Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Cell Biochem Biophys ; 81(3): 395-408, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37395856

ABSTRACT

Several decades of research and clinical trials have conclusively provided proof of concept on the usefulness of monoclonal antibodies in the armamentarium against cancer. There are numerous mAbs approved for both, the treatment of solid tumors as well as hematological malignancies. These have ranked in the top ten best-selling drugs in recent years and one such mAb, pembrolizumab, is slated to be the highest revenue-generating drug by 2024. A large proportion of the mAbs in oncology have been approved by regulatory agencies in just the past decade and many professionals working in the field have been unable to keep abreast with the latest mAbs available and their mechanism of action. In this review, we aim to provide a systematic compilation of the various mAbs in oncology, approved by the US FDA in the past decade. It also elaborates on the mechanism of action of the newly approved mAbs to provide an overall update of the same. For this purpose, we have referred to the Drugs at FDA and relevant articles from PubMed from the year 2010 to date.


Subject(s)
Antineoplastic Agents, Immunological , Neoplasms , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Humans , Neoplasms/drug therapy , Antigens, Neoplasm/metabolism , Tumor Microenvironment , Receptors, Growth Factor/antagonists & inhibitors , Antigens, CD/metabolism , Immune Checkpoint Proteins/metabolism , Animals
2.
Br J Oral Maxillofac Surg ; 61(4): 320-326, 2023 05.
Article in English | MEDLINE | ID: mdl-37061418

ABSTRACT

The immune checkpoint inhibitor (ICI), nivolumab, has revolutionised the treatment of recurrent and metastatic oral cancer. However, the response rate to ICIs remains low, and identifying predictors of nivolumab response is critical. Although the neutrophil-to-lymphocyte ratio (NLR) has been suggested as a predictive marker of nivolumab response in patients with various types of cancer, its utility in oral squamous cell carcinoma (OSCC) has not been elucidated. In this retrospective multicentre cohort study, we evaluated the association between NLR and outcome of nivolumab treatment in 64 patients with OSCC treated between 2017 and 2020. The objective response and disease control rates were 25.1% and 32.9%, respectively. The rates for complete and partial responses were 15.7% (10/64) and 9.4% (6/64), respectively; stable and progressive disease rates were 7.8% (5/64) and 67.1% (43/64), respectively. Complete and partial responses were classified as responders, and stable and progressive diseases were classified as non-responders. The median (range) pre-treatment NLR among responders was 4.3 (2.8-8.0), which decreased to 4.0 (2.6-6.3) after nivolumab treatment, and the median (range) pre-treatment NLR among non-responders was 5.1 (2.7-7.9), which increased to 6.4 (4.0-14.0) with tumour growth. Moreover, overall survival was significantly worse in the group with a higher post-treatment NLR (≥5) than in the group with a lower NLR (<5). Patients with a post-treatment NLR of ≥6 had worse outcomes for salvage chemotherapy following nivolumab treatment. Thus, post-treatment NLR could be a useful marker for predicting the response to nivolumab treatment or salvage chemotherapy in patients with OSCC.


Subject(s)
Antineoplastic Agents, Immunological , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Nivolumab/therapeutic use , Nivolumab/metabolism , Carcinoma, Squamous Cell/pathology , Neutrophils/metabolism , Neutrophils/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Cohort Studies , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/metabolism , Prognosis , Retrospective Studies , Mouth Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Lymphocytes/pathology , Chronic Disease , Head and Neck Neoplasms/pathology
3.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34969858

ABSTRACT

Brain metastases are a leading cause of death in patients with breast cancer. The lack of clinical trials and the presence of the blood-brain barrier limit therapeutic options. Furthermore, overexpression of the human epidermal growth factor receptor 2 (HER2) increases the incidence of breast cancer brain metastases (BCBM). HER2-targeting agents, such as the monoclonal antibodies trastuzumab and pertuzumab, improved outcomes in patients with breast cancer and extracranial metastases. However, continued BCBM progression in breast cancer patients highlighted the need for novel and effective targeted therapies against intracranial metastases. In this study, we engineered the highly migratory and brain tumor tropic human neural stem cells (NSCs) LM008 to continuously secrete high amounts of functional, stable, full-length antibodies against HER2 (anti-HER2Ab) without compromising the stemness of LM008 cells. The secreted anti-HER2Ab impaired tumor cell proliferation in vitro in HER2+ BCBM cells by inhibiting the PI3K-Akt signaling pathway and resulted in a significant benefit when injected in intracranial xenograft models. In addition, dual HER2 blockade using anti-HER2Ab LM008 NSCs and the tyrosine kinase inhibitor tucatinib significantly improved the survival of mice in a clinically relevant model of multiple HER2+ BCBM. These findings provide compelling evidence for the use of HER2Ab-secreting LM008 NSCs in combination with tucatinib as a promising therapeutic regimen for patients with HER2+ BCBM.


Subject(s)
Antineoplastic Agents, Immunological/metabolism , Brain Neoplasms , Neoplasms, Experimental , Neural Stem Cells , Oxazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2 , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
4.
Nat Med ; 27(12): 2200-2211, 2021 12.
Article in English | MEDLINE | ID: mdl-34893775

ABSTRACT

In a recent phase 3 randomized trial of 700 patients with advanced urothelial cancer (JAVELIN Bladder 100; NCT02603432 ), avelumab/best supportive care (BSC) significantly prolonged overall survival relative to BSC alone as maintenance therapy after first-line chemotherapy. Exploratory biomarker analyses were performed to identify biological pathways that might affect survival benefit. Tumor molecular profiling by immunohistochemistry, whole-exome sequencing and whole-transcriptome sequencing revealed that avelumab survival benefit was positively associated with PD-L1 expression by tumor cells, tumor mutational burden, APOBEC mutation signatures, expression of genes underlying innate and adaptive immune activity and the number of alleles encoding high-affinity variants of activating Fcγ receptors. Pathways connected to tissue growth and angiogenesis might have been associated with reduced survival benefit. Individual biomarkers did not comprehensively identify patients who could benefit from therapy; however, multi-parameter models incorporating genomic alteration, immune responses and tumor growth showed promising predictive utility. These results characterize the complex biologic pathways underlying survival benefit from immune checkpoint inhibition in advanced urothelial cancer and suggest that multiple biomarkers might be needed to identify patients who would benefit from treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/metabolism , Biomarkers, Tumor/metabolism , Urinary Bladder Neoplasms/drug therapy , Humans , Mutation , Urinary Bladder Neoplasms/pathology
5.
Sci Rep ; 11(1): 22966, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824364

ABSTRACT

High tumor regulatory T (Treg) cell infiltration is associated with poor prognosis of many cancers. CD25 is highly expressed on tumor Treg cells and is a potential target for Treg deletion. Previously characterized anti-CD25 antibodies appear to have limited efficacy in tumor inhibition. Here we identified two human anti-CD25 antibodies, BA9 and BT942, which did not prevent the activation of IL-2R signaling pathway by IL-2. BT942 had weaker binding and cytotoxic activity to human CD25-expressing cell lines than BA9. But both demonstrated significant tumor growth inhibition in early and late-stage animal cancer models. BT942 resulted in a higher expansion of CD8+ T cells and CD4+ T cells in tumor microenvironment in mouse MC38 model compared to BA9. BT942 also demonstrated significant higher tumor growth inhibition and higher expansion of CD8+ T cells and CD4+ T cells in combination with an anti-PD1 antibody. Pharmacokinetic study of BT942 in cynomolgus monkeys demonstrated a half-life of 206.97 ± 19.03 h. Structural analysis by cryo-EM revealed that BT942 recognizes an epitope on opposite side of the CD25-IL-2 binding site, consistent with no IL-2 signaling blockade in vitro. BT942 appears to be an excellent candidate for cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological , Interleukin-2 Receptor alpha Subunit/immunology , Neoplasms , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/pharmacology , Cell Line , Cryoelectron Microscopy , Disease Models, Animal , Humans , Immunotherapy/methods , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Macaca fascicularis , Mice , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
6.
Cell Rep ; 37(5): 109953, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731630

ABSTRACT

Receptor clustering is the first and critical step to activate apoptosis by death receptor-5 (DR5). The recent discovery of the autoinhibitory DR5 ectodomain has challenged the long-standing view of its mechanistic activation by the natural ligand Apo2L. Because the autoinhibitory residues have remained unknown, here we characterize a crucial patch of positively charged residues (PPCR) in the highly variable domain of DR5. The PPCR electrostatically separates DR5 receptors to autoinhibit their clustering in the absence of ligand and antibody binding. Mutational substitution and antibody-mediated PPCR interference resulted in increased apoptotic cytotoxic function. A dually specific antibody that enables sustained tampering with PPCR function exceptionally enhanced DR5 clustering and apoptotic activation and distinctively improved the survival of animals bearing aggressive metastatic and recurrent tumors, whereas clinically tested DR5 antibodies without PPCR blockade function were largely ineffective. Our study provides mechanistic insights into DR5 activation and a therapeutic analytical design for potential clinical success.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , A549 Cells , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Specificity , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Epitopes , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Int Immunopharmacol ; 100: 108112, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34521023

ABSTRACT

More than 100 monoclonal antibodies (mAbs) have been approved by FDA. The mechanism of action (MoA) involves in neutralization of a specific target via the Fab region and Fc effector functions through Fc region, while the latter include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). ADCP has been recognized one of the most important MoAs, especially for anti-cancer mAbs in recent years. However, traditional bioassays measuring ADCP always introduced primary macrophages and flow cytometry, which are difficult to handle and highly variable. In this study, we engineered a monoclonal Jurkat/NFAT/CD32a-FcεRIγ effector cell line that stably expresses CD32a-FcεRIγ chimeric receptor and NFAT-controlled luciferase. The corresponding mAb could bind with the membrane antigens on the target cells with its Fab fragment and CD32a-FcεRIγ on the effector cells with its Fc fragment, leading to the crosslinking of CD32a-FcεRIγ and the resultant expression of subsequent NFAT-controlled luciferase, which represents the bioactivity of ADCP based on the MoA of the mAb. With rituximab as the model mAb, Raji cells as the target cells, and Jurkat/NFAT/CD32a-FcεRIγ cells as the effector cells, we adopted the strategy of Design of Experiment (DoE) to optimize the bioassay. Then we fully validated the established bioassay according to ICH-Q2(R1), which proved the good assay performance characteristics of the bioassay, including specificity, accuracy, precision, linearity, stability and robustness. This RGA can be applied to evaluate the -ADCP bioactivity for anti-CD20 mAbs in lot release, stability testing as well as biosimilar comparability. The engineered cells may also potentially be used to evaluate the ADCP bioactivity of mAbs with other targets.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents, Immunological/pharmacology , Biological Assay , Genes, Reporter , Lymphoma, B-Cell/drug therapy , Phagocytosis/drug effects , Rituximab/pharmacology , Antineoplastic Agents, Immunological/metabolism , Humans , Jurkat Cells , Luciferases/genetics , Luciferases/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Receptors, IgE/genetics , Receptors, IgE/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Reproducibility of Results , Rituximab/metabolism
8.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1625-1639, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34586349

ABSTRACT

Mucin 1 (MUC1) has been regarded as an ideal target for cancer treatment, since it is overexpressed in a variety of different cancers including the majority of breast cancer. However, there are still no approved monoclonal antibody drugs targeting MUC1. In this study, we generated a humanized MUC1 (HzMUC1) antibody from our previously developed MUC1 mouse monoclonal antibody that only recognizes MUC1 on the surface of tumor cells. Furthermore, an antibody-drug conjugate (ADC) was generated by conjugating HzMUC1 with monomethyl auristatin (MMAE), and the efficacy of HzMUC1-MMAE on the MUC1-positive HER2+ breast cancer in vitro and in 'Xenograft' model was tested. Results from western blot analysis and immunoprecipitation revealed that the HzMUC1 antibody did not recognize cell-free MUC1-N in sera from breast cancer patients. Confocal microscopy analysis showed that HzMUC1 antibody bound to MUC1 on the surface of breast cancer cells. Results from mapping experiments suggested that HzMUC1 may recognize an epitope present in the interaction region between MUC1-N and MUC1-C. Results from colony formation assay and flow cytometry demonstrated that HzMUC1-MMAE significantly inhibited cell growth by inducing G2/M cell cycle arrest and apoptosis in trastuzumab-resistant HER2-positive breast cancer cells. Meanwhile, HzMUC1-MMAE significantly reduced the growth of HCC1954 xenograft tumors by inhibiting cell proliferation and enhancing cell death. In conclusion, our results indicate that HzMUC1-ADC is a novel therapeutic drug that can overcome trastuzumab resistance of breast cancer. HzMUC1-ADC should also be an effective therapeutic drug for the treatment of different MUC1-positive cancers in clinic.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Immunoconjugates/pharmacology , Mucin-1/metabolism , Trastuzumab/pharmacology , Animals , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/immunology , Epitopes , Humans , Immunoconjugates/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Mucin-1/blood , Mucin-1/chemistry , Mucin-1/immunology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
9.
Biomed Pharmacother ; 141: 111913, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34328096

ABSTRACT

BACKGROUND: B7-H4 is a member of the B7 superfamily that is expressed on the surface of tumors and exhibits limited expression on normal tissue. B7-H4 negatively regulates tumor immunity by interacting with the B7-H4 receptor, which is expressed by activated CD8 + T cells. Hence, we sought to generate an immunomodulatory antibody that targets B7-H4 and blocks the immunosuppressive activity of B7-H4. METHODS: Anti-B7-H4 antibodies were generated using the hybridoma technique and screened by a binding assay based on B7-H4-expressing tumor cells. The B7-H4 antagonistic antibodies were further screened based on their checkpoint blockade activity using a SEB-stimulated peripheral blood mononuclear cell (PBMC) assay, which comprised B7-H4-expressing antigen presenting cells (APCs) and activated T cells. To assess the immunomodulatory activity of anti-B7-H4 antibodies, activated human CD8+ T cells were cultured in B7-H4 protein-coated plates, and the production of IL-2 and the proliferation rate of CD8+ T cells were measured. In addition, we evaluated the ADCC effect of anti-B7-H4 antibodies against tumor cell lines. The in vivo antitumor efficacy of the anti-B7-H4 antibody was also evaluated in human T cell-engrafted NOG mice. RESULTS: A panel of anti-B7-H4 antibodies was generated. The top 23 antibodies were screened to identify antibodies that disabled B7-H4-mediated inhibition. Antibody 17 exhibited the greatest induction of the production of IL-2 and IFN-gamma in SEB-stimulated PBMCs. Antibody 17 was constructed as a chimeric antibody (CH17) with a human IgG1 constant domain. CH17 showed high affinity for human B7-H4 and fully cross-reacted with cynomolgus B7-H4. Additionally, CH17 mediated potent antibody-dependent cell cytotoxicity (ADCC) against different B7-H4-positive tumor cell lines. More importantly, CH17 relieved B7-H4-mediated T cell suppression by enhancing IL2 production and promoting T cell proliferation. In an MDA-MB-468-bearing mouse model in which human pan-T cells were engrafted, CH17 delayed tumor growth by engaging T cells and exerted a synergistic effect in combination with an anti-human PD-1 antibody. CONCLUSIONS: We successfully generated an immunomodulatory antibody targeting B7-H4 that possesses both T cell immune checkpoint inhibitory activity and ADCC activity in B7-H4-positive tumors. B7-H4-targeting antibodies might represent a promising immunotherapy for B7-H4-expressing tumors.


Subject(s)
Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/antagonists & inhibitors , V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology , Animals , Antineoplastic Agents, Immunological/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Immunity, Cellular/drug effects , Immunotherapy/methods , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Xenograft Model Antitumor Assays/methods
10.
Mol Biotechnol ; 63(11): 1016-1029, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34185248

ABSTRACT

Plants are promising drug-production platforms with high economic efficiency, stability, and convenience in mass production. However, studies comparing the equivalency between the original antibodies and those produced in plants are limited. Amino acid sequences that constitute the Fab region of an antibody are diverse, and the post-transcriptional modifications that occur according to these sequences in animals and plants are also highly variable. In this study, rituximab, a blockbuster antibody drug used in the treatment of non-Hodgkin's lymphoma, was produced in Nicotiana benthamiana leaves and Arabidopsis thaliana callus, and was compared to the original rituximab produced in CHO cells. Interestingly, the epitope recognition and antigen-binding abilities of rituximab from N. benthamiana leaves were almost lost. In the case of rituximab produced in A. thaliana callus, the specific binding ability and CD20 capping activity were maintained, but the binding affinity was less than 50% of that of original rituximab from CHO cells. These results suggest that different plant species exhibit different binding affinities. Accordingly, in addition to the differences in PTMs between mammals and plants, the differences between the species must also be considered in the process of producing antibodies in plants.


Subject(s)
Antigens, CD20/metabolism , Arabidopsis/metabolism , Nicotiana/metabolism , Plant Leaves/chemistry , Rituximab/metabolism , Animals , Antibody Affinity , Antigens, CD20/chemistry , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/metabolism , Arabidopsis/genetics , Cricetinae , Humans , Plant Leaves/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Rituximab/biosynthesis , Rituximab/genetics , Rituximab/isolation & purification , Nicotiana/genetics
11.
Theranostics ; 11(13): 6293-6314, 2021.
Article in English | MEDLINE | ID: mdl-33995659

ABSTRACT

Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.


Subject(s)
Radioimmunotherapy/methods , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Chelating Agents/administration & dosage , Chelating Agents/metabolism , Click Chemistry , Clinical Trials as Topic , Dose Fractionation, Radiation , Drug Delivery Systems , Forecasting , Humans , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/therapeutic use , Lymphoma, Non-Hodgkin/radiotherapy , Mice , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/radiotherapy , Organ Specificity , Precision Medicine , Radiation Tolerance , Radiopharmaceuticals/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/therapeutic use , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/therapeutic use , Yttrium Radioisotopes/administration & dosage , Yttrium Radioisotopes/therapeutic use
12.
FEBS Lett ; 595(11): 1587-1603, 2021 06.
Article in English | MEDLINE | ID: mdl-33792041

ABSTRACT

OX40 is a costimulatory molecule that belongs to the tumor necrosis factor receptor (TNFR) superfamily. OX40 agonist-based combinations are emerging as promising candidates for novel cancer immunotherapy. Clinical trials have shown that OX40 agonist antibodies could lead to better results in cancer patients. Using a hybridoma platform and three different types of immunization strategies, namely recombinant protein, DNA, and overexpressing cells, we identified a chimeric anti-OX40 antibody (mAb035-hIgG1 from DNA immunization) that shows excellent binding specificity, and slightly stronger activation of human memory CD4+ T cells and similar potent antitumor activity compared with BMS 986178, an anti-OX40 antibody currently being evaluated for the treatment of solid tumors. This paper further systematically investigates the antigen-specific immune response, the number of binders, epitope bins, and functional activities of antibodies among different immunization strategies. Interestingly, we found that different immunization strategies affect the biological activity of monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Immunization/methods , Receptors, OX40/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibody Affinity , Antibody Specificity , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/metabolism , Biological Assay , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CHO Cells , Cricetulus , Female , Freund's Adjuvant/administration & dosage , Gene Expression , Genes, Reporter , HEK293 Cells , Humans , Hybridomas/chemistry , Hybridomas/immunology , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin Fc Fragments/isolation & purification , Immunoglobulin Fc Fragments/pharmacology , Jurkat Cells , Luciferases/genetics , Luciferases/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification
13.
Chem Commun (Camb) ; 57(28): 3457-3460, 2021 Apr 11.
Article in English | MEDLINE | ID: mdl-33687404

ABSTRACT

A novel enzyme cleavable linker for antibody-drug conjugates is reported. The 3-O-sulfo-ß-galactose linker is cleaved sequentially by two lysosomal enzymes - arylsulfatase A and ß-galactosidase - to release the payload in targeted cells. An α-HER2 antibody-drug conjugate synthesised using this highly hydrophilic dual-cleavable linker exhibited excellent cytotoxicity and selectivity.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cerebroside-Sulfatase/chemistry , Immunoconjugates/chemistry , Trastuzumab/pharmacology , beta-Galactosidase/chemistry , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/metabolism , Cell Survival/drug effects , Cerebroside-Sulfatase/metabolism , Dose-Response Relationship, Drug , Humans , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/metabolism , Molecular Structure , Trastuzumab/chemistry , Trastuzumab/metabolism , beta-Galactosidase/metabolism
14.
Commun Biol ; 4(1): 310, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33686177

ABSTRACT

Fc-less bispecific T-cell engagers have reached the immuno-oncology market but necessitate continual infusion due to rapid clearance from the circulation. This work introduces a programmable serum half-life extension platform based on fusion of human albumin sequences engineered with either null (NB), wild type (WT) or high binding (HB) FcRn affinity combined with a bispecific T-cell engager. We demonstrate in a humanised FcRn/albumin double transgenic mouse model (AlbuMus) the ability to tune half-life based on the albumin sequence fused with a BiTE-like bispecific (anti-EGFR nanobody x anti-CD3 scFv) light T-cell engager (LiTE) construct [(t½ 0.6 h (Fc-less LiTE), t½ 19 hours (Albu-LiTE-NB), t½ 26 hours (Albu-LiTE-WT), t½ 37 hours (Albu-LiTE-HB)]. We show in vitro cognate target engagement, T-cell activation and discrimination in cellular cytotoxicity dependent on EGFR expression levels. Furthermore, greater growth inhibition of EGFR-positive BRAF mutated tumours was measured following a single dose of Albu-LiTE-HB construct compared to the Fc-less LiTE format and a full-length anti-EGFR monoclonal antibody in a new AlbuMus RAG1 knockout model introduced in this work. Programmable half-life extension facilitated by this albumin platform potentially offers long-lasting effects, better patient compliance and a method to tailor pharmacokinetics to maximise therapeutic efficacy and safety of immuno-oncology targeted biologics.


Subject(s)
Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Histocompatibility Antigens Class I/metabolism , Neoplasms/drug therapy , Receptors, Fc/metabolism , Serum Albumin, Human/pharmacokinetics , T-Lymphocytes/drug effects , 3T3 Cells , Animals , Antibodies, Bispecific/metabolism , Antineoplastic Agents, Immunological/metabolism , CHO Cells , Cricetulus , Drug Compounding , Female , HEK293 Cells , HT29 Cells , Half-Life , Homeodomain Proteins/genetics , Humans , Jurkat Cells , Lymphocyte Activation/drug effects , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/pathology , Proof of Concept Study , Protein Binding , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Serum Albumin, Human/genetics , Serum Albumin, Human/metabolism , T-Lymphocytes/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Cancer Med ; 10(6): 2125-2136, 2021 03.
Article in English | MEDLINE | ID: mdl-33629544

ABSTRACT

BACKGROUND: CD47 is a widely expressed transmembrane protein located on the surface of somatic cells. It mediates a variety of cellular processes including apoptosis, proliferation, adhesion, and migration. An important role for CD47 is the transmission of a "Don't eat me" signal by interacting with SIRPα on the macrophage surface membrane, thereby preventing the phagocytosis of normal cells. However, cancer cells can take advantage of this autogenous signal to protect themselves from phagocytosis, thus enabling immune escape. Blocking the interaction between CD47 and SIRPα has proven to be effective in removing cancer cells. The treatment of various cancers with CD47 monoclonal antibodies has also been validated. METHODS: We designed and synthesized a peptide (RS17), which can specifically bind to CD47 and block CD47-SIRPα signaling. The affinity of RS17 for CD47-expressing tumor cells was determined, while the inhibition of CD47-SIRPα signaling was evaluated in vitro and in vivo. RESULTS: The results indicated that RS17 significantly promotes the phagocytosis of tumor cells by macrophages and had a similar therapeutic effect compared with a positive control (CD47 monoclonal antibodies). In addition, a cancer xenograft mouse model was established using CD47-expressing HepG2 cells to evaluate the effect of RS17 on tumor growth in vivo. Using ex vivo and in vivo mouse models, RS17 demonstrated a high inhibitory effect on tumor growth. CONCLUSIONS: Based on our results, RS17 may represent a novel therapeutic peptide for cancer therapy.


Subject(s)
Antigens, Differentiation/metabolism , Antineoplastic Agents, Immunological/metabolism , CD47 Antigen/metabolism , Macrophages/metabolism , Peptides/metabolism , Phagocytosis , Receptors, Immunologic/metabolism , Amino Acids/metabolism , Animals , Antibodies, Monoclonal , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/chemistry , Carcinoma, Squamous Cell/metabolism , Cell Culture Techniques , Cell Line, Tumor , Female , Flow Cytometry , Hep G2 Cells/metabolism , Humans , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Peptides/chemical synthesis , Peptides/therapeutic use , Phagocytosis/immunology , Skin Neoplasms/metabolism , Software , Tumor Escape , Xenograft Model Antitumor Assays
16.
J Cell Physiol ; 236(6): 4303-4312, 2021 06.
Article in English | MEDLINE | ID: mdl-33421131

ABSTRACT

The overexpression of epidermal growth factor receptor (EGFR) could result in the development of solid tumors of prostate, breast, gastric, colorectal, ovarian, and head and neck, leading to carcinoma. Antibody therapies are ideal methods to overcome malignant diseases. However, immunoribonucleases are a new generation of antibodies in which an RNase binds to a specific antibody and shows a stronger ability to terminate cancer cells. In this study, we engineered Rana pipiens RNase to bind to the scFv of human antiepidermal growth factor receptor antibody. The molecular dynamic simulations confirmed protein stability and the ability of scFv-ranpirnase (rantoxin) to bind to epidermal growth factor receptor protein. Then, the rantoxin construct was synthesized in a pCDNA 3.1 Neo vector. CHO-K1 cells were used as expression hosts and the construct was transfected. Cells were selected by antibiotic therapies using neomycin, 120 mg/ml, and the high-yield colony was screened by real-time polymerase chain reaction (PCR) methods. Then, the recombinant protein production was confirmed using the sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blot analyses. The molecular dynamic simulation (MDS) confirmed that the I467, S468, Q408, and H409 amino acids of EGFR bonded well to rantoxin. As revealed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analyses, the rantoxin production and PCR analysis showed that the T3 colony can produce rantoxin messenger RNA fourfold higher than the GAPDH gene. The immunotoxin function was assessed in A431 cancer cells and EGFR-negative HEK293 cells, and IC50  values were estimated to be 22.4 ± 3 and >620.4 ± 5 nM, respectively. The results indicated that the immunotoxins produced in this study had the potential for use as anticancer drugs.


Subject(s)
Amphibian Proteins/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunotoxins/pharmacology , Protein Engineering , Ribonucleases/pharmacology , Single-Chain Antibodies/pharmacology , Skin Neoplasms/drug therapy , Amphibian Proteins/genetics , Amphibian Proteins/metabolism , Animals , Antineoplastic Agents, Immunological/metabolism , Apoptosis/drug effects , Binding Sites, Antibody , CHO Cells , Cell Line, Tumor , Cricetulus , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , ErbB Receptors/metabolism , HEK293 Cells , Humans , Immunotoxins/genetics , Immunotoxins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Rana pipiens , Ribonucleases/genetics , Ribonucleases/metabolism , Single-Chain Antibodies/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/pathology
17.
Med Sci (Paris) ; 37(1): 47-52, 2021 Jan.
Article in French | MEDLINE | ID: mdl-33492218

ABSTRACT

Research on viruses, bacteria and protozoa-based immunotherapy has been on the rise for several years. The antitumoral efficacy of these microorganisms relies on three main mechanisms: Destruction of tumor cells, stimulation of the immune response and reprogramming of the tumor microenvironment. In order to optimize their immunotherapeutic action, these microorganisms can be genetically engineered to enhance their tumor-targeting efficacy or to vectorize immunostimulating molecules and/or antibodies. To this aim, molecular engineering allows the design of new antibody formats optimizing their functions. From whole antibodies to tandem single-chain variable fragments, various antibody formats can be vectorized by microorganisms to target receptors such as immune checkpoints or recruit immune effector cells within the tumor. Such possibilities broaden the arsenal of immunotherapeutic cancer treatment. This review focuses on these innovations and their advantages for immunotherapy.


TITLE: Micro-organismes anti-cancéreux et armement - Le couteau suisse de l'immunothérapie. ABSTRACT: Depuis plusieurs années, la recherche sur les micro-organismes pour une utilisation à des fins d'immunothérapie antitumorale est en plein essor. L'efficacité antitumorale de ces micro-organismes repose sur trois mécanismes principaux : la destruction des cellules tumorales, la stimulation du système immunitaire et la reprogrammation du microenvironnement tumoral. Afin d'optimiser leur action immunothérapeutique, ces micro-organismes peuvent être génétiquement modifiés pour les rendre capables de vectoriser des molécules immunostimulantes ou des anticorps. Par ingénierie moléculaire, il est désormais possible de diversifier les formats et fonctions de ces anticorps afin d'inhiber les points de contrôle immunitaire ou encore de recruter les cellules immunitaires effectrices au site de la tumeur. Cette Synthèse s'intéresse particulièrement à ces innovations et à leurs avantages en immunothérapie.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Immunotherapy , Microorganisms, Genetically-Modified/physiology , Animals , Antineoplastic Agents, Immunological/metabolism , Genetic Therapy/methods , Genetic Therapy/trends , Genetic Vectors/therapeutic use , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/genetics , Immunologic Factors/metabolism , Immunotherapy/methods , Immunotherapy/trends , Microorganisms, Genetically-Modified/genetics , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/therapy , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
18.
Biomed Pharmacother ; 133: 111053, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378959

ABSTRACT

Trastuzumab is considered to be a fundamental drug for treatment of breast cancer with Her-2 overexpression (Her-2 positive cells). Trastuzumab is a monoclonal antibody that targets the Her-2 receptor. Trastuzumab treatment used in breast cancer therapy require a visualization to validate their delivery and response. The objective of this study was to investigate Trastuzumab-dendrimer-fluorine drug delivery system by synthesis and characterization of a series of fluorinated dendrimers. MATERIALS AND METHODS: Trastuzumab-dendrimer-fluorine drug delivery system is a covalent attachment of Trastuzumab to fluorinated dendrimers. We design synthesis and evaluate main product by using electrophoresis, HPLC and LC-MS techniques. We prepared three-dimensional breast cancer cell culture in bioreactor device. For the cell culture we used MCF-7 cells with Her-2 overexpression to study Trastuzumab-dendrimer-fluorine drug delivery system efficacy. We evaluate efficacy by Magnetic Resonance Imaging (MRI) relaxation time. RESULTS: An analytical analysis showed that synthesis of Trastuzumab-dendrimer-fluorine drug delivery system is possible to obtain with a good yield. The results obtained indicated potential of Trastuzumab-dendrimer-fluorine drug delivery system is more efficient than trastuzumab alone. Chromatographic and electrophoretic separations showed that the synthetized conjugates were a Trastuzumab-dendrimer-fluorine drug delivery systems. The hight synthesis efficiency was found. The presence of molecules with lower masses than trastuzumab can have influence on efficiency. CONCLUSIONS: Trastuzumab-dendrimer-fluorine drug delivery system is a new form of Trastuzumab to treat breast cancer cells in vitro. Due to presence of 19F nuclei the system can be monitored by MRI measurements.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Dendrimers/chemistry , Drug Carriers , Fluorine/chemistry , Trastuzumab/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/metabolism , Bioreactors , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Cell Survival/drug effects , Coculture Techniques , Drug Compounding , Female , Humans , MCF-7 Cells , Magnetic Resonance Imaging , Trastuzumab/chemistry , Trastuzumab/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.
Cancer Biol Med ; 17(4): 1026-1038, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33299651

ABSTRACT

Objective: Natural killer (NK) cells have gained considerable attention due to their potential in treating "cold tumors," and are therefore considered as one of the new strategies for curing cancer, by using worldwide development of their new possibilities and interventions with NK cell-related therapeutic products. Methods: We constructed a trispecific killer engager (TriKE) consisting of anti-CD16, IL-15, and anti-CD19. This TriKE was designed to attract CD19+ tumor cells to CD16+ NK cells, whereas IL-15 sustained the proliferation, development, and survival of NK cells. Results: Treatment with 161519 TriKE in the presence of CD19+ targets upregulated expression of CD69, CD107a, TRAIL, IFN-γ, and TNF-α in NK cells, and significantly improved the proliferation and cytotoxicity of NK cells. NK cells "armed" with 161519 TriKE showed stronger cytolysis against CD19+ targets compared with that of "unarmed" NK cells. A preclinical model of B-cell lymphoma in human peripheral blood mononuclear cell-reconstituted xenograft mice showed significant inhibition of tumor growth and prolonged overall survival after treatment with 161519 TriKE, when compared with that in control mice or mice treated with 1619 BiKE. Combined use of IL-2 was a more effective treatment with 1619 BiKE, when compared with that using 161519 TriKE. Conclusions: The newly generated 161519 TriKE enhanced the proliferation, activation, cytokine secretion, and cytotoxicity of NK cells in the presence of CD19+ tumor cells. The 161519 TriKE aided inhibition of tumor growth and prolonged the overall survival of murine xenografts, and could be used to treat CD19-positive cancers.


Subject(s)
Antigens, CD19/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Killer Cells, Natural/immunology , Animals , Antineoplastic Agents, Immunological/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred NOD , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, IgG/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...