Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 9(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34599020

RESUMO

BACKGROUND: T cell checkpoint immunotherapies have shown promising results in the clinic, but most patients remain non-responsive. CD47-signal regulatory protein alpha (SIRPα) myeloid checkpoint blockade has shown early clinical activity in hematologic malignancies. However, CD47 expression on peripheral blood limits αCD47 antibody selectivity and thus efficacy in solid tumors. METHODS: To improve the antibody selectivity and therapeutic window, we developed a novel affinity-tuned bispecific antibody targeting CD47 and programmed death-ligand 1 (PD-L1) to antagonize both innate and adaptive immune checkpoint pathways. This PD-L1-targeted CD47 bispecific antibody was designed with potent affinity for PD-L1 and moderate affinity for CD47 to achieve preferential binding on tumor and myeloid cells expressing PD-L1 in the tumor microenvironment (TME). RESULTS: The antibody design reduced binding on red blood cells and enhanced selectivity to the TME, improving the therapeutic window compared with αCD47 and its combination with αPD-L1 in syngeneic tumor models. Mechanistically, both myeloid and T cells were activated and contributed to antitumor activity of αCD47/PD-L1 bispecific antibody. Distinct from αCD47 and αPD-L1 monotherapies or combination therapies, single-cell RNA sequencing (scRNA-seq) and gene expression analysis revealed that the bispecific treatment resulted in unique innate activation, including pattern recognition receptor-mediated induction of type I interferon pathways and antigen presentation in dendritic cells and macrophage populations. Furthermore, treatment increased the Tcf7+ stem-like progenitor CD8 T cell population in the TME and promoted its differentiation to an effector-like state. Consistent with mouse data, the compounds were well tolerated and demonstrated robust myeloid and T cell activation in non-human primates (NHPs). Notably, RNA-seq analysis in NHPs provided evidence that the innate activation was mainly contributed by CD47-SIRPα but not PD-L1-PD-1 blockade from the bispecific antibody. CONCLUSION: These findings provide novel mechanistic insights into how myeloid and T cells can be uniquely modulated by the dual innate and adaptive checkpoint antibody and demonstrate its potential in clinical development (NCT04881045) to improve patient outcomes over current PD-(L)1 and CD47-targeted therapies.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antígeno CD47/metabolismo , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata , Imunoterapia/métodos , Macaca fascicularis , Camundongos , Microambiente Tumoral
2.
J Biol Chem ; 297(4): 101102, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34419446

RESUMO

CD27 is a tumor necrosis factor (TNF) receptor, which stimulates lymphocytes and promotes their differentiation upon activation by TNF ligand CD70. Activation of the CD27 receptor provides a costimulatory signal to promote T cell, B cell, and NK cell activity to facilitate antitumor and anti-infection immunity. Aberrant increased and focused expression of CD70 on many tumor cells renders CD70 an attractive therapeutic target for direct tumor killing. However, despite their use as drug targets to treat cancers, the molecular basis and atomic details of CD27 and CD70 interaction remain elusive. Here we report the crystal structure of human CD27 in complex with human CD70. Analysis of our structure shows that CD70 adopts a classical TNF ligand homotrimeric assembly to engage CD27 receptors in a 3:3 stoichiometry. By combining structural and rational mutagenesis data with reported disease-correlated mutations, we identified the key amino acid residues of CD27 and CD70 that control this interaction. We also report increased potency for plate-bound CD70 constructs compared with solution-phase ligand in a functional activity to stimulate T-cells in vitro. These findings offer new mechanistic insight into this critical costimulatory interaction.


Assuntos
Ligante CD27/química , Complexos Multiproteicos/química , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/química , Ligante CD27/genética , Ligante CD27/imunologia , Cristalografia por Raios X , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Estrutura Quaternária de Proteína , Linfócitos T/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
3.
Cancer Immunol Res ; 9(10): 1141-1157, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34376502

RESUMO

The use of cytokines for immunotherapy shows clinical efficacy but is frequently accompanied by severe adverse events caused by excessive and systemic immune activation. Here, we set out to address these challenges by engineering a fusion protein of a single, potency-reduced, IL15 mutein and a PD1-specific antibody (anti-PD1-IL15m). This immunocytokine was designed to deliver PD1-mediated, avidity-driven IL2/15 receptor stimulation to PD1+ tumor-infiltrating lymphocytes (TIL) while minimally affecting circulating peripheral natural killer (NK) cells and T cells. Treatment of tumor-bearing mice with a mouse cross-reactive fusion, anti-mPD1-IL15m, demonstrated potent antitumor efficacy without exacerbating body weight loss in B16 and MC38 syngeneic tumor models. Moreover, anti-mPD1-IL15m was more efficacious than an IL15 superagonist, an anti-mPD-1, or the combination thereof in the B16 melanoma model. Mechanistically, anti-PD1-IL15m preferentially targeted CD8+ TILs and single-cell RNA-sequencing analyses revealed that anti-mPD1-IL15m treatment induced the expansion of an exhausted CD8+ TIL cluster with high proliferative capacity and effector-like signatures. Antitumor efficacy of anti-mPD1-IL15m was dependent on CD8+ T cells, as depletion of CD8+ cells resulted in the loss of antitumor activity, whereas depletion of NK cells had little impact on efficacy. The impact of anti-hPD1-IL15m on primary human TILs from patients with cancer was also evaluated. Anti-hPD1-IL15m robustly enhanced the proliferation, activation, and cytotoxicity of CD8+ and CD4+ TILs from human primary cancers in vitro, whereas tumor-derived regulatory T cells were largely unaffected. Taken together, our findings showed that anti-PD1-IL15m exhibits a high translational promise with improved efficacy and safety of IL15 for cancer immunotherapy via targeting PD1+ TILs.See related Spotlight by Felices and Miller, p. 1110.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/terapia , Imunoterapia , Interleucina-15/uso terapêutico , Melanoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-15/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/imunologia , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
4.
MAbs ; 13(1): 1871171, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33557687

RESUMO

T-cell engaging biologics is a class of novel and promising immune-oncology compounds that leverage the immune system to eradicate cancer. Here, we compared and contrasted a bispecific diabody-Fc format, which displays a relatively short antigen-binding arm distance, with our bispecific IgG platform. By generating diverse panels of antigen-expressing cells where B cell maturation antigen is either tethered to the cell membrane or located to the juxtamembrane region and masked by elongated structural spacer units, we presented a systematic approach to investigate the role of antigen epitope location and molecular formats in immunological synapse formation and cytotoxicity. We demonstrated that diabody-Fc is more potent for antigen epitopes located in the membrane distal region, while bispecific IgG is more efficient for membrane-proximal epitopes. Additionally, we explored other parameters, including receptor density, antigen-binding affinity, and kinetics. Our results show that molecular format and antigen epitope location, which jointly determine the intermembrane distance between target cells and T cells, allow decoupling of cytotoxicity and cytokine release, while antigen-binding affinities appear to be positively correlated with both readouts. Our work offers new insight that could potentially lead to a wider therapeutic window for T-cell engaging biologics in general.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antígeno de Maturação de Linfócitos B/metabolismo , Produtos Biológicos/farmacologia , Epitopos , Imunoglobulina G/farmacologia , Engenharia de Proteínas , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/metabolismo , Citotoxicidade Celular Dependente de Anticorpos , Reações Antígeno-Anticorpo , Antígeno de Maturação de Linfócitos B/imunologia , Sítios de Ligação de Anticorpos , Produtos Biológicos/imunologia , Produtos Biológicos/metabolismo , Complexo CD3/imunologia , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Mapeamento de Epitopos , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Sinapses Imunológicas/efeitos dos fármacos , Sinapses Imunológicas/imunologia , Sinapses Imunológicas/metabolismo , Cinética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tirosina Quinase 3 Semelhante a fms/imunologia , Tirosina Quinase 3 Semelhante a fms/metabolismo
5.
Nat Commun ; 12(1): 710, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514714

RESUMO

Antibody-based therapeutics have experienced a rapid growth in recent years and are now utilized in various modalities spanning from conventional antibodies, antibody-drug conjugates, bispecific antibodies to chimeric antigen receptor (CAR) T cells. Many next generation antibody therapeutics achieve enhanced potency but often increase the risk of adverse events. Antibody scaffolds capable of exhibiting inducible affinities could reduce the risk of adverse events by enabling a transient suspension of antibody activity. To demonstrate this, we develop conditionally activated, single-module CARs, in which tumor antigen recognition is directly modulated by an FDA-approved small molecule drug. The resulting CAR T cells demonstrate specific cytotoxicity of tumor cells comparable to that of traditional CARs, but the cytotoxicity is reversibly attenuated by the addition of the small molecule. The exogenous control of conditional CAR T cell activity allows continual modulation of therapeutic activity to improve the safety profile of CAR T cells across all disease indications.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva/métodos , Metotrexato/administração & dosagem , Neoplasias/terapia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Terapia Combinada/métodos , Feminino , Células HEK293 , Humanos , Imunoterapia Adotiva/efeitos adversos , Camundongos , Neoplasias/imunologia , Cultura Primária de Células , Receptores de Antígenos Quiméricos/imunologia , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T/efeitos dos fármacos , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mol Cancer Ther ; 18(11): 2008-2020, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31434693

RESUMO

The restricted expression pattern of B-cell maturation antigen (BCMA) makes it an ideal tumor-associated antigen (TAA) for the treatment of myeloma. BCMA has been targeted by both CD3 bispecific antibody and antibody-drug conjugate (ADC) modalities, but a true comparison of modalities has yet to be performed. Here we utilized a single BCMA antibody to develop and characterize both a CD3 bispecific and 2 ADC formats (cleavable and noncleavable) and compared activity both in vitro and in vivo with the aim of generating an optimal therapeutic. Antibody affinity, but not epitope was influential in drug activity and hence a high-affinity BCMA antibody was selected. Both the bispecific and ADCs were potent in vitro and in vivo, causing dose-dependent cell killing of myeloma cell lines and tumor regression in orthotopic myeloma xenograft models. Primary patient cells were effectively lysed by both CD3 bispecific and ADCs, with the bispecific demonstrating improved potency, maximal cell killing, and consistency across patients. Safety was evaluated in cynomolgus monkey toxicity studies and both modalities were active based on on-target elimination of B lineage cells. Distinct nonclinical toxicity profiles were seen for the bispecific and ADC modalities. When taken together, results from this comparison of BCMA CD3 bispecific and ADC modalities suggest better efficacy and an improved toxicity profile might be achieved with the bispecific modality. This led to the advancement of a bispecific candidate into phase I clinical trials.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Antígeno de Maturação de Linfócitos B/metabolismo , Complexo CD3/imunologia , Imunoconjugados/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Animais , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/farmacologia , Afinidade de Anticorpos , Antígeno de Maturação de Linfócitos B/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/farmacologia , Camundongos , Mieloma Múltiplo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 9(1): 8420, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182754

RESUMO

Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.


Assuntos
Anticorpos Biespecíficos/imunologia , Complexo CD3/imunologia , Claudinas/imunologia , Imunoconjugados/uso terapêutico , Neoplasias Pancreáticas/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/terapia , Animais , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Humanos , Imunoconjugados/sangue , Camundongos , Neoplasias Pancreáticas/sangue , Ratos , Neoplasias Gástricas/sangue
9.
Oncotarget ; 9(71): 33446-33458, 2018 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-30323890

RESUMO

Epidermal growth factor receptor (EGFR) is a clinically validated target and often overexpressed in some solid tumors. Both EGFR tyrosine kinase inhibitors and ligand-blocking antibodies have been approved for treatment of NSCLC, head and neck cancers and colorectal cancers. However, clinical response is limited and often accompanied by significant toxicities due to normal tissue expression. To improve the effectiveness of targeting EGFR while minimizing the toxicities on normal tissues, we developed a low-affinity anti-EGFR antibody drug conjugate (ADC), RN765C. Potent in vitro cytotoxicity of RN765C, with nanomolar to subnanomolar EC50, was observed on a panel of cancer cell lines expressing moderate to high level of EGFR. In contrast, RN765C was less effective in killing normal human keratinocytes, presumably due to its lower receptor expression. Mechanistically, RN765C has multiple modes of action: inducing payload mediated mitotic arrest and cell death, blocking EGFR pathway signal and mediating antibody dependent cell cytotoxicity. In preclinical studies, a single dose of RN765C at 1.5-3 mg/kg was generally sufficient to induce tumor regression in multiple cell line and patient-derived xenograft models, including those that are resistant to EGFR-directed tyrosine kinase inhibitors. Our data support further investigation of RN765C in the clinic to treat EGFR expressing solid tumors.

11.
MAbs ; 6(4): 1059-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24830649

RESUMO

Nerve growth factor (NGF) is indispensable during normal embryonic development and critical for the amplification of pain signals in adults. Intervention in NGF signaling holds promise for the alleviation of pain resulting from human diseases such as osteoarthritis, cancer and chronic lower back disorders. We developed a fast, high-fidelity method to convert a hybridoma-derived NGF-targeted mouse antibody into a clinical candidate. This method, termed Library Scanning Mutagenesis (LSM), resulted in the ultra-high affinity antibody tanezumab, a first-in-class anti-hyperalgesic specific for an NGF epitope. Functional and structural comparisons between tanezumab and the mouse 911 precursor antibody using neurotrophin-specific cell survival assays and X-ray crystal structures of both Fab-antigen complexes illustrated high fidelity retention of the NGF epitope. These results suggest the potential for wide applicability of the LSM method for optimization of well-characterized antibodies during humanization.


Assuntos
Anticorpos Monoclonais Murinos , Complexo Antígeno-Anticorpo , Epitopos , Mutagênese , Fator de Crescimento Neural , Adulto , Animais , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/genética , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/genética , Anticorpos Monoclonais Murinos/imunologia , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Complexo Antígeno-Anticorpo/imunologia , Células Cultivadas , Cristalografia por Raios X , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Feminino , Humanos , Dor Lombar/tratamento farmacológico , Dor Lombar/imunologia , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Fator de Crescimento Neural/antagonistas & inibidores , Fator de Crescimento Neural/química , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/imunologia , Osteoartrite/tratamento farmacológico , Osteoartrite/imunologia , Manejo da Dor/métodos , Estrutura Quaternária de Proteína , Anticorpos de Cadeia Única
12.
J Immunol Methods ; 382(1-2): 101-16, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22609372

RESUMO

Label-free biosensors are often used in the discovery of therapeutic antibodies to characterize the epitope binding regions of a panel of monoclonal antibodies that target a specific antigen, thus facilitating their organization into epitope groups or "bins". When tested in a pairwise combinatorial manner, two antibodies that compete with one another for binding to a specific antigen may be grouped into the same epitope bin - that is, they recognize similar or overlapping epitopes - whereas two antibodies that bind simultaneously to the antigen are placed into different epitope bins. However, depending on the assay format used, results from such experiments can sometimes contradict one another. Here, we provide two examples that illustrate how antigen heterogeneity, either inherent in an antigen sample, or induced by the assay conditions, can confound the interpretation of epitope binning results and, in some cases, lead to erroneous conclusions. We highlight why assays that employ solution antigen are often more reliable than those that employ immobilized antigen and, by corroborating our binning results with assays that utilize native antigen, we determine which subpopulations of our heterogeneous antigen samples are biologically relevant and thus improve the correlation between epitope bins and functional activity. Furthermore, we provide recommendations for performing definitive binning assays and a diagnostic assay procedure that can be followed when antigen heterogeneity is suspected.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos/genética , Técnicas Biossensoriais , Epitopos/imunologia , Reações Antígeno-Anticorpo , Antígenos/imunologia , Proteínas de Ligação a Ácido Graxo/análise , Proteínas de Ligação a Ácido Graxo/imunologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/imunologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Dados de Sequência Molecular , Progranulinas
13.
J Mol Biol ; 420(3): 204-19, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22543237

RESUMO

Bispecific antibodies and antibody fragments are a new class of therapeutics increasingly utilized in the clinic for T cell recruitment (catumaxomab anti-EpCAM/CD3 and blinatumomab anti-CD19/CD3), increase in the selectivity of targeting, or simultaneous modulation of multiple cellular pathways. While the clinical potential for certain bispecific antibody formats is clear, progress has been hindered because they are often difficult to manufacture, may suffer from suboptimal pharmacokinetic properties, and may be limited due to potential immunogenicity issues. Current state-of-the-art human IgG-like bispecific technologies require co-expression of two heavy chains with a single light chain, use crossover domains to segregate light chains, or utilize scFv (single-chain fragment variable)-Fc fusion. We have engineered both human IgG1 and IgG2 subtypes, with minimal point mutations, to form full-length bispecific human antibodies with high efficiency and in high purity. In our system, the two antibodies of interest can be expressed and purified separately, mixed together under appropriate redox conditions, resulting in a formation of a stable bispecific antibody with high yields. With this approach, it is not necessary to generate new antibodies that share a common light chain, therefore allowing the immediate use of an existing antibody regardless of whether it has been generated via standard hybridoma or display methods. We demonstrate the generality of the approach and show that these bispecific antibodies have properties similar to those of wild-type IgGs, and we further demonstrate the utility of the technology with an example of a CD3/CD20 bispecific antibody that effectively depletes B cells in vitro and in vivo.


Assuntos
Anticorpos Biespecíficos/imunologia , Imunoglobulina G/metabolismo , Engenharia de Proteínas/métodos , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/isolamento & purificação , Anticorpos Biespecíficos/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Especificidade de Anticorpos , Antígenos CD20/imunologia , Linfócitos B/imunologia , Complexo CD3/imunologia , Cetuximab , Citotoxicidade Imunológica , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação Puntual , Ratos , Ratos Sprague-Dawley , Receptores Fc/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Linfócitos T/imunologia
14.
PLoS One ; 7(4): e36261, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558410

RESUMO

Therapeutic antibodies are often engineered or selected to have high on-target binding affinities that can be challenging to determine precisely by most biophysical methods. Here, we explore the dynamic range of the kinetic exclusion assay (KinExA) by exploiting the interactions of an anti-DKK antibody with a panel of DKK antigens as a model system. By tailoring the KinExA to each studied antigen, we obtained apparent equilibrium dissociation constants (K(D) values) spanning six orders of magnitude, from approximately 100 fM to 100 nM. Using a previously calibrated antibody concentration and working in a suitable concentration range, we show that a single experiment can yield accurate and precise values for both the apparent K(D) and the apparent active concentration of the antigen, thereby increasing the information content of an assay and decreasing sample consumption. Orthogonal measurements obtained on Biacore and Octet label-free biosensor platforms further validated our KinExA-derived affinity and active concentration determinations. We obtained excellent agreement in the apparent affinities obtained across platforms and within the KinExA method irrespective of the assay orientation employed or the purity of the recombinant or native antigens.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Bioensaio/métodos , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Animais , Técnicas Biossensoriais , Calibragem , Linhagem Celular Tumoral , Humanos , Camundongos , Ratos
15.
Anal Biochem ; 411(1): 139-51, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21168382

RESUMO

Here we demonstrate methods to expand the throughput of the ProteOn XPR36 biosensor allowing for the simultaneous kinetic characterization of several multiplexed formats, such as 36 disparate antibodies targeting the same antigen, and facilitating detailed epitope binning and mapping studies. The kinetic rate constants determined by these methods correlated with those obtained on Biacore 2000 and the absolute parameter values obtained on the ProteOn's alginate-based GLC chip agreed closer with those from Biacore's flat C1 chip than Biacore's dextran-based CM4 chip. Pairwise epitope binning data from the ProteOn 36-ligand array format and those generated on an orthogonal array-based biosensor, the Octet QK384, gave similar results. In an epitope mapping study using biotinylated peptides, all three biosensor platforms were similar in their ability to identify antibodies that bound to linear epitopes. We apply alternative formats of the ProteOn array that enable a significantly higher number of assays to be conducted simultaneously than previously anticipated on this platform.


Assuntos
Técnicas Biossensoriais/métodos , Análise Serial de Proteínas/métodos , Animais , Anticorpos Monoclonais/metabolismo , Mapeamento de Epitopos , Epitopos/imunologia , Proteínas Imobilizadas/metabolismo , Cinética , Ligantes , Biblioteca de Peptídeos , Ligação Proteica , Ratos , Titulometria
16.
Anal Biochem ; 352(2): 208-21, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16564019

RESUMO

To explore the reliability of Biacore-based assays, 22 study participants measured the binding of prostate-specific antigen (PSA) to a monoclonal antibody (mAb). Each participant was provided with the same reagents and a detailed experimental protocol. The mAb was immobilized on the sensor chip at three different densities and a two-step assay was used to determine the kinetic and affinity parameters of the PSA/mAb complex. First, PSA was tested over a concentration range of 2.5-600 nM to obtain k(a) information. Second, to define the k(d) of this stable antigen/antibody complex accurately, the highest PSA concentration was retested with the dissociation phase of each binding cycle monitored for 1h. All participants collected data that could be analyzed to obtain kinetic parameters for the interaction. The association and the extended-dissociation data derived from the three antibody surfaces were globally fit using a simple 1:1 interaction model. The average k(a) and k(d) for the PSA/mAb interaction as calculated from the 22 analyses were (4.1+/-0.6) x 10(4) M(-1) s(-1) and (4.5+/-0.6) x 10(-5) s(-1), respectively. Overall, the experimental standard errors in the rate constants were only approximately 14%. Based on the kinetic rate constants, the affinity (K(D)) of the PSA/mAb interaction was 1.1+/-0.2 nM.


Assuntos
Anticorpos Monoclonais/química , Reações Antígeno-Anticorpo , Técnicas Biossensoriais/métodos , Antígeno Prostático Específico/química , Técnicas Biossensoriais/instrumentação , Técnicas Biossensoriais/normas , Humanos , Cinética , Ligantes , Antígeno Prostático Específico/análise , Antígeno Prostático Específico/imunologia , Ligação Proteica , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Ressonância de Plasmônio de Superfície/instrumentação , Ressonância de Plasmônio de Superfície/métodos , Ressonância de Plasmônio de Superfície/normas , Fatores de Tempo
17.
Biochemistry ; 44(27): 9507-19, 2005 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-15996105

RESUMO

Soluble oligosaccharide mimetics of natural selectin ligands act as competitive inhibitors of leukocyte adhesion in models of inflammation. We quantified the binding of simple oligosaccharides based on sialyl Lewis-X (sLe(X)) and complex molecules with the core-2 structure to L- and P-selectin, under both static and fluid flow conditions. Isolated human neutrophils were employed to mimic the physiological valency of selectins and selectin ligands. Surface plasmon resonance studies quantified binding kinetics. We observed the following: (i) The functional group at the anomeric position of carbohydrates plays an important role during selectin recognition, since sLe(X) and sialyl Lewis-a (sLe(a)) were approximately 5-7-fold poorer inhibitors of L-selectin mediated cell adhesion compared to their methyl glycosides. (ii) Despite their homology to physiological glycans, the putative carbohydrate epitopes of GlyCAM-1 and PSGL-1 bound selectins with low affinity comparable to that of sLe(X)-selectin interactions. Thus, besides the carbohydrate portion, the protein core of GlyCAM-1 or the presentation of carbohydrates in clusters on this glycoprotein may contribute to selectin recognition. (iii) A compound Galbeta1,4(Fucalpha1,3)GlcNAcbeta1,6(GalNAcbeta1,3)GalNAcalpha-OMe was identified which blocked L- and P-selectin binding at 30-100-fold lower doses than sLe(X). (iv) Surface plasmon resonance experiments determined that an sLe(X) analogue (TBC1269) competitively inhibited, via steric/allosteric mechanisms, the binding of two anti-P-selectin function blocking antibodies that recognized different epitopes of P-selectin. (v) TBC1269 bound P-selectin via both calcium-dependent and -independent mechanisms, with K(D) of approximately 111.4 microM. The measured on- and off-rates were high (k(off) > 3 s(-)(1), k(on) > 27,000 M(-)(1) s(-)(1)). Similar binding kinetics are expected for sLe(X)-selectin interactions. Taken together, our study provides new insight into the kinetics and mechanisms of carbohydrate interaction with selectins.


Assuntos
Selectina L/metabolismo , Antígenos do Grupo Sanguíneo de Lewis/química , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Selectina-P/metabolismo , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Compostos de Bifenilo/química , Compostos de Bifenilo/metabolismo , Células CHO , Sequência de Carboidratos , Agregação Celular/efeitos dos fármacos , Agregação Celular/fisiologia , Cricetinae , Humanos , Cinética , Selectina L/genética , Ligantes , Manose/análogos & derivados , Manosídeos/química , Manosídeos/metabolismo , Dados de Sequência Molecular , N-Acetilglucosaminiltransferases/química , N-Acetilglucosaminiltransferases/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/fisiologia , Selectina-P/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Reologia , Antígeno Sialil Lewis X , Ressonância de Plasmônio de Superfície , Trissacarídeos/química , Trissacarídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...