Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35511740

RESUMO

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Assuntos
Imunoconjugados , Proteínas ADAM , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Imunoconjugados/química , Proteínas de Membrana/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Haematologica ; 104(8): 1633-1639, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30733273

RESUMO

Antibody-drug conjugates (ADC) are a novel way to deliver potent cytotoxic compounds to cells expressing a specific antigen. Four ADC targeting CD19, including SAR3419 (coltuximab ravtansine), have entered clinical development. Here, we present huB4-DGN462, a novel ADC based on the SAR3419 anti-CD19 antibody linked via sulfo-SPDB to the potent DNA-alkylating agent DGN462. huB4-DGN462 had improved in vitro anti-proliferative and cytotoxic activity compared to SAR3419 across multiple B-cell lymphoma and human acute lymphoblastic leukemia cell lines. In vivo experiments using lymphoma xenografts models confirmed the in vitro data. The response of B-cell lymphoma lines to huB4-DGN462 was not correlated with CD19 expression, the presence of BCL2 or MYC translocations, TP53 inactivation or lymphoma histology. In conclusion, huB4-DGN462 is an attractive candidate for clinical investigation in patients with B-cell malignancies.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD19/metabolismo , Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Leucemia/metabolismo , Linfoma/metabolismo , Maitansina/análogos & derivados , Animais , Anticorpos Monoclonais Humanizados/química , Antineoplásicos/química , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoconjugados/química , Leucemia/tratamento farmacológico , Leucemia/patologia , Linfoma/tratamento farmacológico , Linfoma/patologia , Maitansina/química , Maitansina/farmacologia , Camundongos , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Neoplasia ; 19(9): 661-671, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28753442

RESUMO

Naratuximab emtansine (IMGN529) is an investigational antibody-drug conjugate consisting of a CD37-targeting antibody conjugated to the maytansine-derived microtuble disruptor, DM1. IMGN529 has shown promising preclinical and clinical activity in non-Hodgkin lymphoma, including diffuse large B-cell lymphoma (DLBCL). Due to the aggressive nature of the disease, DLBCL is often treated with combination therapies to maximize clinical outcomes; therefore, we investigated the potential of combining IMGN529 with both standard-of-care and emerging therapies against multiple oncology-relevant targets and pathways. The strongest enhancement in potency was seen with anti-CD20 antibodies, including rituximab. The combination of IMGN529 and rituximab was more potent than either agent alone, and this combinatorial benefit was associated with increased apoptotic induction and cell death. Additional studies revealed that rituximab treatment increased the internalization and degradation of the CD37-targeting antibody moiety of IMGN529. The combination of IMGN529 and rituximab was highly efficacious in multiple xenograft models, with superior antitumor efficacy seen compared to either agent alone or treatment with R-CHOP therapy. These findings suggest a novel mechanism whereby the potency of IMGN529 can be enhanced by CD20 binding, which results in the increased internalization and degradation of IMGN529 leading to the generation of greater amounts of cytotoxic catabolite. Overall, these data provide a biological rationale for the enhanced activity of IMGN529 in combination with rituximab and support the ongoing clinical evaluation of IMGN529 in combination with rituximab in patients with relapsed and/or refractory DLBCL.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Linfoma não Hodgkin/tratamento farmacológico , Rituximab/farmacologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Humanos , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/patologia , Camundongos , Terapia de Alvo Molecular , Proteólise , Ensaios Antitumorais Modelo de Xenoenxerto
4.
MAbs ; 8(3): 513-23, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26752675

RESUMO

Antibody-drug conjugates (ADCs) are of great interest as targeted cancer therapeutics. Preparation of ADCs for early stage screening is constrained by purification and biochemical analysis techniques that necessitate burdensome quantities of antibody. Here we describe a method, developed for the maytansinoid class of ADCs, enabling parallel conjugation of antibodies in 96-well format. The method utilizes ∼ 100 µg of antibody per well and requires <5 µg of ADC for characterization. We demonstrate the capabilities of this system using model antibodies. We also provide multiple examples applying this method to early-stage screening of maytansinoid ADCs. The method can greatly increase the throughput with which candidate ADCs can be screened in cell-based assays, and may be more generally applicable to high-throughput preparation and screening of different types of protein conjugates.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Maitansina/farmacologia , Neoplasias/tratamento farmacológico , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Antineoplásicos/imunologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Imunoconjugados/imunologia , Neoplasias/imunologia
5.
MAbs ; 6(4): 957-67, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24758837

RESUMO

Antibody combination therapeutics (ACTs) are polyvalent biopharmaceuticals that are uniquely suited for the control of complex diseases, including antibiotic resistant infectious diseases, autoimmune disorders and cancers. However, ACTs also represent a distinct manufacturing challenge because the independent manufacture and subsequent mixing of monoclonal antibodies quickly becomes cost prohibitive as more complex mixtures are envisioned. We have developed a virus-free recombinant protein expression platform based on adeno-associated viral (AAV) elements that is capable of rapid and consistent production of complex antibody mixtures in a single batch format. Using both multiplexed immunoassays and cation exchange (CIEX) chromatography, cell culture supernatants generated using our system were assessed for stability of expression and ratios of the component antibodies over time. Cultures expressing combinations of three to ten antibodies maintained consistent expression levels and stable ratios of component antibodies for at least 60 days. Cultures showed remarkable reproducibility following cell banking, and AAV-based cultures showed higher stability and productivity than non-AAV based cultures. Therefore, this non-viral AAV-based expression platform represents a predictable, reproducible, quick and cost effective method to manufacture or quickly produce for preclinical testing recombinant antibody combination therapies and other recombinant protein mixtures.


Assuntos
Dependovirus , Expressão Gênica , Anticorpos de Cadeia Única/biossíntese , Linhagem Celular , Cromatografia por Troca Iônica/métodos , Técnicas de Cocultura , Quimioterapia Combinada , Humanos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/isolamento & purificação
6.
Cell Host Microbe ; 10(1): 9-20, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21767808

RESUMO

Several pathogenic bacteria utilize type III secretion systems (TTSS) to deliver into host cells bacterial virulence proteins with the capacity to modulate a variety of cellular pathways. Once delivered into host cells, the accurate targeting of bacterial effectors to specific locations is critical for their proper function. However, little is known about the mechanisms these virulence effectors use to reach their subcellular destination. Here we show that the Salmonella TTSS effector proteins SspH2 and SseI are localized to the plasma membrane of host cells, a process dependent on S-palmitoylation of a conserved cysteine residue within their N-terminal domains. We also show that effector protein lipidation is mediated by a specific subset of host-cell palmitoyltransferases and that lipidation is critical for effector function. This study describes a remarkable mechanism by which a pathogen exploits host-cell machinery to properly target its virulence factors.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Salmonella typhimurium/patogenicidade , Fatores de Virulência/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos , Membrana Celular/metabolismo , Sequência Conservada , Cisteína , Células HeLa/microbiologia , Humanos , Lipoilação , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Processamento de Proteína Pós-Traducional , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Fatores de Virulência/genética
7.
J Biol Chem ; 286(21): 19014-23, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21464135

RESUMO

S-Palmitoylation of G protein-coupled receptors (GPCRs) is a prevalent modification, contributing to the regulation of receptor function. Despite its importance, the palmitoylation status of the ß(1)-adrenergic receptor, a GPCR critical for heart function, has never been determined. We report here that the ß(1)-adrenergic receptor is palmitoylated on three cysteine residues at two sites in the C-terminal tail. One site (proximal) is adjacent to the seventh transmembrane domain and is a consensus site for GPCRs, and the other (distal) is downstream. These sites are modified in different cellular compartments, and the distal palmitoylation site contributes to efficient internalization of the receptor following agonist stimulation. Using a bioorthogonal palmitate reporter to quantify palmitoylation accurately, we found that the rates of palmitate turnover at each site are dramatically different. Although palmitoylation at the proximal site is remarkably stable, palmitoylation at the distal site is rapidly turned over. This is the first report documenting differential dynamics of palmitoylation sites in a GPCR. Our results have important implications for function and regulation of the clinically important ß(1)-adrenergic receptor.


Assuntos
Cisteína/metabolismo , Lipoilação/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Receptores Adrenérgicos beta 1/metabolismo , Cisteína/genética , Células HEK293 , Humanos , Estrutura Terciária de Proteína , Receptores Adrenérgicos beta 1/genética
8.
Proc Natl Acad Sci U S A ; 106(12): 4864-9, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19273841

RESUMO

Processes as diverse as receptor binding and signaling, cytoskeletal dynamics, and programmed cell death are manipulated by mimics of host proteins encoded by pathogenic bacteria. We show here that the Salmonella virulence factor SspH2 belongs to a growing class of bacterial effector proteins that harness and subvert the eukaryotic ubiquitination pathway. This virulence protein possesses ubiquitination activity that depends on a conserved cysteine residue. A crystal structure of SspH2 reveals a canonical leucine-rich repeat (LRR) domain that interacts with a unique E3 ligase [which we have termed NEL for Novel E3 Ligase] C-terminal fold unrelated to previously observed HECT or RING-finger E3 ligases. Moreover, the LRR domain sequesters the catalytic cysteine residue contained in the NEL domain, and we suggest a mechanism for activation of the ligase requiring a substantial conformational change to release the catalytic domain for function. We also show that the N-terminal domain targets SspH2 to the apical plasma membrane of polarized epithelial cells and propose a model whereby binding of the LRR to proteins at the target site releases the ligase domain for site-specific function.


Assuntos
Proteínas de Bactérias/metabolismo , Salmonella typhimurium/enzimologia , Salmonella typhimurium/patogenicidade , Ubiquitina-Proteína Ligases/metabolismo , Fatores de Virulência/metabolismo , Proteínas de Bactérias/química , Domínio Catalítico , Cristalografia por Raios X , Ativação Enzimática , Células HeLa , Humanos , Modelos Moleculares , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Ubiquitina-Proteína Ligases/química , Ubiquitinação
9.
Mol Biol Cell ; 17(12): 5346-55, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17050738

RESUMO

The peripheral Golgi protein golgin-160 is induced during 3T3L1 adipogenesis and is primarily localized to the Golgi cisternae distinct from the trans-Golgi network (TGN) in a general distribution similar to p115. Small interfering RNA (siRNA)-mediated reduction in golgin-160 protein resulted in an increase accumulation of the insulin-responsive amino peptidase (IRAP) and the insulin-regulated glucose transporter (GLUT4) at the plasma membrane concomitant with enhanced glucose uptake in the basal state. The redistribution of GLUT4 was rescued by expression of a siRNA-resistant golgin-160 cDNA. The basal state accumulation of plasma membrane GLUT4 occurred due to an increased rate of exocytosis without any significant effect on the rate of endocytosis. This GLUT4 trafficking to the plasma membrane in the absence of golgin-160 was independent of TGN/Golgi sorting, because it was no longer inhibited by the expression of a dominant-interfering Golgi-localized, gamma-ear-containing ARF-binding protein mutant and displayed reduced binding to the lectin wheat germ agglutinin. Moreover, expression of the amino terminal head domain (amino acids 1-393) had no significant effect on the distribution or insulin-regulated trafficking of GLUT4 or IRAP. In contrast, expression of carboxyl alpha helical region (393-1498) inhibited insulin-stimulated GLUT4 and IRAP translocation, but it had no effect on the sorting of constitutive membrane trafficking proteins, the transferrin receptor, or vesicular stomatitis virus G protein. Together, these data demonstrate that golgin-160 plays an important role in directing insulin-regulated trafficking proteins toward the insulin-responsive compartment in adipocytes.


Assuntos
Adipócitos/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Complexo de Golgi/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Células 3T3-L1 , Fatores de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Adipócitos/citologia , Animais , Cistinil Aminopeptidase/metabolismo , Endocitose/fisiologia , Glicosilação , Proteínas da Matriz do Complexo de Golgi , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/deficiência , Camundongos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Precursores de Proteínas/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA