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










Base de dados
Intervalo de ano de publicação
1.
ACS Cent Sci ; 9(6): 1241-1251, 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37396859

RESUMO

Molecularly targeted radionuclide therapies (TRTs) struggle with balancing efficacy and safety, as current strategies to increase tumor absorption often alter drug pharmacokinetics to prolong circulation and normal tissue irradiation. Here we report the first covalent protein TRT, which, through reacting with the target irreversibly, increases radioactive dose to the tumor without altering the drug's pharmacokinetic profile or normal tissue biodistribution. Through genetic code expansion, we engineered a latent bioreactive amino acid into a nanobody, which binds to its target protein and forms a covalent linkage via the proximity-enabled reactivity, cross-linking the target irreversibly in vitro, on cancer cells, and on tumors in vivo. The radiolabeled covalent nanobody markedly increases radioisotope levels in tumors and extends tumor residence time while maintaining rapid systemic clearance. Furthermore, the covalent nanobody conjugated to the α-emitter actinium-225 inhibits tumor growth more effectively than the noncovalent nanobody without causing tissue toxicity. Shifting the protein-based TRT from noncovalent to covalent mode, this chemical strategy improves tumor responses to TRTs and can be readily scaled to diverse protein radiopharmaceuticals engaging broad tumor targets.

2.
Chem Sci ; 14(29): 7913-7921, 2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37502323

RESUMO

The proximity-enabled sulfur(vi) fluoride exchange (SuFEx) reaction generates specific covalent linkages between proteins in cells and in vivo, which opens innovative avenues for studying elusive protein-protein interactions and developing potent covalent protein drugs. To exploit the power and expand the applications of covalent proteins, covalent linkage formation between proteins is the critical step, for which fundamental kinetic and essential properties remain unexplored. Herein, we systematically studied SuFEx kinetics in different proteins and conditions. In contrast to in small molecules, SuFEx in interacting proteins conformed with a two-step mechanism involving noncovalent binding, followed by covalent bond formation, exhibiting nonlinear rate dependence on protein concentration. The protein SuFEx rate consistently changed with protein binding affinity as well as chemical reactivity of the functional group and was impacted by target residue identity and solution pH. In addition, kinetic analyses of nanobody SR4 binding with SARS-CoV-2 spike protein revealed that viral target mutations did not abolish covalent binding but decreased the SuFEx rate with affinity decrease. Moreover, off-target cross-linking of a SuFEx-capable nanobody in human serum was not detected, and the SuFEx-generated protein linkage was stable at cellular acidic pHs, suggesting SuFEx suitability for in vivo usage. These results advanced our understanding of SuFEx reactivity and kinetics in proteins, which is invaluable for ongoing exploration of SuFEx-enabled covalent proteins for basic biological research and creative biotherapeutics.

3.
Chem Commun (Camb) ; 58(48): 6861-6864, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35621237

RESUMO

The introduction of new covalent bonds into proteins is affording novel avenues for protein research and applications, yet it remains difficult to generate covalent linkages at all possible sites and across diverse protein classes. Herein, we genetically encoded meta-fluorosulfate-L-tyrosine (mFSY) to selectively react with lysine, tyrosine, and histidine via proximity-enabled SuFEx reaction. mFSY was able to target residues that were elusive for previous unnatural amino acids, and permitted engineering of various proteins including affibody, nanobody, and Fab into covalent binders that irreversibly cross-linked EGFR and HER2. mFSY is thus valuable for developing covalent proteins for biological research, synthetic biology, and biotherapeutics.


Assuntos
Proteínas , Tirosina , Aminoácidos/química , Histidina/química , Lisina/química , Proteínas/química , Tirosina/química
4.
J Am Chem Soc ; 143(27): 10341-10351, 2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34213894

RESUMO

Genetically introducing novel chemical bonds into proteins provides innovative avenues for biochemical research, protein engineering, and biotherapeutic applications. Recently, latent bioreactive unnatural amino acids (Uaas) have been incorporated into proteins to covalently target natural residues through proximity-enabled reactivity. Aryl fluorosulfate is particularly attractive due to its exceptional biocompatibility and multitargeting capability via sulfur(VI) fluoride exchange (SuFEx) reaction. Thus far, fluorosulfate-l-tyrosine (FSY) is the only aryl fluorosulfate-containing Uaa that has been genetically encoded. FSY has a relatively rigid and short side chain, which restricts the diversity of proteins targetable and the scope of applications. Here we designed and genetically encoded a new latent bioreactive Uaa, fluorosulfonyloxybenzoyl-l-lysine (FSK), in E. coli and mammalian cells. Due to its long and flexible aryl fluorosulfate-containing side chain, FSK was particularly useful in covalently linking protein sites that are unreachable with FSY, both intra- and intermolecularly, in vitro and in live cells. In addition, we created covalent nanobodies that irreversibly bound to epidermal growth factor receptors (EGFR) on cells, with FSK and FSY targeting distinct positions on EGFR to counter potential mutational resistance. Moreover, we established the use of FSK and FSY for genetically encoded chemical cross-linking to capture elusive enzyme-substrate interactions in live cells, allowing us to target residues aside from Cys and to cross-link at the binding periphery. FSK complements FSY to expand target diversity and versatility. Together, they provide a powerful, genetically encoded, latent bioreactive SuFEx system for creating covalent bonds in diverse proteins in vitro and in vivo, which will be widely useful for biological research and applications.


Assuntos
Receptores ErbB/metabolismo , Engenharia de Proteínas/métodos , Proteínas/química , Animais , Proteínas de Bactérias , Reagentes de Ligações Cruzadas , Receptores ErbB/química , Escherichia coli , Proteínas de Fluorescência Verde , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica
5.
Bioorg Med Chem ; 29: 115896, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33285408

RESUMO

Drugs with a covalent mechanism of action benefit from enhanced potency, selectivity, and in vivo efficacy. Historically, the only covalent drugs on the market have been covalent small molecules. However, many proteins and protein-protein interactions cannot be targeted by small molecules due to their lack of small molecule binding pockets, and are thus deemed "undruggable." In order to drug the undruggable, peptide and protein therapeutics that can better bind to flat protein surfaces have been developed. Until recently, peptide and protein therapeutics have had noncovalent mechanisms of action. The recent advancement of unnatural amino acid chemistry, along with the development of better and more specific electrophilic warheads, has allowed for the application of covalent mechanisms to peptide and protein drugs. Covalent peptide and protein therapeutics have the potential to benefit from the same advantages that covalent small molecules have over their noncovalent counterparts. Here we provide a brief overview of the chemistry that makes this advancement possible, as well as examples of covalent peptides and the first covalent protein drug. These examples successfully crosslink their target proteins and have beneficial therapeutic effects.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Preparações Farmacêuticas/química , Proteínas/química , Proteínas/farmacologia , Acrilamidas/química , Sequência de Aminoácidos , Animais , Química Click , Descoberta de Drogas , Humanos , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Ácidos Sulfínicos/química , Sulfonamidas/química
6.
J Am Chem Soc ; 142(40): 17057-17068, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32915556

RESUMO

Site-specific modification of proteins with functional molecules provides powerful tools for researching and engineering proteins. Here we report a new chemical conjugation method which photocages highly reactive but chemically selective moieties, enabling the use of protein-inert amines for selective protein modification. New amino acids FnbY and FmnbY, bearing photocaged quinone methides (QMs), were genetically incorporated into proteins. Upon light activation, they generated highly reactive QM, which rapidly reacted with amine derivatives. This method features a rare combination of desired properties including fast kinetics, small and stable linkage, compatibility with low temperature, photocontrollability, and widely available reagents. Moreover, labeling via FnbY occurs on the ß-carbon, affording the shortest linkage to protein backbone which is essential for advanced studies involving orientation and distance. We installed various functionalities onto proteins and attached a spin label as close as possible to the protein backbone, achieving high resolution in double electron-electron paramagnetic resonance distance measurements.


Assuntos
Aminas/química , Indolquinonas/química , Proteínas/química , Coloração e Rotulagem/métodos , Aminoácidos/química , Sítios de Ligação , Espectroscopia de Ressonância de Spin Eletrônica , Cinética , Processos Fotoquímicos , Conformação Proteica , Processamento de Proteína Pós-Traducional , Solventes/química , Marcadores de Spin , Compostos de Sulfidrila/química , Temperatura
7.
Cell ; 182(1): 85-97.e16, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32579975

RESUMO

Small molecule covalent drugs provide desirable therapeutic properties over noncovalent ones for treating challenging diseases. The potential of covalent protein drugs, however, remains unexplored due to protein's inability to bind targets covalently. We report a proximity-enabled reactive therapeutics (PERx) approach to generate covalent protein drugs. Through genetic code expansion, a latent bioreactive amino acid fluorosulfate-L-tyrosine (FSY) was incorporated into human programmed cell death protein-1 (PD-1). Only when PD-1 interacts with PD-L1 did the FSY react with a proximal histidine of PD-L1 selectively, enabling irreversible binding of PD-1 to only PD-L1 in vitro and in vivo. When administrated in immune-humanized mice, the covalent PD-1(FSY) exhibited strikingly more potent antitumor effect over the noncovalent wild-type PD-1, attaining therapeutic efficacy equivalent or superior to anti-PD-L1 antibody. PERx should provide a general platform technology for converting various interacting proteins into covalent binders, achieving specific covalent protein targeting for biological studies and therapeutic capability unattainable with conventional noncovalent protein drugs.


Assuntos
Preparações Farmacêuticas/metabolismo , Proteínas/uso terapêutico , Sequência de Aminoácidos , Animais , Antineoplásicos/metabolismo , Antígeno B7-H1/química , Antígeno B7-H1/metabolismo , Membrana Celular/metabolismo , Proliferação de Células , Células Dendríticas/metabolismo , Humanos , Cinética , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Monócitos/metabolismo , Fenótipo , Proteínas/química , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Am Chem Soc ; 139(1): 255-261, 2017 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-27935689

RESUMO

We describe an unprecedented DNA-catalyzed DNA cleavage process in which a radical-based reaction pathway cleanly results in excision of most atoms of a specific guanosine nucleoside. Two new deoxyribozymes (DNA enzymes) were identified by in vitro selection from N40 or N100 random pools initially seeking amide bond hydrolysis, although they both cleave simple single-stranded DNA oligonucleotides. Each deoxyribozyme generates both superoxide (O2-• or HOO•) and hydrogen peroxide (H2O2) and leads to the same set of products (3'-phosphoglycolate, 5'-phosphate, and base propenal) as formed by the natural product bleomycin, with product assignments by mass spectrometry and colorimetric assay. We infer the same mechanistic pathway, involving formation of the C4' radical of the guanosine nucleoside that is subsequently excised. Consistent with a radical pathway, glutathione fully suppresses catalysis. Conversely, adding either superoxide or H2O2 from the outset strongly enhances catalysis. The mechanism of generation and involvement of superoxide and H2O2 by the deoxyribozymes is not yet defined. The deoxyribozymes do not require redox-active metal ions and function with a combination of Zn2+ and Mg2+, although including Mn2+ increases the activity, and Mn2+ alone also supports catalysis. In contrast to all of these observations, unrelated DNA-catalyzed radical DNA cleavage reactions require redox-active metals and lead to mixtures of products. This study reports an intriguing example of a well-defined, DNA-catalyzed, radical reaction process that cleaves single-stranded DNA and requires only redox-inactive metal ions.


Assuntos
Clivagem do DNA , DNA Catalítico/metabolismo , Peróxido de Hidrogênio/metabolismo , Superóxidos/metabolismo , DNA Catalítico/química , Peróxido de Hidrogênio/química , Espectrometria de Massas , Superóxidos/química
9.
J Am Chem Soc ; 138(7): 2106-9, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26854515

RESUMO

DNA catalysts (deoxyribozymes) for a variety of reactions have been identified by in vitro selection. However, for certain reactions this identification has not been achieved. One important example is DNA-catalyzed amide hydrolysis, for which a previous selection experiment instead led to DNA-catalyzed DNA phosphodiester hydrolysis. Subsequent efforts in which the selection strategy deliberately avoided phosphodiester hydrolysis led to DNA-catalyzed ester and aromatic amide hydrolysis, but aliphatic amide hydrolysis has been elusive. In the present study, we show that including modified nucleotides that bear protein-like functional groups (any one of primary amino, carboxyl, or primary hydroxyl) enables identification of amide-hydrolyzing deoxyribozymes. In one case, the same deoxyribozyme sequence without the modifications still retains substantial catalytic activity. Overall, these findings establish the utility of introducing protein-like functional groups into deoxyribozymes for identifying new catalytic function. The results also suggest the longer-term feasibility of deoxyribozymes as artificial proteases.


Assuntos
Amidas/química , DNA/química , Catálise , Hidrólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...