Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
MAbs ; 15(1): 2289681, 2023.
Article in English | MEDLINE | ID: mdl-38084840

ABSTRACT

Gremlin-1, a high-affinity antagonist of bone morphogenetic proteins (BMP)-2, -4, and -7, is implicated in tumor initiation and progression. Increased gremlin-1 expression, and therefore suppressed BMP signaling, correlates with poor prognosis in a range of cancer types. A lack of published work using therapeutic modalities has precluded the testing of the hypothesis that blocking the gremlin-1/BMP interaction will provide benefits to patients. To address this shortfall, we developed ginisortamab (UCB6114), a first-in-class clinical anti-human gremlin-1 antibody, currently in clinical development for the treatment of cancer, along with its murine analog antibody Ab7326 mouse immunoglobulin G1 (mIgG1). Surface plasmon resonance assays revealed that ginisortamab and Ab7326 mIgG1 had similar affinities for human and mouse gremlin-1, with mean equilibrium dissociation constants of 87 pM and 61 pM, respectively. The gremlin-1/Ab7326 antigen-binding fragment (Fab) crystal structure revealed a gremlin-1 dimer with a Fab molecule bound to each monomer that blocked BMP binding. In cell culture experiments, ginisortamab fully blocked the activity of recombinant human gremlin-1, and restored BMP signaling pathways in human colorectal cancer (CRC) cell lines. Furthermore, in a human CRC - fibroblast co-culture system where gremlin-1 is produced by the fibroblasts, ginisortamab restored BMP signaling in both the CRC cells and fibroblasts, demonstrating its activity in a relevant human tumor microenvironment model. The safety and efficacy of ginisortamab are currently being evaluated in a Phase 1/2 clinical trial in patients with advanced solid tumors (NCT04393298).


Subject(s)
Neoplasms , Signal Transduction , Humans , Animals , Mice , Cell Line , Neoplasms/drug therapy , Tumor Microenvironment
2.
Cells ; 9(7)2020 07 07.
Article in English | MEDLINE | ID: mdl-32645919

ABSTRACT

Basophils are involved in manifestations of hypersensitivity, however, the current understanding of their propensity for activation and their prognostic value in cancer patients remains unclear. As in healthy and atopic individuals, basophil populations were identified in blood from ovarian cancer patients (n = 53) with diverse tumor histologies and treatment histories. Ex vivo basophil activation was measured by CD63 expression using the basophil activation test (BAT). Irrespective of prior treatment, basophils could be activated by stimulation with IgE- (anti-FcεRI and anti-IgE) and non-IgE (fMLP) mediated triggers. Basophil activation was detected by ex vivo exposure to paclitaxel, but not to other anti-cancer therapies, in agreement with a clinical history of systemic hypersensitivity reactions to paclitaxel. Protein and gene expression analyses support the presence of basophils (CCR3, CD123, FcεRI) and activated basophils (CD63, CD203c, tryptase) in ovarian tumors. Greater numbers of circulating basophils, cells with greater capacity for ex vivo stimulation (n = 35), and gene signatures indicating the presence of activated basophils in tumors (n = 439) were each associated with improved survival in ovarian cancer. Circulating basophils in cancer patients respond to IgE- and non-IgE-mediated signals and could help identify hypersensitivity to therapeutic agents. Activated circulating and tumor-infiltrating basophils may be potential biomarkers in oncology.


Subject(s)
Basophils/metabolism , Ovarian Neoplasms/metabolism , Basophils/immunology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Female , Flow Cytometry , Humans , Immunoglobulin E/metabolism , Immunophenotyping , Ovarian Neoplasms/immunology , Tetraspanin 30/metabolism
3.
Nat Rev Clin Oncol ; 16(3): 185-204, 2019 03.
Article in English | MEDLINE | ID: mdl-30514977

ABSTRACT

Most cancer-related deaths are a result of metastasis, and thus the importance of this process as a target of therapy cannot be understated. By asking 'how can we effectively treat cancer?', we do not capture the complexity of a disease encompassing >200 different cancer types - many consisting of multiple subtypes - with considerable intratumoural heterogeneity, which can result in variable responses to a specific therapy. Moreover, we have much less information on the pathophysiological characteristics of metastases than is available for the primary tumour. Most disseminated tumour cells that arrive in distant tissues, surrounded by unfamiliar cells and a foreign microenvironment, are likely to die; however, those that survive can generate metastatic tumours with a markedly different biology from that of the primary tumour. To treat metastasis effectively, we must inhibit fundamental metastatic processes and develop specific preclinical and clinical strategies that do not rely on primary tumour responses. To address this crucial issue, Cancer Research UK and Cancer Therapeutics CRC Australia formed a Metastasis Working Group with representatives from not-for-profit, academic, government, industry and regulatory bodies in order to develop recommendations on how to tackle the challenges associated with treating (micro)metastatic disease. Herein, we describe the challenges identified as well as the proposed approaches for discovering and developing anticancer agents designed specifically to prevent or delay the metastatic outgrowth of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Development/organization & administration , Neoplasm Metastasis/drug therapy , Animals , Antineoplastic Agents/pharmacology , Humans , Molecular Targeted Therapy , United Kingdom , Xenograft Model Antitumor Assays
4.
Cancer Res ; 77(5): 1127-1141, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28096174

ABSTRACT

IgE antibodies are key mediators of antiparasitic immune responses, but their potential for cancer treatment via antibody-dependent cell-mediated cytotoxicity (ADCC) has been little studied. Recently, tumor antigen-specific IgEs were reported to restrict cancer cell growth by engaging high-affinity Fc receptors on monocytes and macrophages; however, the underlying therapeutic mechanisms were undefined and in vivo proof of concept was limited. Here, an immunocompetent rat model was designed to recapitulate the human IgE-Fcε receptor system for cancer studies. We also generated rat IgE and IgG mAbs specific for the folate receptor (FRα), which is expressed widely on human ovarian tumors, along with a syngeneic rat tumor model expressing human FRα. Compared with IgG, anti-FRα IgE reduced lung metastases. This effect was associated with increased intratumoral infiltration by TNFα+ and CD80+ macrophages plus elevated TNFα and the macrophage chemoattractant MCP-1 in lung bronchoalveolar lavage fluid. Increased levels of TNFα and MCP-1 correlated with IgE-mediated tumor cytotoxicity by human monocytes and with longer patient survival in clinical specimens of ovarian cancer. Monocytes responded to IgE but not IgG exposure by upregulating TNFα, which in turn induced MCP-1 production by monocytes and tumor cells to promote a monocyte chemotactic response. Conversely, blocking TNFα receptor signaling abrogated induction of MCP-1, implicating it in the antitumor effects of IgE. Overall, these findings show how antitumor IgE reprograms monocytes and macrophages in the tumor microenvironment, encouraging the clinical use of IgE antibody technology to attack cancer beyond the present exclusive reliance on IgG. Cancer Res; 77(5); 1127-41. ©2017 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Folate Receptor 1/immunology , Macrophages/immunology , Ovarian Neoplasms/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Line, Tumor , Female , Folate Receptor 1/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Rats , Rats, Wistar , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
5.
Clin Cancer Res ; 18(14): 3912-23, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22781553

ABSTRACT

PURPOSE: Deregulated phosphatidylinositol 3-kinase pathway signaling through AGC kinases including AKT, p70S6 kinase, PKA, SGK and Rho kinase is a key driver of multiple cancers. The simultaneous inhibition of multiple AGC kinases may increase antitumor activity and minimize clinical resistance compared with a single pathway component. EXPERIMENTAL DESIGN: We investigated the detailed pharmacology and antitumor activity of the novel clinical drug candidate AT13148, an oral ATP-competitive multi-AGC kinase inhibitor. Gene expression microarray studies were undertaken to characterize the molecular mechanisms of action of AT13148. RESULTS: AT13148 caused substantial blockade of AKT, p70S6K, PKA, ROCK, and SGK substrate phosphorylation and induced apoptosis in a concentration and time-dependent manner in cancer cells with clinically relevant genetic defects in vitro and in vivo. Antitumor efficacy in HER2-positive, PIK3CA-mutant BT474 breast, PTEN-deficient PC3 human prostate cancer, and PTEN-deficient MES-SA uterine tumor xenografts was shown. We show for the first time that induction of AKT phosphorylation at serine 473 by AT13148, as reported for other ATP-competitive inhibitors of AKT, is not a therapeutically relevant reactivation step. Gene expression studies showed that AT13148 has a predominant effect on apoptosis genes, whereas the selective AKT inhibitor CCT128930 modulates cell-cycle genes. Induction of upstream regulators including IRS2 and PIK3IP1 as a result of compensatory feedback loops was observed. CONCLUSIONS: The clinical candidate AT13148 is a novel oral multi-AGC kinase inhibitor with potent pharmacodynamic and antitumor activity, which shows a distinct mechanism of action from other AKT inhibitors. AT13148 will now be assessed in a first-in-human phase I trial.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Drug Discov Today ; 17(5-6): 227-31, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22207222

ABSTRACT

Recent changes to non-clinical cancer guidelines offer a golden opportunity to expedite the translation of new anticancer drugs into the clinic. In this review we look at how these guidelines can be implemented and how they can be integrated with non-clinical and clinical study design to produce robust and safe clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic/legislation & jurisprudence , Drug Discovery/legislation & jurisprudence , Neoplasms/drug therapy , Animals , European Union , Guidelines as Topic , Humans , United Kingdom
7.
Bioorg Med Chem Lett ; 20(19): 5803-6, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20732811

ABSTRACT

Following the discovery of 4-(substituted amino)-1-alkyl-pyrazolo[3,4-b]pyridine-5-carboxamides as potent and selective phosphodiesterase 4B inhibitors, [Hamblin, J. N.; Angell, T.; Ballentine, S., et al. Bioorg. Med. Chem. Lett.2008, 18, 4237] the SAR of the 5-position was investigated further. A range of substituted heterocycles showed good potencies against PDE4. Optimisation using X-ray crystallography and computational modelling led to the discovery of 16, with sub-nM inhibition of LPS-induced TNF-α production from isolated human peripheral blood mononuclear cells.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Heterocyclic Compounds/chemistry , Phosphodiesterase Inhibitors/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Binding Sites , Computer Simulation , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Humans , Models, Chemical , Models, Molecular , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
8.
Bioorg Med Chem Lett ; 18(14): 4237-41, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18539455

ABSTRACT

Optimisation of a high-throughput screening hit resulted in the discovery of 4-(substituted amino)-1-alkyl-pyrazolo[3,4-b]pyridine-5-carboxamides as potent and selective inhibitors of Phosphodiesterase 4 (PDE4). Herein, we describe early SAR studies around this novel template highlighting preferred substituents and rationalization of SAR through X-ray crystal structures of analogues bound to the PDE4 active site. Pyrazolopyridine 20a was found to be a potent and selective PDE4 inhibitor which also inhibits LPS induced TNF-alpha production from isolated human peripheral blood mononuclear cells and has an encouraging rat PK profile suitable for oral dosing.


Subject(s)
Chemistry, Pharmaceutical/methods , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Phosphodiesterase 4 Inhibitors , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Benzamides/pharmacology , Crystallography, X-Ray/methods , Cyclopropanes/pharmacology , Drug Design , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Models, Biological , Models, Chemical , Pyrrolidinones/chemistry , Rats , Rolipram/pharmacology , Tumor Necrosis Factor-alpha/metabolism
10.
Org Lett ; 5(24): 4631-4, 2003 Nov 27.
Article in English | MEDLINE | ID: mdl-14627401

ABSTRACT

[reaction: see text] In this, the second of two Letters, the optimization of the pyrrolidine-5,5-trans-lactam template (exemplified by 1a) as a mechanism-based inhibitor of hepatitis C NS3/4A protease is described. "Right Box" analysis of cassette dosing screening pharmacokinetic data was used to rapidly categorize the compounds. GW0014 (compound 4d) emerged as the compound displaying an optimal balance of biochemical and replicon potency, along with low i.v. clearance in the dog.


Subject(s)
Lactams/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Pyrrolidines/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Inhibitory Concentration 50 , Molecular Structure , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/metabolism
11.
Bioorg Med Chem Lett ; 13(10): 1657-60, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12729635

ABSTRACT

Using the pyrrolidine-5,5-trans-lactam template, we have designed small, neutral, mechanism-based inhibitors of hepatitis C NS3/4A protease. Compound 2b, with a spiro-cyclobutyl P1 substituent and an isopropyl carbonyl substituent at the lactam nitrogen, has an IC(50) value in the replicon cell-based assay of 3 microM.


Subject(s)
Lactams/pharmacology , Protease Inhibitors/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Drug Design , Drug Stability , Inhibitory Concentration 50 , Lactams/chemical synthesis , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacology , Structure-Activity Relationship
12.
Eur J Med Chem ; 38(4): 339-43, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12750019

ABSTRACT

The pyrrolidine-5,5-trans-lactam template was used to design small, neutral, mechanism-based inhibitors of hepatitis C NS3/4A protease displaying potent activity in the replicon cell-based assay. The activity of this series is not dependent upon its chemical reactivity and molecules have been synthesised which combine enhanced biochemical potency with improved plasma stability. Promising initial pharmacokinetic data indicating the potential for further optimisation of this series into low molecular weight, drug-like inhibitors is presented.


Subject(s)
Drug Design , Hepacivirus/enzymology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Drug Stability , Hepacivirus/drug effects , Humans , Injections, Intravenous , Lactams/chemical synthesis , Lactams/pharmacokinetics , Lactams/pharmacology , Models, Biological , Molecular Structure , Protease Inhibitors/pharmacokinetics , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Rats , Viral Nonstructural Proteins/metabolism
13.
Org Lett ; 4(25): 4475-8, 2002 Dec 12.
Article in English | MEDLINE | ID: mdl-12465916

ABSTRACT

[reaction: see text] In this, the first of two letters, we outline the use of the pyrrolidine-5,5-trans-lactam template to design small, neutral, mechanism-based inhibitors of hepatitis C NS3/4A protease. The hitherto unreported reaction of the acyl iminium ion precursor 4 with dialkyl-substituted silyl ketene acetals (e.g., 8b) is described. Compound 12b, with a spirocyclobutyl P1 substituent and a cyclopropylacyl substituent on the lactam nitrogen, has a k(obs)/I of 400 M(-)(1) s(-)(1) and demonstrates activity in a replicon cell-based surrogate HCV assay.


Subject(s)
Hepacivirus/enzymology , Lactams/chemical synthesis , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Pyrrolidines/chemical synthesis , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Cell Line , Drug Design , Drug Stability , Hepacivirus/drug effects , Humans , Lactams/chemistry , Molecular Structure , Protease Inhibitors/chemistry , Pyrrolidines/chemistry , Viral Nonstructural Proteins/metabolism
14.
SELECTION OF CITATIONS
SEARCH DETAIL