Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Methods Mol Biol ; 2793: 41-54, 2024.
Article in English | MEDLINE | ID: mdl-38526722

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Bacteriophages , Cetuximab , Point Mutation , ErbB Receptors/metabolism , Bacteriophages/metabolism , Antibodies, Monoclonal, Humanized/genetics
2.
Prep Biochem Biotechnol ; 53(3): 239-246, 2023.
Article in English | MEDLINE | ID: mdl-35579623

ABSTRACT

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.


Subject(s)
Antigens, CD20 , Single-Chain Antibodies , Humans , Antigens, CD20/genetics , Antigens, CD20/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/metabolism , Single-Chain Antibodies/genetics
3.
Dis Markers ; 2022: 3132941, 2022.
Article in English | MEDLINE | ID: mdl-36051360

ABSTRACT

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.


Subject(s)
Aminoglycosides , Leukemia, Myeloid, Acute , Sialic Acid Binding Ig-like Lectin 3 , Aminoglycosides/therapeutic use , Antibodies, Monoclonal, Humanized/genetics , Gemtuzumab/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Polymorphism, Single Nucleotide , Sialic Acid Binding Ig-like Lectin 3/genetics
4.
Front Immunol ; 13: 865875, 2022.
Article in English | MEDLINE | ID: mdl-35693833

ABSTRACT

Psoriasis is a worldwide chronic inflammatory skin disease. The treatment of disease is usually designed according to its severity. In this research, RNA-seq was performed on 37 patients with psoriasis treated with guselkumab before and after treatment, and the patients were divided into fast responder and slow responder according to PASI score to analyze the differentially expressed genes (DEGs) between them. Moreover, The biological mechanism of psoriasis was explored by Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Gene Ontology (GO) analysis, and Gene Set Enrichment Analysis (GSEA) analysis. And then, this protein-protein interaction network was constructed and 17 DEGs including IL-1ß, CXCL8, S100A12 and MMP9 were analyzed by GSVA. DEGs were detected by GO and KEGG analysis of target genes, which were primarily associated with immune response, neutrophil activation, neutrophil degranulation. GSEA reminded that fast responders were mainly involved in low-density neutrophils and abundant NK cells. And the GSVA showed that the DEGs were down-regulated after the early stage of the fast responder and the reverse in the slow responder by GSVA analysis. On the whole, these results indicated that these DEGs may serve as a psoriasis potential diagnostic and predictive biomarkers after been treated by guselkumab.


Subject(s)
Neutrophils , Psoriasis , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Gene Expression , Humans , Neutrophils/metabolism , Psoriasis/drug therapy , Psoriasis/genetics , Psoriasis/metabolism
5.
Cell Mol Biol Lett ; 27(1): 20, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236282

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Interleukin-1/genetics , Interleukin-1/pharmacology , Interleukin-1/therapeutic use , Mutation
6.
Sci Rep ; 11(1): 22365, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34785671

ABSTRACT

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.


Subject(s)
Amino Acid Substitution , Antibodies, Monoclonal, Humanized/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Variable Region/chemistry , Transforming Growth Factor beta2/chemistry , Transforming Growth Factor beta3/chemistry , Animals , Antibodies, Monoclonal, Humanized/genetics , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Variable Region/genetics , Protein Structure, Quaternary , Rabbits , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta3/genetics
7.
Sci Rep ; 11(1): 19500, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593880

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Fungi/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/isolation & purification , Antibody Specificity/immunology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Resistance, Fungal/drug effects , Enzyme-Linked Immunosorbent Assay , Genetic Engineering , Humans , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains/genetics , Mice , Microbial Sensitivity Tests , Phagocytosis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
8.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576184

ABSTRACT

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.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Cricetulus , Cytokines/genetics , Cytokines/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Fluorescent Antibody Technique , Immunotherapy , Mice, Inbred BALB C , Mutation , Protein Structure, Secondary , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/genetics
9.
Sci Rep ; 11(1): 18630, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34545109

ABSTRACT

While achieving rapid developments in recent years, bispecific antibodies are still difficult to design and manufacture, due to mispair of both heavy and light chains. Here we report a novel technology to make bispecific molecules. The knob-into-hole method was used to pair two distinct heavy chains as a heterodimer. IgG4 S228P CH1-CL interface was then partially replaced by T-cell receptor α/ß constant domain to increase the efficiency of cognate heavy and light chain pairing. Following expression and purification, the bispecific antibody interface exchange was confirmed by Western blotting and LC-MS/MS. To ensure its validity, we combined a monovalent bispecific antibody against PD-1 (sequence from Pembrolizumab) and LAG3 (sequence from Relatlimab). The results showed that the molecule could be assembled correctly at a ratio of 95% in cells. In vitro functional assay demonstrated that the purified bispecific antibody exhibits an enhanced agonist activity compared to that of the parental antibodies. Low immunogenicity was predicted by an open-access software and ADA test.


Subject(s)
Antibodies, Bispecific/biosynthesis , Immunoglobulin G/biosynthesis , Amino Acid Substitution , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antigens, CD/immunology , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , In Vitro Techniques , Male , Models, Molecular , Mutagenesis, Site-Directed , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Engineering/methods , Protein Multimerization , Protein Stability , Rats , Rats, Sprague-Dawley , Static Electricity , Lymphocyte Activation Gene 3 Protein
10.
Sci Rep ; 11(1): 15694, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344952

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Gene Expression , Genetic Therapy , Palivizumab/genetics , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses , Animals , Dependovirus/genetics , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Injections, Intramuscular , Lentivirus/genetics , Mice , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Transduction, Genetic , Treatment Outcome
11.
Microb Cell Fact ; 20(1): 134, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34261490

ABSTRACT

Monoclonal antibodies, antibody fragments and fusion proteins derived thereof have revolutionized the practice of medicine. Major challenges faced by the biopharmaceutical industry are however high production costs, long processing times and low productivities associated with their production in mammalian cell lines. The yeast Saccharomyces cerevisiae, a well-characterized eukaryotic cell factory possessing the capacity of post-translational modifications, has been industrially exploited as a secretion host for production of a range of products, including pharmaceuticals. However, due to the incompatible surface glycosylation, few antibody molecules have been functionally expressed in S. cerevisiae. Here, three non-glycosylated antibody fragments from human and the Camelidae family were chosen for expression in a S. cerevisiae strain (HA) previously evolved for high α-amylase secretion. These included the Fab fragment Ranibizumab (Ran), the scFv peptide Pexelizumab (Pex), and a nanobody consisting of a single V-type domain (Nan). Both secretion and biological activities of the antibody fragments were confirmed. In addition, the secretion level of each protein was compared in the wild type (LA) and two evolved strains (HA and MA) with different secretory capacities. We found that the secretion of Ran and Nan was positively correlated with the strains' secretory capacity, while Pex was most efficiently secreted in the parental strain. To investigate the mechanisms for different secretion abilities in these selected yeast strains for the different antibody fragments, RNA-seq analysis was performed. The results showed that several bioprocesses were significantly enriched for differentially expressed genes when comparing the enriched terms between HA.Nan vs. LA.Nan and HA.Pex vs. LA.Pex, including amino acid metabolism, protein synthesis, cell cycle and others, which indicates that there are unique physiological needs for each antibody fragment secretion.


Subject(s)
Gene Expression , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/immunology , Single-Domain Antibodies/genetics , Antibodies, Monoclonal, Humanized/genetics , Glycosylation , Protein Processing, Post-Translational , Protein Transport , Ranibizumab/genetics , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/metabolism , Single-Chain Antibodies/genetics
12.
Signal Transduct Target Ther ; 6(1): 236, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34158475

ABSTRACT

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.


Subject(s)
Breast Neoplasms/drug therapy , HLA-G Antigens/genetics , Receptor, ErbB-2/genetics , Receptors, KIR2DL4/genetics , Trastuzumab/pharmacology , Adult , Aged , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Humans , Interferon-gamma/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Middle Aged , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Trastuzumab/adverse effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
13.
Nanomedicine ; 36: 102424, 2021 08.
Article in English | MEDLINE | ID: mdl-34174417

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Cytotoxins , D-Amino-Acid Oxidase , Fibronectins , Neoplasm Proteins , Neoplasms/drug therapy , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Cytotoxins/chemistry , Cytotoxins/pharmacology , D-Amino-Acid Oxidase/chemistry , D-Amino-Acid Oxidase/genetics , D-Amino-Acid Oxidase/pharmacology , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology
14.
Clin Cancer Res ; 27(17): 4685-4689, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34083238

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Drug Approval , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Adult , Child , Humans , Retrospective Studies , United States
15.
Mol Immunol ; 135: 320-328, 2021 07.
Article in English | MEDLINE | ID: mdl-33971510

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Recombinant Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibody Affinity/immunology , CHO Cells , Cell Line, Tumor , Cricetulus , Female , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Molecular Docking Simulation , Protein Conformation , Protein Transport/genetics , Protein Transport/physiology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Recombinant Proteins/immunology
16.
Monoclon Antib Immunodiagn Immunother ; 40(2): 52-59, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33900821

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Forkhead Transcription Factors/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized/genetics , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/genetics , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunotherapy/trends , Neoplasms/immunology , Neoplasms/therapy , Th1 Cells/immunology , Th17 Cells/immunology
17.
Exp Biol Med (Maywood) ; 246(8): 940-951, 2021 04.
Article in English | MEDLINE | ID: mdl-33475433

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Arthritis, Experimental/drug therapy , Interleukin-9/pharmacology , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal, Humanized/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Drug Delivery Systems , Drug Evaluation , Interleukin-9/genetics , Male , Mice , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics
18.
MAbs ; 13(1): 1868066, 2021.
Article in English | MEDLINE | ID: mdl-33404287

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Killer Cells, Natural/drug effects , Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Disease Progression , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
19.
Front Immunol ; 11: 604318, 2020.
Article in English | MEDLINE | ID: mdl-33365032

ABSTRACT

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.


Subject(s)
Amino Acids/metabolism , Antibodies, Monoclonal, Humanized/biosynthesis , Antineoplastic Agents, Immunological/metabolism , Biotechnology , Immunoglobulin E/biosynthesis , Immunoglobulin Variable Region , Protein Engineering , Protein Sorting Signals , Trastuzumab/biosynthesis , Antibodies, Monoclonal, Humanized/genetics , Culture Media/metabolism , HEK293 Cells , Humans , Immunoglobulin E/genetics , Immunoglobulin Variable Region/genetics , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Protein Sorting Signals/genetics , Recombinant Proteins/biosynthesis , Trastuzumab/genetics , Workflow
20.
MAbs ; 12(1): 1829335, 2020.
Article in English | MEDLINE | ID: mdl-33103593

ABSTRACT

The early phase of protein drug development has traditionally focused on target binding properties leading to a desired mode of therapeutic action. As more protein therapeutics pass through the development pipeline; however, it is clear that non-optimal biophysical properties can emerge, particularly as proteins are formulated at high concentrations, causing aggregation or polyreactivity. Such late-stage "developability" problems can lead to delay or failure in traversing the development process. Aggregation propensity is also correlated with increased immunogenicity, resulting in expensive, late-stage clinical failures. Using nucleases-directed integration, we have constructed large mammalian display libraries where each cell contains a single antibody gene/cell inserted at a single locus, thereby achieving transcriptional normalization. We show a strong correlation between poor biophysical properties and display level achieved in mammalian cells, which is not replicated by yeast display. Using two well-documented examples of antibodies with poor biophysical characteristics (MEDI-1912 and bococizumab), a library of variants was created based on surface hydrophobic and positive charge patches. Mammalian display was used to select for antibodies that retained target binding and permitted increased display level. The resultant variants exhibited reduced polyreactivity and reduced aggregation propensity. Furthermore, we show in the case of bococizumab that biophysically improved variants are less immunogenic than the parental molecule. Thus, mammalian display helps to address multiple developability issues during the earliest stages of lead discovery, thereby significantly de-risking the future development of protein drugs.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibody Affinity/genetics , Cell Surface Display Techniques , HEK293 Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...