Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Cell Rep Phys Sci ; 4(2): 101258, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36741337

RESUMO

The sensitivity of therapeutic antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral "escape" mutations has inspired efforts to develop treatment strategies that are still effective in the face of rapidly mutating viral surface proteins. Here, we demonstrate a chemical strategy that enforces viral opsonization by natural serum antibodies. This strategy uses chimeric molecules that we call covalent viral opsonizers, which covalently label viral surface proteins, with synthetic antibody-binding ligands. As a proof of concept, we develop covalent viral opsonizers that covalently label the spike protein on SARS-CoV-2 using a "mutation-proof" small-molecule-binding ligand for anti-dinitrophenyl serum antibodies. In model assays, we observe that covalent viral opsonizers can rapidly and selectively covalently label the receptor-binding domain of both native and mutant spike proteins, leading to antibody opsonization. Opsonization mediated by this strategy is able to efficiently block the key binding domain interactions, in contrast to non-covalent analogs. We also show that covalent viral opsonizers enact targeted anti-viral phagocytotic immune function. This strategy has potential general utility for the rapid deployment of anti-viral synthetic immunotherapeutics at the onset of a new pandemic to reinforce vaccination and antibody engineering efforts.

3.
Biochemistry ; 60(19): 1447-1458, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33930269

RESUMO

Antibody recruiting molecules (ARMs) represent an important class of "proximity-inducing" chemical tools with therapeutic potential. ARMs function by simultaneously binding to a hapten-specific serum antibody (Ab) (e.g., anti-dinitrophenyl (DNP)) and a cancer cell surface protein, enforcing their proximity. ARM anticancer efficacy depends on the formation of ARM:Ab complexes on the cancer cell surface, which activate immune cell recognition and elimination of the cancer cell. Problematically, ARM function in human patients may be limited by conditions that drive the dissociation of ARM:Ab complexes, namely, intrinsically low binding affinity and/or low concentrations of anti-hapten antibodies in human serum. To address this potential limitation, we previously developed a covalent ARM (cARM) chemical tool that eliminates the ARM:antibody equilibrium through a covalent linkage. In the current study, we set out to determine to what extent maximizing the stability of ARM:antibody complexes via cARMs enhances target immune recognition. We observe cARMs significantly increase target immune recognition relative to ARMs across a range of therapeutically relevant antibody concentrations. These results demonstrate that ARM therapeutic function can be dramatically enhanced by increasing the kinetic stability of ARM:antibody complexes localized on cancer cells. Our findings suggest that a) high titres/concentrations of target antibody in human serum are not neccessary and b) saturative antibody recruitment to cancer cells not sufficient, to achieve maximal ARM therapeutic function.


Assuntos
Anticorpos/química , Imunoterapia/métodos , Neoplasias/imunologia , Anticorpos/uso terapêutico , Formação de Anticorpos , Haptenos/química , Haptenos/imunologia , Humanos , Imunoglobulinas , Cinética , Neoplasias/tratamento farmacológico , Ligação Proteica/imunologia
4.
Curr Protoc Chem Biol ; 12(4): e88, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33326159

RESUMO

The emergence of covalent inhibitors and chemoproteomic probes in translational chemical biology research requires the development of robust biophysical and analytical methods to characterize their complex interactions with target biomolecules. Importantly, these methods must efficiently assess target selectivity and accurately discern noncovalent binding from the formation of resultant covalent adducts. One recently reported covalent chemical tool used in tumor immune oncology, covalent immune recruiters (CIRs), increases the proximity of immune cells and cancer cells, promoting immune recognition and response. Herein we describe biolayer interferometry (BLI) biosensor, flow cytometry, and solution fluorescence-based assay approaches to characterize CIR:antibody binding and CIR-antibody covalent-labeling kinetics. BLI technology, akin to surface plasmon resonance, provides the unique opportunity to investigate molecular binding and labeling kinetics both on a solid surface (Basic Protocol 1) and in solution (Alternate Protocol 1). Here, recruitment of mass-containing proteins to the BLI probe via CIR is measured with high sensitivity and is used as a readout of CIR labeling activity. Further, CIR technology is used to label antibodies with a fluorescent handle. In this system, labeling is monitored via SDS-PAGE with a fluorescence gel imager, where increased fluorescence intensity of a sample reflects increased labeling (Basic Protocol 2). Analysis of CIR:antibody target-specific immune activation is demonstrated with a flow cytometry-based antibody-dependent cellular phagocytosis (ADCP) assay (Basic Protocol 3). This ADCP protocol may be further used to discern CIR:antibody binding from covalent adduct formation (Alternate Protocol 3). For the protocols described, each method may be used to analyze characteristics of any covalent-tagging or antibody-recruiting small molecule or protein-based technology. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Determining "on-probe" reaction kinetics of CIR1/CIR4 via biolayer interferometry with Octet RED96 Alternate Protocol 1: Determining "in-solution" reaction kinetics of prostate-specific membrane antigen targeting CIR (CIR3) via biolayer interferometry with Octet RED96 Basic Protocol 2: Reaction kinetics of covalently labeled antibodies via fluorescence SDS-PAGE Basic Protocol 3: Small molecule-directed antibody-dependent cellular phagocytosis on live human cells measured via flow cytometry Alternate Protocol 2: Kinetic analysis of CIR3:antibody labeling via antibody-dependent cellular phagocytosis on flow cytometry Support Protocol 1: Activation of U937 monocytes with interferon γ Support Protocol 2: Labeling streptavidin beads with biotinylated prostate-specific membrane antigen receptor.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas Repressoras/imunologia , Sítios de Ligação , Técnicas Biossensoriais , Citometria de Fluxo , Humanos , Cinética , Imagem Óptica
5.
ACS Chem Biol ; 15(4): 1089-1095, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32100991

RESUMO

Unprecedented progress made in the treatment of cancer using the body's own immune system has encouraged the development of synthetic molecule based immunotherapeutics. An emerging class of these compounds, called Antibody Recruiting Molecules (ARMs) or Antibody Engagers (AEs), functions by reversibly binding antibodies naturally present in human serum and recruiting these to cancer cells. The recruited antibodies then engage immune cells to form quaternary complexes that drive cancer erradication. Despite their promise, the requirement to form quaternary complexes governed by multiple equilibria complicates an understanding of their in vivo efficacy. Particularly problematic are low endogenous serum antibody concentrations and rapid clearance of AEs from circulation. Here we describe a new class of trifunctional chemical tools we call covalent immune recruiters (CIRs). CIRs covalently label specific serum antibodies in a selective manner with a target protein binding ligand. CIRs thereby exert well-defined control over antibody recruitment and simplify quaternary complex equilibium, enabling probing of the resultant effects on immune recognition. We demonstrate CIRs can selectively covalently label anti-DNP IgG, a natural human antibody, directly in human serum to drive efficient immune cell recognition of targets. We expect CIRs will be useful tools to probe how quaternary complex stability impacts the immune recognition of cancer in vivo, revealing new design principles to guide the development of future AEs.


Assuntos
Imidazóis/química , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Sondas Moleculares/química , Receptores Imunológicos/metabolismo , Animais , Antígenos de Superfície/metabolismo , Biotina/análogos & derivados , Biotina/química , Biotina/metabolismo , Glutamato Carboxipeptidase II/metabolismo , Células HEK293 , Humanos , Imunoglobulina G/sangue , Camundongos , Simulação de Acoplamento Molecular , Sondas Moleculares/metabolismo , Fagocitose/efeitos dos fármacos , Ligação Proteica , Estreptavidina/metabolismo , Ureia/análogos & derivados , Ureia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...