Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Methods Mol Biol ; 2793: 41-54, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38526722

RESUMEN

Resistance to therapeutic antibodies caused by on-target point mutations is a major obstacle in anticancer therapy, creating an "unmet clinical need." To tackle this problem, researchers are developing new generations of antibody drugs that can overcome the resistance mechanisms of existing agents. We have previously reported a structure-guided and phage-assisted evolution (SGAPAE) approach to evolve cetuximab, a therapeutic antibody, to effectively reverse the resistance driven by EGFRS492R or EGFRG465R mutations, without changing the binding epitope or compromising the antibody efficacy. In this protocol, we provide detailed instructions on how to use the SGAPAE approach to evolve cetuximab, which can also be applied to other therapeutic antibodies for reversing on-target point mutation-mediated resistance. The protocol consists of four steps: structure preparation, computational prediction, phage display library construction, and antibody candidate selection.


Asunto(s)
Anticuerpos Monoclonales , Bacteriófagos , Cetuximab , Mutación Puntual , Receptores ErbB/metabolismo , Bacteriófagos/metabolismo , Anticuerpos Monoclonales Humanizados/genética
2.
Prep Biochem Biotechnol ; 53(3): 239-246, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35579623

RESUMEN

Several monoclonal antibodies targeting the CD20 have been produced and Ofatumumab is a case in point. Although whole antibodies target cancer cells effectively, their applications are restricted in some ways. Single-chain fragment variable antibodies, rather than employing the entire structure of antibodies, have proven a practical approach for creating completely functional antigen-binding fragments. In current research, the DNA coding sequence of VL and VH of the wild and mutant forms of ofatumumab were joined with a flexible linker (GGGGS)3 separately. Using the E. coli BL21 (DE3) expression system, the VL-linker-VH genes were cloned into the pET-28 a (+), and the associated recombinant proteins were produced. Purified and refolded scFvs (scFv-C and scFv-V3) represented a concentration of around 0.7 mg/ml from 1 L of initial E. coli culture with a molecular weight of about 27 kDa. Affinity measurement disclosed anti-CD20 scFv-V3 possesses a higher affinity constant compared to anti-CD20 scFv-C. The recombinant scFvs exclusively attach to Raji cells but not to Jurkat cells, according to a cell-ELISA analysis. The MTT test signified anti-CD20 scFvs could affect cell viability in Raji cells but had no impact on Jurkat cells and also, Raji cells viability was affected more significantly by anti-CD20 scFv-V3.


Asunto(s)
Antígenos CD20 , Anticuerpos de Cadena Única , Humanos , Antígenos CD20/genética , Antígenos CD20/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos de Cadena Única/genética
3.
Dis Markers ; 2022: 3132941, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36051360

RESUMEN

Gemtuzumab ozogamicin (GO) is a conjugate of a monoclonal antibody and calicheamicin, which has been reapproved for the treatment of acute myeloid leukemia (AML). AML patients with the CD33 rs12459419 CC genotype might benefit from the addition of GO to intensive treatment in contrast to patients with CT/TT genotypes. Nevertheless, contradictory results have been reported. We sought to shed light on the prediction of GO response in AML patients with rs12459419 polymorphism who were treated with GO in the consolidation (n = 70) or reinduction (n = 20) phase. The frequency distribution of the rs12459419 polymorphism in the complete cohort of patients was 44.4% (n = 40), 50% (n = 45), and 5.6% (n = 5) for CC, CT, and TT genotypes, respectively. Regarding the patients treated with GO for consolidation, we performed a Kaplan-Meier analysis of overall survival and relapse-free survival according to the rs12459419 polymorphism (CC vs. CT/TT patients) and genetic risk using the European Leukemia Net (ELN) 2010 risk score. We also carried out a Cox regression analysis for the prediction of overall survival, with age and ELN 2010 as covariates. We found no statistical significance in the univariate or multivariate analysis. Additionally, we performed a global Kaplan-Meier analysis for the patients treated with GO for reinduction and did not find significant differences; however, our cohort was too small to draw any conclusion from this analysis. The use of GO in consolidation treatment is included in the approval of the compound; however, evidence regarding its efficacy in this setting is lacking. Rs12459419 polymorphism could help in the selection of patients who might benefit from GO. Regrettably, in our cohort, the rs12459419 polymorphism does not seem to be an adequate tool for the selection of patients who might benefit from the addition of GO in consolidation cycles.


Asunto(s)
Aminoglicósidos , Leucemia Mieloide Aguda , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Aminoglicósidos/uso terapéutico , Anticuerpos Monoclonales Humanizados/genética , Gemtuzumab/uso terapéutico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Polimorfismo de Nucleótido Simple , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética
4.
Cell Mol Biol Lett ; 27(1): 20, 2022 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-35236282

RESUMEN

BACKGROUND: EGFR targeting is currently the main treatment strategy for metastatic colorectal cancer (mCRC). Results of different clinical trials show that patients with wild-type KRAS and BRAF benefit from anti-EGFR monoclonal antibodies (moAbs) cetuximab (CTX) or panitumumab. Unfortunately, despite initial response, patients soon became refractory. Tumor heterogeneity and multiple escaping routes have been addressed as the main culprit, and, behind genomic alterations already described, changes in signaling pathways induced by drug pressure are emerging as mechanisms of acquired resistance. We previously reported an association between reduced sensitivity to CTX and increased expression of IL-1. However, how IL-1 mediates CTX resistance in mCRC is still unclear. METHODS: Under CTX treatment, the upregulation of IL-1R1 expression and a senescence program in sensitive colorectal cancer (CRC) cell lines is examined over time using qPCR, immunoblotting, and immunofluorescence. RESULTS: In sensitive CRC cells, IL-1 appeared responsible for a CTX-mediated G0 phase arrest. On the contrary, CTX-resistant CRC cells (CXR) maintained high mRNA levels of IL-1R1 and a post-senescence reprogramming, as indicated by increased SNAIL expression. Interestingly, treatment of CXR cells with a recombinant decoy, able to sequester the soluble form of IL-1, pushed CTX-resistant CRC cells back into a stage of senescence, thus blocking their proliferation. Our model suggests a trans-regulatory mechanism mediated by IL-1 on EGFR signaling. By establishing senescence and regulating EGFR activity and expression, IL-1 exposure ultimately bestows resistance. CONCLUSIONS: To sum up, our findings point to the combined blockage of IL-1R and EGFR as a promising therapeutical approach to restore sensitivity to EGFR-targeting monoclonal antibodies.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Interleucina-1/genética , Interleucina-1/farmacología , Interleucina-1/uso terapéutico , Mutación
5.
Sci Rep ; 11(1): 22365, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34785671

RESUMEN

Antibody function is typically entirely dictated by the Complementarity Determining Regions (CDRs) that directly bind to the antigen, while the framework region acts as a scaffold for the CDRs and maintains overall structure of the variable domain. We recently reported that the rabbit monoclonal antibody 4A11 (rbt4A11) disrupts signaling through both TGFß2 and TGFß3 (Sun et al. in Sci Transl Med, 2021. https://doi.org/10.1126/scitranslmed.abe0407 ). Here, we report a dramatic, unexpected discovery during the humanization of rbt4A11 where, two variants of humanized 4A11 (h4A11), v2 and v7 had identical CDRs, maintained high affinity binding to TGFß2/3, yet exhibited distinct differences in activity. While h4A11.v7 completely inhibited TGFß2/3 signaling like rbt4A11, h4A11.v2 did not. We solved crystal structures of TGFß2 complexed with Fab fragments of h4A11.v2 or h4A11.v7 and identified a novel interaction between the two heavy chain molecules in the 2:2 TGFb2:h4A11.v2-Fab complex. Further characterization revealed that framework residue variations at either position 19, 79 or 81 (Kabat numbering) of the heavy chain strikingly converts h4A11.v2 into an inhibitory antibody. Our work suggests that in addition to CDRs, framework residues and interactions between Fabs in an antibody could be engineered to further modulate activity of antibodies.


Asunto(s)
Sustitución de Aminoácidos , Anticuerpos Monoclonales Humanizados/química , Fragmentos Fab de Inmunoglobulinas/química , Región Variable de Inmunoglobulina/química , Factor de Crecimiento Transformador beta2/química , Factor de Crecimiento Transformador beta3/química , Animales , Anticuerpos Monoclonales Humanizados/genética , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina/genética , Estructura Cuaternaria de Proteína , Conejos , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta3/genética
6.
Sci Rep ; 11(1): 19500, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593880

RESUMEN

Invasive fungal infections mainly affect patients undergoing transplantation, surgery, neoplastic disease, immunocompromised subjects and premature infants, and cause over 1.5 million deaths every year. The most common fungi isolated in invasive diseases are Candida spp., Cryptococcus spp., and Aspergillus spp. and even if four classes of antifungals are available (Azoles, Echinocandins, Polyenes and Pyrimidine analogues), the side effects of drugs and fungal acquired and innate resistance represent the major hurdles to be overcome. Monoclonal antibodies are powerful tools currently used as diagnostic and therapeutic agents in different clinical contexts but not yet developed for the treatment of invasive fungal infections. In this paper we report the development of the first humanized monoclonal antibody specific for ß-1,3 glucans, a vital component of several pathogenic fungi. H5K1 has been tested on C. auris, one of the most urgent threats and resulted efficient both alone and in combination with Caspofungin and Amphotericin B showing an enhancement effect. Our results support further preclinical and clinical developments for the use of H5K1 in the treatment of patients in need.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Hongos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/aislamiento & purificación , Especificidad de Anticuerpos/inmunología , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Farmacorresistencia Fúngica/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Ingeniería Genética , Humanos , Cadenas Pesadas de Inmunoglobulina , Cadenas Ligeras de Inmunoglobulina/genética , Ratones , Pruebas de Sensibilidad Microbiana , Fagocitosis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
7.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34576184

RESUMEN

Antibody-cytokine fusion proteins (immunocytokines) are gaining importance for cancer therapy, but those products are often limited by systemic toxicity related to the activity of the cytokine payload in circulation and in secondary lymphoid organs. Tumor necrosis factor (TNF) is used as a pro-inflammatory payload to trigger haemorrhagic necrosis and boost anti-cancer immunity at the tumor site. Here we describe a depotentiated version of TNF (carrying the single point mutation I97A), which displayed reduced binding affinity to its cognate receptor tumor necrosis factor receptor 1 (TNFR-1) and lower biocidal activity. The fusion of the TNF(I97A) mutant to the L19 antibody promoted restoration of anti-tumor activity upon accumulation on the cognate antigen, the alternatively spliced EDB domain of fibronectin. In vivo administration of high doses (375 µg/Kg) of the fusion protein showed a potent anti-tumor effect without apparent toxicity compared with the wild type protein. L19-TNFI97A holds promise for the targeted delivery of TNF activity to neoplastic lesions, helping spare normal tissues.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/metabolismo , Cricetulus , Citocinas/genética , Citocinas/metabolismo , Femenino , Fibronectinas/genética , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunoterapia , Ratones Endogámicos BALB C , Mutación , Estructura Secundaria de Proteína , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/genética
8.
Sci Rep ; 11(1): 15694, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34344952

RESUMEN

Respiratory syncytial virus (RSV) infection is a common cause of hospitalisation in infants and the elderly. Palivizumab prophylaxis is the only approved treatment modality but is costly and only offered to select vulnerable populations. Here, we investigated gene delivery approaches via recombinant adeno-associated virus (rAAV2/8) and simian immunodeficiency virus (rSIV.F/HN) vectors to achieve sustained in vivo production of palivizumab in a murine model. Delivery of palivizumab-expressing vectors 28 days prior to RSV challenge resulted in complete protection from RSV-induced weight loss. This approach offers prophylaxis against RSV infection, allowing for wider use and reduction in treatment costs in vulnerable populations.


Asunto(s)
Anticuerpos Monoclonales Humanizados/genética , Expresión Génica , Terapia Genética , Palivizumab/genética , Infecciones por Virus Sincitial Respiratorio/terapia , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Inyecciones Intramusculares , Lentivirus/genética , Ratones , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Transducción Genética , Resultado del Tratamiento
9.
Clin Cancer Res ; 27(17): 4685-4689, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34083238

RESUMEN

The FDA approved pembrolizumab on June 16, 2020, for the treatment of adult and pediatric patients with unresectable or metastatic tumor mutational burden-high [TMB-H; ≥10 mutations/megabase (mut/Mb)] solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options. FDA granted the approval based on a clinically important overall response rate (29%; 95% confidence interval, 21-39) and duration of response (57% of responses lasting ≥ 12 months) in the subset of patients with TMB-H solid tumors (n = 102) spanning nine different tumor types enrolled in a multicenter single-arm trial (KEYNOTE-158). The efficacy of pembrolizumab was supported by the results of whole-exome sequencing (WES) analyses of TMB in additional patients enrolled across multiple pembrolizumab clinical trials, and a scientific understanding of the effects of PD-1 inhibition. Overall, the adverse event profile of pembrolizumab was similar to the adverse event profile observed in prior trials that supported the approval of pembrolizumab in other indications. This approval of pembrolizumab is the first time that the FDA has approved a cancer treatment for an indication based on TMB, and the fourth based on the presence of a biomarker rather than the primary site of origin.


Asunto(s)
Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Aprobación de Drogas , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Adulto , Niño , Humanos , Estudios Retrospectivos , Estados Unidos
10.
Nanomedicine ; 36: 102424, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34174417

RESUMEN

In order to generate an antibody directed enzyme prodrug therapy, here we designed a chimeric protein by fusing the F8 antibody that recognizes the EDA of fibronectin (expressed on the tumor neovasculature) and an evolved variant of the ROS-generating enzyme D-amino acid oxidase (DAAO). The F8(scFv)-DAAO-Q144R recombinant protein is expressed by both CHO-S and E. coli cells. The F8(scFv)-DAAO-Q144R from E. coli cells is fully soluble, shows a high specific activity, is more thermostable in blood than the native DAAO, possesses a binding affinity for EDA well suited for efficient tumor accumulation, and localizes in tumor tissues. Notably, the F8(scFv)-DAAO-Q144R conjugate generates a stronger cytotoxicity to tumor cells than the native enzyme, especially when an inhibitor of heme oxygenase-1 (HO-1) is used, making it a promising candidate for a selective antitumor oxidative therapy controlled by the substrate addition, in the so called "activity on demand", thus sparing normal tissue from damage.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antineoplásicos , Citotoxinas , D-Aminoácido Oxidasa , Fibronectinas , Proteínas de Neoplasias , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Células CHO , Células COS , Chlorocebus aethiops , Cricetulus , Citotoxinas/química , Citotoxinas/farmacología , D-Aminoácido Oxidasa/química , D-Aminoácido Oxidasa/genética , D-Aminoácido Oxidasa/farmacología , Fibronectinas/antagonistas & inhibidores , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología
11.
Signal Transduct Target Ther ; 6(1): 236, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34158475

RESUMEN

Despite the successful use of the humanized monoclonal antibody trastuzumab (Herceptin) in the clinical treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer, the frequently occurring drug resistance remains to be overcome. The regulatory mechanisms of trastuzumab-elicited immune response in the tumor microenvironment remain largely uncharacterized. Here, we found that the nonclassical histocompatibility antigen HLA-G desensitizes breast cancer cells to trastuzumab by binding to the natural killer (NK) cell receptor KIR2DL4. Unless engaged by HLA-G, KIR2DL4 promotes antibody-dependent cell-mediated cytotoxicity and forms a regulatory circuit with the interferon-γ (IFN-γ) production pathway, in which IFN-γ upregulates KIR2DL4 via JAK2/STAT1 signaling, and then KIR2DL4 synergizes with the Fcγ receptor to increase IFN-γ secretion by NK cells. Trastuzumab treatment of neoplastic and NK cells leads to aberrant cytokine production characterized by excessive tumor growth factor-ß (TGF-ß) and IFN-γ, which subsequently reinforce HLA-G/KIR2DL4 signaling. In addition, TGF-ß and IFN-γ impair the cytotoxicity of NK cells by upregulating PD-L1 on tumor cells and PD-1 on NK cells. Blockade of HLA-G/KIR2DL4 signaling improved the vulnerability of HER2-positive breast cancer to trastuzumab treatment in vivo. These findings provide novel insights into the mechanisms underlying trastuzumab resistance and demonstrate the applicability of combined HLA-G and PD-L1/PD-1 targeting in the treatment of trastuzumab-resistant breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Antígenos HLA-G/genética , Receptor ErbB-2/genética , Receptores KIR2DL4/genética , Trastuzumab/farmacología , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Femenino , Humanos , Interferón gamma/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Persona de Mediana Edad , Receptores de Células Asesinas Naturales/genética , Receptores de Células Asesinas Naturales/inmunología , Trastuzumab/efectos adversos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
12.
Mol Immunol ; 135: 320-328, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33971510

RESUMEN

Using antibody drug conjugates (ADC) which can exclusively bind to their target cells and upon internalization release their toxic agent, is one of the most effective methods for killing tumor cells. Therefore, increasing the internalization rate is an important factor for tumor treatment in this case. The aim of the present study was to develop a new variant of pertuzumab (an anti-ErbB2 humanized antibody) with higher internalization rate that can be a good candidate for the production of ADC. To this end, the Human Immunodeficiency Virus TAT Protein Transduction Domain (TAT-PTD) was replaced into the structure of the pertuzumab. At first, the best site in antibody heavy chain constant region for the replacement of TAT-PTD was predicted through computational methods. Then, the resulting recombinant antibody, of which TAT-PTD was located at amino acid position 130-140 and named Tatibody, was produced in CHO-S cell line. Finally, its physicochemical properties and biological activities were evaluated and compared with pertuzumab. Results showed that the binding ability of Tatibody to the ErbB2 receptor is similar to that of pertuzumab, but its internalization potency is 3.6 fold higher and can be used as a good candidate for ADC construction.


Asunto(s)
Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Recombinantes/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Afinidad de Anticuerpos/inmunología , Células CHO , Línea Celular Tumoral , Cricetulus , Femenino , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Simulación del Acoplamiento Molecular , Conformación Proteica , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/inmunología , Proteínas Recombinantes/inmunología
13.
Monoclon Antib Immunodiagn Immunother ; 40(2): 52-59, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33900821

RESUMEN

DNAM-1 is an activating immunoreceptor expressed on hematopoietic cells, including both CD4+ and CD8+ T cells, natural killer cells, and platelets. Since DNAM-1 is involved in the pathogenesis of various inflammatory diseases and cancers in humans as well as mouse models, it is a potential target for immunotherapy for these diseases. In this study, we generated a humanized neutralizing antihuman DNAM-1 monoclonal antibody (mAb), named TNAX101A, which contains an engineered Fc portion of human IgG1 to reduce Fc-mediated effector functions. We show that TNAX101A efficiently interfered the binding of DNAM-1 to its ligand CD155 and showed unique functions; it decreased production of the inflammatory cytokines such as interferon-gamma, tumor necrosis factor alpha, interleukin (IL)-6, IL-17A, and IL-17F by anti-CD3 antibody-stimulated or alloantigen-stimulated T cells and increased FOXP3 expression in anti-CD3-stimulated regulatory T (Treg) cells. These dual functions of TNAX101A may be advantageous for the treatment of T cell-mediated inflammatory diseases through both downregulation of effector T cell function and upregulation of Treg cell function.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Factores de Transcripción Forkhead/genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales Humanizados/genética , Antígenos de Diferenciación de Linfocitos T/efectos de los fármacos , Antígenos de Diferenciación de Linfocitos T/genética , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoterapia/tendencias , Neoplasias/inmunología , Neoplasias/terapia , Células TH1/inmunología , Células Th17/inmunología
14.
MAbs ; 13(1): 1868066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33404287

RESUMEN

LIGHT is a member of the tumor necrosis factor superfamily, which has been claimed to mediate anti-tumor activity on the basis of cancer cures observed in immunocompetent mice bearing transgenic LIGHT-expressing tumors. The preclinical development of a LIGHT-based therapeutic has been hindered by the lack of functional stability exhibited by this protein. Here, we describe the cloning, expression, and characterization of five antibody-LIGHT fusion proteins, directed against the alternatively spliced extra domain A of fibronectin, a conserved tumor-associated antigen. Among the five tested formats, only the sequential fusion of the F8 antibody in single-chain diabody format, followed by the LIGHT homotrimer expressed as a single polypeptide, yielded a protein (termed "F8-LIGHT") that was not prone to aggregation. A quantitative biodistribution analysis in tumor-bearing mice, using radio-iodinated protein preparations, confirmed that F8-LIGHT was able to preferentially accumulate at the tumor site, with a tumor-to-blood ratio of ca. five to one 24 hours after intravenous administration. Tumor therapy experiments, performed in two murine tumor models (CT26 and WEHI-164), featuring different levels of lymphocyte infiltration into the neoplastic mass, revealed that F8-LIGHT could significantly reduce tumor-cell growth and was more potent than a similar fusion protein (KSF-LIGHT), directed against hen egg lysozyme and serving as negative control of irrelevant specificity in the mouse. At a mechanistic level, the activity of F8-LIGHT was mainly due to an intratumoral expansion of natural killer cells, whereas there was no evidence of expansion of CD8 + T cells, neither in the tumor, nor in draining lymph nodes. Abbreviations: CTLA-4: Cytotoxic T-lymphocytes-associated protein 4; EGFR: Epidermal growth factor receptor; HVEM: Herpesvirus entry mediator; IFNγ: Interferon-gamma; LIGHT: Lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes; LTßR: Lymphotoxin beta receptor; NF-κB: Nuclear factor "kappa-light-chain-enhancer" of activated B cells; NK: Natural killer cells; PD-1: Programmed cell death protein 1; PD-L1: Programmed death-ligand 1; TNF: Tumor necrosis factor.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Células Asesinas Naturales/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/metabolismo , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Progresión de la Enfermedad , Humanos , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
15.
Exp Biol Med (Maywood) ; 246(8): 940-951, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33475433

RESUMEN

Interleukin-9 is a cytokine with multiple functions, including the ability to activate group 2 innate lymphoid cells, which has been postulated to be therapeutically active in mouse models of arthritis. Similarly, interleukin-9 has been suggested to play an important role in tumor immunity. Here, we describe the cloning, expression, and characterization of three fusion proteins based on murine interleukin-9 and the F8 antibody, specific to the alternatively spliced EDA domain of fibronectin. EDA is strongly expressed in cancer and in various arthritic conditions, while being undetectable in the majority of healthy organs. Interleukin-9-based fusion proteins with an irrelevant antibody specific to hen egg lysozyme served as negative control in our study. The fusion proteins were characterized by quantitative biodistribution analysis in tumor-bearing mice using radioiodinated protein preparations. The highest tumor uptake and best tumor:organ ratios were observed for a format, in which the interleukin-9 moiety was flanked by two units of the F8 antibody in single-chain Fv format. Biological activity of interleukin-9 was retained when the payload was fused to antibodies. However, the targeted delivery of interleukin-9 to the disease site resulted in a modest anti-tumor activity in three different murine models of cancer (K1735M2, CT26, and F9), while no therapeutic benefit was observed in a collagen induced model of arthritis. Collectively, these results confirm the possibility to deliver interleukin-9 to the site of disease but cast doubts about the alleged therapeutic activity of this cytokine in cancer and arthritis, which has been postulated in previous publications.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Artritis Experimental/tratamiento farmacológico , Interleucina-9/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Animales , Anticuerpos Monoclonales Humanizados/genética , Artritis Experimental/genética , Artritis Experimental/metabolismo , Sistemas de Liberación de Medicamentos , Evaluación de Medicamentos , Interleucina-9/genética , Masculino , Ratones , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética
16.
Front Immunol ; 11: 604318, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33365032

RESUMEN

Boosting the production of recombinant therapeutic antibodies is crucial in both academic and industry settings. In this work, we investigated the usage of varying signal peptides by antibody V-genes and their roles in recombinant transient production, systematically comparing myeloma and the native signal peptides of both heavy and light chains in 168 antibody permutation variants. We found that amino acids count and types (essential or non-essential) were important factors in a logistic regression equation model for predicting transient co-transfection protein production rates. Deeper analysis revealed that the culture media were often incomplete and that the supplementation of essential amino acids can improve the recombinant protein yield. While these findings are derived from transient HEK293 expression, they also provide insights to the usage of the large repertoire of antibody signal peptides, where by varying the number of specific amino acids in the signal peptides attached to the variable regions, bottlenecks in amino acid availability can be mitigated.


Asunto(s)
Aminoácidos/metabolismo , Anticuerpos Monoclonales Humanizados/biosíntesis , Antineoplásicos Inmunológicos/metabolismo , Biotecnología , Inmunoglobulina E/biosíntesis , Región Variable de Inmunoglobulina , Ingeniería de Proteínas , Señales de Clasificación de Proteína , Trastuzumab/biosíntesis , Anticuerpos Monoclonales Humanizados/genética , Medios de Cultivo/metabolismo , Células HEK293 , Humanos , Inmunoglobulina E/genética , Región Variable de Inmunoglobulina/genética , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Señales de Clasificación de Proteína/genética , Proteínas Recombinantes/biosíntesis , Trastuzumab/genética , Flujo de Trabajo
17.
Protein Sci ; 29(9): 1924-1930, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32683733

RESUMEN

Cathepsin L (CTSL) is a cysteine protease involved in a variety of physiological and pathological processes. Potent inhibitors against CTSL have long been sought for drug development. Due to insufficient specificity and suboptimal pharmacological properties for current CTSL inhibitors, novel agents are still required for selectively blocking CTSL activity. Here we generated a humanized antibody inhibitor of CTSL by genetically fusing the inhibitory propeptide of procathepsin L to the N-terminus of the light chain of a humanized antibody. The resulting antibody fusion could be stably expressed and displays highly potent inhibition activity and specificity toward CTSL. This work demonstrates a new approach for the rapid generation of antibody inhibitors of CTSL. It can possibly be extended to create inhibitory antibodies targeting other cathepsin proteases, providing novel research and therapeutic tools.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Catepsina L/antagonistas & inhibidores , Catepsina L/química , Inhibidores de Cisteína Proteinasa/química , Anticuerpos Monoclonales Humanizados/genética , Catepsina L/genética , Humanos
18.
Cancer Immunol Immunother ; 69(12): 2561-2569, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32583154

RESUMEN

Many cancer cells express CD47 as a 'don't eat me' signal to mask their presences from immune recognition and destruction. Such a signal is transmitted when CD47 binds to the signal regulatory protein-α (SIRPα) on macrophages to cut the phagocytic reaction. Most recent studies have focused on developing CD47 blocking agents with different affinities and avidities in order to optimize the therapeutic window between efficacy and toxicities involving normal cells expressing CD47. We described in this study a new design to fuse one CD47 binding domain of SIRPα with a pharmacokinetics modifying domain F8. The resulted single valent long-acting CD47 antagonist SIRPα-F8 was able to bind to CD47 and disrupt CD47-SIRPα axis. However, by itself it cannot trigger endocytosis and has no effect on tumor growth. Only when used in combination with the anti-CD20 mAbs, there were greatly improved phagocytic activities towards CD20 positive cancer cells. In vivo the combination also resulted in better tumor growth inhibition comparing to the vehicle control group. In addition, we showed that the F8 fusion bound to hFcRn only inside endosomes at pH 6.0, enabled hFcRn mediated recycling and thus greatly extended the circulation half-life in hFcRn knock-in mice. Taken together, the SIRPα-F8 design may suggest a new option to improve the therapeutic index of antibody treatment in clinical use towards tumors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Antígeno CD47/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/farmacología , Antígenos de Diferenciación/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno CD47/inmunología , Células CHO , Línea Celular Tumoral , Cricetulus , Técnicas de Sustitución del Gen , Células HEK293 , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inyecciones Intralesiones , Macrófagos , Ratones Transgénicos , Monocitos , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/inmunología , Receptores Fc/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Biomol NMR ; 74(6-7): 311-319, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32415582

RESUMEN

Monoclonal antibody (mAb) drugs are clinically important for the treatment of various diseases. TTAC-0001 is under development as a new anti-cancer antibody drug targeting VEGFR-2. As the less severe toxicity of TTAC-0001 compared to Bevacizumab, likely due to the decreased in vivo half-life, seems to be related to its structural flexibility, it is important to map the exact flexible regions. Although the 13C/15N-labeled protein is required for NMR analyses, it is difficult to obtain antibody fragments (Fab and scFv) containing disulfide bonds through general cytosolic expression in Escherichia coli (E. coli). Here, we notably increased the periplasmic expression of the 13C/15N-labeled TTAC-0001-Fab (13C/15N-TTAC-Fab) through simple isopropyl ß-D-1-thiogalactopyranoside (IPTG)-induction at an increased optical density (1.5 OD600nm). Through NMR triple resonance experiments, two loop insertions (LI-1 between the VH and CH1; LI-2 between the VL and CL) were confirmed to be highly flexible. The additional LIs could be another way to engineer the antibody by changing the pharmacokinetic properties.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Anticuerpos Monoclonales Humanizados/genética , Isótopos de Carbono , Disulfuros/química , Escherichia coli/genética , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Isopropil Tiogalactósido , Marcaje Isotópico , Isótopos de Nitrógeno , Periplasma/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Clin Cancer Res ; 26(14): 3694-3706, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32273277

RESUMEN

PURPOSE: The murine Lym-1 mAb targets a discontinuous epitope (Lym-1 epitope) on several subtypes of HLA-DR, which is upregulated in a majority of human B-cell lymphomas and leukemias. Unlike CD19, the Lym-1 epitope does not downregulate upon crosslinking, which may provide an advantage as a target for CAR T-cell therapy. Lym-1 CAR T cells with a conventional 4-1BB and CD3ζ (BB3z) signaling domain exhibited impaired ex vivo expansion. This study aimed to identify the underlying mechanisms and develop strategies to overcome this effect. EXPERIMENTAL DESIGN: A functional humanized Lym-1 antibody (huLym-1-B) was identified and its scFv form was used for CAR design. To overcome observed impaired expansion in vitro, a huLym-1-B CAR using DAP10 and DAP12 (DAP) signaling domains was evaluated for ex vivo expansion and in vivo function. RESULTS: Impaired expansion in huLym-1-B-BB3z CAR T cells was shown to be due to ligand-dependent suboptimal CAR signaling caused by interaction of the CAR binding domain and the surface of human T cells. Using the novel DAP signaling domain construct, the effects of suboptimal CAR signaling were overcome to produce huLym-1-B CAR T cells with improved expansion ex vivo and function in vivo. In addition, the Lym-1 epitope does not significantly downregulate in response to huLym-1-B-DAP CAR T cells both ex vivo and in vivo. CONCLUSIONS: DAP intracellular domains can serve as signaling motifs for CAR, and this new construct enables nonimpaired production of huLym-1-B CAR T cells with potent in vivo antitumor efficacy.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales de Origen Murino/genética , Línea Celular Tumoral , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Dominios Proteicos/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA