Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Anal Chem ; 95(8): 3922-3931, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36791402

RESUMEN

Characterization of antibody binding epitopes is an important factor in therapeutic drug discovery, as the binding site determines and drives antibody pharmacology and pharmacokinetics. Here, we present a novel application of carbene chemical footprinting with mass spectrometry for identification of antibody binding epitopes at the single-residue level. Two different photoactivated diazirine reagents provide complementary labeling information allowing structural refinement of the antibody binding interface. We applied this technique to map the epitopes of multiple MICA and CTLA-4 antibodies and validated the findings with X-ray crystallography and yeast surface display epitope mapping. The characterized epitopes were used to understand biolayer interferometry-derived competitive binding results at the structural level. We show that carbene footprinting provides fast and high-resolution epitope information critical in the antibody selection process and enables mechanistic understanding of function to accelerate the drug discovery process.


Asunto(s)
Anticuerpos , Metano , Epítopos/química , Mapeo Epitopo/métodos
2.
Bioconjug Chem ; 33(4): 576-585, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35344340

RESUMEN

N-linked glycosylation is one of the most common and complex posttranslational modifications that govern the biological functions and physicochemical properties of therapeutic antibodies. We evaluated thermal and metabolic stabilities of antibody-drug conjugates (ADCs) with payloads attached to the C'E loop in the immunoglobulin G (IgG) Fc CH2 domain, comparing the glycosylated and aglycosylated Fc ADC variants. Our study revealed that introduction of small-molecule drugs into an aglycosylated antibody can compensate for thermal destabilization originating from structural distortions caused by elimination of N-linked glycans. Depending on the conjugation site, glycans had both positive and negative effects on plasma stability of ADCs. The findings highlight the importance of consideration for selection of conjugation site to achieve desirable physicochemical properties and plasma stability.


Asunto(s)
Inmunoconjugados , Inmunoglobulina G , Glicosilación , Inmunoconjugados/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional
3.
Anal Chem ; 93(13): 5371-5376, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33750099

RESUMEN

Traditionally the biotransformation of antibody drug conjugates (ADCs) has been evaluated by affinity capture on streptavidin magnetic beads coated with a biotinylated capture reagent. To reduce the complexity of the analyte, the affinity captured ADCs are digested with enzymes ("on-bead" or after elution), and/or interchain disulfides are reduced to generate LC and HC fragments prior to mass spectrometry analysis. The "on-bead" enzymatic digestion with IdeS and PNGase F is not efficient and requires longer incubation times to achieve complete Fc and N-glycan removal. This results in a prolonged sample preparation time (7-18 h) and is not suitable for labile ADCs due to the possibility of assay-induced artifacts. To address these challenges, we developed an affinity capture method, where the ADCs are first captured onto streptavidin cartridges coated with a biotinylated generic capture reagent, followed by a 15 min "on-cartridge" digestion with IdeS or PNGase F. The ADCs are then eluted and directly analyzed by LC-HRMS. This method was successfully applied for the biotransformation assessment of site-specific ADCs with payload conjugated on the Fab or Fc. The reduced complexity of the analyte (Fc and N-glycan removal) combined with HRMS enabled sensitive and accurate identification of minor mass change catabolites and changes in the DAR distribution. This automated cartridge-based affinity capture method is fast with a total sample preparation time of less than 4 h (hands-on time of less than 1 h) and can be utilized for any human mAb/ADC independent of isotype (IgG1, IgG2, and IgG4).


Asunto(s)
Inmunoconjugados , Biotransformación , Disulfuros , Humanos , Inmunoglobulina G , Espectrometría de Masas
4.
Anal Chem ; 93(49): 16474-16480, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34854675

RESUMEN

Identification of antibodies targeting diverse functional epitopes on an antigen is highly crucial for discovering effective therapeutic candidates. Employing a traditional stepwise antibody "screening funnel" as well as prioritizing affinity-based selections over epitope-based selections, result in lead antibody panels lacking epitope diversity. In the present study, we employed an array-based surface plasmon resonance (SPR) platform to perform high-throughput epitope binning analysis on a large number of monoclonal antibodies (mAbs) generated in the early drug discovery process. The mAb panel contained clones from different antibody generation techniques and diverse transgenic mouse strains. The epitope binning results were analyzed in unique ways using various visualizations in the form of dendrograms and network plots, which assisted in determining diversity and redundancy in the mAb sample set. The binning data were further integrated with affinity information to evaluate the performance of seven different transgenic mouse strains. The combination of epitope binning results with binding kinetics and sequence analysis provided an effective and efficient way of selecting high affinity antibodies representing a diverse set of sequence families and epitopes.


Asunto(s)
Anticuerpos Monoclonales , Antineoplásicos Inmunológicos , Animales , Epítopos , Ratones , Resonancia por Plasmón de Superficie
5.
Anal Chem ; 92(15): 10709-10716, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32639723

RESUMEN

Bispecific antibodies (BsAbs), with a unique mechanism of recognizing two different epitopes or antigens, have shown potential in various therapeutic areas. Molecular characterization of BsAbs' epitopes not only allows for detailed understanding of their mechanism of actions but also guides the design and selection of drug candidate molecules. In this study, we illustrate the practical utility of an integrated approach, including size exclusion chromatography with multiangle light scattering and native mass spectrometry (MS) for the biophysical characterization of complex formation of a BsAb with two target antigens, cluster of differentiation 3 (CD3) and B-cell maturation antigen (BCMA). MS-based protein footprinting strategies, including hydrogen/deuterium exchange MS, fast photochemical oxidation of proteins, and carboxyl group footprinting with glycine ethyl ester, were further applied to determine BsAb's binding epitopes. This combination approach provides molecular details on the binding mechanisms of BsAb to the two distinct antigens with rapid output and high resolution.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígenos/inmunología , Cromatografía en Gel , Mapeo Epitopo/métodos , Espectrometría de Masas , Huella de Proteína , Anticuerpos Biespecíficos/química , Modelos Moleculares , Conformación Proteica
6.
Anal Chem ; 92(2): 2065-2073, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31860282

RESUMEN

Antibody drug conjugates (ADCs) can undergo in vivo biotransformation (e.g., payload metabolism, deconjugation) leading to reduced or complete loss of activity. The location/site of conjugation of payload-linker can have an effect on ADC stability and hence needs to be carefully optimized. Affinity capture LC-MS of intact ADCs or ADC subfragments has been extensively used to evaluate ADC biotransformation. However, the current methods have certain limitations such as the requirement of specific capture reagents, limited mass resolution of low mass change metabolites, low sensitivity, and use of capillary or nanoflow LC-MS. To address these challenges, we developed a generic affinity capture LC-MS assay that can be utilized to evaluate the biotransformation of any site-specific ADC independent of antibody type and site of conjugation (Fab and Fc) in preclinical studies. The method involves a combination of some or all of these steps: (1) "mono capture" or "dual capture" of ADCs from serum with streptavidin magnetic beads coated with a generic biotinylated antihuman capture reagent, (2) "on-bead" digestion with IdeS and/or PNGase F, and (3) reduction of interchain disulfide bonds to generate ∼25 kDa ADC subfragments, which are finally analyzed by LC-HRMS on a TOF mass spectrometer. The advantages of this method are that it can be performed using commercially available generic reagents and requires sample preparation time of less than 7 h. Furthermore, by reducing the size of intact ADC (∼150 kDa) to subfragments (∼25 kDa), the identification of conjugated payload and its metabolites can be achieved with excellent sensitivity and resolution (hydrolysis and other small mass change metabolites). This method was successfully applied to evaluate the in vitro and in vivo biotransformation of ADCs conjugated at different sites (LC, HC-Fab, and HC-Fc) with various classes of payload-linkers.


Asunto(s)
Biotransformación , Inmunoconjugados/sangre , Inmunoconjugados/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Cromatografía Liquida , Humanos , Espectrometría de Masas
7.
Nat Immunol ; 9(7): 777-84, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18516039

RESUMEN

It remains unclear whether gammadelta T cell antigen receptors (TCRs) detect antigens in a way similar to antibodies or alphabeta TCRs. Here we show that reactivity between the G8 and KN6 gammadelta TCRs and the major histocompatibility complex class Ib molecule T22 could be recapitulated, with retention of wild-type ligand affinity, in an alphabeta TCR after grafting of a G8 or KN6 complementarity-determining region 3-delta (CDR3delta) loop in place of the CDR3alpha loop of an alphabeta TCR. We also found that a shared sequence motif in CDR3delta loops of all T22-reactive gammadelta TCRs bound T22 in energetically distinct ways, and that T10(d), which bound G8 with weak affinity, was converted into a high-affinity ligand by a single point mutation. Our results demonstrate unprecedented autonomy of a single CDR3 loop in antigen recognition.


Asunto(s)
Regiones Determinantes de Complementariedad/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Sitios de Unión , Dicroismo Circular , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Unión Proteica , Estructura Cuaternaria de Proteína , Receptores de Antígenos de Linfocitos T gamma-delta/química , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie
8.
Bioconjug Chem ; 31(4): 1199-1208, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32178516

RESUMEN

Antibody-drug conjugates (ADCs) are a therapeutic modality that traditionally enable the targeted delivery of highly potent cytotoxic agents to specific cells such as tumor cells. More recently, antibodies have been used to deliver molecules such as antibiotics, antigens, and adjuvants to bacteria or specific immune cell subsets. Site-directed mutagenesis of proteins permits more precise control over the site and stoichiometry of their conjugation, giving rise to homogeneous chemically defined ADCs. Identification of favorable sites for conjugation in antibodies is essential as reaction efficiency and product stability are influenced by the tertiary structure of immunoglobulin G (IgG). Current methods to evaluate potential conjugation sites are time-consuming and labor intensive, involving multistep processes for individually produced reactions. Here, we describe a highly efficient method for identification of conjugatable genetic variants by analyzing pooled ADC libraries using mass spectrometry. This approach provides a versatile platform to rapidly uncover new conjugation sites for site-specific ADCs.


Asunto(s)
Inmunoconjugados/química , Inmunoconjugados/genética , Variación Genética , Inmunoglobulina G/química , Espectrometría de Masas , Estructura Terciaria de Proteína
9.
J Biol Chem ; 291(27): 13974-13986, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27129258

RESUMEN

The prominent role of voltage-gated sodium channel 1.7 (Nav1.7) in nociception was revealed by remarkable human clinical and genetic evidence. Development of potent and subtype-selective inhibitors of this ion channel is crucial for obtaining therapeutically useful analgesic compounds. Microproteins isolated from animal venoms have been identified as promising therapeutic leads for ion channels, because they naturally evolved to be potent ion channel blockers. Here, we report the engineering of highly potent and selective inhibitors of the Nav1.7 channel based on tarantula ceratotoxin-1 (CcoTx1). We utilized a combination of directed evolution, saturation mutagenesis, chemical modification, and rational drug design to obtain higher potency and selectivity to the Nav1.7 channel. The resulting microproteins are highly potent (IC50 to Nav1.7 of 2.5 nm) and selective. We achieved 80- and 20-fold selectivity over the closely related Nav1.2 and Nav1.6 channels, respectively, and the IC50 on skeletal (Nav1.4) and cardiac (Nav1.5) sodium channels is above 3000 nm The lead molecules have the potential for future clinical development as novel therapeutics in the treatment of pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Manejo del Dolor/métodos , Ingeniería de Proteínas , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Células HEK293 , Humanos , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Técnicas de Placa-Clamp , Filogenia , Venenos de Araña/química
10.
J Biol Chem ; 291(23): 12254-70, 2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27129281

RESUMEN

Purinergic homomeric P2X3 and heteromeric P2X2/3 receptors are ligand-gated cation channels activated by ATP. Both receptors are predominantly expressed in nociceptive sensory neurons, and an increase in extracellular ATP concentration under pathological conditions, such as tissue damage or visceral distension, induces channel opening, membrane depolarization, and initiation of pain signaling. Hence, these receptors are considered important therapeutic targets for pain management, and development of selective antagonists is currently progressing. To advance the search for novel analgesics, we have generated a panel of monoclonal antibodies directed against human P2X3 (hP2X3). We have found that these antibodies produce distinct functional effects, depending on the homomeric or heteromeric composition of the target, its kinetic state, and the duration of antibody exposure. The most potent antibody, 12D4, showed an estimated IC50 of 16 nm on hP2X3 after short term exposure (up to 18 min), binding to the inactivated state of the channel to inhibit activity. By contrast, with the same short term application, 12D4 potentiated the slow inactivating current mediated by the heteromeric hP2X2/3 channel. Extending the duration of exposure to ∼20 h resulted in a profound inhibition of both homomeric hP2X3 and heteromeric hP2X2/3 receptors, an effect mediated by efficient antibody-induced internalization of the channel from the plasma membrane. The therapeutic potential of mAb12D4 was assessed in the formalin, complete Freund's adjuvant, and visceral pain models. The efficacy of 12D4 in the visceral hypersensitivity model indicates that antibodies against P2X3 may have therapeutic potential in visceral pain indications.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X2/inmunología , Receptores Purinérgicos P2X3/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Línea Celular Tumoral , Células Cultivadas , Femenino , Adyuvante de Freund , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/prevención & control , Canales Iónicos/química , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones Endogámicos BALB C , Microscopía Confocal , Dolor/inducido químicamente , Dolor/metabolismo , Dolor/prevención & control , Multimerización de Proteína/inmunología , Ratas , Receptores Purinérgicos P2X2/química , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2X3/química , Receptores Purinérgicos P2X3/metabolismo , Ácido Trinitrobencenosulfónico , Dolor Visceral/inducido químicamente , Dolor Visceral/metabolismo , Dolor Visceral/prevención & control
11.
Bioconjug Chem ; 28(4): 1102-1114, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28151644

RESUMEN

Antibody drug conjugates (ADCs) provide an efficacious and relatively safe means by which chemotherapeutic agents can be specifically targeted to cancer cells. In addition to the selection of antibody targets, ADCs offer a modular design that allows selection of ADC characteristics through the choice of linker chemistries, toxins, and conjugation sites. Many studies have indicated that release of toxins bound to antibodies via noncleavable linker chemistries relies on the internalization and intracellular trafficking of the ADC. While this can make noncleavable ADCs more stable in the serum, it can also result in lower efficacy when their respective targets are not internalized efficiently or are recycled back to the cell surface following internalization. Here, we show that a lysosomally targeted ADC against the protein APLP2 mediates cell killing, both in vitro and in vivo, more effectively than an ADC against Trop2, a protein with less efficient lysosomal targeting. We also engineered a bispecific ADC with one arm targeting HER2 for the purpose of directing the ADC to tumors, and the other arm targeting APLP2, whose purpose is to direct the ADC to lysosomes for toxin release. This proof-of-concept bispecific ADC demonstrates that this technology can be used to shift the intracellular trafficking of a constitutively recycled target by directing one arm of the antibody against a lysosomally delivered protein. Our data also show limitations of this approach and potential future directions for development.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inmunoconjugados/farmacología , Lisosomas/metabolismo , Transcitosis , Precursor de Proteína beta-Amiloide/inmunología , Precursor de Proteína beta-Amiloide/uso terapéutico , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Inmunoconjugados/metabolismo , Ratones Desnudos , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/uso terapéutico , Receptor ErbB-2/inmunología , Receptor ErbB-2/uso terapéutico
12.
Transfusion ; 56(2): 421-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26426339

RESUMEN

BACKGROUND: Routine storage of red blood cells (RBCs) results in the progressive accumulation of storage lesions. While the clinical relevance of these lesions is still a matter of debate, alterations to RBC morphology and biochemistry, especially in terms of energy and redox homeostasis, are likely to affect RBC physiology and functionality at a minimum. Identification of oxidative modifications that accumulate on key RBC proteins will help bridge the gap between storage induced alterations and post-transfusion RBC viability. STUDY DESIGN AND METHODS: Five AS-3 units were analyzed during routine storage via one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis-nano-high-performance liquid chromatography coupled online with tandem mass spectrometry and advanced database searches. RESULTS: We identified oxidative modifications to functional residues of hemoglobin (Hb) beta chain, including proximal histidine, cysteine beta 94 (counting initiator methionine in the sequence), and histidine 144. Semiquantitative analysis indicates that up to approximately 20% of total Hb could be targeted by these oxidative modifications that are overlooked by standard proteomics approaches using routine database search conditions. Progressive accumulation of oxidized residues in stored RBCs and selective accumulation in vesicles was observed, further substantiating the hypothesis that vesiculation represents a self-protective mechanism in ageing RBCs. CONCLUSION: Several of the oxidized residues identified play well-established roles in heme iron coordination, 2,3-diphosphoglycerate binding, and nitric oxide homeostasis. Further functional and structural studies are necessary to determine possible associations between these modifications and impaired gas transport homeostasis in RBCs from old units.


Asunto(s)
Conservación de la Sangre , Eritrocitos/química , Hemoglobinas/química , Eritrocitos/citología , Femenino , Humanos , Masculino , Oxidación-Reducción
13.
Bioconjug Chem ; 26(4): 650-9, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25643134

RESUMEN

The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in determining VC-PABC linker stability in mouse plasma, and that the stability of the linker positively correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degradation pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclinical stages of ADC programs. The divergence of linker metabolism in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.


Asunto(s)
Aminobenzoatos/química , Anticuerpos Monoclonales/química , Antineoplásicos/química , Inmunoconjugados/química , Oligopéptidos/química , Neoplasias Pancreáticas/tratamiento farmacológico , Aminobenzoatos/sangre , Aminobenzoatos/farmacocinética , Aminobenzoatos/farmacología , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Carbamatos/química , Catepsina B/química , Catepsina B/metabolismo , Línea Celular Tumoral , Dipéptidos/química , Sistemas de Liberación de Medicamentos/métodos , Estabilidad de Medicamentos , Femenino , Humanos , Inmunoconjugados/sangre , Inmunoconjugados/farmacocinética , Inmunoconjugados/farmacología , Ratones , Ratones Desnudos , Modelos Moleculares , Oligopéptidos/sangre , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Bioconjug Chem ; 25(5): 855-62, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24694238

RESUMEN

Although microbial transglutaminases (mTGs) were initially discovered to offset the cost of producing mammalian transglutaminases for food applications, they have quickly become important tools in research and biotechnology. Today, mTGs are utilized for a large number of applications to conjugate proteins and peptides to small molecules, polymers, surfaces, and DNA, as well as to other proteins. It is important to know how to maximize the advantages of the enzymatic approach and avoid undesired cross-linking. This review focuses on the versatility of transglutaminases in the field of bioconjugation and covers recent developments in utilizing mTG for generating antibody drug conjugates (ADCs) for therapeutic applications.


Asunto(s)
Neoplasias/tratamiento farmacológico , Streptomyces/enzimología , Transglutaminasas/metabolismo , Anticuerpos/química , Anticuerpos/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Biotecnología , ADN/química , ADN/metabolismo , Neoplasias/metabolismo , Polímeros/química , Polímeros/metabolismo , Especificidad por Sustrato , Transglutaminasas/química
15.
Bioconjug Chem ; 25(2): 240-50, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24359082

RESUMEN

Antibody drug conjugates (ADCs) are becoming an important new class of therapeutic agents for the treatment of cancer. ADCs are produced through the linkage of a cytotoxic small molecule (drug) to monoclonal antibodies that target tumor cells. Traditionally, most ADCs rely on chemical conjugation methods that yield heterogeneous mixtures of varying number of drugs attached at different positions. The potential benefits of site-specific drug conjugation in terms of stability, manufacturing, and improved therapeutic index has recently led to the development of several new site-specific conjugation technologies. However, detailed characterization of the degree of site specificity is currently lacking. In this study we utilize mass spectrometry to characterize the extent of site-specificity of an enzyme-based site-specific antibody-drug conjugation technology that we recently developed. We found that, in addition to conjugation of the engineered site, a small amount of aglycosylated antibody present in starting material led to conjugation at position Q295, resulting in approximately 1.3% of off-target conjugation. Based on our detection limits, we show that Q295N mutant eliminates the off-target conjugation yielding highly homogeneous conjugates that are better than 99.8% site-specific. Our study demonstrates the importance of detailed characterization of ADCs and describes methods that can be utilized to characterize not only our enzyme based conjugates, but also ADCs generated by other conjugation technologies.


Asunto(s)
Anticuerpos/química , Preparaciones Farmacéuticas/química , Espectrometría de Masas en Tándem/métodos , Transglutaminasas/química , Cromatografía Liquida
16.
J Biol Chem ; 287(14): 11090-7, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22294692

RESUMEN

Target-mediated clearance and high antigen load can hamper the efficacy and dosage of many antibodies. We show for the first time that the mouse, cynomolgus, and human cross-reactive, antagonistic anti-proprotein convertase substilisin kexin type 9 (PCSK9) antibodies J10 and the affinity-matured and humanized J16 exhibit target-mediated clearance, resulting in dose-dependent pharmacokinetic profiles. These antibodies prevent the degradation of low density lipoprotein receptor, thus lowering serum levels of LDL-cholesterol and potently reducing serum cholesterol in mice, and selectively reduce LDL-cholesterol in cynomolgus monkeys. In order to increase the pharmacokinetic and efficacy of this promising therapeutic for hypercholesterolemia, we engineered pH-sensitive binding to mouse, cynomolgus, and human PCSK9 into J16, resulting in J17. This antibody shows prolonged half-life and increased duration of cholesterol lowering in two species in vivo by binding to endogenous PCSK9 in mice and cynomolgus monkeys, respectively. The proposed mechanism of this pH-sensitive antibody is that it binds with high affinity to PCSK9 in the plasma at pH 7.4, whereas the antibody-antigen complex dissociates at the endosomal pH of 5.5-6.0 in order to escape from target-mediated degradation. Additionally, this enables the antibody to bind to another PCSK9 and therefore increase the antigen-binding cycles. Furthermore, we show that this effect is dependent on the neonatal Fc receptor, which rescues the dissociated antibody in the endosome from degradation. Engineered pH-sensitive antibodies may enable less frequent or lower dosing of antibodies hampered by target-mediated clearance and high antigen load.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticolesterolemiantes/farmacología , Anticolesterolemiantes/farmacocinética , Proproteína Convertasas/inmunología , Ingeniería de Proteínas , Serina Endopeptidasas/inmunología , Animales , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacología , Anticolesterolemiantes/sangre , Anticolesterolemiantes/inmunología , Regiones Determinantes de Complementariedad/química , Semivida , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Macaca fascicularis , Masculino , Ratones , Proproteína Convertasa 9 , Receptores Fc/metabolismo
17.
Bioinformatics ; 28(24): 3282-9, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23093611

RESUMEN

MOTIVATION: Structural characterization of protein interactions is necessary for understanding and modulating biological processes. On one hand, X-ray crystallography or NMR spectroscopy provide atomic resolution structures but the data collection process is typically long and the success rate is low. On the other hand, computational methods for modeling assembly structures from individual components frequently suffer from high false-positive rate, rarely resulting in a unique solution. RESULTS: Here, we present a combined approach that computationally integrates data from a variety of fast and accessible experimental techniques for rapid and accurate structure determination of protein-protein complexes. The integrative method uses atomistic models of two interacting proteins and one or more datasets from five accessible experimental techniques: a small-angle X-ray scattering (SAXS) profile, 2D class average images from negative-stain electron microscopy micrographs (EM), a 3D density map from single-particle negative-stain EM, residue type content of the protein-protein interface from NMR spectroscopy and chemical cross-linking detected by mass spectrometry. The method is tested on a docking benchmark consisting of 176 known complex structures and simulated experimental data. The near-native model is the top scoring one for up to 61% of benchmark cases depending on the included experimental datasets; in comparison to 10% for standard computational docking. We also collected SAXS, 2D class average images and 3D density map from negative-stain EM to model the PCSK9 antigen-J16 Fab antibody complex, followed by validation of the model by a subsequently available X-ray crystallographic structure.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Complejos Multiproteicos/química , Complejo Antígeno-Anticuerpo/química , Cristalografía por Rayos X , Microscopía Electrónica , Dispersión del Ángulo Pequeño , Programas Informáticos , Difracción de Rayos X
18.
MAbs ; 15(1): 2253788, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37675979

RESUMEN

The clinical successes of immune checkpoint blockade have invigorated efforts to activate T cell-mediated responses against cancer. Targeting members of the PVR family, consisting of inhibitory receptors TIGIT, CD96, and CD112R, has been an active area of clinical investigation. In this study, the binding interactions and molecular assemblies of the PVR family receptors and ligands have been assessed in vitro. Furthermore, the anti-TIGIT monoclonal antibody BMS-986207 crystal structure in complex with TIGIT was determined and shows that the antibody binds an epitope that is commonly targeted by the CD155 ligand as well as other clinical anti-TIGIT antibodies. In contrast to previously proposed models, where TIGIT outcompetes costimulatory receptor CD226 for binding to CD155 due to much higher affinity (nanomolar range), our data rather suggest that PVR family members all engage in interactions with relatively weak affinity (micromolar range), including TIGIT and CD155 interactions. Thus, TIGIT and other PVR inhibitory receptors likely elicit immune suppression via increased surface expression rather than inherent differences in affinity. This work provides an improved foundational understanding of the PVR family network and mechanistic insight into therapeutic antibody intervention.


Asunto(s)
Neoplasias , Receptores Inmunológicos , Humanos , Linfocitos T/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Ligandos
19.
J Pharmacol Exp Ther ; 340(2): 228-36, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22019884

RESUMEN

Proprotein convertase substilisin/kexin type 9 (PCSK9) promotes the degradation of low-density lipoprotein (LDL) receptor (LDLR) and thereby increases serum LDL-cholesterol (LDL-C). We have developed a humanized monoclonal antibody that recognizes the LDLR binding domain of PCSK9. This antibody, J16, and its precursor mouse antibody, J10, potently inhibit PCSK9 binding to the LDLR extracellular domain and PCSK9-mediated down-regulation of LDLR in vitro. In vivo, J10 effectively reduces serum cholesterol in C57BL/6 mice fed normal chow. J16 reduces LDL-C in healthy and diet-induced hypercholesterolemic cynomologous monkeys, but does not significantly affect high-density lipoprotein-cholesterol. Furthermore, J16 greatly lowered LDL-C in hypercholesterolemic monkeys treated with the HMG-CoA reductase inhibitor simvastatin. Our data demonstrate that anti-PCSK9 antibody is a promising LDL-C-lowering agent that is both efficacious and potentially additive to current therapies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , LDL-Colesterol/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Primates , Proproteína Convertasas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Dominio Catalítico/inmunología , Línea Celular Tumoral , Colesterol/sangre , Colesterol en la Dieta/administración & dosificación , Colesterol en la Dieta/farmacología , HDL-Colesterol/sangre , HDL-Colesterol/efectos de los fármacos , LDL-Colesterol/sangre , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/métodos , Epítopos/inmunología , Femenino , Fluorobencenos/farmacología , Fluorobencenos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipercolesterolemia/sangre , Hipercolesterolemia/inducido químicamente , Hipercolesterolemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Proproteína Convertasa 9 , Proproteína Convertasas/inmunología , Proproteína Convertasas/farmacología , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Receptores de LDL/metabolismo , Rosuvastatina Cálcica , Serina Endopeptidasas/sangre , Serina Endopeptidasas/inmunología , Serina Endopeptidasas/farmacología , Simvastatina/farmacología , Simvastatina/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
20.
Cancer Immunol Res ; 10(10): 1175-1189, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-35981087

RESUMEN

Novel therapeutic approaches combining immune-checkpoint inhibitors are needed to improve clinical outcomes for patients with cancer. Lymphocyte-activation gene 3 (LAG-3) is an immune-checkpoint molecule that inhibits T-cell activity and antitumor immune responses, acting through an independent mechanism from that of programmed death-1 (PD-1) and cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4). Here, we describe the development and preclinical characterization of relatlimab, a human antibody that binds to human LAG-3 with high affinity and specificity to block the interaction of LAG-3 with the ligands MHC II and fibrinogen-like protein-1, and to reverse LAG-3-mediated inhibition of T-cell function in vitro. Consistent with previous reports, in mouse models, the combined blockade of LAG-3 and PD-1 with surrogate antibodies resulted in enhanced antitumor activity greater than the individual blockade of either receptor. In toxicity studies in cynomolgus monkeys, relatlimab was generally well tolerated when combined with nivolumab. These results are consistent with findings from the RELATIVITY-047 phase II/III trial showing that relatlimab combined with nivolumab is a well-tolerated regimen that demonstrates superior progression-free survival compared with nivolumab monotherapy in patients with unresectable or metastatic melanoma.


Asunto(s)
Melanoma , Nivolumab , Animales , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4 , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Fibrinógeno/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico , Macaca fascicularis , Melanoma/patología , Ratones , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA