Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
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.
Curr Protoc Immunol ; 129(1): e95, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32294319

RESUMO

Multispecific antibodies bind two or more different antigens and enable new therapeutic applications that cannot be replicated with conventional monoclonal antibodies, such as bridging different cells or bringing soluble proteins in close proximity. The DART and TRIDENT platforms enable the engineering of such antibodies. A DART molecule combines two independent antigen-binding sites in a stabilized, diabody-like structure. A DART molecule can be expressed with or without an Fc domain and thus can be tailored to have a long or short half-life in vivo and to induce or ablate effector function. Linking two DART units or a DART unit and a Fab domain (the latter structure is called TRIDENT format) via an Fc domain creates a monospecific, bispecific, trispecific, or tetraspecific molecule with up to tetravalent targeting of antigens. This article focuses on the design of DART and TRIDENT molecules that target two or three different antigens. © 2020 by John Wiley & Sons, Inc. Basic Protocol 1: Design and generation of expression plasmids encoding DART and TRIDENT molecules Basic Protocol 2: Expression of DART and TRIDENT molecules by transient transfection of CHO cells Basic Protocol 3: Purification of DART and TRIDENT molecules from CHO cell supernatants.


Assuntos
Anticorpos Monoclonais/metabolismo , Engenharia Genética/métodos , Animais , Anticorpos Monoclonais/genética , Especificidade de Anticorpos/genética , Células CHO , Técnicas de Cultura de Células , Cricetulus , Humanos , Fragmentos Fc das Imunoglobulinas/genética
3.
Front Immunol ; 11: 713, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373131

RESUMO

Mother-to-child transmission of HIV-1 remains a major global health challenge. Currently, HIV-1-infected infants require strict lifelong adherence to antiretroviral therapy to prevent replication of virus from reservoirs of infected cells, and to halt progression of disease. There is a critical need for immune interventions that can be deployed shortly after infection to eliminate HIV-1-infected cells in order to promote long-term remission of viremia, or to potentially cure pediatric HIV-1-infection. Bispecific HIV × CD3 DART® molecules able to co-engage the HIV-1 envelope protein on the surface of infected cells and CD3 on cytolytic T cells have been previously shown to eliminate HIV-1 infected cells in vitro and are candidates for passive immunotherapy to reduce the virus reservoir. However, their potential utility as therapy for infant HIV-1 infection is unclear as the ability of these novel antibody-based molecules to work in concert with cells of the infant immune system had not been assessed. Here, we use human umbilical cord blood as a model of the naïve neonatal immune system to evaluate the ability of HIV x CD3 DART molecules to recruit and redirect neonatal effector cells for elimination of autologous CD4+ T cells infected with HIV-1 encoding an envelope gene sequenced from a mother-to-child transmission event. We found that HIV × CD3 DART molecules can redirect T cells present in cord blood for elimination of HIV-infected CD4+ T cells. However, we observed reduced killing by T cells isolated from cord blood when compared to cells isolated from adult peripheral blood-likely due to the absence of the memory and effector CD8+ T cells that are most cytolytic when redirected by bispecific DART molecules. We also found that newly developed HIV × CD16 DART molecules were able to recruit CD16-expressing natural killer cells from cord blood to eliminate HIV-infected cells, and the activity of cord blood natural killer cells could be substantially increased by priming with IL-15. Our results support continued development of HIV-specific DART molecules using relevant preclinical animal models to optimize strategies for effective use of this immune therapy to reduce HIV-1 infection in pediatric populations.


Assuntos
Anticorpos Biespecíficos/imunologia , Linfócitos T CD4-Positivos/imunologia , Sangue Fetal/citologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/transmissão , HIV-1/imunologia , Imunização Passiva/métodos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Células Matadoras Naturais/imunologia , Linfócitos T Citotóxicos/imunologia , Adulto , Citotoxicidade Celular Dependente de Anticorpos , Doadores de Sangue , Células Cultivadas , Feminino , Infecções por HIV/virologia , Humanos , Interleucina-15/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
4.
Cell Rep Med ; 1(9): 100163, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33377134

RESUMO

Combination immunotherapy with antibodies directed against PD-1 and CTLA-4 shows improved clinical benefit across cancer indications compared to single agents, albeit with increased toxicity. Leveraging the observation that PD-1 and CTLA-4 are co-expressed by tumor-infiltrating lymphocytes, an investigational PD-1 x CTLA-4 bispecific DART molecule, MGD019, is engineered to maximize checkpoint blockade in the tumor microenvironment via enhanced CTLA-4 blockade in a PD-1-binding-dependent manner. In vitro, MGD019 mediates the combinatorial blockade of PD-1 and CTLA-4, confirming dual inhibition via a single molecule. MGD019 is well tolerated in non-human primates, with evidence of both PD-1 and CTLA-4 blockade, including increases in Ki67+CD8 and ICOS+CD4 T cells, respectively. In the ongoing MGD019 first-in-human study enrolling patients with advanced solid tumors (NCT03761017), an analysis undertaken following the dose escalation phase revealed acceptable safety, pharmacodynamic evidence of combinatorial blockade, and objective responses in multiple tumor types typically unresponsive to checkpoint inhibitor therapy.


Assuntos
Anticorpos/uso terapêutico , Antígeno CTLA-4/imunologia , Imunoterapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/métodos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Receptor de Morte Celular Programada 1/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
5.
Biochem Biophys Res Commun ; 366(2): 314-20, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18042463

RESUMO

Bilateral frontoparietal polymicrogyria (BFPP) is a rare genetic disease characterized by cortical malformation associated with GPR56 mutations of frameshift, splicing, and point mutations (Science 303:2033). All the missense point mutations are located in the regions predicted to be exposed at the cell surface, e.g. the N-terminal extracellular domain (ECD), the proteolytic site (GPS), and the extracellular loops of transmembrane domain (TM), implying functionally important interaction among these domains. Wild type GPR56 protein is cleaved at the GPCR protein cleavage site (GPS) and gives rise to two subunits (ECD and TM), which are transported to cell surface. We have shown that GPR56 GPS mutant protein is defective in cleavage and surface localization, while non-GPS mutant proteins are cleaved normally but still defective in surface localization. Furthermore, all the mutant proteins demonstrated different glycosylation pattern from that of wild-type protein. PNGase F and Endo H sensitivity assays suggests that the mutant proteins are trapped in endoplasmic reticulum (ER), preventing them from trafficking to Golgi where further glycosylation modification usually occurs before destination to cell surface. Therefore, the loss-of-function of all these missense mutations is primarily caused by their failure to localize to cell surface.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Animais , Predisposição Genética para Doença/genética , Humanos , Camundongos , Mutação , Células NIH 3T3 , Receptores Acoplados a Proteínas G/química
6.
FEBS J ; 273(17): 3871-85, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16934030

RESUMO

Hepatitis C virus (HCV) exists in different forms in the circulation of infected people: lipoprotein bound and lipoprotein free, enveloped and nonenveloped. Viral particles with the highest infectivity are associated with lipoproteins, whereas lipoprotein-free virions are poorly infectious. The detection of HCV's envelope proteins E1 and E2 in lipoprotein-associated virions has been challenging. Because lipoproteins are readily endocytosed, some forms of HCV might utilize their association with lipoproteins rather than E1 and E2 for cell attachment and internalization. However, vaccination of chimpanzees with recombinant envelope proteins protected the animals from hepatitis C infection, suggesting an important role for E1 and E2 in cell entry. It seems possible that different forms of HCV use different receptors to attach to and enter cells. The putative receptors and the assays used for their validation are discussed in this review.


Assuntos
Hepacivirus/patogenicidade , Hepatite C/patologia , Hepatite C/virologia , Hepacivirus/química , Hepacivirus/genética , Hepacivirus/isolamento & purificação , Hepatite C/metabolismo , Humanos
7.
MAbs ; 8(3): 454-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26854859

RESUMO

CD73 (ecto-5'-nucleotidase) has recently been established as a promising immuno-oncology target. Given its role in activating purinergic signaling pathways to elicit immune suppression, antagonizing CD73 (i.e., releasing the brake) offers a complimentary pathway to inducing anti-tumor immune responses. Here, we describe the mechanistic activity of a new clinical therapeutic, MEDI9447, a human monoclonal antibody that non-competitively inhibits CD73 activity. Epitope mapping, structural, and mechanistic studies revealed that MEDI9447 antagonizes CD73 through dual mechanisms of inter-CD73 dimer crosslinking and/or steric blocking that prevent CD73 from adopting a catalytically active conformation. To our knowledge, this is the first report of an antibody that inhibits an enzyme's function through 2 distinct modes of action. These results provide a finely mapped epitope that can be targeted for selective, potent, and non-competitive inhibition of CD73, as well as establish a strategy for inhibiting enzymes that function in both membrane-bound and soluble states.


Assuntos
5'-Nucleotidase , Monofosfato de Adenosina , Anticorpos Monoclonais/química , Antineoplásicos/química , Inibidores Enzimáticos/química , 5'-Nucleotidase/antagonistas & inibidores , 5'-Nucleotidase/química , Monofosfato de Adenosina/química , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/química , Humanos , Hidrólise
8.
Oncoimmunology ; 5(8): e1208875, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27622077

RESUMO

MEDI9447 is a human monoclonal antibody that is specific for the ectoenzyme CD73 and currently undergoing Phase I clinical trials. Here we show that MEDI9447 is a potent inhibitor of CD73 ectonucleotidase activity, with wide ranging immune regulatory consequences. MEDI9447 results in relief from adenosine monophosphate (AMP)-mediated lymphocyte suppression in vitro and inhibition of mouse syngeneic tumor growth in vivo. In contrast with other cancer immunotherapy agents such as checkpoint inhibitors or T-cell agonists, MEDI9447 drives changes in both myeloid and lymphoid infiltrating leukocyte populations within the tumor microenvironment of mouse models. Changes include significant alterations in a number of tumor micro-environmental subpopulations including increases in CD8(+) effector cells and activated macrophages. Furthermore, these changes correlate directly with responder and non-responder subpopulations within animal studies using syngeneic tumors. Combination data showing additive activity between MEDI9447 and anti-PD-1 antibodies using human cells in vitro and mouse tumor models further demonstrate the potential value of relieving adenosine-mediated immunosuppression. Based on these data, a Phase I study to test the safety, tolerability, and clinical activity of MEDI9447 in cancer patients was initiated (NCT02503774).

9.
J Immunol Methods ; 365(1-2): 118-25, 2011 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-21223970

RESUMO

We describe an assay and data evaluation technique for sorting a panel of murine monoclonal antibodies according to epitope specificities. The assay analyzes the simultaneous binding (pairing) of antibodies to an antigen and groups together antibodies with similar pairing profiles. Similar profiles indicate that the antibodies bind to the same or closely related epitopes. The assay works well with crude hybridoma supernatants and can be multiplexed. These features make the assay particularly suitable for the early phase of hybridoma/antibody screening when antibodies are available only as low volume culture harvest samples.


Assuntos
Anticorpos Monoclonais/metabolismo , Reações Antígeno-Anticorpo/fisiologia , Epitopos/metabolismo , Imunoensaio/métodos , Animais , Especificidade de Anticorpos , Ensaio de Imunoadsorção Enzimática/métodos , Hibridomas/imunologia , Técnicas In Vitro , Camundongos
10.
Clin Cancer Res ; 16(16): 4226-35, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20664024

RESUMO

PURPOSE: p95HER2 is an NH(2)-terminally truncated form of HER2 that lacks the trastuzumab binding site and is therefore thought to confer resistance to trastuzumab treatment. In this report, we introduce a new antibody that has enabled the first direct quantitative measurement of p95HER2 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissues. We sought to show that quantitative p95HER2 levels would correlate with outcome in trastuzumab-treated HER2-positive metastatic breast cancer. EXPERIMENTAL DESIGN: The novel p95HER2 antibody used here was characterized for sensitivity, specificity, and selectivity over full-length HER2. Quantitative p95HER2 levels were measured in 93 metastatic breast tumors using a VeraTag FFPE assay to determine the correlation of p95HER2 levels with outcomes. RESULTS: Within a cohort of trastuzumab-treated metastatic breast cancer patients, high levels of p95HER2 were found to correlate with shorter progression-free survival [hazard ratio (HR), 1.9; P = 0.017] and overall survival (HR, 2.2; P = 0.012) in patients with tumors selected to be HER2 positive by the VeraTag HER2 assay. For those with tumors found to be fluorescence in situ hybridization positive, elevated p95HER2 correlated similarly with shorter progression-free survival (HR, 1.8; P = 0.022) and overall survival (HR, 2.2; P = 0.009). CONCLUSIONS: We have successfully generated an antibody that can specifically detect p95HER2, and developed an assay to quantify expression in FFPE tumor specimens. Using this novel assay, we have identified a group of HER2-positive patients expressing p95HER2 that have a worse outcome while on trastuzumab. As p95HER2 retains sensitivity to kinase inhibitors, measurement of p95HER2 in breast tumor sections may be useful in guiding treatment for patients with HER2-positive breast cancer.


Assuntos
Anticorpos Monoclonais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Receptor ErbB-2/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Especificidade de Anticorpos , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/análise , Western Blotting , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Separação Celular , Estudos de Coortes , Intervalo Livre de Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Formaldeído , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Inclusão em Parafina , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/imunologia , Receptor ErbB-2/imunologia , Sensibilidade e Especificidade , Fixação de Tecidos , Trastuzumab , Resultado do Tratamento
11.
Biochemistry ; 41(49): 14539-45, 2002 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-12463753

RESUMO

Tapasin is a type I membrane glycoprotein involved with other accessory proteins in the assembly of class I MHC-beta(2)m-peptide complexes in the endoplasmic reticulum. We have probed the three-dimensional structure of the lumenal region of human tapasin (residues 1-392) tagged with a (His)(6) sequence at its C-terminus using biochemical and biophysical techniques. The far-UV circular dichroism spectrum revealed that tapasin possesses well-defined secondary structural elements corresponding predominantly to beta-sheets. A thermal denaturation curve recorded at 216 nm showed a midpoint transition centered at approximately 45 degrees C. Sedimentation analysis showed that tapasin is monomeric in solution with a sedimentation coefficient, S degrees (20,w), of 2.68 S. This value of S degrees (20,w) combined with the value of the molar mass obtained by MALDI mass spectrometry (44.2 kDa) yielded a frictional ratio, f/f(0), of 1.47. Assuming tapasin is a prolate ellipsoid, we calculated an apparent length of 22.5 nm and a diameter of 2.62 nm, consistent with an elongated molecular shape. Controlled proteolysis using various enzymes revealed that a narrow region of tapasin near residue 90 is highly susceptible to digestion, resulting in two fragments that are resistant to further cleavage. The identity of these fragments was determined by amino acid sequencing and MALDI mass spectrometry and revealed a 9 kDa N-terminal fragment and a 34 kDa C-terminal fragment. Collectively, these results suggest that tapasin is comprised of two core domains of different sizes loosely linked by a flexible region.


Assuntos
Antiporters/química , Imunoglobulinas/química , Antiporters/biossíntese , Antiporters/genética , Antiporters/isolamento & purificação , Quimotripsina/química , Dicroísmo Circular , Humanos , Hidrólise , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Imunoglobulinas/isolamento & purificação , Proteínas de Membrana Transportadoras , Elastase Pancreática/química , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/isolamento & purificação , Mapeamento de Peptídeos/métodos , Desnaturação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Análise de Sequência de Proteína , Solubilidade , Termolisina/química , Ultracentrifugação
12.
J Biol Chem ; 277(48): 46415-23, 2002 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-12235131

RESUMO

Glycosylation analysis was used to probe the sequence of events accompanying the binding of antigenic peptides to the major histocompatibility complex class I heavy chains. Free heavy chains were isolated from the beta(2)-microglobulin-negative cell line Daudi and from the B-lymphoblastoid cell line Raji. Heavy chains were also isolated from Raji cells in multimolecular complexes (peptide loading complexes) containing the transporter associated with antigen processing, tapasin and ERp57 with and without the lectin-like folding chaperone, calreticulin. Calreticulin is a soluble protein that recognizes primarily the terminal glucose of Glc(1)Man(7-9)GlcNAc(2) glycans. This paper shows that monoglucosylated glycoforms of heavy chain, which exist transiently in the endoplasmic reticulum in the initial stages of the glycosylation processing pathway, are present in the peptide loading complex. The data are consistent with a model in which the release of peptide-loaded major histocompatibility complex class I molecules from calreticulin, induced by deglucosylation of the heavy chain N-linked glycan, signals the dissociation of the complex. This is consistent with the hypothesis that the class I loading process is an adaptation of the quality control mechanism involving calreticulin and ERp57.


Assuntos
Calreticulina/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Isomerases/metabolismo , Isoformas de Proteínas/metabolismo , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Antígenos de Histocompatibilidade Classe I/química , Humanos , Isomerases de Dissulfetos de Proteínas , Isoformas de Proteínas/química , Controle de Qualidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA