidad Inmunológica , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Antineoplásicos/genética , Afinidad de Anticuerpos , Antígenos CD/genética , Antígenos CD/inmunología , Antígeno de Maduración de Linfocitos B/genética , Antígeno de Maduración de Linfocitos B/inmunología , Células CHO , Técnicas de Cocultivo , Cricetulus , Expresión Génica , Humanos , Células Asesinas Naturales/citología , Activación de Linfocitos , Cultivo Primario de Células , Unión Proteica , Receptores de IgG/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
12.
Front Oncol ; 7: 100, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28596941

RESUMEN

To harness the cytotoxic capacity of immune cells for the treatment of solid tumors, we developed tetravalent, bispecific tandem diabody (TandAb) antibodies that recognize EGFRvIII, the deletion variant III of the epidermal growth factor receptor (EGFR), and CD3 on T-cells, thereby directing immune cells to eliminate EGFRvIII-positive tumor cells. Using phage display, we identified scFv antibodies selectively binding to EGFRvIII. These highly EGFRvIII-specific, fully human scFv were substantially improved by affinity maturation, achieving KDs in the picomolar range, and were used to construct a set of bispecific EGFRvIII-targeting TandAbs with a broad range of binding and cytotoxic properties. These antibodies exhibited an exquisite specificity for a distinguished epitope in the N-terminal portion of EGFRvIII, as shown on recombinant antigen in Western Blot, SPR, and ELISA, as well as on antigen-expressing cells in FACS assays, and did not bind to the wild-type EGFR. High-affinity EGFRvIII/CD3 TandAbs were most potent in killing assays, displaying cytotoxicity toward EGFRvIII-expressing CHO, F98 glioma, or human DK-MG cells with EC50 values in the range of 1-10 pM in vitro. They also demonstrated dose-dependent growth control in vivo in an EGFRvIII-positive subcutaneous xenograft tumor model. Together with the tumor-exclusive expression of EGFRvIII, the EGFRvIII/CD3 TandAbs' high specificity and strictly target-dependent activation with no off-target activity provide an opportunity to target tumor cells and spare normal tissues, thereby reducing the side effects associated with other anti-EGFR therapies. In summary, EGFRvIII/CD3 TandAbs are highly attractive therapeutic antibody candidates for selective immunotherapy of EGFRvIII-positive tumors.

13.
J Alzheimers Dis ; 49(3): 645-57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26484914

RESUMEN

The neuronal perturbations in Alzheimer's disease are attributed to the formation of extracellular amyloid-ß (Aß) neuritic plaques, composed predominantly of the neurotoxic Aß42 isoform. Although the plaques have demonstrated a role in synaptic dysfunction, neuronal cytotoxicity has been attributed to soluble Aß42 oligomers. The 37kDa/67kDa laminin receptor has been implicated in Aß42 shedding and Aß42-induced neuronal cytotoxicity, as well as internalization of this neurotoxic peptide. As the cellular prion protein binds to both LRP/LR and Aß42, the mechanism underlying this cytotoxicity may be indirectly due to the PrPc-Aß42 interaction with LRP/LR. The effects of this interaction were investigated by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assays. PrPc overexpression significantly enhanced Aß42 cytotoxicity in vitro, while PrP-/-  cells were more resistant to the cytotoxic effects of Aß42 and exhibited significantly less cell death than PrPc expressing N2a cells. Although anti-LRP/LR specific antibody IgG1-iS18 significantly enhanced cell viability in both pSFV1-huPrP1-253 transfected and non-transfected cells treated with exogenous Aß42, it failed to have any cell rescuing effect in PrP-/-  HpL3-4 cells. These results suggest that LRP/LR plays a significant role in Aß42-PrPc mediated cytotoxicity and that anti-LRP/LR specific antibodies may serve as potential therapeutic tools for Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Anticuerpos/farmacología , Fragmentos de Péptidos/toxicidad , Priones/metabolismo , Receptores de Laminina/inmunología , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ratones , Fragmentos de Péptidos/metabolismo , Priones/genética , Proteínas Ribosómicas , Transfección
14.
Clin Cancer Res ; 22(23): 5829-5838, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27189165

RESUMEN

PURPOSE: Randomized studies with gemtuzumab ozogamicin have validated CD33 as a target for antigen-specific immunotherapy of acute myelogenous leukemia (AML). Here, we investigated the potential of CD33/CD3-directed tandem diabodies (TandAbs) as novel treatment approach for AML. These tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a bivalent antibody and have a molecular weight exceeding the renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs. EXPERIMENTAL DESIGN: We constructed a series of TandAbs composed of anti-CD33 and anti-CD3 variable domains of diverse binding affinities and profiled their functional properties in CD33+ human leukemia cell lines, xenograft models, and AML patient samples. RESULTS: Our studies demonstrated that several CD33/CD3 TandAbs could induce potent, dose-dependent cytolysis of CD33+ AML cell lines. This effect was modulated by the effector-to-target cell ratio and strictly required the presence of T cells. Activation and proliferation of T cells and maximal AML cell cytolysis correlated with high avidity to both CD33 and CD3. High-avidity TandAbs were broadly active in primary specimens from patients with newly diagnosed or relapsed/refractory AML in vitro, with cytotoxic properties independent of CD33 receptor density and cytogenetic risk. Tumor growth delay and inhibition were observed in both prophylactic and established HL-60 xenograft models in immunodeficient mice. CONCLUSIONS: Our data show high efficacy of CD33/CD3 TandAbs in various preclinical models of human AML. Together, these findings support further study of CD33/CD3 TandAbs as novel immunotherapeutics for patients with AML. Clin Cancer Res; 22(23); 5829-38. ©2016 AACR.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Complejo CD3/inmunología , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Aminoglicósidos/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Sitios de Unión/inmunología , Línea Celular Tumoral , Gemtuzumab , Semivida , Humanos , Inmunoterapia/métodos , Ratones , Linfocitos T/inmunología
15.
J Mol Biol ; 330(1): 99-111, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12818205

RESUMEN

Bispecific single-chain Fv antibodies comprise four covalently linked immunoglobulin variable (VH and VL) domains of two different specificities. Depending on the order of the VH and VL domains and on the length of peptides separating them, the single-chain molecule either forms two single-chain Fv (scFv) modules from the adjacent domains of the same specificity, a so-called scFv-scFv tandem [(scFv)(2)], or folds head-to-tail with the formation of a diabody-like structure, a so-called bispecific single-chain diabody (scBsDb). We generated a number of four-domain constructs composed of the same VH and VL domains specific either for human CD19 or CD3, but arranged in different orders. When expressed in bacteria, all (scFv)(2) variants appeared to be only half-functional, binding to CD19 and demonstrating no CD3-binding activity. Only the diabody-like scBsDb could bind both antigens. Comparison of the scBsDb with a structurally similar non-covalent dimer (diabody) demonstrated a stabilizing effect of the linker in the middle of the scBsDb molecule. We demonstrated that the mechanism of inactivation of CD19xCD3 diabody under physiological conditions is initiated by a dissociation of the weaker (anti-CD3) VH/VL interface followed by domain swapping with the formation of non-active homodimers. The instability of one homodimer makes the process of diabody dissociation/reassociation irreversible, thus gradually decreasing the fraction of active molecules. The structural parameters influencing the formation of functional bispecific single-chain antibodies are indicated and ways of making relatively stable bispecific molecules are proposed.


Asunto(s)
Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/metabolismo , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Secuencia de Aminoácidos , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/farmacología , Antígenos/metabolismo , Antígenos CD19/genética , Complejo CD3/genética , Dimerización , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Escherichia coli/genética , Humanos , Sueros Inmunes , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/farmacología , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/farmacología , Datos de Secuencia Molecular , Ingeniería de Proteínas/métodos , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Relación Estructura-Actividad , Células Tumorales Cultivadas
16.
PLoS One ; 10(11): e0141618, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26545108

RESUMEN

Cancer has become a major problem worldwide due to its increasing incidence and mortality rates. Both the 37kDa/67kDa laminin receptor (LRP/LR) and telomerase are overexpressed in cancer cells. LRP/LR enhances the invasiveness of cancer cells thereby promoting metastasis, supporting angiogenesis and hampering apoptosis. An essential component of telomerase, hTERT is overexpressed in 85-90% of most cancers. hTERT expression and increased telomerase activity are associated with tumor progression. As LRP/LR and hTERT both play a role in cancer progression, we investigated a possible correlation between LRP/LR and telomerase. LRP/LR and hTERT co-localized in the perinuclear compartment of tumorigenic breast cancer (MDA_MB231) cells and non-tumorigenic human embryonic kidney (HEK293) cells. FLAG® Co-immunoprecipitation assays confirmed an interaction between LRP/LR and hTERT. In addition, flow cytometry revealed that both cell lines displayed high cell surface and intracellular LRP/LR and hTERT levels. Knock-down of LRP/LR by RNAi technology significantly reduced telomerase activity. These results suggest for the first time a novel function of LRP/LR in contributing to telomerase activity. siRNAs targeting LRP/LR may act as a potential alternative therapeutic tool for cancer treatment by (i) blocking metastasis (ii) promoting angiogenesis (iii) inducing apoptosis and (iv) impeding telomerase activity.


Asunto(s)
Técnicas de Silenciamiento del Gen , Receptores de Laminina/deficiencia , Receptores de Laminina/genética , Proteínas Ribosómicas/deficiencia , Proteínas Ribosómicas/genética , Telomerasa/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , Receptores de Laminina/metabolismo , Proteínas Ribosómicas/metabolismo
17.
MAbs ; 7(3): 584-604, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875246

RESUMEN

To harness the potent tumor-killing capacity of T cells for the treatment of CD19(+) malignancies, we constructed AFM11, a humanized tetravalent bispecific CD19/CD3 tandem diabody (TandAb) consisting solely of Fv domains. The molecule exhibits good manufacturability and stability properties. AFM11 has 2 binding sites for CD3 and 2 for CD19, an antigen that is expressed from early B cell development through differentiation into plasma cells, and is an attractive alternative to CD20 as a target for the development of therapeutic antibodies to treat B cell malignancies. Comparison of the binding and cytotoxicity of AFM11 with those of a tandem scFv bispecific T cell engager (BiTE) molecule targeting the same antigens revealed that AFM11 elicited more potent in vitro B cell lysis. Though possessing high affinity to CD3, the TandAb mediates serial-killing of CD19(+) cells with little dependence of potency or efficacy upon effector:target ratio, unlike the BiTE. The advantage of the TandAb over the BiTE was most pronounced at lower effector:target ratios. AFM11 mediated strictly target-dependent T cell activation evidenced by CD25 and CD69 induction, proliferation, and cytokine release, notwithstanding bivalent CD3 engagement. In a NOD/scid xenograft model, AFM11 induced dose-dependent growth inhibition of Raji tumors in vivo, and radiolabeled TandAb exhibited excellent localization to tumor but not to normal tissue. After intravenous administration in mice, half-life ranged from 18.4 to 22.9 h. In a human ex vivo B-cell chronic lymphocytic leukemia study, AFM11 exhibited substantial cytotoxic activity in an autologous setting. Thus, AFM11 may represent a promising therapeutic for treatment of CD19(+) malignancies with an advantageous safety risk profile and anticipated dosing regimen.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Antineoplásicos/farmacología , Antígenos CD19/inmunología , Complejo CD3/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Anticuerpos de Cadena Única/farmacología , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/inmunología , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Células Jurkat , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Protein Eng Des Sel ; 17(4): 357-66, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15126676

RESUMEN

Bispecific single-chain Fv antibodies comprise four covalently linked immunoglobulin variable (V(H) and V(L)) domains of two different specificities connected by three linkers. When assembled in the order V(H)(A)-linker(1)-V(L)(B)-linker(2)-V(H)(B)-linker(3)-V(L)(A), the single-chain molecule either folds head-to-tail with the formation of a diabody-like structure, a so-called bispecific single-chain diabody, or forms a homodimer that is twice as large, a so-called tandem diabody. The formation of the tandem diabody is determined by the association of complementary V(H) and V(L) domains located on different polypeptide chains, and depends on the length and probably the amino acid composition of the three linkers joining the variable domains. We generated a number of single-chain constructs using four V(H) and V(L) domains specific either for human CD3, a component of T-cell receptor (TCR) complex, or for CD19, a human B-cell antigen, separated by different rationally designed peptide linkers of 6-27 amino acid residues. The generated bispecific constructs were expressed in bacterial periplasm and their molecular forms, antigen-binding properties, stability, and T-cell proliferative and anti-tumor activities were compared. Using peripheral blood mononuclear cell cultures from patients suffering from B-cell chronic lymphocytic leukemia, we demonstrated that the tandab-mediated activation of autologous T cells and depletion of malignant cells correlates with the stability of the recombinant molecule and with the distance between the CD19 and CD3 binding sites.


Asunto(s)
Región Variable de Inmunoglobulina/química , Secuencia de Aminoácidos , Antígenos CD19/inmunología , Secuencia de Bases , Complejo CD3/inmunología , Cartilla de ADN , Citometría de Flujo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
19.
J Immunol Methods ; 285(1): 111-27, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14871540

RESUMEN

The mouse anti-human CD3 monoclonal antibody OKT3 is a potent immunosuppressive agent used in clinical transplantation. However, OKT3 therapy is associated with unpleasant and often serious side effects which appear to result from cytokine release, complement activation and a human anti-mouse antibody (HAMA) response. To decrease these adverse side effects, we constructed antibody fragments comprising OKT3 variable domains without any constant domains. Single-chain Fv (scFv) monomers, dimers and trimers were generated by changing the linker length between the V(H) and V(L) domains. The linkers used were the natural extensions of the V(H) into the C(H)1 domain. The dimeric molecules (diabodies) demonstrated the best CD3-binding activity. The diabody with the six amino acid linker was produced in bacteria with a tenfold higher yield than other scFvs and possessed CD3-binding affinity approaching that of the parental mAb. In contrast to OKT3 mAb, the anti-CD3 diabody and scFv monomer did not cause any T-cell activation and cytokine release in vitro, while demonstrating CD3 modulation. In mixed lymphocyte cultures, both diabody and scFv, but not the monoclonal antibody OKT3, were able to suppress T-cell activation and secretion of IL-2 and IFN-gamma in a dose-dependent manner. The anti-CD3 diabody may provide a potent immunosuppressive drug with low toxicity and immunogenicity.


Asunto(s)
Complejo CD3/inmunología , Región Variable de Inmunoglobulina/farmacología , Inmunosupresores/farmacología , Muromonab-CD3/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , División Celular , Citocinas/biosíntesis , ADN Complementario/genética , Dimerización , Diseño de Fármacos , Estabilidad de Medicamentos , Humanos , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/genética , Inmunosupresores/química , Células Jurkat , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Muromonab-CD3/química , Muromonab-CD3/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-2/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología
20.
Sci Rep ; 4: 5556, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24990253

RESUMEN

Neuronal loss is a major neuropathological hallmark of Alzheimer's disease (AD). The associations between soluble Aß oligomers and cellular components cause this neurotoxicity. The 37 kDa/67 kDa laminin receptor (LRP/LR) has recently been implicated in Aß pathogenesis. In this study the mechanism underlying the pathological role of LRP/LR was elucidated. Försters Resonance Energy Transfer (FRET) revealed that LRP/LR and Aß form a biologically relevant interaction. The ability of LRP/LR to form stable associations with endogenously shed Aß was confirmed by pull down assays and Aß-ELISAs. Antibody blockade of this association significantly lowered Aß42 induced apoptosis. Furthermore, antibody blockade and shRNA mediated downregulation of LRP/LR significantly hampered Aß42 internalization. These results suggest that LRP/LR is a receptor for Aß42 internalization, mediating its endocytosis and contributing to the cytotoxicity of the neuropeptide by facilitating intra-cellular Aß42 accumulation. These findings recommend anti-LRP/LR specific antibodies and shRNAs as potential therapeutic tools for AD treatment.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Apoptosis , Endocitosis , Células HEK293 , Humanos , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Receptores de Laminina/metabolismo , Proteínas Ribosómicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA