Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Peptides ; 152: 170760, 2022 06.
Article in English | MEDLINE | ID: mdl-35150805

ABSTRACT

A new strategy of peptide half-life extension has been evaluated. We investigated libraries of a small and very stable protein scaffold called Nanofitin, capable of high affinity for protein targets. We have identified Nanofitins targeting Human and mouse Serum Albumin, which could significantly improve the pharmacokinetics of an active associated peptide, mobilizing the patient's own albumin without external source. To demonstrate the impact of this approach on half-life extension, a genetic fusion of an Exenatide peptide with an Albumin Binding Nanofitin (ABNF) was performed. Specific activity of Exenatide-ABNF was measured and unaffected by the fusion. In vivo mice results provided convincing data (t½ of 8 min for Exenatide peptide compared to 20 h for Exenatide-ABNF) with sustained pharmacological activity over 3 days. This study constitutes a proof-of-concept of in vivo half-life extension of a biologic using an ABNF. Besides, the absence of cysteine in the Nanofitin scaffold, which is therefore devoid of structuring disulfide bonds, allows manufacturing in microbial cost effective systems.


Subject(s)
Biological Products , Peptides , Albumins , Animals , Exenatide , Half-Life , Mice , Peptides/chemistry
2.
Nat Cancer ; 1(1): 86-98, 2020 01.
Article in English | MEDLINE | ID: mdl-35121834

ABSTRACT

Despite the significant therapeutic advances provided by immune-checkpoint blockade and chimeric antigen receptor T cell treatments, many malignancies remain unresponsive to immunotherapy. Bispecific antibodies targeting tumor antigens and activating T cell receptor signaling have shown some clinical efficacy; however, providing co-stimulatory signals may improve T cell responses against tumors. Here, we developed a trispecific antibody that interacts with CD38, CD3 and CD28 to enhance both T cell activation and tumor targeting. The engagement of both CD3 and CD28 affords efficient T cell stimulation, whereas the anti-CD38 domain directs T cells to myeloma cells, as well as to certain lymphomas and leukemias. In vivo administration of this antibody suppressed myeloma growth in a humanized mouse model and also stimulated memory/effector T cell proliferation and reduced regulatory T cells in non-human primates at well-tolerated doses. Collectively, trispecific antibodies represent a promising platform for cancer immunotherapy.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Antibodies, Bispecific/therapeutic use , CD28 Antigens , Mice , Multiple Myeloma/drug therapy , Receptors, Antigen, T-Cell , T-Lymphocytes
3.
Nat Med ; 25(9): 1402-1407, 2019 09.
Article in English | MEDLINE | ID: mdl-31501610

ABSTRACT

Natalizumab (NZM), a humanized monoclonal IgG4 antibody to α4 integrins, is used to treat patients with relapsing-remitting multiple sclerosis (MS)1,2, but in about 6% of the cases persistent neutralizing anti-drug antibodies (ADAs) are induced leading to therapy discontinuation3,4. To understand the basis of the ADA response and the mechanism of ADA-mediated neutralization, we performed an in-depth analysis of the B and T cell responses in two patients. By characterizing a large panel of NZM-specific monoclonal antibodies, we found that, in both patients, the response was polyclonal and targeted different epitopes of the NZM idiotype. The neutralizing activity was acquired through somatic mutations and correlated with a slow dissociation rate, a finding that was supported by structural data. Interestingly, in both patients, the analysis of the CD4+ T cell response, combined with mass spectrometry-based peptidomics, revealed a single immunodominant T cell epitope spanning the FR2-CDR2 region of the NZM light chain. Moreover, a CDR2-modified version of NZM was not recognized by T cells, while retaining binding to α4 integrins. Collectively, our integrated analysis identifies the basis of T-B collaboration that leads to ADA-mediated therapeutic resistance and delineates an approach to design novel deimmunized antibodies for autoimmune disease and cancer treatment.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Epitopes, T-Lymphocyte/immunology , Multiple Sclerosis/drug therapy , Natalizumab/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Neutralizing/chemistry , Antibody Formation/drug effects , Antibody Formation/immunology , B-Lymphocytes/drug effects , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Integrin alpha4/antagonists & inhibitors , Integrin alpha4/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Protein Conformation/drug effects , T-Lymphocytes/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
J Med Chem ; 58(1): 362-75, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25369539

ABSTRACT

The Aurora family of serine/threonine kinases is essential for mitosis. Their crucial role in cell cycle regulation and aberrant expression in a broad range of malignancies have been demonstrated and have prompted intensive search for small molecule Aurora inhibitors. Indeed, over 10 of them have reached the clinic as potential anticancer therapies. We report herein the discovery and optimization of a novel series of tricyclic molecules that has led to SAR156497, an exquisitely selective Aurora A, B, and C inhibitor with in vitro and in vivo efficacy. We also provide insights into its mode of binding to its target proteins, which could explain its selectivity.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Benzimidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/chemistry , Aurora Kinase A/metabolism , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/chemistry , Aurora Kinase B/metabolism , Aurora Kinase C/antagonists & inhibitors , Aurora Kinase C/chemistry , Aurora Kinase C/metabolism , Aurora Kinases/chemistry , Aurora Kinases/metabolism , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Female , HCT116 Cells , Humans , Mice, SCID , Models, Chemical , Models, Molecular , Molecular Structure , Neoplasms/drug therapy , Neoplasms/pathology , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Quinolones/chemistry , Quinolones/metabolism , Sf9 Cells , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 20(17): 4574-83, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24987056

ABSTRACT

PURPOSE: The CD38 cell surface antigen is expressed in diverse hematologic malignancies including multiple myeloma, B-cell non-Hodgkin lymphoma (NHL), B-cell chronic lymphocytic leukemia, B-cell acute lymphoblastic leukemia (ALL), and T-cell ALL. Here, we assessed the antitumor activity of the anti-CD38 antibody SAR650984. EXPERIMENTAL DESIGN: Activity of SAR650984 was examined on lymphoma, leukemia and multiple myeloma cell lines, primary multiple myeloma samples, and multiple myeloma xenograft models in immunodeficient mice. RESULTS: We identified a humanized anti-CD38 antibody with strong proapoptotic activity independent of cross-linking agents, and potent effector functions including complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and antibody-dependent cellular phagocytosis (ADCP), equivalent in vitro to rituximab in CD20+ and CD38+ models. This unique antibody, termed SAR650984, inhibited the ADP-ribosyl cyclase activity of CD38, likely through an allosteric antagonism as suggested by 3D structure analysis of the complex. In vivo, SAR650984 was active in diverse NHL, ALL, and multiple myeloma CD38+ tumor xenograft models. SAR650984 demonstrated single-agent activity comparable with rituximab or cyclophosphamide in Daudi or SU-DHL-8 lymphoma xenograft models with induction of the proapoptotic marker cleaved capase-7. In addition, SAR650984 had more potent antitumor activity than bortezomib in NCI-H929 and Molp-8 multiple myeloma xenograft studies. Consistent with its mode of action, SAR650984 demonstrated potent proapoptotic activity against CD38+ human primary multiple myeloma cells. CONCLUSION: These results validate CD38 as a therapeutic target and support the current evaluation of this unique CD38-targeting functional antibody in phase I clinical trials in patients with CD38+ B-cell malignancies.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Antibodies, Monoclonal, Humanized/administration & dosage , Hematologic Neoplasms/drug therapy , Lymphoma, B-Cell/drug therapy , Membrane Glycoproteins/genetics , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Drug-Related Side Effects and Adverse Reactions , Hematologic Neoplasms/pathology , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Multiple Myeloma/pathology , Rituximab , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...