Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 725
Filtrar
1.
Acta Crystallogr F Struct Biol Commun ; 80(Pt 7): 154-163, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38958188

RESUMO

The third complementary-determining regions of the heavy-chain (CDR3H) variable regions (VH) of some cattle antibodies are highly extended, consisting of 48 or more residues. These `ultralong' CDR3Hs form ß-ribbon stalks that protrude from the surface of the antibody with a disulfide cross-linked knob region at their apex that dominates antigen interactions over the other CDR loops. The structure of the Fab fragment of a naturally paired bovine ultralong antibody (D08), identified by single B-cell sequencing, has been determined to 1.6 Šresolution. By swapping the D08 native light chain with that of an unrelated antigen-unknown ultralong antibody, it is shown that interactions between the CDR3s of the variable domains potentially affect the fine positioning of the ultralong CDR3H; however, comparison with other crystallographic structures shows that crystalline packing is also a major contributor. It is concluded that, on balance, the exact positioning of ultralong CDR3H loops is most likely to be due to the constraints of crystal packing.


Assuntos
Regiões Determinantes de Complementaridade , Fragmentos Fab das Imunoglobulinas , Cadeias Pesadas de Imunoglobulinas , Cadeias Leves de Imunoglobulina , Modelos Moleculares , Animais , Bovinos , Cadeias Pesadas de Imunoglobulinas/química , Cristalografia por Raios X , Cadeias Leves de Imunoglobulina/química , Cadeias Leves de Imunoglobulina/genética , Regiões Determinantes de Complementaridade/química , Fragmentos Fab das Imunoglobulinas/química , Sequência de Aminoácidos , Conformação Proteica
2.
Elife ; 122024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38921957

RESUMO

Accurate prediction of the structurally diverse complementarity determining region heavy chain 3 (CDR-H3) loop structure remains a primary and long-standing challenge for antibody modeling. Here, we present the H3-OPT toolkit for predicting the 3D structures of monoclonal antibodies and nanobodies. H3-OPT combines the strengths of AlphaFold2 with a pre-trained protein language model and provides a 2.24 Å average RMSDCα between predicted and experimentally determined CDR-H3 loops, thus outperforming other current computational methods in our non-redundant high-quality dataset. The model was validated by experimentally solving three structures of anti-VEGF nanobodies predicted by H3-OPT. We examined the potential applications of H3-OPT through analyzing antibody surface properties and antibody-antigen interactions. This structural prediction tool can be used to optimize antibody-antigen binding and engineer therapeutic antibodies with biophysical properties for specialized drug administration route.


Assuntos
Regiões Determinantes de Complementaridade , Aprendizado Profundo , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/imunologia , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Modelos Moleculares , Conformação Proteica , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia , Humanos
3.
PLoS Comput Biol ; 20(6): e1012239, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38913733

RESUMO

As of now, more than 60 years have passed since the first determination of protein structures through crystallography, and a significant portion of protein structures can be predicted by computers. This is due to the groundbreaking enhancement in protein structure prediction achieved through neural network training utilizing extensive sequence and structure data. However, substantial challenges persist in structure prediction due to limited data availability, with antibody structure prediction standing as one such challenge. In this paper, we propose a novel neural network architecture that effectively enables structure prediction by reflecting the inherent combinatorial nature involved in protein structure formation. The core idea of this neural network architecture is not solely to track and generate a single structure but rather to form a community of multiple structures and pursue accurate structure prediction by exchanging information among community members. Applying this concept to antibody CDR H3 loop structure prediction resulted in improved structure sampling. Such an approach could be applied in the structural and functional studies of proteins, particularly in exploring various physiological processes mediated by loops. Moreover, it holds potential in addressing various other types of combinatorial structure prediction and design problems.


Assuntos
Biologia Computacional , Aprendizado Profundo , Modelos Moleculares , Conformação Proteica , Biologia Computacional/métodos , Regiões Determinantes de Complementaridade/química , Redes Neurais de Computação , Anticorpos/química , Bases de Dados de Proteínas , Humanos , Algoritmos
4.
Arch Biochem Biophys ; 758: 110068, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38909835

RESUMO

Affinity maturation increases antigen-binding affinity and specificity of antibodies by somatic hypermutation. Various monoclonal antibodies against (4-hydroxy-3-nitrophenyl)acetyl (NP) were obtained during affinity maturation. Among them, highly matured anti-NP antibodies, such as E11 and E3, possess Cys96H and Cys100H in the complementarity-determining region 3 of the heavy chain, which would form a disulfide bond. In this study, we evaluated the effects of disulfide bonds on antigen binding by generating single-chain Fv (scFv) antibodies of E11 and its mutants, E11_C96KH/C100EH and E11_C96KH/C100QH, and determined their antigen-binding thermodynamics and kinetics. The binding affinities of the Cys mutants were lower than that of E11 scFv, indicating that the disulfide bond contributed to antigen binding, especially for stable complex formation. This was also supported by the decreased affinity of E11 scFv in the presence of a reducing agent. The crystal structures of NP-free and NP-bound E11 scFvs were determined at high resolution, showing the existence of a disulfide bond between Cys96H and Cys100H, and the antigen recognition mechanism, which could be compared with those of other anti-NP antibodies, such as germline-type N1G9 and matured-type C6, as reported previously. These structures could explain the molecular basis of changes in antigen-binding affinity and thermal stability in the absence or presence of antigens. Small-angle X-ray scattering further showed a local conformational change in E11 scFv upon antigen binding in solution.


Assuntos
Afinidade de Anticorpos , Regiões Determinantes de Complementaridade , Dissulfetos , Anticorpos de Cadeia Única , Dissulfetos/química , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/genética , Regiões Determinantes de Complementaridade/química , Humanos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Animais , Termodinâmica , Cinética , Cristalografia por Raios X , Modelos Moleculares
5.
ACS Chem Biol ; 19(7): 1583-1592, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38916527

RESUMO

The binding affinity of antibodies to specific antigens stems from a remarkably broad repertoire of hypervariable loops known as complementarity-determining regions (CDRs). While recognizing the pivotal role of the heavy-chain 3 CDRs (CDR-H3s) in maximizing antibody-antigen affinity and specificity, the key structural determinants responsible for their adaptability to diverse loop sequences, lengths, and noncanonical structures are hitherto unknown. To address this question, we achieved a de novo synthesis of bulged CDR-H3 mimics excised from their full antibody context. CD and NMR data revealed that these stable standalone ß-hairpin scaffolds are well-folded and retain many of the native bulge CDR-H3 features in water. In particular, the tryptophan residue, highly conserved across CDR-H3 sequences, was found to extend the kinked base of these ß-bulges through a combination of stabilizing intramolecular hydrogen bond and CH/π interaction. The structural ensemble consistent with our NMR observations exposed the dynamic nature of residues at the base of the loop, suggesting that ß-bulges act as molecular hinges connecting the rigid stem to the more flexible loops of CDR-H3s. We anticipate that this deeper structural understanding of CDR-H3s will lay the foundation to inform the design of antibody drugs broadly and engineer novel CDR-H3 peptide scaffolds as therapeutics.


Assuntos
Regiões Determinantes de Complementaridade , Regiões Determinantes de Complementaridade/química , Modelos Moleculares , Cadeias Pesadas de Imunoglobulinas/química , Humanos , Sequência de Aminoácidos
6.
Int J Mol Sci ; 25(10)2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38791221

RESUMO

Snakebite accidents, neglected tropical diseases per the WHO, pose a significant public health threat due to their severity and frequency. Envenomation by Bothrops genus snakes leads to severe manifestations due to proteolytic enzymes. While the antibothropic serum produced by the Butantan Institute saves lives, its efficacy is limited as it fails to neutralize certain serine proteases. Hence, developing new-generation antivenoms, like monoclonal antibodies, is crucial. This study aimed to explore the inhibitory potential of synthetic peptides homologous to the CDR3 regions of a monoclonal antibody targeting a snake venom thrombin-like enzyme (SVTLE) from B. atrox venom. Five synthetic peptides were studied, all stable against hydrolysis by venoms and serine proteases. Impressively, four peptides demonstrated uncompetitive SVTLE inhibition, with Ki values ranging from 10-6 to 10-7 M. These findings underscore the potential of short peptides homologous to CDR3 regions in blocking snake venom toxins, suggesting their promise as the basis for new-generation antivenoms. Thus, this study offers potential advancements in combatting snakebites, addressing a critical public health challenge in tropical and subtropical regions.


Assuntos
Anticorpos Monoclonais , Bothrops , Peptídeos , Serina Proteases , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Serina Proteases/química , Serina Proteases/metabolismo , Antivenenos/química , Antivenenos/imunologia , Antivenenos/farmacologia , Regiões Determinantes de Complementaridade/química , Venenos de Crotalídeos/antagonistas & inibidores , Venenos de Crotalídeos/imunologia , Venenos de Crotalídeos/enzimologia , Venenos de Crotalídeos/química , Sequência de Aminoácidos , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia
7.
Nat Commun ; 15(1): 4301, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773089

RESUMO

The vaccine elicitation of HIV tier-2-neutralization antibodies has been a challenge. Here, we report the isolation and characterization of a CD4-binding site (CD4bs) specific monoclonal antibody, HmAb64, from a human volunteer immunized with a polyvalent DNA prime-protein boost HIV vaccine. HmAb64 is derived from heavy chain variable germline gene IGHV1-18 and light chain germline gene IGKV1-39. It has a third heavy chain complementarity-determining region (CDR H3) of 15 amino acids. On a cross-clade panel of 208 HIV-1 pseudo-virus strains, HmAb64 neutralized 20 (10%), including tier-2 strains from clades B, BC, C, and G. The cryo-EM structure of the antigen-binding fragment of HmAb64 in complex with a CNE40 SOSIP trimer revealed details of its recognition; HmAb64 uses both heavy and light CDR3s to recognize the CD4-binding loop, a critical component of the CD4bs. This study demonstrates that a gp120-based vaccine can elicit antibodies capable of tier 2-HIV neutralization.


Assuntos
Vacinas contra a AIDS , Anticorpos Neutralizantes , Antígenos CD4 , Anticorpos Anti-HIV , HIV-1 , Humanos , Vacinas contra a AIDS/imunologia , HIV-1/imunologia , Anticorpos Anti-HIV/imunologia , Anticorpos Neutralizantes/imunologia , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Vacinas de DNA/imunologia , Anticorpos Monoclonais/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/imunologia , Infecções por HIV/virologia , Microscopia Crioeletrônica , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp120 do Envelope de HIV/química , Sítios de Ligação , Regiões Determinantes de Complementaridade/imunologia , Regiões Determinantes de Complementaridade/química
8.
Protein J ; 43(3): 405-424, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38724751

RESUMO

As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.


Assuntos
Aminoácidos , Regiões Determinantes de Complementaridade , Engenharia de Proteínas , Aminoácidos/química , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Humanos , Engenharia de Proteínas/métodos , Anticorpos/química , Anticorpos/imunologia , Anticorpos/metabolismo , Afinidade de Anticorpos , Animais
9.
Nat Commun ; 15(1): 3974, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730230

RESUMO

Antibodies are engineerable quantities in medicine. Learning antibody molecular recognition would enable the in silico design of high affinity binders against nearly any proteinaceous surface. Yet, publicly available experiment antibody sequence-binding datasets may not contain the mutagenic, antigenic, or antibody sequence diversity necessary for deep learning approaches to capture molecular recognition. In part, this is because limited experimental platforms exist for assessing quantitative and simultaneous sequence-function relationships for multiple antibodies. Here we present MAGMA-seq, an integrated technology that combines multiple antigens and multiple antibodies and determines quantitative biophysical parameters using deep sequencing. We demonstrate MAGMA-seq on two pooled libraries comprising mutants of nine different human antibodies spanning light chain gene usage, CDR H3 length, and antigenic targets. We demonstrate the comprehensive mapping of potential antibody development pathways, sequence-binding relationships for multiple antibodies simultaneously, and identification of paratope sequence determinants for binding recognition for broadly neutralizing antibodies (bnAbs). MAGMA-seq enables rapid and scalable antibody engineering of multiple lead candidates because it can measure binding for mutants of many given parental antibodies in a single experiment.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Fragmentos Fab das Imunoglobulinas , Mutação , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Engenharia de Proteínas/métodos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/química , Afinidade de Anticorpos , Antígenos/imunologia , Antígenos/genética
10.
Biomolecules ; 14(4)2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38672440

RESUMO

This study assessed the suitability of the complementarity-determining region 2 (CDR2) of the nanobody (Nb) as a template for the derivation of nanobody-derived peptides (NDPs) targeting active-state ß2-adrenergic receptor (ß2AR) conformation. Sequences of conformationally selective Nbs favoring the agonist-occupied ß2AR were initially analyzed by the informational spectrum method (ISM). The derived NDPs in complex with ß2AR were subjected to protein-peptide docking, molecular dynamics (MD) simulations, and metadynamics-based free-energy binding calculations. Computational analyses identified a 25-amino-acid-long CDR2-NDP of Nb71, designated P4, which exhibited the following binding free-energy for the formation of the ß2AR:P4 complex (ΔG = -6.8 ± 0.8 kcal/mol or a Ki = 16.5 µM at 310 K) and mapped the ß2AR:P4 amino acid interaction network. In vitro characterization showed that P4 (i) can cross the plasma membrane, (ii) reduces the maximum isoproterenol-induced cAMP level by approximately 40% and the isoproterenol potency by up to 20-fold at micromolar concentration, (iii) has a very low affinity to interact with unstimulated ß2AR in the cAMP assay, and (iv) cannot reduce the efficacy and potency of the isoproterenol-mediated ß2AR/ß-arrestin-2 interaction in the BRET2-based recruitment assay. In summary, the CDR2-NDP, P4, binds preferentially to agonist-activated ß2AR and disrupts Gαs-mediated signaling.


Assuntos
Peptídeos , Receptores Adrenérgicos beta 2 , Anticorpos de Domínio Único , Humanos , Sequência de Aminoácidos , Regiões Determinantes de Complementaridade/química , AMP Cíclico/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/química , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia , Anticorpos de Domínio Único/metabolismo
11.
Bioinformatics ; 40(3)2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38449295

RESUMO

MOTIVATION: Antibodies are proteins that the immune system produces in response to foreign pathogens. Designing antibodies that specifically bind to antigens is a key step in developing antibody therapeutics. The complementarity determining regions (CDRs) of the antibody are mainly responsible for binding to the target antigen, and therefore must be designed to recognize the antigen. RESULTS: We develop an antibody design model, AbFlex, that exhibits state-of-the-art performance in terms of structure prediction accuracy and amino acid recovery rate. Furthermore, >38% of newly designed antibody models are estimated to have better binding energies for their antigens than wild types. The effectiveness of the model is attributed to two different strategies that are developed to overcome the difficulty associated with the scarcity of antibody-antigen complex structure data. One strategy is to use an equivariant graph neural network model that is more data-efficient. More importantly, a new data augmentation strategy based on the flexible definition of CDRs significantly increases the performance of the CDR prediction model. AVAILABILITY AND IMPLEMENTATION: The source code and implementation are available at https://github.com/wsjeon92/AbFlex.


Assuntos
Complexo Antígeno-Anticorpo , Regiões Determinantes de Complementaridade , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/metabolismo , Sequência de Aminoácidos , Modelos Moleculares , Complexo Antígeno-Anticorpo/química , Antígenos
12.
MAbs ; 16(1): 2322533, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38477253

RESUMO

Antibodies have increasingly been developed as drugs with over 100 now licensed in the US or EU. During development, it is often necessary to increase or reduce the affinity of an antibody and rational attempts to do so rely on having a structure of the antibody-antigen complex often obtained by modeling. The antigen-binding site consists primarily of six loops known as complementarity-determining regions (CDRs), and an open question has been whether these loops change their conformation when they bind to an antigen. Existing surveys of antibody-antigen complex structures have only examined CDR conformational change in case studies or small-scale surveys. With an increasing number of antibodies where both free and complexed structures have been deposited in the Protein Data Bank, a large-scale survey of CDR conformational change during binding is now possible. To this end, we built a dataset, AbAgDb, that currently includes 177 antibodies with high-quality CDRs, each of which has at least one bound and one unbound structure. We analyzed the conformational change of the Cα backbone of each CDR upon binding and found that, in most cases, the CDRs (other than CDR-H3) show minimal movement, while 70.6% and 87% of CDR-H3s showed global Cα RMSD ≤ 1.0Å and ≤ 2.0Å, respectively. We also compared bound CDR conformations with the conformational space of unbound CDRs and found most of the bound conformations are included in the unbound conformational space. In future, our results will contribute to developing insights into antibodies and new methods for modeling and docking.


Assuntos
Antígenos , Regiões Determinantes de Complementaridade , Sequência de Aminoácidos , Modelos Moleculares , Conformação Proteica , Regiões Determinantes de Complementaridade/química , Complexo Antígeno-Anticorpo/química , Sítios de Ligação de Anticorpos
13.
Front Immunol ; 15: 1352703, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38482007

RESUMO

Deep learning models have been shown to accurately predict protein structure from sequence, allowing researchers to explore protein space from the structural viewpoint. In this paper we explore whether "novel" features, such as distinct loop conformations can arise from these predictions despite not being present in the training data. Here we have used ABodyBuilder2, a deep learning antibody structure predictor, to predict the structures of ~1.5M paired antibody sequences. We examined the predicted structures of the canonical CDR loops and found that most of these predictions fall into the already described CDR canonical form structural space. We also found a small number of "new" canonical clusters composed of heterogeneous sequences united by a common sequence motif and loop conformation. Analysis of these novel clusters showed their origins to be either shapes seen in the training data at very low frequency or shapes seen at high frequency but at a shorter sequence length. To evaluate explicitly the ability of ABodyBuilder2 to extrapolate, we retrained several models whilst withholding all antibody structures of a specific CDR loop length or canonical form. These "starved" models showed evidence of generalisation across CDRs of different lengths, but they did not extrapolate to loop conformations which were highly distinct from those present in the training data. However, the models were able to accurately predict a canonical form even if only a very small number of examples of that shape were in the training data. Our results suggest that deep learning protein structure prediction methods are unable to make completely out-of-domain predictions for CDR loops. However, in our analysis we also found that even minimal amounts of data of a structural shape allow the method to recover its original predictive abilities. We have made the ~1.5 M predicted structures used in this study available to download at https://doi.org/10.5281/zenodo.10280181.


Assuntos
Regiões Determinantes de Complementaridade , Aprendizado Profundo , Regiões Determinantes de Complementaridade/química , Conformação Proteica , Modelos Moleculares , Anticorpos
14.
Biol Chem ; 405(7-8): 461-470, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-38373142

RESUMO

In this work we have generated cattle-derived chimeric ultralong CDR-H3 antibodies targeting tumor necrosis factor α (TNF-α) via immunization and yeast surface display. We identified one particular ultralong CDR-H3 paratope that potently neutralized TNF-α. Interestingly, grafting of the knob architecture onto a peripheral loop of the CH3 domain of the Fc part of an IgG1 resulted in the generation of a TNF-α neutralizing Fc (Fcknob) that did not show any potency loss compared with the parental chimeric IgG format. Eventually, grafting this knob onto the CH3 region of adalimumab enabled the engineering of a novel TNF-α targeting antibody architecture displaying augmented TNF-α inhibition.


Assuntos
Adalimumab , Fator de Necrose Tumoral alfa , Adalimumab/imunologia , Adalimumab/farmacologia , Adalimumab/química , Animais , Bovinos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/farmacologia , Regiões Determinantes de Complementaridade/imunologia , Regiões Determinantes de Complementaridade/química
15.
MAbs ; 16(1): 2309685, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38356181

RESUMO

Rabbits produce robust antibody responses and have unique features in their antibody repertoire that make them an attractive alternative to rodents for in vivo discovery. However, the frequent occurrence of a non-canonical disulfide bond between complementarity-determining region (CDR) H1 (C35a) and CDRH2 (C50) is often seen as a liability for therapeutic antibody development, despite limited reports of its effect on antibody binding, function, and stability. Here, we describe the discovery and humanization of a human-mouse cross-reactive anti-programmed cell death 1 (PD-1) monoclonal rabbit antibody, termed h1340.CC, which possesses this non-canonical disulfide bond. Initial removal of the non-canonical disulfide resulted in a loss of PD-1 affinity and cross-reactivity, which led us to explore protein engineering approaches to recover these. First, guided by the sequence of a related clone and the crystal structure of h1340.CC in complex with PD-1, we generated variant h1340.SA.LV with a potency and cross-reactivity similar to h1340.CC, but only partially recovered affinity. Side-by-side developability assessment of both h1340.CC and h1340.SA.LV indicate that they possess similar, favorable properties. Next, and prompted by recent developments in machine learning (ML)-guided protein engineering, we used an unbiased ML- and structure-guided approach to rapidly and efficiently generate a different variant with recovered affinity. Our case study thus indicates that, while the non-canonical inter-CDR disulfide bond found in rabbit antibodies does not necessarily constitute an obstacle to therapeutic antibody development, combining structure- and ML-guided approaches can provide a fast and efficient way to improve antibody properties and remove potential liabilities.


Assuntos
Anticorpos , Receptor de Morte Celular Programada 1 , Coelhos , Animais , Camundongos , Humanos , Regiões Determinantes de Complementaridade/química , Engenharia de Proteínas/métodos
16.
Biochem Genet ; 62(1): 530-546, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37392243

RESUMO

With lung cancer remaining a challenging disease, new approaches to biomarker discovery and therapy development are needed. Recent immunogenomics, adaptive immune receptor approaches have indicated that it is very likely that B cells play an important role in mediating better overall outcomes. As such, we assessed physicochemical features of lung adenocarcinoma resident IGL complementarity determining region-3 (CDR3) amino acid (AA) sequences and determined that hydrophobic CDR3 AA sequences were associated with a better disease-free survival (DFS) probability. Further, using a recently developed chemical complementarity scoring algorithm particularly suitable for the evaluation of large patient datasets, we determined that IGL CDR3 chemical complementarity with certain cancer testis antigens was associated with better DFS. Chemical complementarity scores for IGL CDR3-MAGEC1 represented a gender bias, with an overrepresentation of males among the higher IGL-CDR3-CTA complementarity scores that were in turn associated with better DFS (logrank p < 0.065). Overall, this study pointed towards potential biomarkers for prognoses that, in some cases are likely gender-specific; and towards biomarkers for guiding therapy, e.g., IGL-based opportunities for antigen targeting in the lung cancer setting.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Masculino , Feminino , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/química , Intervalo Livre de Doença , Sexismo , Neoplasias Pulmonares/genética , Biomarcadores
17.
Nat Biomed Eng ; 8(1): 30-44, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37550425

RESUMO

Conventional methods for humanizing animal-derived antibodies involve grafting their complementarity-determining regions onto homologous human framework regions. However, this process can substantially lower antibody stability and antigen-binding affinity, and requires iterative mutational fine-tuning to recover the original antibody properties. Here we report a computational method for the systematic grafting of animal complementarity-determining regions onto thousands of human frameworks. The method, which we named CUMAb (for computational human antibody design; available at http://CUMAb.weizmann.ac.il ), starts from an experimental or model antibody structure and uses Rosetta atomistic simulations to select designs by energy and structural integrity. CUMAb-designed humanized versions of five antibodies exhibited similar affinities to those of the parental animal antibodies, with some designs showing marked improvement in stability. We also show that (1) non-homologous frameworks are often preferred to highest-homology frameworks, and (2) several CUMAb designs that differ by dozens of mutations and that use different human frameworks are functionally equivalent.


Assuntos
Anticorpos , Regiões Determinantes de Complementaridade , Animais , Humanos , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Anticorpos/química
18.
J Biol Chem ; 300(1): 105555, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38072062

RESUMO

Discovery and optimization of a biotherapeutic monoclonal antibody requires a careful balance of target engagement and physicochemical developability properties. To take full advantage of the sequence diversity provided by different antibody discovery platforms, a rapid and reliable process for humanization of antibodies from nonhuman sources is required. Canonically, maximizing homology of the human variable region (V-region) to the original germline was believed to result in preservation of binding, often without much consideration for inherent molecular properties. We expand on this approach by grafting the complementary determining regions (CDRs) of a mouse anti-LAG3 antibody into an extensive matrix of human variable heavy chain (VH) and variable light chain (VL) framework regions with substantially broader sequence homology to assess the impact on complementary determining region-framework compatibility through progressive evaluation of expression, affinity, biophysical developability, and function. Specific VH and VL framework sequences were associated with major expression and purification phenotypes. Greater VL sequence conservation was correlated with retained or improved affinity. Analysis of grafts that bound the target demonstrated that initial developability criteria were significantly impacted by VH, but not VL. In contrast, cell binding and functional characteristics were significantly impacted by VL, but not VH. Principal component analysis of all factors identified multiple grafts that exhibited more favorable antibody properties, notably with nonoptimal sequence conservation. Overall, this study demonstrates that modern throughput systems enable a more thorough, customizable, and systematic analysis of graft-framework combinations, resulting in humanized antibodies with improved global properties that may progress through development more quickly and with a greater probability of success.


Assuntos
Anticorpos Monoclonais Humanizados , Anticorpos Monoclonais , Animais , Humanos , Camundongos , Anticorpos Monoclonais Humanizados/química , Afinidade de Anticorpos , Regiões Determinantes de Complementaridade/química
19.
Pharm Res ; 40(12): 3087-3098, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37936013

RESUMO

PURPOSE: Monoclonal antibodies (mAbs), like other protein therapeutics, are prone to various forms of degradation, some of which are difficult to distinguish from the native form yet may alter potency. A generalizable LC-MS approach was developed to enable quantitative analysis of isoAsp. In-depth understanding of product quality attributes (PQAs) enables optimization of the manufacturing process, better formulation selection, and decreases risk associated with product handling in the clinic or during shipment. METHODS: Reversed-phase chromatographic peak splitting was observed when a mAb was exposed to elevated temperatures. Multiple LC-MS based methods were applied to identify the reason for peak splitting. The approach involved the use of complementary HPLC columns, multiple enzymatic digestions and different MS/MS ion dissociation methods. In addition, mAb potency was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: The split peaks had identical masses, and the root cause of the peak splitting was identified as isomerization of an aspartic acid located in the complementarity-determining region (CDR) of the light chain. And the early eluting and late eluting peaks were collected and performed enzymatic digestion to confirm the isoAsp enrichment in the early eluting peak. In addition, decreased potency was observed in the same heat-stressed sample, and the increased isoAsp levels in the CDR correlate well with a decrease of potency. CONCLUSION: Liquid chromatography-mass spectrometry (LC-MS) has been utilized extensively to assess PQAs of biological therapeutics. In this study, a generalizable LC-MS-based approach was developed to enable identification and quantitation of the isoAsp-containing peptides.


Assuntos
Anticorpos Monoclonais , Espectrometria de Massas em Tandem , Anticorpos Monoclonais/química , Cromatografia Líquida , Espectrometria de Massas em Tandem/métodos , Espectrometria de Massa com Cromatografia Líquida , Cromatografia Líquida de Alta Pressão/métodos , Regiões Determinantes de Complementaridade/química
20.
Protein Eng Des Sel ; 362023 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-38015984

RESUMO

The Fv region of the antibody (comprising VH and VL domains) is the area responsible for target binding and thus the antibody's specificity. The orientation, or packing, of these two domains relative to each other influences the topography of the Fv region, and therefore can influence the antibody's binding affinity. We present abYpap, an improved method for predicting the packing angle between the VH and VL domains. With the large data set now available, we were able to expand greatly the number of features that could be used compared with our previous work. The machine-learning model was tuned for improved performance using 37 selected residues (previously 13) and also by including the lengths of the most variable 'complementarity determining regions' (CDR-L1, CDR-L2 and CDR-H3). Our method shows large improvements from the previous version, and also against other modeling approaches, when predicting the packing angle.


Assuntos
Regiões Determinantes de Complementaridade , Cadeias Pesadas de Imunoglobulinas , Cadeias Pesadas de Imunoglobulinas/química , Modelos Moleculares , Regiões Determinantes de Complementaridade/química , Anticorpos , Cadeias Leves de Imunoglobulina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...