Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioconjug Chem ; 33(6): 1210-1221, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35658441

ABSTRACT

Inhibition of intracellular nicotinamide phosphoribosyltransferase (NAMPT) represents a new mode of action for cancer-targeting antibody-drug conjugates (ADCs) with activity also in slowly proliferating cells. To extend the repertoire of available effector chemistries, we have developed a novel structural class of NAMPT inhibitors as ADC payloads. A structure-activity relationship-driven approach supported by protein structural information was pursued to identify a suitable attachment point for the linker to connect the NAMPT inhibitor with the antibody. Optimization of scaffolds and linker structures led to highly potent effector chemistries which were conjugated to antibodies targeting C4.4a (LYPD3), HER2 (c-erbB2), or B7H3 (CD276) and tested on antigen-positive and -negative cancer cell lines. Pharmacokinetic studies, including metabolite profiling, were performed to optimize the stability and selectivity of the ADCs and to evaluate potential bystander effects. Optimized NAMPTi-ADCs demonstrated potent in vivo antitumor efficacy in target antigen-expressing xenograft mouse models. This led to the development of highly potent NAMPT inhibitor ADCs with a very good selectivity profile compared with the corresponding isotype control ADCs. Moreover, we demonstrate─to our knowledge for the first time─the generation of NAMPTi payload metabolites from the NAMPTi-ADCs in vitro and in vivo. In conclusion, NAMPTi-ADCs represent an attractive new payload class designed for use in ADCs for the treatment of solid and hematological cancers.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Nicotinamide Phosphoribosyltransferase , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7 Antigens , Cell Line, Tumor , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
2.
J Med Chem ; 63(20): 11639-11662, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32969660

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a rare and devastating chronic lung disease of unknown etiology. Despite the approved treatment options nintedanib and pirfenidone, the medical need for a safe and well-tolerated antifibrotic treatment of IPF remains high. The human prostaglandin F receptor (hFP-R) is widely expressed in the lung tissue and constitutes an attractive target for the treatment of fibrotic lung diseases. Herein, we present our research toward novel quinoline-based hFP-R antagonists, including synthesis and detailed structure-activity relationship (SAR). Starting from a high-throughput screening (HTS) hit of our corporate compound library, multiple parameter improvements-including increase of the relative oral bioavailability Frel from 3 to ≥100%-led to a highly potent and selective hFP-R antagonist with complete oral absorption from suspension. BAY-6672 (46) represents-to the best of our knowledge-the first reported FP-R antagonist to demonstrate in vivo efficacy in a preclinical animal model of lung fibrosis, thus paving the way for a new treatment option in IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Quinolines/chemical synthesis , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Animals , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Molecular Structure , Quinolines/chemistry , Quinolines/therapeutic use , Rats , Rats, Wistar , Structure-Activity Relationship
3.
Bioconjug Chem ; 31(8): 1893-1898, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32667786

ABSTRACT

Several antibody-drug conjugates (ADCs) have failed to achieve a sufficiently large therapeutic window in patients due to toxicity induced by unspecific payload release in the circulation or ADC uptake into healthy organs. Herein, we describe the successful engineering of ADCs consisting of novel linkers, which are efficiently and selectively cleaved by the tumor-associated protease legumain. ADCs generated via this approach demonstrate high potency and a preferential activation in tumors compared to healthy tissue, thus providing an additional level of safety. A remarkable tolerance of legumain for different linker peptides, including those with just a single asparagine residue, together with a modifier of the physicochemical metabolite profile, proves the broad applicability of this approach for a tailored design of ADCs.


Subject(s)
Immunoconjugates/chemistry , Kinesins/antagonists & inhibitors , Animals , Cysteine Endopeptidases/metabolism , Drug Delivery Systems , Humans , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Prodrugs , Structure-Activity Relationship , Urologic Neoplasms/drug therapy , Urothelium
4.
Chemistry ; 25(35): 8208-8213, 2019 Jun 21.
Article in English | MEDLINE | ID: mdl-30869180

ABSTRACT

Many antibody-drug conjugates (ADCs) have failed to achieve a sufficient therapeutic window in clinical studies either due to target-mediated or off-target toxicities. To achieve an additional safety level, a new class of antibody-prodrug conjugates (APDCs) directed against different targets in solid tumors is here described. The tumor-associated lysosomal endopeptidase legumain with a unique cleavage sequence was utilized for APDC metabolism. Legumain-activatable APDCs were as potent as their cathepsin B-activatable analogues. The peptide sequence susceptible to legumain cleavage was optimized for further discrimination of the formation of active metabolites within tumor cells versus healthy tissues, leveraging different tissue-specific legumain activities. Optimized APDCs with slow legumain-mediated conversion reduced preclinically the levels of active metabolite in healthy organs while retaining high activity against different TWEAKR- and B7H3-expressing tumors.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/chemistry , Cysteine Endopeptidases/metabolism , Immunoconjugates/chemistry , Kinesins/antagonists & inhibitors , Oligopeptides/chemistry , Prodrugs/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , B7 Antigens/genetics , B7 Antigens/immunology , B7 Antigens/metabolism , Cell Line, Tumor , Heterografts , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/metabolism , Mice
5.
Monoclon Antib Immunodiagn Immunother ; 33(6): 369-77, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25545205

ABSTRACT

Monoclonal antibodies are produced in cultured hybridoma cell lines, but these cells tend to be unstable; it is therefore necessary to rescue the corresponding genetic information. Here we describe an improved method for the amplification of antibody variable gene (V-gene) information from murine hybridoma cells using a panel of specific, non-degenerate primers. This primer set allows sequences to be rescued from all murine V-genes, except the lambda light chain genes, which rarely contribute to murine immune diversity. We tested the primers against a range of antibodies and recovered specific amplification products in all cases. The heavy and light chain variable regions were subsequently joined by a two-step cloning strategy or by splice overlap extension PCR.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/genetics , DNA Primers/genetics , Hybridomas/immunology , Immunoglobulin Variable Region/genetics , Recombinant Proteins/genetics , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Base Sequence , Blotting, Western , Cell Line , Cloning, Molecular/methods , DNA, Complementary/biosynthesis , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genetic Vectors/genetics , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/immunology , Sequence Analysis, DNA
6.
BMC Cancer ; 9: 200, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19552806

ABSTRACT

BACKGROUND: RAI3 is an orphan G-protein coupled receptor (GPCR) that has been associated with malignancy and may play a role in the proliferation of breast cancer cells. Although its exact function in normal and malignant cells remains unclear and evidence supporting its role in oncogenesis is controversial, its abundant expression on the surface of cancer cells would make it an interesting target for the development of antibody-based therapeutics. To investigate the link with cancer and provide more evidence for its role, we carried out a systematic analysis of RAI3 expression in a large set of human breast cancer specimens. METHODS: We expressed recombinant human RAI3 in bacteria and reconstituted the purified protein in liposomes to raise monoclonal antibodies using classical hybridoma techniques. The specific binding activity of the antibodies was confirmed by enzyme-linked immunosorbent assay (ELISA), western blot and immunocytochemistry. We carried out a systematic immunohistochemical analysis of RAI3 expression in human invasive breast carcinomas (n = 147) and normal breast tissues (n = 44) using a tissue microarray. In addition, a cDNA dot blot hybridisation assay was used to investigate a set of matched normal and cancerous breast tissue specimens (n = 50) as well as lymph node metastases (n = 3) for RAI3 mRNA expression. RESULTS: The anti-RAI3 monoclonal antibodies bound to recombinant human RAI3 protein with high specificity and affinity, as shown by ELISA, western blot and ICC. The cDNA dot blot and immunohistochemical experiments showed that both RAI3 mRNA and RAI3 protein were abundantly expressed in human breast carcinoma. However, there was no association between RAI3 protein expression and prognosis based on overall and recurrence-free survival. CONCLUSION: We have generated a novel, highly-specific monoclonal antibody that detects RAI3 in formaldehyde-fixed paraffin-embedded tissue. This is the first study to report a systematic analysis of RAI3 expression in normal and cancerous human breast tissue at both the mRNA and protein levels.


Subject(s)
Antibodies, Monoclonal/metabolism , Breast Neoplasms/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Enzyme-Linked Immunosorbent Assay/methods , Genetic Techniques , Humans , Immunohistochemistry/methods , Liposomes/chemistry , Liposomes/metabolism , Microscopy, Confocal/methods , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...