Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cancer Immunol Res ; 12(2): 195-213, 2024 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-38091375

RESUMO

The costimulatory receptor CD137 (also known as TNFRSF9 or 4-1BB) sustains effective cytotoxic T-cell responses. Agonistic anti-CD137 cancer immunotherapies are being investigated in clinical trials. Development of the first-generation CD137-agonist monotherapies utomilumab and urelumab was unsuccessful due to low antitumor efficacy mediated by the epitope recognized on CD137 or hepatotoxicity mediated by Fcγ receptors (FcγR) ligand-dependent CD137 activation, respectively. M9657 was engineered as a tetravalent bispecific antibody (mAb2) in a human IgG1 backbone with LALA mutations to reduce binding to FCγRs. Here, we report that M9657 selectively binds to mesothelin (MSLN) and CD137 with similar affinity in humans and cynomolgus monkeys. In a cellular functional assay, M9657 enhanced CD8+ T cell-mediated cytotoxicity and cytokine release in the presence of tumor cells, which was dependent on both MSLN expression and T-cell receptor/CD3 activation. Both FS122m, a murine surrogate with the same protein structure as M9657, and chimeric M9657, a modified M9657 antibody with the Fab portion replaced with an anti-murine MSLN motif, demonstrated in vivo antitumor efficacy against various tumors in wild-type and human CD137 knock-in mice, and this was accompanied by activated CD8+ T-cell infiltration in the tumor microenvironment. The antitumor immunity of M9657 and FS122m depended on MSLN expression density and the mAb2 structure. Compared with 3H3, a murine surrogate of urelumab, FS122m and chimeric M9657 displayed significantly lower on-target/off-tumor toxicity. Taken together, M9657 exhibits a promising profile for development as a tumor-targeting immune agonist with potent anticancer activity without systemic immune activation and associated hepatotoxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Neoplasias , Humanos , Animais , Camundongos , Mesotelina , Inflamação , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral , Microambiente Tumoral
2.
Sci Rep ; 13(1): 20412, 2023 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-37989777

RESUMO

TL-895 (formerly known as M7583) is a potent, highly selective, adenosine triphosphate (ATP)-competitive, second-generation, irreversible inhibitor of Bruton's tyrosine kinase (BTK). We characterized its biochemical and cellular effects in in vitro and in vivo models. TL-895 was evaluated preclinically for potency against BTK using IC50 concentration-response curves; selectivity using a 270-kinase panel; BTK phosphorylation in Ramos Burkitt's lymphoma cells by ProteinSimple Wes analysis of one study; anti-proliferative effects in primary chronic lymphocytic leukemia (CLL) blasts; cell viability effects in diffuse large B-cell lymphoma (DLBCL) and mantle-cell lymphoma (MCL) cell lines; effects on antibody-dependent cell-mediated cytotoxicity (ADCC) from Daudi cells and chromium-51 release from human tumor cell lines; and efficacy in vivo using four MCL xenograft model and 21 DLBCL patient-derived xenograft (PDX) models (subtypes: 9 ABC, 11 GCB, 1 Unclassified). TL-895 was active against recombinant BTK (average IC50 1.5 nM) and inhibited only three additional kinases with IC50 within tenfold of BTK activity. TL-895 inhibited BTK auto-phosphorylation at the Y223 phosphorylation site (IC50 1-10 nM). TL-895 inhibited the proliferation of primary CLL blasts in vitro and inhibited growth in a subset of activated DLBCL and MCL cell lines. TL-895 inhibited the ADCC mechanism of therapeutic antibodies only at supra-clinical exposure levels. TL-895 significantly inhibited tumor growth in the Mino MCL xenograft model and in 5/21 DLBCL PDX models relative to vehicle controls. These findings demonstrate the potency of TL-895 for BTK and its efficacy in models of B-cell lymphoma despite its refined selectivity.


Assuntos
Leucemia Linfocítica Crônica de Células B , Linfoma Difuso de Grandes Células B , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Linfócitos B/metabolismo , Tirosina Quinase da Agamaglobulinemia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/química , Linfoma Difuso de Grandes Células B/patologia
3.
J Immunother Cancer ; 10(7)2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35858707

RESUMO

BACKGROUND: Bintrafusp alfa (BA) is a bifunctional fusion protein designed for colocalized, simultaneous inhibition of two immunosuppressive pathways, transforming growth factor-ß (TGF-ß) and programmed death-ligand 1 (PD-L1), within the tumor microenvironment (TME). We hypothesized that targeting PD-L1 to the tumor by BA colocalizes the TGF-ß trap (TGF-ßRII) to the TME, enabling it to sequester TGF-ß in the tumor more effectively than systemic TGF-ß blockade, thereby enhancing antitumor activity. METHODS: Multiple technologies were used to characterize the TGF-ß trap binding avidity. BA versus combinations of anti-PD-L1 and TGF-ß trap or the pan-TGF-ß antibody fresolimumab were compared in proliferation and two-way mixed lymphocyte reaction assays. Immunophenotyping of tumor-infiltrating lymphocytes (TILs) and RNA sequencing (RNAseq) analysis assessing stromal and immune landscape following BA or the combination therapy were performed in MC38 tumors. TGF-ß and PD-L1 co-expression and their associated gene signatures in MC38 tumors and human lung carcinoma tissue were studied with single-cell RNAseq (scRNAseq) and immunostaining. BA-induced internalization, degradation, and depletion of TGF-ß were investigated in vitro. RESULTS: BA and fresolimumab had comparable intrinsic binding to TGF-ß1, but there was an ~80× avidity-based increase in binding affinity with BA. BA inhibited cell proliferation in TGF-ß-dependent and PD-L1-expressing cells more potently than TGF-ß trap or fresolimumab. Compared with the combination of anti-PD-L1 and TGF-ß trap or fresolimumab, BA enhanced T cell activation in vitro and increased TILs in MC38 tumors, which correlated with efficacy. BA induced distinct gene expression in the TME compared with the combination therapy, including upregulation of immune-related gene signatures and reduced activities in TGF-ß-regulated pathways, such as epithelial-mesenchymal transition, extracellular matrix deposition, and fibrosis. Regulatory T cells, macrophages, immune cells of myeloid lineage, and fibroblasts were key PD-L1/TGF-ß1 co-expressing cells in the TME. scRNAseq analysis suggested BA modulation of the macrophage phenotype, which was confirmed by histological assessment. PD-L1/TGF-ß1 co-expression was also seen in human tumors. Finally, BA induced TGF-ß1 internalization and degradation in the lysosomes. CONCLUSION: BA more effectively blocks TGF-ß by targeting TGF-ß trap to the tumor via PD-L1 binding. Such colocalized targeting elicits distinct and superior antitumor responses relative to single agent combination therapy.


Assuntos
Neoplasias Pulmonares , Fator de Crescimento Transformador beta , Antígeno B7-H1 , Humanos , Fatores Imunológicos , Receptor de Morte Celular Programada 1 , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Microambiente Tumoral
4.
Arch Biochem Biophys ; 526(2): 219-25, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22426455

RESUMO

Certain combinations of non-competitive anti-EGFR antibodies have been reported to produce new effects on cells compared to either antibody used separately. New and enhanced combination-activity includes increased inhibition of signaling, increased receptor internalization and degradation, reduced proliferation of tumor cell lines and induction of complement-dependent cytotoxicity (CDC) effector function. To test requirements and mechanisms to elicit enhanced combination-activity with different EGFR binding domains, we created an anti-EGFR biparatopic antibody. A biparatopic antibody interacts through two different antigen-binding sites to a single antigen. A heterodimeric antibody with one binding domain derived from the C225 antibody and one binding domain derived from the humanized 425 (hu425) antibody was built on the strand-exchange engineered domain (SEED) scaffold. This anti-EGFR biparatopic-SEED antibody was compared to parental antibodies used alone and in combination, and to the corresponding monovalent anti-EGFR-SEED antibodies used alone or in combination. We found that the anti-EGFR biparatopic-SEED had enhanced activity, similar to the combination of the two parental antibodies. Combinations of monovalent anti-EGFR-SEED antibodies did not produce enhanced effectiveness in cellular assays. Our results show that the anti-EGFR biparatopic antibody created using the SEED scaffold has enhanced combination-activity in a single molecule. Furthermore, these data suggest that the potential to cross-link the two different epitopes is an important requirement in the mechanism of enhanced combination-activity.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais/imunologia , Receptores ErbB/imunologia , Anticorpos Biespecíficos/química , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados/química , Sítios de Ligação de Anticorpos , Linhagem Celular Tumoral , Proliferação de Células , Cetuximab , Epitopos/imunologia , Humanos , Estrutura Terciária de Proteína
5.
Protein Eng Des Sel ; 24(5): 447-54, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21498564

RESUMO

The strand-exchange engineered domain (SEED) platform was designed to generate asymmetric and bispecific antibody-like molecules, a capability that expands therapeutic applications of natural antibodies. This new protein engineered platform is based on exchanging structurally related sequences of immunoglobulin within the conserved CH3 domains. Alternating sequences from human IgA and IgG in the SEED CH3 domains generate two asymmetric but complementary domains, designated AG and GA. The SEED design allows efficient generation of AG/GA heterodimers, while disfavoring homodimerization of AG and GA SEED CH3 domains. Using a clinically validated antibody (C225), we tested whether Fab derivatives constructed on the SEED platform retain desirable therapeutic antibody features such as in vitro and in vivo stability, favorable pharmacokinetics, ligand binding and effector functions including antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. In addition, we tested SEED with combinations of binder domains (scFv, VHH, Fab). Mono- and bivalent Fab-SEED fusions retain full binding affinity, have excellent biochemical and biophysical stability, and retain desirable antibody-like characteristics conferred by Fc domains. Furthermore, SEED is compatible with different combinations of Fab, scFv and VHH domains. Our assessment shows that the new SEED platform expands therapeutic applications of natural antibodies by generating heterodimeric Fc-analog proteins.


Assuntos
Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Especificidade de Anticorpos , Engenharia de Proteínas/métodos , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Afinidade de Anticorpos , Linhagem Celular Tumoral , Proteínas do Sistema Complemento/imunologia , Receptores ErbB/imunologia , Meia-Vida , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/genética , Masculino , Camundongos , Multimerização Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
6.
Ultrason Sonochem ; 18(1): 356-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20675175

RESUMO

Ultrasound irradiation is used for the deposition of silver nanoparticles with an average size of 40 nm on the surface of a stainless steel plate. The sonochemical reduction of an AgNO(3) solution (water, ethylene glycol and aqueous ammonia) was carried out at 30°C. Irradiation was done with a high intensity ultrasonic horn (Ti horn from Sonics and Materials VCX600, 20 kHz, 600 W at 70% efficiency) under an argon flow. The influence of the distance of the stainless steel plate from the sonicator tip varied, and a homogeneous coating without agglomeration was achieved at a distance of 1 cm. By controlling the reaction conditions and the distance from the tip we could achieve a homogeneous monolayer coating of silver nanoparticles on the stainless steel surface. The silver-deposited stainless steel plates were analysed by UV-visible, XRD, SEM and FIB analyses.


Assuntos
Nanopartículas Metálicas/química , Prata/química , Aço Inoxidável/química , Ultrassom , Tamanho da Partícula , Propriedades de Superfície
7.
Hum Mol Genet ; 16(9): 1017-29, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17341488

RESUMO

Membrane palmitoylated protein 4 (Mpp4) is a member of the membrane-associated guanylate kinase family. We show that Mpp4 localizes specifically to the plasma membrane of photoreceptor synaptic terminals. Plasma membrane Ca(2+) ATPases (PMCAs), the Ca(2+) extrusion pumps, interact with an Mpp4-dependent presynaptic membrane protein complex that includes Veli3 and PSD95. In mice lacking Mpp4, PMCAs were lost from rod photoreceptor presynaptic membranes. Synaptic ribbons were enlarged, a phenomenon known to correlate with higher Ca(2+). SERCA2 (sarcoplasmic-endoplasmic reticulum Ca(2+) ATPase, type 2), which pumps cytosolic Ca(2+) into intracellular Ca(2+) stores and localizes next to the ribbons, was increased. The distribution of IP(3)RII (InsP(3) receptor, type 2), which releases Ca(2+) from the stores, was shifted away from the synaptic terminals. Synaptic transmission to second-order neurons was maintained but was reduced in amplitude. These data suggest that loss of Mpp4 disrupts a Ca(2+) extrusion mechanism at the presynaptic membranes, with ensuing adaptive responses by the photoreceptor to restore Ca(2+) homeostasis. We propose that Mpp4 organizes a presynaptic protein complex that includes PMCAs and has a role in modulating Ca(2+) homeostasis and synaptic transmission in rod photoreceptors.


Assuntos
Cálcio/metabolismo , Proteínas de Membrana/fisiologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Terminações Pré-Sinápticas/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína 4 Homóloga a Disks-Large , Imunofluorescência , Deleção de Genes , Guanilato Quinases , Homeostase , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Modelos Biológicos , Complexos Multiproteicos/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Terminações Pré-Sinápticas/ultraestrutura , Ligação Proteica , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Técnicas do Sistema de Duplo-Híbrido
8.
Invest Ophthalmol Vis Sci ; 47(5): 2185-94, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16639031

RESUMO

PURPOSE: To investigate the impact of aryl hydrocarbon receptor-interacting protein-like (AIPL)-1 on photoreception in rods. METHODS: Photoresponses of mouse rods expressing lowered amounts of AIPL1 were studied by single-cell and electroretinogram (ERG) recordings. Phototransduction protein levels and enzymatic activities were determined in biochemical assays. Ca2+ dynamics were probed with a fluorescent dye. Comparisons were made to rods expressing mutant Y99C guanylate cyclase activating protein (GCAP)-1, to understand which effects arose from elevated dark levels of cGMP and Ca2+. RESULTS: Except for PDE, transduction protein levels were normal in low-AIPL1 retinas, as were guanylate cyclase (GC), rhodopsin kinase (RK), and normalized phosphodiesterase (PDE) activities. Y99C and low-AIPL1 rods were more sensitive to flashes than normal, but flash responses of low-AIPL1 rods showed an abnormal delay, reduced rate of increase, and longer recovery not present in Y99C rod responses. In addition, low-AIPL1 rods but not Y99C rods failed to reach the normal light-induced minimum in Ca2+ concentration. CONCLUSIONS: Reduced AIPL1 delayed the photoresponse, decreased its amplification constant, slowed a rate-limiting step in its recovery, and limited the light-induced decrease in Ca2+. Not all changes were attributable to decreased PDE or to elevated cGMP and Ca2+ in darkness. Therefore, AIPL1 directly or indirectly affects more than one component of phototransduction.


Assuntos
Proteínas de Transporte/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Visão Ocular/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Cálcio/metabolismo , Cromatografia Gasosa , GMP Cíclico/metabolismo , Eletrorretinografia , Ácidos Graxos/metabolismo , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Guanilato Ciclase/metabolismo , Proteínas Ativadoras de Guanilato Ciclase/metabolismo , Camundongos , Camundongos Transgênicos , Diester Fosfórico Hidrolases/metabolismo , Estimulação Luminosa , Células Fotorreceptoras Retinianas Bastonetes/efeitos da radiação , Transducina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA