Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 29(2): 473-485, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29425028

RESUMEN

THIOMAB antibody technology utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technology has enabled the exploration of different attachment sites on the antibody in combination with small molecules, peptides, or proteins to yield antibody conjugates with unique properties. As reported previously ( Shen , B. Q. , et al. ( 2012 ) Nat. Biotechnol. 30 , 184 - 189 ; Pillow , T. H. , et al. ( 2017 ) Chem. Sci. 8 , 366 - 370 ), the specific location of the site of conjugation on an antibody can impact the stability of the linkage to the engineered cysteine for both thio-succinimide and disulfide bonds. High stability of the linkage is usually desired to maximize the delivery of the cargo to the intended target. In the current study, cysteines were individually substituted into every position of the anti-HER2 antibody (trastuzumab), and the stabilities of drug conjugations at those sites were evaluated. We screened a total of 648 THIOMAB antibody-drug conjugates, each generated from a trastuzamab prepared by sequentially mutating non-cysteine amino acids in the light and heavy chains to cysteine. Each THIOMAB antibody variant was conjugated to either maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E (MC-vc-PAB-MMAE) or pyridyl disulfide monomethyl auristatin E (PDS-MMAE) using a high-throughput, on-bead conjugation and purification method. Greater than 50% of the THIOMAB antibody variants were successfully conjugated to both MMAE derivatives with a drug to antibody ratio (DAR) of >0.5 and <50% aggregation. The relative in vitro plasma stabilities for approximately 750 conjugates were assessed using enzyme-linked immunosorbent assays, and stable sites were confirmed with affinity-capture LC/MS-based detection methods. Highly stable conjugation sites for the two types of MMAE derivatives were identified on both the heavy and light chains. Although the stabilities of maleimide conjugates were shown to be greater than those of the disulfide conjugates, many sites were identified that were stable for both. Furthermore, in vitro stabilities of selected stable sites translated across different cytotoxic payloads and different target antibodies as well as to in vivo stability.


Asunto(s)
Antineoplásicos Inmunológicos/química , Cisteína/química , Disulfuros/química , Inmunoconjugados/química , Maleimidas/química , Trastuzumab/química , Animales , Antineoplásicos Inmunológicos/sangre , Cisteína/sangre , Cisteína/genética , Disulfuros/sangre , Estabilidad de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoconjugados/sangre , Maleimidas/sangre , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oligopéptidos/sangre , Oligopéptidos/química , Agregado de Proteínas , Estabilidad Proteica , Ratas , Trastuzumab/sangre , Trastuzumab/genética
2.
EMBO J ; 32(5): 713-27, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23386062

RESUMEN

The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.


Asunto(s)
Endosomas/microbiología , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Vacuolas/microbiología , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Transporte Biológico , Western Blotting , Endosomas/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Receptores de Imidazolina/genética , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/metabolismo , Lisosomas/microbiología , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Salmonella/microbiología , Técnicas del Sistema de Dos Híbridos , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rac/genética
3.
J Biol Chem ; 290(43): 25834-46, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26342081

RESUMEN

The insulin/insulin-like growth factor (IGF)-1 signaling pathway (ISP) plays a fundamental role in long term health in a range of organisms. Protein kinases including Akt and ERK are intimately involved in the ISP. To identify other kinases that may participate in this pathway or intersect with it in a regulatory manner, we performed a whole kinome (779 kinases) siRNA screen for positive or negative regulators of the ISP, using GLUT4 translocation to the cell surface as an output for pathway activity. We identified PFKFB3, a positive regulator of glycolysis that is highly expressed in cancer cells and adipocytes, as a positive ISP regulator. Pharmacological inhibition of PFKFB3 suppressed insulin-stimulated glucose uptake, GLUT4 translocation, and Akt signaling in 3T3-L1 adipocytes. In contrast, overexpression of PFKFB3 in HEK293 cells potentiated insulin-dependent phosphorylation of Akt and Akt substrates. Furthermore, pharmacological modulation of glycolysis in 3T3-L1 adipocytes affected Akt phosphorylation. These data add to an emerging body of evidence that metabolism plays a central role in regulating numerous biological processes including the ISP. Our findings have important implications for diseases such as type 2 diabetes and cancer that are characterized by marked disruption of both metabolism and growth factor signaling.


Asunto(s)
Glucosa/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Fosfofructoquinasa-2/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Células 3T3-L1 , Animales , Transportador de Glucosa de Tipo 4/metabolismo , Células HeLa , Humanos , Ratones , ARN Interferente Pequeño/genética
4.
Diabetologia ; 58(7): 1587-600, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25899451

RESUMEN

AIMS/HYPOTHESIS: Adipose tissue (AT) distribution is a major determinant of mortality and morbidity in obesity. In mice, intra-abdominal transplantation of subcutaneous AT (SAT) protects against glucose intolerance and insulin resistance (IR), but the underlying mechanisms are not well understood. METHODS: We investigated changes in adipokines, tissue-specific glucose uptake, gene expression and systemic inflammation in male C57BL6/J mice implanted intra-abdominally with either inguinal SAT or epididymal visceral AT (VAT) and fed a high-fat diet (HFD) for up to 17 weeks. RESULTS: Glucose tolerance was improved in mice receiving SAT after 6 weeks, and this was not attributable to differences in adiposity, tissue-specific glucose uptake, or plasma leptin or adiponectin concentrations. Instead, SAT transplantation prevented HFD-induced hepatic triacylglycerol accumulation and normalised the expression of hepatic gluconeogenic enzymes. Grafted fat displayed a significant increase in glucose uptake and unexpectedly, an induction of skeletal muscle-specific gene expression. Mice receiving subcutaneous fat also displayed a marked reduction in the plasma concentrations of several proinflammatory cytokines (TNF-α, IL-17, IL-12p70, monocyte chemoattractant protein-1 [MCP-1] and macrophage inflammatory protein-1ß [ΜIP-1ß]), compared with sham-operated mice. Plasma IL-17 and MIP-1ß concentrations were reduced from as early as 4 weeks after transplantation, and differences in plasma TNF-α and IL-17 concentrations predicted glucose tolerance and insulinaemia in the entire cohort of mice (n = 40). In contrast, mice receiving visceral fat transplants were glucose intolerant, with increased hepatic triacylglycerol content and elevated plasma IL-6 concentrations. CONCLUSIONS/INTERPRETATION: Intra-abdominal transplantation of subcutaneous fat reverses HFD-induced glucose intolerance, hepatic triacylglycerol accumulation and systemic inflammation in mice.


Asunto(s)
Intolerancia a la Glucosa/cirugía , Inflamación/cirugía , Grasa Subcutánea/trasplante , Adipocitos/metabolismo , Adipocitos/ultraestructura , Adiponectina/sangre , Adiposidad , Animales , Composición Corporal , Citocinas/sangre , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Gluconeogénesis , Glucosa/metabolismo , Insulina/sangre , Leptina/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Triglicéridos/metabolismo
5.
J Cell Sci ; 126(Pt 20): 4647-58, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23902685

RESUMEN

Invadopodia-dependent degradation of the basement membrane plays a major role during metastasis of breast cancer cells. Basement membrane degradation is mediated by targeted secretion of various matrix metalloproteinases (MMPs). Specifically, MMP2 and MMP9 (MMP2/9) possess the ability to hydrolyze components of the basement membrane and regulate various aspects of tumor growth and metastasis. However, the membrane transport machinery that mediates targeting of MMP2/9 to the invadopodia during cancer cell invasion remains to be defined. Because Rab GTPases are key regulators of membrane transport, we screened a human Rab siRNA library and identified Rab40b GTPase as a protein required for secretion of MMP2/9. We also have shown that Rab40b functions during at least two distinct steps of MMP2/9 transport. Here, we demonstrate that Rab40b is required for MMP2/9 sorting into VAMP4-containing secretory vesicles. We also show that Rab40b regulates transport of MMP2/9 secretory vesicles during invadopodia formation and is required for invadopodia-dependent extracellular matrix degradation. Finally, we demonstrate that Rab40b is also required for breast cancer cell invasion in vitro. On the basis of these findings, we propose that Rab40b mediates trafficking of MMP2/9 during invadopodia formation and metastasis of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Seudópodos/metabolismo , Seudópodos/patología , Proteínas de Unión al GTP rab/metabolismo , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica , Seudópodos/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección
6.
Pharm Res ; 32(6): 1884-93, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25446772

RESUMEN

PURPOSE: THIOMAB™ drug conjugates (TDCs) with engineered cysteine residues allow site-specific drug conjugation and defined Drug-to-Antibody Ratios (DAR). In order to help elucidate the impact of drug-loading, conjugation site, and subsequent deconjugation on pharmacokinetics and efficacy, we have developed an integrated mathematical model to mechanistically characterize pharmacokinetic behavior and preclinical efficacy of MMAE conjugated TDCs with different DARs. General applicability of the model structure was evaluated with two different TDCs. METHOD: Pharmacokinetics studies were conducted for unconjugated antibody and purified TDCs with DAR-1, 2 and 4 for trastuzumab TDC and Anti-STEAP1 TDC in mice. Total antibody concentrations and individual DAR fractions were measured. Efficacy studies were performed in tumor-bearing mice. RESULTS: An integrated model consisting of distinct DAR species (DAR0-4), each described by a two-compartment model was able to capture the experimental data well. Time series measurements of each Individual DAR species allowed for the incorporation of site-specific drug loss through deconjugation and the results suggest a higher deconjugation rate from heavy chain site HC-A114C than the light chain site LC-V205C. Total antibody concentrations showed multi-exponential decline, with a higher clearance associated with higher DAR species. The experimentally observed effects of TDC on tumor growth kinetics were successfully described by linking pharmacokinetic profiles to DAR-dependent killing of tumor cells. CONCLUSION: Results from the integrated model evaluated with two different TDCs highlight the impact of DAR and site of conjugation on pharmacokinetics and efficacy. The model can be used to guide future drug optimization and in-vivo studies.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Modelos Biológicos , Compuestos de Sulfhidrilo/farmacocinética , Trastuzumab/metabolismo , Administración Intravenosa , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antígenos de Neoplasias/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Disponibilidad Biológica , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cisteína , Femenino , Masculino , Tasa de Depuración Metabólica , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Compuestos de Sulfhidrilo/administración & dosificación , Compuestos de Sulfhidrilo/química , Trastuzumab/administración & dosificación , Trastuzumab/química
7.
Traffic ; 13(10): 1429-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22762500

RESUMEN

Insulin stimulates glucose transport in adipocytes by triggering translocation of GLUT4 glucose transporters to the plasma membrane (PM) and several Rabs including Rab10 have been implicated in this process. To delineate the molecular regulation of this pathway, we conducted a TBC/RabGAP overexpression screen in adipocytes. This identified TBC1D13 as a potent inhibitor of insulin-stimulated GLUT4 translocation without affecting other trafficking pathways. To determine the potential Rab substrate for TBC1D13 we conducted a yeast two-hybrid screen and found that the GTP bound forms of Rabs 1 and 10 specifically interacted with TBC1D13 but not with eight other TBC proteins. Surprisingly, a comprehensive in vitro screen for TBC1D13 GAP activity revealed Rab35 but not Rab10 as a specific substrate. TBC1D13 also displayed in vivo GAP activity towards Rab35. Overexpression of constitutively active Rab35 but not constitutively active Rab10 reversed the block in insulin-stimulated GLUT4 translocation observed with TBC1D13 overexpression. These studies implicate an important role for Rab35 in insulin-stimulated GLUT4 translocation in adipocytes.


Asunto(s)
Adipocitos/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Células 3T3-L1 , Animales , Proteínas Activadoras de GTPasa/genética , Expresión Génica , Células HEK293 , Humanos , Insulina/metabolismo , Ratones , Proteínas Nucleares/genética , Transporte de Proteínas
8.
J Biol Chem ; 287(9): 6128-38, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22207758

RESUMEN

Akt plays a major role in insulin regulation of metabolism in muscle, fat, and liver. Here, we show that in 3T3-L1 adipocytes, Akt operates optimally over a limited dynamic range. This indicates that Akt is a highly sensitive amplification step in the pathway. With robust insulin stimulation, substantial changes in Akt phosphorylation using either pharmacologic or genetic manipulations had relatively little effect on Akt activity. By integrating these data we observed that half-maximal Akt activity was achieved at a threshold level of Akt phosphorylation corresponding to 5-22% of its full dynamic range. This behavior was also associated with lack of concordance or demultiplexing in the behavior of downstream components. Most notably, FoxO1 phosphorylation was more sensitive to insulin and did not exhibit a change in its rate of phosphorylation between 1 and 100 nm insulin compared with other substrates (AS160, TSC2, GSK3). Similar differences were observed between various insulin-regulated pathways such as GLUT4 translocation and protein synthesis. These data indicate that Akt itself is a major amplification switch in the insulin signaling pathway and that features of the pathway enable the insulin signal to be split or demultiplexed into discrete outputs. This has important implications for the role of this pathway in disease.


Asunto(s)
Adipocitos/enzimología , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Simulación por Computador , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Transportador de Glucosa de Tipo 4/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Dinámicas no Lineales , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
9.
J Biol Chem ; 287(49): 40996-1006, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23055524

RESUMEN

The APPL1 and APPL2 proteins (APPL (adaptor protein, phosphotyrosine interaction, pleckstrin homology (PH) domain, and leucine zipper-containing protein)) are localized to their own endosomal subcompartment and interact with a wide range of proteins and small molecules at the cell surface and in the nucleus. They play important roles in signal transduction through their ability to act as Rab effectors. (Rabs are a family of Ras GTPases involved in membrane trafficking.) Both APPL1 and APPL2 comprise an N-terminal membrane-curving BAR (Bin-amphiphysin-Rvs) domain linked to a PH domain and a C-terminal phosphotyrosine-binding domain. The structure and interactions of APPL1 are well characterized, but little is known about APPL2. Here, we report the crystal structure and low resolution solution structure of the BARPH domains of APPL2. We identify a previously undetected hinge site for rotation between the two domains and speculate that this motion may regulate APPL2 functions. We also identified Rab binding partners of APPL2 and show that these differ from those of APPL1, suggesting that APPL-Rab interaction partners have co-evolved over time. Isothermal titration calorimetry data reveal the interaction between APPL2 and Rab31 has a K(d) of 140 nM. Together with other biophysical data, we conclude the stoichiometry of the complex is 2:2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Membrana Celular/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Calorimetría/métodos , Núcleo Celular/metabolismo , Clonación Molecular , Cristalización , Cristalografía por Rayos X/métodos , Dimerización , GTP Fosfohidrolasas/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Fosfatidilinositoles/química , Mapeo de Interacción de Proteínas/métodos , Estructura Terciaria de Proteína , Dispersión de Radiación , Homología de Secuencia de Aminoácido , Transducción de Señal , Solventes/química , Electricidad Estática , Propiedades de Superficie , Rayos X , Proteínas de Unión al GTP rab/metabolismo
10.
MAbs ; 15(1): 2229101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37639687

RESUMEN

The antibody-drug conjugate (ADC) field has undergone a renaissance, with substantial recent developmental investment and subsequent drug approvals over the past 6 y. In November 2022, ElahereTM became the latest ADC to be approved by the US Food and Drug Administration (FDA). To date, over 260 ADCs have been tested in the clinic against various oncology indications. Here, we review the clinical landscape of ADCs that are currently FDA approved (11), agents currently in clinical trials but not yet approved (164), and candidates discontinued following clinical testing (92). These clinically tested ADCs are further analyzed by their targeting tumor antigen(s), linker, payload choices, and highest clinical stage achieved, highlighting limitations associated with the discontinued drug candidates. Lastly, we discuss biologic engineering modifications preclinically demonstrated to improve the therapeutic index that if incorporated may increase the proportion of molecules that successfully transition to regulatory approval.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Estados Unidos , Inmunoconjugados/uso terapéutico , Anticuerpos Monoclonales , United States Food and Drug Administration
11.
Blood ; 114(13): 2721-9, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19633198

RESUMEN

Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a standard-of-care regimen, R-CHOP (ie, rituximab with a single intravenous injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.


Asunto(s)
Antígenos CD79/inmunología , Linfoma no Hodgkin/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Animales , Anticuerpos Antiidiotipos/uso terapéutico , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Inmunoconjugados/uso terapéutico , Linfoma no Hodgkin/patología , Ratones , Ratones Endogámicos ICR , Ratones SCID , Oligopéptidos/química , Resultado del Tratamiento , Carga Tumoral , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Anal Biochem ; 412(1): 56-66, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21216214

RESUMEN

Antibody-drug conjugates (ADCs) are designed to facilitate the targeted delivery of cytotoxic drugs to improve their tumor fighting effects and minimize systemic toxicity. However, efficacy and safety can potentially be compromised due to the release of conjugated drugs from the ADC with time while in circulation, resulting in changes in the drug-to-antibody ratio (DAR). Current understanding of this process is limited because existing methods such as immunoassays fail to distinguish ADCs with different DARs. Here we demonstrate a novel method with bead-based affinity capture and capillary liquid chromatography-mass spectrometry to allow direct measurement of drug release by quantifying DAR distributions of the ADC in plasma/serum. This method successfully identified individual intact conjugated antibody species produced due to drug loss from ADCs (e.g., an engineered site-specific anti-MUC16 THIOMAB-drug conjugate) and measured the corresponding DAR distributions in vitro and in vivo. Information obtained can provide insights into the mechanisms involved in drug loss and help to optimize ADC therapeutics. Other potential applications of the method may include characterization of posttranslational modifications, protein adducts, and immunogenicity.


Asunto(s)
Anticuerpos Monoclonales/sangre , Cromatografía de Afinidad/métodos , Cromatografía Liquida/métodos , Inmunoconjugados/sangre , Espectrometría de Masas/métodos , Preparaciones Farmacéuticas/sangre , Animales , Anticuerpos Monoclonales/química , Biotinilación , Antígeno Ca-125/inmunología , Femenino , Haplorrinos , Humanos , Inmunoconjugados/química , Inmunoconjugados/aislamiento & purificación , Masculino , Proteínas de la Membrana/inmunología , Ratones , Oligopéptidos/química , Preparaciones Farmacéuticas/química , Ratas
13.
Commun Biol ; 4(1): 475, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846513

RESUMEN

COVID-19 is a respiratory illness caused by a novel coronavirus called SARS-CoV-2. The viral spike (S) protein engages the human angiotensin-converting enzyme 2 (ACE2) receptor to invade host cells with ~10-15-fold higher affinity compared to SARS-CoV S-protein, making it highly infectious. Here, we assessed if ACE2 polymorphisms can alter host susceptibility to SARS-CoV-2 by affecting this interaction. We analyzed over 290,000 samples representing >400 population groups from public genomic datasets and identified multiple ACE2 protein-altering variants. Using reported structural data, we identified natural ACE2 variants that could potentially affect virus-host interaction and thereby alter host susceptibility. These include variants S19P, I21V, E23K, K26R, T27A, N64K, T92I, Q102P and H378R that were predicted to increase susceptibility, while variants K31R, N33I, H34R, E35K, E37K, D38V, Y50F, N51S, M62V, K68E, F72V, Y83H, G326E, G352V, D355N, Q388L and D509Y were predicted to be protective variants that show decreased binding to S-protein. Using biochemical assays, we confirmed that K31R and E37K had decreased affinity, and K26R and T92I variants showed increased affinity for S-protein when compared to wildtype ACE2. Consistent with this, soluble ACE2 K26R and T92I were more effective in blocking entry of S-protein pseudotyped virus suggesting that ACE2 variants can modulate susceptibility to SARS-CoV-2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Predisposición Genética a la Enfermedad/genética , Mutación Missense/genética , Polimorfismo Genético , Receptores Virales/genética , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Receptores Virales/química , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
14.
J Immunol Methods ; 332(1-2): 41-52, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18230399

RESUMEN

Cysteines with reactive thiol groups are attractive tools for site-specific labeling of proteins. Engineering a reactive cysteine residue into proteins with multiple disulfide bonds is often a challenging task as it may interfere with structural and functional properties of the protein. Here we developed a phage display-based biochemical assay, PHESELECTOR (Phage ELISA for Selection of Reactive Thiols) to rapidly screen reactive thiol groups on antibody fragments without interfering with their antigen binding, using trastuzumab-Fab (hu4D5Fab) as a model system. The solvent accessibility values for all the amino acid residues in the hu4D5Fab were calculated using available crystal structure information. Serine, alanine and valine residues with highest solvent accessibility values were selected and tested to compare structure-based design with the PHESELECTOR biochemical method. Cysteine substitutions at partially solvent-accessible alanine or valine residues exhibited better thiol reactivity values than substitutions at serine residues. The poor correlation between fractional solvent accessibility and thiol reactivity of the engineered hu4D5Fab variants indicated the value of PHESELECTOR biochemical assay to identify reactive thiol groups on the antibody-Fab surface. Mass spectrometric analysis of biotinylated ThioFab (Fab with engineered cysteine) variants confirmed that conjugation occurred only at the engineered cysteine thiols of either light or heavy chains. ThioFabs with engineered cysteine residues in the constant domains (CL and CH(1)) should allow universal application for site-specific conjugation of antibody-Fabs.


Asunto(s)
Anticuerpos Monoclonales/química , Cisteína/química , Fragmentos Fab de Inmunoglobulinas/química , Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo , Sitios de Unión/genética , Sitios de Unión/inmunología , Biotina/química , Línea Celular Tumoral , Cromatografía Liquida/métodos , Cisteína/genética , Cisteína/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Regulación de la Expresión Génica/genética , Ingeniería Genética , Variación Genética , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/inmunología , Maleimidas/química , Espectrometría de Masas/métodos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Plásmidos/genética , Sensibilidad y Especificidad , Coloración y Etiquetado/métodos , Propiedades de Superficie
15.
ACS Med Chem Lett ; 9(1): 56-60, 2018 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-29348812

RESUMEN

Antibody-drug conjugates (ADCs) represent an important class of emerging cancer therapeutics. Recent ADC development efforts highlighted the use of pyrrolobenzodiazepine (PBD) dimer payload for the treatment of several cancers. We identified the isoquinolidinobenzodiazepine (IQB) payload (D211), a new class of PBD dimer family of DNA damaging payloads. We have successfully synthesized all three IQB stereoisomers, experimentally showed that the purified (S,S)-D211 isomer is functionally more active than (R,R)-D221 and (S,R)-D231 isomers by >50,000-fold and ∼200-fold, respectively. We also synthesized a linker-payload (D212) that uses (S,S)-D211 payload with a cathepsin cleavable linker, a hydrophilic PEG8 spacer, and a thiol reactive maleimide. In addition, homogeneous ADCs generated using D212 linker-payload exhibited ideal physicochemical properties, and anti-CD33 ADC displayed a robust target-specific potency on AML cell lines. These results demonstrate that D212 linker-payload described here can be utilized for developing novel ADC therapeutics for targeted cancer therapy.

16.
Blood Adv ; 2(14): 1738-1749, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30037800

RESUMEN

The current standard of care for acute myeloid leukemia (AML) is largely ineffective with very high relapse rates and low survival rates, mostly due to the inability to eliminate a rare population of leukemic stem cells (LSCs) that initiate tumor growth and are resistant to standard chemotherapy. RNA-sequencing analysis on isolated LSCs confirmed C-type lectin domain family 12 member A (CLL1, also known as CLEC12A) to be highly expressed on LSCs but not on normal hematopoietic stem cells (HSCs) or other healthy organ tissues. Expression of CLL1 was consistent across different types of AML. We developed CLT030 (CLL1-ADC), an antibody-drug conjugate (ADC) based on a humanized anti-CLL1 antibody with 2 engineered cysteine residues linked covalently via a cleavable linker to a highly potent DNA-binding payload, thus resulting in a site-specific and homogenous ADC product. The ADC is designed to be stable in the bloodstream and to release its DNA-binding payload only after the ADC binds to CLL1-expressing tumor cells, is internalized, and the linker is cleaved in the lysosomal compartment. CLL1-ADC inhibits in vitro LSC colony formation and demonstrates robust in vivo efficacy in AML cell tumor models and tumor growth inhibition in the AML patient-derived xenograft model. CLL1-ADC demonstrated a reduced effect on differentiation of healthy normal human CD34+ cells to various lineages as observed in an in vitro colony formation assay and in an in vivo xenotransplantation model as compared with CD33-ADC. These results demonstrate that CLL1-ADC could be an effective ADC therapeutic for the treatment of AML.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunoconjugados/farmacología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda , Proteínas de Neoplasias/inmunología , Células Madre Neoplásicas , Receptores Mitogénicos/inmunología , Animales , Femenino , Células HL-60 , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Masculino , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Células Madre Neoplásicas/patología , Receptores Mitogénicos/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Mol Cancer Ther ; 17(3): 638-649, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29282299

RESUMEN

Luminal A (hormone receptor-positive) breast cancer constitutes 70% of total breast cancer patients. In an attempt to develop a targeted therapeutic for this cancer indication, we have identified and characterized Glial cell line-Derived Neurotrophic Factor (GDNF) Family Receptor Alpha 1 (GFRA1) antibody-drug conjugates (ADC) using a cleavable valine-citrulline-MMAE (vcMMAE) linker-payload. RNAseq and IHC analysis confirmed the abundant expression of GFRA1 in luminal A breast cancer tissues, whereas minimal or no expression was observed in most normal tissues. Anti-GFRA-vcMMAE ADC internalized to the lysosomes and exhibited target-dependent killing of GFRA1-expressing cells both in vitro and in vivo The ADCs using humanized anti-GFRA1 antibodies displayed robust therapeutic activity in clinically relevant cell line-derived (MCF7 and KPL-1) tumor xenograft models. The lead anti-GFRA1 ADC cross-reacts with rodent and cynomolgus monkey GFRA1 antigen and showed optimal pharmacokinetic properties in both species. These properties subsequently enabled a target-dependent toxicity study in rats. Anti-GFRA1 ADC is well tolerated in rats, as seen with other vcMMAE linker-payload based ADCs. Overall, these data suggest that anti-GFRA1-vcMMAE ADC may provide a targeted therapeutic opportunity for luminal A breast cancer patients. Mol Cancer Ther; 17(3); 638-49. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/antagonistas & inhibidores , Inmunoconjugados/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos/química , Anticuerpos/inmunología , Anticuerpos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/inmunología , Células HEK293 , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Células MCF-7 , Macaca fascicularis , Ratones Desnudos , Ratones SCID , Ratas Sprague-Dawley , Receptores de Esteroides/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética
18.
Eur J Cell Biol ; 86(8): 417-31, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17628206

RESUMEN

Rab11-FIP3 is a Rab11-binding protein that has been implicated in regulating cytokinesis in mammalian cells. FIP3 functions by simultaneously interacting with Rab11 as well as Arf GTPases. However, unlike the interaction between Rab11 and FIP3, the structural basis of FIP3 binding to Arf GTPases has not yet been determined. The specificity of interaction between FIP3 and Arf GTPases remains controversial. While it was reported that FIP3 preferentially binds to Arf6 some data suggest that FIP3 can also interact with Arf5 and even possibly Arf4. The Arf-interaction motif on FIP3 also remains to be determined. Finally, the importance of Arf binding to FIP3 in regulating cell division and other cellular functions remains unclear. Here we used a combination of various biochemical techniques to measure the affinity of FIP3 binding to various Arfs and to demonstrate that FIP3 predominantly interacts with Arf6 in vitro and in vivo. In addition, we identified the motifs mediating Arf6 and FIP3 interaction and demonstrated that FIP3 binds to the Arf6 C-terminus rather than switch motifs. Finally we show that FIP3 and Arf6 binding is required for the targeting of Arf6 to the cleavage furrow during cytokinesis. Thus, we propose that FIP3 is a scaffolding protein that, in addition to regulating endosome targeting to the cleavage furrow, also is required for Arf6 recruitment to the midbody during late telophase.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Portadoras/metabolismo , Factor 6 de Ribosilación del ADP , Sitios de Unión , Proteínas Portadoras/química , Citocinesis , Endosomas/metabolismo , Células HeLa , Humanos , Proteínas Mutantes/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Transfección , Técnicas del Sistema de Dos Híbridos
19.
Mol Biol Cell ; 15(8): 3530-41, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15181150

RESUMEN

Rab 11 GTPase is an important regulator of endocytic membrane traffic. Recently, we and others have identified a novel family of Rab11 binding proteins, known as Rab11-family interacting proteins (FIPs). One of the family members, Rab coupling protein (RCP), was identified as a protein binding to both Rab4 and Rab11 GTPases. RCP was therefore suggested to serve a dual function as Rab4 and Rab11 binding protein. In this study, we characterized the cellular functions of RCP and mapped its interactions with Rab4 and Rab11. Our data show that RCP interacts only weakly with Rab4 in vitro and does not play the role of coupling Rab11 and Rab4 in vivo. Furthermore, our data indicate that the RCP-Rab11 complex regulates the sorting of transferrin receptors from the degradative to the recycling pathway. We therefore propose that RCP functions primarily as a Rab11 binding protein that regulates protein sorting in tubular endosomes.


Asunto(s)
Proteínas Portadoras/metabolismo , Endocitosis/fisiología , Proteínas de la Membrana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Endocitosis/efectos de los fármacos , Endocitosis/genética , Endosomas/inmunología , Endosomas/fisiología , Células HeLa , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión al GTP rab4/metabolismo
20.
Mol Biol Cell ; 15(5): 2218-29, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15004230

RESUMEN

Rab GTPases are localized to various intracellular compartments and are known to play important regulatory roles in membrane trafficking. Here, we report the subcellular distribution and function of Rab14. By immunofluorescence and immunoelectron microscopy, both endogenous as well as overexpressed Rab14 were localized to biosynthetic (rough endoplasmic reticulum, Golgi, and trans-Golgi network) and endosomal compartments (early endosomal vacuoles and associated vesicles). Notably overexpression of Rab14Q70L shifted the distribution toward the early endosome associated vesicles, whereas the S25N and N124I mutants induced a shift toward the Golgi region. A similar, although less pronounced, redistribution of the transferrin receptor was also observed in cells overexpressing Rab14 mutants. Impairment of Rab14 function did not however affect transferrin uptake or recycling kinetics. Together, these findings suggest that Rab14 is involved in the biosynthetic/recycling pathway between the Golgi and endosomal compartments.


Asunto(s)
Endosomas/fisiología , Aparato de Golgi/fisiología , Proteínas de Unión al GTP rab/fisiología , Animales , Anticuerpos/inmunología , Línea Celular , Membrana Celular/fisiología , Endosomas/ultraestructura , Expresión Génica , Aparato de Golgi/ultraestructura , Proteínas Fluorescentes Verdes/análisis , Humanos , Espacio Intracelular/ultraestructura , Ratones , Microscopía Fluorescente , Mutación Puntual/genética , Transporte de Proteínas/fisiología , Ratas , Receptores de Transferrina/análisis , Receptores de Transferrina/metabolismo , Transferrina/metabolismo , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión al GTP rab/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA