Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Invest New Drugs ; 40(4): 747-755, 2022 08.
Article in English | MEDLINE | ID: mdl-35404015

ABSTRACT

Ephrin type-A 2 (EphA2) is a transmembrane receptor expressed in epithelial cancers. We report on a phase I dose escalation and biodistribution study of DS-8895a, an anti-EphA2 antibody, in patients with advanced EphA2 positive cancers. DS-8895a was administered at 1, 3, 10 or 20 mg/kg every 2 weeks to determine safety, pharmacokinetics and anti-tumor efficacy. All patients underwent 89Zr trace-labelled infusion of DS-8895a (89Zr-DS-8995a) positron emission tomography imaging to determine the biodistribution of DS-8895a, and correlate findings with EphA2 expression, receptor saturation and response. Nine patients were enrolled on study. Of patients enrolled, seven patients received at least one infusion of DS-8895a: four patients received 1 mg/kg dose (Cohort 1) and three patients received 3 mg/kg dose (Cohort 2). Median age was 67.0 years (range 52-81), majority male (71%), and median number of prior systemic therapies was three (range 0-8). The primary cancer diagnosis was colorectal cancer (two patients) and one patient each had gastric, head and neck, high-grade serous adenocarcinoma, lung, and pancreatic cancers. No dose-limiting toxicities or treatment-related adverse events reported. The best response for the patients in Cohort 1 was stable disease and in Cohort 2 was progressive disease. 89Zr-DS-8895a demonstrated no normal tissue uptake and specific low-grade uptake in most tumours. DS-8895a had limited therapeutic efficacy at doses evaluated and 89Zr-DS-8895a demonstrated low tumour uptake. The biodistribution data from this study were key in halting further development of DS-8895a, highlighting the importance of biodistribution studies in drug development. (Trial registration: ClinicalTrials.gov Identifier NCT02252211).


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Neoplasms , Aged , Aged, 80 and over , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Ephrin-A2/immunology , Humans , Male , Middle Aged , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Receptor, EphA2/drug effects , Tissue Distribution
2.
Clin Cancer Res ; 26(18): 4785-4794, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32591465

ABSTRACT

PURPOSE: Taletrectinib (DS-6051b/AB-106) is an oral, tyrosine kinase inhibitor of ROS1 and NTRK with potent preclinical activity against ROS1 G2032R solvent-front mutation among others. We report the first-in-human U.S. phase I results of taletrectinib. PATIENTS AND METHODS: Patients ≥18 years old with neuroendocrine tumors, with tumor-induced pain, or tumors harboring ROS1/NTRK rearrangements were eligible. Accelerated titration followed by modified continuous reassessment method and escalation with overdose control was used (50-1,200 mg once daily or 400 mg twice daily). Primary objectives were safety/tolerability, and MTD determination. Secondary objectives were food-effect pharmacokinetics and antitumor activity. RESULTS: A total of 46 patients were enrolled. Steady-state peak concentration (C max) and exposure (AUC0-8) increased dose dependently from 50-mg to 800-mg once-daily doses. The ratio of the geometric mean of AUC0-24 between low-fat-diet-fed/fasted state was 123% (90% confidence interval, 104%-149%). Dose-limiting toxicities (grade 3 transaminases increase) occurred in two patients (1,200-mg once-daily dose). MTD was 800 mg once daily. Most common treatment-related adverse events were nausea (47.8%), diarrhea (43.5%), and vomiting (32.6%). Pain score reductions were observed in the 800-mg once-daily dose cohort. Confirmed objective response rate was 33.3% among the six patients with RECIST-evaluable crizotinib-refractory ROS1+ NSCLC. One patient with TPM3-NTRK1 differentiated thyroid cancer achieving a confirmed partial response of 27 months at data cutoff. We identified a cabozantinib-sensitive ROS1 L2086F as an acquired taletrectinib-resistance mutation. CONCLUSIONS: Taletrectinib has manageable toxicities at the MTD of 800 mg daily. Preliminary efficacy was observed in patients with crizotinib-refractory ROS1+ NSCLC.


Subject(s)
Food-Drug Interactions , Imidazoles/adverse effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Pyridazines/adverse effects , Adult , Aged , Female , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridazines/administration & dosage , Pyridazines/pharmacokinetics , Receptor, trkA/antagonists & inhibitors , Response Evaluation Criteria in Solid Tumors , Young Adult
3.
Invest New Drugs ; 37(1): 147-158, 2019 02.
Article in English | MEDLINE | ID: mdl-30056611

ABSTRACT

U3-1565 is a monoclonal antibody directed against heparin-binding epidermal growth factor-like growth factor (HB-EGF), which mediates angiogenesis via induction of vascular endothelial growth factor (VEGF-A). This first-in-human study characterized the safety, tolerability, efficacy, pharmacokinetics, and pharmacodynamics of U3-1565 in subjects with advanced solid tumors. In Part 1 (dose escalation following a modified 3 + 3 design), Cohorts 1-4, U3-1565 was administered at 2, 8, 16, and 24 mg/kg every 3 weeks for Cycle 1 and every 2 weeks thereafter. In Part 1, Cohort 5, and in Part 2 (dose expansion), U3-1565 was administered at 24 mg/kg every week. Thirty-six subjects were enrolled and treated (15 in Part 1; 21 in Part 2). No subject experienced dose limiting toxicity and maximum tolerated dose was not reached. All drug-related events were Grade 1 or 2 in severity, with fatigue and rash predominating. Following treatment with U3-1565, 1 subject with metastatic colorectal cancer experienced partial response and 6 subjects achieved stable disease. Four subjects completed the study main phase (first 12 cycles) and entered the extension phase. Of the 6/36 subjects with high (> 1500 pg/ml) baseline VEGF-A levels, all showed a decrease in VEGF-A (median - 60% [-22% to -97%]). Of the remaining subjects, only 19/30 showed a decrease (median - 18% [-2% to -82%]). Subjects with high VEGF-A baseline levels remained on treatment longer (3/6 entered study extension phase versus 1/30), and were more likely to show disease control (3/6 versus 4/30). In conclusion, U3-1565 demonstrates both proof of mechanism and clinical activity across different tumor types.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Heparin-binding EGF-like Growth Factor/chemistry , Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/pharmacokinetics , Female , Heparin-binding EGF-like Growth Factor/immunology , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Tissue Distribution
4.
Clin Drug Investig ; 38(11): 1001-1009, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30171457

ABSTRACT

BACKGROUND AND OBJECTIVES: Mirogabalin is an α2δ ligand being developed to treat neuropathic pain. A small fraction of mirogabalin is metabolized by the liver, where hepatic impairment may affect exposure. The objective of this phase I, open-label single-dose study was to determine if mild or moderate hepatic impairment alters the pharmacokinetics of mirogabalin. METHODS: Serial blood samples were collected for determination of maximum observed concentration, time to maximum concentration, and area under the concentration-time curve until the last quantifiable concentration of the active free form (A200-700) and inactive lactam metabolite (A204-4455) of mirogabalin. RESULTS: The A200-700 maximum observed concentration was similar in subjects with mild hepatic impairment but lower in subjects with moderate hepatic impairment vs. control subjects. The A204-4455 maximum observed concentration was lower in subjects with mild and moderate hepatic impairment vs. control groups. The A200-700 area under the concentration-time curve until the last quantifiable concentration was slightly lower in subjects with mild hepatic impairment and slightly higher in subjects with moderate hepatic impairment vs. control subjects. Peak A204-4455 levels were approximately 22% and 31% lower for subjects with mild and moderate hepatic impairment vs. control individuals, respectively. Exposure to A204-4455 was approximately 37% lower in subjects with mild hepatic impairment but unaffected in subjects with moderate hepatic impairment vs. control groups. Two subjects in the mild hepatic impairment group reported a treatment-emergent adverse event of mild somnolence. No serious or severe treatment-emergent adverse events, discontinuations as a result of treatment-emergent adverse events, or deaths were reported. CONCLUSIONS: Mild hepatic impairment resulted in lower A200-700 and A204-4455 exposure, while moderate hepatic impairment did not affect A200-700 exposure. Overall, mild-to-moderate hepatic impairment did not have a significant effect on mirogabalin exposure. A single 15-mg dose of mirogabalin was well tolerated by subjects with mild or moderate hepatic impairment.


Subject(s)
Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/pharmacokinetics , Liver Diseases/blood , Liver Diseases/drug therapy , Administration, Oral , Adult , Aged , Area Under Curve , Female , Humans , Male , Middle Aged , Young Adult
5.
Theranostics ; 8(15): 4199-4209, 2018.
Article in English | MEDLINE | ID: mdl-30128047

ABSTRACT

B7-H3 is a transmembrane protein widely expressed in a variety of cancers and has been shown to play a role in anti-tumor immunity. This study aims to develop a molecular imaging probe to identify B7-H3 expression in tumors and to develop 89Zr-DS-5573a as a theranostic that could aid patient selection in clinical Phase I studies. Methods: The anti-B7-H3 humanised monoclonal antibody DS-5573a was labeled with zirconium-89 (89Zr-), and assessed for radiochemical purity, immunoreactivity (Lindmo analysis), antigen binding affinity (Scatchard analysis), and serum stability in vitro. In vivo biodistribution and imaging studies were performed with positron emission tomography and magnetic resonance imaging (PET/MRI) studies to identify and quantitate 89Zr-DS-5573a tumor uptake in a B7-H3-positive breast cancer model (MDA-MB-231) and a B7-H3-negative murine colon cancer model (CT26). Imaging and biodistribution studies were also performed in MDA-MB-231 tumor-bearing SCID mice in the absence and presence of therapeutic DS-5573a antibody dose (3 mg/kg DS-5573a). Results:89Zr-DS-5573a showed high and specific binding to B7-H3-expressing MDA-MB-231 cells (immunoreactivity on day 0, 75.0 ± 2.9%), and low binding to B7-H3-negative CT26 cells (immunoreactivity on day 0, 10.85 ± 0.11%) in vitro. 89Zr-DS-5573a demonstrated good serum stability in vitro with 57.2 ± 2.0% of immunoreactivity remaining on day 7. In vivo biodistribution studies showed high uptake of 89Zr-DS-5573a in B7-H3-expressing MDA-MB-231 tumor-bearing mice, achieving 32.32 ± 6.55 %ID/g on day 7 post injection in BALB/c nu/nu mice and 25.76 ± 1.79 %ID/g in SCID mice, with minimal evidence of non-specific uptake in normal tissues, and excellent tumor localization on PET/MRI. In a combined imaging/therapy study, receptor saturation was demonstrated in tumors responding to therapy. Conclusion:89Zr-DS-5573a demonstrates specific and prolonged targeting of B7-H3-expressing tumors in vivo. Saturation of binding sites was demonstrated in tumors responding to DS-5573a therapy. These results indicate that 89Zr-DS-5573a has potential to target B7-H3-expressing tumors in cancer patients. Furthermore 89Zr-DS-5573a has the potential to provide important insights into T cell biology through its specific binding to B7-H3.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , B7 Antigens/analysis , Breast Neoplasms/diagnosis , Colonic Neoplasms/diagnosis , Molecular Imaging/methods , Radioisotopes/administration & dosage , T-Lymphocytes/chemistry , Zirconium/administration & dosage , Animals , Disease Models, Animal , Humans , Magnetic Resonance Imaging/methods , Mice, Inbred BALB C , Mice, SCID , Positron-Emission Tomography/methods , Theranostic Nanomedicine/methods
6.
J Clin Pharmacol ; 58(12): 1557-1565, 2018 12.
Article in English | MEDLINE | ID: mdl-29746725

ABSTRACT

DS-2969b is a novel GyrB inhibitor in development for the treatment of Clostridium difficile infection. The aim of this study was to assess the safety, tolerability, pharmacokinetics, and effects on normal gastrointestinal microbiota groups of single daily oral ascending doses of DS-2969b in healthy subjects. The study enrolled 6 sequential ascending dose cohorts (6 mg, 20 mg, 60 mg, 200 mg, 400 mg, and 600 mg). In each cohort, 6 subjects were administered DS-2969b and 2 subjects were administered matching placebo. DS-2969b was safe and well tolerated at all dose levels examined. All adverse events related to DS-2969b were mild to moderate in severity and predominantly related to the gastrointestinal tract. DS-2969a (free form of DS-2969b) plasma concentrations increased with increasing doses; however, both the maximum serum concentration and area under the plasma concentration-time curve generally increased less than dose proportionally. DS-2969a was predominantly eliminated in the urine, with feces as a minor route of elimination. While the overall proportion of DS-2969a eliminated in the feces was low, target fecal levels sufficient for C. difficile eradication were achieved within 24 hours of administration with doses of 60 mg or higher. In exploratory analyses, DS-2969b appeared to reduce bacterial counts in 8 of the 25 groups of normal intestinal microbiota examined, suggesting that DS-2969b has only a mild effect on intestinal microbiota. Data from this study support and encourage further development of DS-2969b as a novel treatment for C. difficile infection.


Subject(s)
Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Piperidines/adverse effects , Piperidines/pharmacokinetics , Thiadiazoles/adverse effects , Thiadiazoles/pharmacokinetics , Topoisomerase II Inhibitors/adverse effects , Topoisomerase II Inhibitors/pharmacokinetics , Adolescent , Adult , Area Under Curve , Bacteria/drug effects , Dose-Response Relationship, Drug , Female , Gastrointestinal Microbiome/drug effects , Half-Life , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Male , Middle Aged , Molecular Structure , Piperidines/administration & dosage , Piperidines/blood , Thiadiazoles/administration & dosage , Thiadiazoles/blood , Topoisomerase II Inhibitors/administration & dosage , Topoisomerase II Inhibitors/blood , Young Adult
7.
Article in English | MEDLINE | ID: mdl-29439973

ABSTRACT

DS-2969b is a novel GyrB inhibitor in development for the treatment of Clostridium difficile infection (CDI). The aim of this study was to assess the safety, tolerability, pharmacokinetics, and effects on the normal gastrointestinal microbiota of multiple daily oral ascending doses of DS-2969b in healthy subjects. The study enrolled three sequential ascending-dose cohorts (60 mg, 200 mg, and 400 mg). In each cohort, subjects received an oral dose of DS-2969b or placebo (six subjects received DS-2969b, and two received placebo) each morning for 14 days. DS-2969b was safe and well tolerated at all dose levels examined. All adverse events related to DS-2969b were mild and predominantly related to the gastrointestinal tract. DS-2969a (free form of DS-2969b) plasma concentrations increased with increasing doses; however, both the maximum concentration of drug in serum (Cmax) and the area under the concentration-time curve (AUC) increased less than dose proportionally. In all cohorts, sufficient fecal levels of DS-2969a were achieved within 24 h following the administration of the first dose and maintained for at least 17 days. Following treatment with DS-2969b, clear reductions in the populations of Clostridium coccoides and Bifidobacterium groups were observed. However, populations of three other bacterial groups examined (Bacteroides fragilis, Clostridium leptum, and Prevotella) were not affected. Data from this study support and encourage the further development of DS-2969b as a novel treatment for CDI.


Subject(s)
Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Topoisomerase II Inhibitors/pharmacokinetics , Administration, Oral , Adolescent , Adult , Bacteroides fragilis/drug effects , Bacteroides fragilis/metabolism , Bifidobacterium/drug effects , Bifidobacterium/pathogenicity , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Clostridium Infections/metabolism , Double-Blind Method , Drug Administration Schedule , Female , Gastrointestinal Microbiome/drug effects , Healthy Volunteers , Humans , Male , Middle Aged , Prevotella/drug effects , Prevotella/pathogenicity , Topoisomerase II Inhibitors/adverse effects , Young Adult
8.
J Clin Pharmacol ; 57(9): 1221-1230, 2017 09.
Article in English | MEDLINE | ID: mdl-28464321

ABSTRACT

DS-3801b is an orally active, nonmacrolide, selective motilin receptor agonist. The aim of this 2-part first-in-human study was to assess the safety, tolerability, pharmacokinetics, and pharmacodynamic effects on proximal and distal gastrointestinal (GI) motility of single oral doses of DS-3801b in healthy subjects. The 13 C-octanoate breath test was used to assess gastric emptying (GE), a measure of proximal GI motility. The time to first bowel movement (TTFBM) and the consistency of the first bowel movement according to the Bristol Stool Scale (BSS) were recorded to assess distal GI motility. In part A, 48 subjects received single oral doses of DS-3801b from 1 to 100 mg or placebo (6 DS-3801b, 2 placebo per cohort). In part B, 12 subjects received 50 mg of DS-3801b or placebo to assess GE. DS-3801b is safe and generally well tolerated after doses up to 50 mg, resulting in mild, predominantly GI adverse events. DS-3801a plasma concentrations increase with increasing doses; however, Cmax increases greater than dose-proportionally, whereas AUC increases less than dose-proportionally. The double peaks observed are consistent with multiple absorption sites. Results of the 13 C-octanoate breath test indicate that DS-3801b accelerates GE. Fifty milligrams of DS-3801b resulted in a 20.8% median reduction in GE T1/2 and a 20.6% median reduction in GE Tlag compared with placebo. However, this increase in proximal GI motility was not accompanied by an effect on distal GI motility, as indicated by no significant differences in TTFBM and BSS values across DS-3801b dose levels or compared with placebo.


Subject(s)
Cyclohexanes , Gastrointestinal Motility/drug effects , Piperazines , Receptors, Gastrointestinal Hormone/agonists , Receptors, Neuropeptide/agonists , Adult , Cross-Over Studies , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
9.
Invest New Drugs ; 35(3): 298-306, 2017 06.
Article in English | MEDLINE | ID: mdl-28050790

ABSTRACT

Background DR5 is a transmembrane receptor that transduces extracellular ligand-binding to activate apoptosis signaling cascades. This phase 1 study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics of a new monoclonal antibody potent DR5 agonist, DS-8273a, in subjects with advanced solid tumors. Methods The study comprised dose escalation and dose expansion cohorts. The dose escalation cohorts intended to determine the safety and to identify the maximum tolerated dose (MTD) or maximum administered dose (MAD) and to characterize the pharmacokinetics and pharmacodynamics by a conventional 3 + 3 design (starting at 2 mg/kg and escalating through 8, 16 and 24 mg/kg once every 3 weeks). In the dose expansion cohort, additional subjects were treated at the MAD for further evaluation of PK and safety. Results Thirty two subjects were enrolled and treated, 16 in the dose escalation cohorts and 16 in the dose expansion cohort. No subjects experienced a dose limiting toxicity (DLT). Treatment emergent adverse events were observed in 29 (91%) subjects, 14 (44%) of which were attributed to study-drug; all drug-related events were grade 1 and 2 in severity, and were mainly fatigue, nausea, vomiting and diarrhea. Measures of plasma exposure increased dose-proportionally and the mean terminal elimination half-life was 11 days. Blood samples available from a subset of patients treated at 24 mg/kg revealed declines in myeloid derived suppressor cells (MDSC) at 2 weeks. No objective responses were observed in any subjects. Conclusions DS-8273a was well tolerated and demonstrated linear pharmacokinetics. Decreases in MDSC were temporally associated with DS-8273a exposure. This agent could be studied further in combination with other agents, pending further proof-of-target-engagement.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Cell Count , Diarrhea/chemically induced , Fatigue/chemically induced , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Myeloid-Derived Suppressor Cells/drug effects , Nausea/chemically induced , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Vomiting/chemically induced
10.
Theranostics ; 6(12): 2225-2234, 2016.
Article in English | MEDLINE | ID: mdl-27924159

ABSTRACT

Background: DS-8273a, an anti-human death receptor 5 (DR5) agonistic antibody, has cytotoxic activity against human cancer cells and induces apoptosis after specific binding to DR5. DS-8273a is currently being used in clinical Phase I trials. This study evaluated the molecular imaging of DR5 expression in vivo in mouse tumor models using SPECT/CT and PET/MRI, as a tool for drug development and trial design. Methods: DS-8273a was radiolabeled with indium-111 and zirconium-89. Radiochemical purity, immunoreactivity, antigen binding affinity and serum stability were assessed in vitro. In vivo biodistribution and pharmacokinetic studies were performed, including SPECT/CT and PET/MR imaging. A dose-escalation study using a PET/MR imaging quantitative analysis was also performed to determine DR5 receptor saturability in a mouse model. Results:111In-CHX-A″-DTPA-DS-8273a and 89Zr-Df-Bz-NCS-DS-8273a showed high immunoreactivity (100%), high serum stability, and bound to DR5 expressing cells with high affinity (Ka, 1.02-1.22 × 1010 M-1). The number of antibodies bound per cell was 32,000. In vivo biodistribution studies showed high and specific uptake of 111In-CHX-A″-DTPA-DS-8273a and 89Zr-Df-Bz-NCS-DS-8273a in DR5 expressing COLO205 xenografts, with no specific uptake in normal tissues or in DR5-negative CT26 xenografts. DR5 receptor saturation was observed in vivo by biodistribution studies and quantitative PET/MRI analysis. Conclusion:89Zr-Df-Bz-NCS-DS-8273a is a potential novel PET imaging reagent for human bioimaging trials, and can be used for effective dose assessment and patient response evaluation in clinical trials.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/therapy , Antibodies/administration & dosage , Radioisotopes/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Theranostic Nanomedicine/methods , Zirconium/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Indium/administration & dosage , Indium/pharmacokinetics , Magnetic Resonance Imaging , Mice, Inbred BALB C , Positron-Emission Tomography , Radioisotopes/pharmacokinetics , Radiotherapy/methods , Tomography, Emission-Computed, Single-Photon , Treatment Outcome , Zirconium/pharmacokinetics
11.
J Nucl Med ; 57(6): 974-80, 2016 06.
Article in English | MEDLINE | ID: mdl-26940768

ABSTRACT

UNLABELLED: Subtype A2 of the erythropoietin-producing hepatocellular tyrosine kinase (EphA2) cell surface receptor is expressed in a range of epithelial cancers. This study evaluated the molecular imaging of EphA2 expression in vivo in mouse tumor models using SPECT/MR and PET/MR and a humanized anti-EphA2 antibody, DS-8895a. METHODS: DS-8895a was labeled with (111)In, (125)I, and (89)Zr and assessed for radiochemical purity, immunoreactivity (Lindmo analysis), antigen-binding affinity (Scatchard analysis), and serum stability in vitro. In vivo biodistribution, imaging, and pharmacokinetic studies were performed with SPECT/MR and PET/MR. A dose-escalation study was also performed to determine EphA2 receptor saturability through tissue and imaging quantitative analysis. RESULTS: All conjugates demonstrated good serum stability and specific binding to EphA2-expressing cells in vitro. In vivo biodistribution studies showed high uptake of (111)In-CHX-A″-DTPA-DS-8895a and (89)Zr-Df-Bz-NCS-DS-8895a in EphA2-expressing xenograft models, with no specific uptake in normal tissues. In comparison, retention of (125)I-DS-8895a in tumors was lower because of internalization of the radioconjugate and dehalogenation. These results were confirmed by SPECT/MR and PET/MR. EphA2 receptor saturation was observed at the 30 mg/kg dose. CONCLUSION: Molecular imaging of tumor uptake of DS-8895a allows noninvasive measurement of EphA2 expression in tumors in vivo and determination of receptor saturation. (89)Zr-Df-Bz-NCS-DS-8895a is suited for human bioimaging trials on the basis of superior imaging characteristics and will inform DS-8895a dose assessment and patient response evaluation in clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Cell Transformation, Neoplastic , Positron-Emission Tomography/methods , Radioisotopes , Receptor, EphA2/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Zirconium/chemistry , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Cell Line, Tumor , Deferoxamine/analogs & derivatives , Deferoxamine/chemistry , Female , Humans , Isothiocyanates/chemistry , Mice , Pentetic Acid/chemistry , Quality Control , Receptor, EphA2/immunology , Tissue Distribution
12.
Eur J Immunol ; 34(9): 2356-64, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15307168

ABSTRACT

The transcription factor IFN regulatory factor-1 (IRF-1) regulates production and activity of many inflammatory mediators and cells. Here, we investigated the role of IRF-1 in intestinal inflammation using clinical and histologic scores; inflammatory mediators were also measured in colonic tissue. Dextran sulfate sodium (DSS) or trinitrobenzene sulfonic acid (TNBS) was administered to wild-type (WT) or IRF-1 knockout (KO) mice. DSS or TNBS led to a dramatic increase in lethality and colitis severity in IRF-1 KO compared with WT mice. Reduced levels of IFN-gamma and IL-18-binding protein (IL-18BP) were observed in the colon of IRF-1 KO mice, whereas levels of inducible nitric oxide synthase, cyclooxygenase-2, phosphorylated STAT-3, chemokines, TNF-alpha, IL-1beta, IL-15, and IL-18 were not significantly changed. Intestinal inflammation was not altered in IFN-gamma KO mice or in WT mice given neutralizing anti-IFN-gamma antibodies, but was increased in mice lacking TCR gamma delta lymphocytes, a population significantly decreased in the intestine of IRF-1-deficient mice. Administration of IL-18BP reversed the increased susceptibility of IRF-1 KO mice to DSS. These results suggest a protective role for IRF-1 in intestinal inflammation, with a possible anti-inflammatory and/or restorative role. IL-18BP and TCR gamma delta cells appear to be critical factors in the anti-inflammatory effects of IRF-1.


Subject(s)
Colitis/etiology , DNA-Binding Proteins/physiology , Glycoproteins/physiology , Phosphoproteins/physiology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocytes/physiology , Animals , DNA-Binding Proteins/genetics , Female , Intercellular Signaling Peptides and Proteins , Interferon Regulatory Factor-1 , Interferon-gamma/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics
13.
Neuroimmunomodulation ; 11(4): 224-32, 2004.
Article in English | MEDLINE | ID: mdl-15249728

ABSTRACT

OBJECTIVE: This comparative in vitro study examined the effects of all known gp130 cytokines on murine corticotroph AtT-20 cell function. METHODS: Cytokines were tested at equimolar concentrations from 0.078 to 10 nM. Tyrosine phosphorylation of the signal transducer and activator of transcription (STAT)3 and STAT1, the STAT-dependent suppressor of cytokine signaling (SOCS)-3 promoter activity, SOCS-3 gene expression, STAT-dependent POMC promoter activity and adrenocorticotropic hormone (ACTH) secretion were determined. RESULTS: Leukemia inhibitory factor (LIF), human oncostatin M (OSM) and cardiotrophin (CT)-1 (LIFR/gp130 ligands), as well as ciliary neurotrophic factor (CNTF) and novel neurotrophin-1/B-cell stimulating factor-3 (CNTFR alpha/LIFR/gp130 ligands) are potent stimuli of corticotroph cells in vitro. In comparison, interleukin (IL)-6 (IL-6R/gp130 ligand) and IL-11 (IL-11R/gp130 ligand) exhibited only modest direct effects on corticotrophs, while murine OSM (OSMR/gp130 ligand) showed no effect. CONCLUSION: (i) CNTFR complex ligands are potent stimuli of corticotroph function, comparable to LIFR complex ligands; (ii) IL-6 and IL-11 are relatively weak direct stimuli of corticotroph function; (iii) differential effects of human and murine OSM suggest that LIFR/gp130 (OSMR type I) but not OSMR/gp130 (OSMR type II) are involved in corticotroph signaling. (iv) CT-1 has the hitherto unknown ability to stimulate corticotroph function, and (v) despite redundant immuno-neuroendocrine effects of different gp130 cytokines, corticotroph cells are preferably activated through the LIFR and CNTFR complexes.


Subject(s)
Antigens, CD/metabolism , Cytokines/pharmacology , Hypothalamo-Hypophyseal System/immunology , Membrane Glycoproteins/metabolism , Neuroimmunomodulation/immunology , Pituitary Gland, Anterior/immunology , Adrenocorticotropic Hormone/metabolism , Animals , Antigens, CD/drug effects , Antigens, CD/immunology , Cell Line , Cytokine Receptor gp130 , Cytokines/immunology , Cytokines/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression/immunology , Hypothalamo-Hypophyseal System/drug effects , Leukemia Inhibitory Factor Receptor alpha Subunit , Ligands , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/immunology , Mice , Phosphorylation/drug effects , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Pro-Opiomelanocortin/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/immunology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Ciliary Neurotrophic Factor/drug effects , Receptor, Ciliary Neurotrophic Factor/immunology , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Cytokine/drug effects , Receptors, Cytokine/immunology , Receptors, Cytokine/metabolism , Receptors, OSM-LIF , Repressor Proteins/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Tyrosine/metabolism
14.
Methods Mol Med ; 98: 171-90, 2004.
Article in English | MEDLINE | ID: mdl-15064439

ABSTRACT

Tumor necrosis factor alpha(TNFalpha) is a crucial mediator involved in the communications between immune and nervous systems in physiological conditions, and its relevance is amplified during disease. Considered originally detrimental and a target for therapeutic intervention, recently it has also gained attention for its protective role, especially in central nervous system (CNS) confined diseases. Thus, TNFalpha has become the key molecule illustrating the peculiar and still not completely understood pathways by which inflammatory and immune reactions occur in the brain. Several human pathologies that lack an efficient therapy and that carry enormous social costs rely on these mechanisms. Thus, further research is needed to improve our knowledge and to allow the identification of therapeutic targets or strategies for immune-mediated inflammatory disease of the CNS in which TNFalpha is primarily involved. We describe here how to induce experimental autoimmune encephalomyelitis, cerebral malaria, and brain ischemia in rodents, and some protocols to analyze them. The application of innovative research strategies or original therapeutic approaches to these experimental models may be rewarding in terms of advancement in a field that is crucial for the management of many human patients.


Subject(s)
Central Nervous System Diseases/metabolism , Disease Models, Animal , Tumor Necrosis Factor-alpha/physiology , Animals , Brain/pathology , Brain Ischemia/metabolism , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Ischemia , Malaria, Cerebral/metabolism , Mice , Mice, Transgenic , Perfusion , Tumor Necrosis Factor-alpha/metabolism
15.
Cytokine ; 25(3): 119-26, 2004 Feb 07.
Article in English | MEDLINE | ID: mdl-14698138

ABSTRACT

We studied the effects of pretreatment with granulocyte-colony stimulating factor (G-CSF) on the production of pro- and anti-inflammatory cytokines induced by lipopolysaccharide (LPS). Mice received G-CSF or control saline once a day for 7 days or once at 1 h before the injection of LPS. Cytokines were measured by enzyme-linked immunosorbent assay or antibody-based electrochemiluminescence assay and cytokine mRNA was measured by RNAse protection assay. Mice pretreated with G-CSF for 7 days before LPS had lower serum levels of LPS-induced interferon-gamma (IFN-gamma) and higher levels of interleukin (IL)-6 and IL-10 than controls. G-CSF-pretreated mice also had lower mRNA levels of IFN-gamma and higher mRNA levels of IL-6 and IL-10 in the spleen and/or liver than controls. G-CSF-pretreated mice had serum levels of tumor necrosis factor, IL-12 p70 and IL-12 p40 similar to controls. G-CSF-pretreated mice had lower levels of spleen IL-18 than controls-serum IL-18 being undetectable in mice after LPS-and lower levels of IL-18 mRNA in the spleen. Mice pretreated with G-CSF 1 h before LPS had lower levels of serum IFN-gamma and spleen IL-18 than controls. G-CSF pretreatment alters the expression of LPS-induced cytokines with a decrease in pro-inflammatory IFN-gamma and an increase in anti-inflammatory IL-6 and IL-10. G-CSF decrease of IL-18 production may be a major mechanism explaining the effects of G-CSF on the production of IFN-gamma.


Subject(s)
Granulocyte Colony-Stimulating Factor/physiology , Interferon-gamma/metabolism , Interleukin-18/metabolism , Lipopolysaccharides/pharmacology , Animals , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-12/blood , Interleukin-12/genetics , Interleukin-12 Subunit p40 , Interleukin-18/analysis , Interleukin-18/genetics , Interleukin-6/blood , Interleukin-6/genetics , Leukocyte Count , Liver/chemistry , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Protein Subunits/blood , Protein Subunits/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins , Spleen/chemistry , Spleen/drug effects , Spleen/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics
16.
Eur Cytokine Netw ; 15(4): 323-6, 2004.
Article in English | MEDLINE | ID: mdl-15627641

ABSTRACT

Previous reports have indicated that the administration of granulocyte colony-stimulating factor (G-CSF) decreases ex vivo tumor necrosis factor (TNF) production in humans. In this study, we report that daily pretreatment of mice with G-CSF for three days decreases ex vivo lipopolysaccharide (LPS)-induced TNF production in whole blood. Conversely, production of interleukin-10 (IL-10) and prostaglandin E(2) (PGE(2)) is increased. The inhibitory effect of G-CSF pretreatment on TNF production is partially reversed by addition of an anti-IL-10 antibody, and completely reversed by combined addition of anti-IL-10 antibody and the cyclooxygenase (COX) inhibitor, ketoprofen. These results suggest that G-CSF decreases TNF production in this experimental model by increasing production of IL-10 and PGE(2), which are both known inhibitors of TNF production.


Subject(s)
Blood Cells/physiology , Dinoprostone/biosynthesis , Granulocyte Colony-Stimulating Factor/administration & dosage , Interleukin-10/biosynthesis , Lipopolysaccharides/toxicity , Tumor Necrosis Factors/biosynthesis , Animals , Cells, Cultured , In Vitro Techniques , Injections, Subcutaneous , Mice
17.
Clin Immunol ; 109(3): 347-54, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14697750

ABSTRACT

The interaction between receptor activator of nuclear factor-kappaB ligand (RANKL) and RANK has been reported to regulate immunity in addition to bone metabolism. The aim of this study was to determine if osteoprotegerin (OPG), an inhibitor of the RANKL-RANK interaction and possibly a new drug against osteoporosis, would adversely affect immunity. OPG was used to treat mice developing different models of cellular and humoral immune responses and also in vitro in T and B cell assays. In mice, OPG does not affect cell-mediated reactions such as contact hypersensitivity to the hapten oxazolone and liver damage, granuloma formation, and infectious load induced by mycobacterial infection. However, OPG increases humoral reactions such as the production of IgM, IgG, and IgE against the T cell dependent antigen keyhole limpet hemocyanin and the production of IgM against the T cell independent antigen Pneumovax. In vitro, OPG modestly co-stimulates T cells but does not affect the proliferation of B cells. OPG has modest immunoregulatory effects that seem to be confined to the humoral response to specific antigens.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Glycoproteins/immunology , Immunity, Cellular/immunology , Receptors, Cytoplasmic and Nuclear/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Division/drug effects , Cell Division/immunology , Female , Hemocyanins/immunology , Humans , Hypersensitivity, Delayed/immunology , Immunity, Cellular/drug effects , Immunoglobulins/blood , Immunoglobulins/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Mice , Mice, Inbred BALB C , Mycobacterium bovis/drug effects , Mycobacterium bovis/immunology , Osteoprotegerin , Oxazolone/immunology , Pneumococcal Vaccines/immunology , Receptors, Tumor Necrosis Factor , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
18.
Br J Haematol ; 123(5): 869-78, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14632778

ABSTRACT

Cytokines of the gp130 family, particularly interleukin 6 (IL-6), play a central role in the growth and survival of malignant plasma cells. Recently, novel neurotrophin-1 (NNT-1)/B cell-stimulating factor-3 (BSF-3), also reported as cardiotrophin-like cytokine (CLC), was identified as a cytokine belonging to the gp130 family. BSF-3, similar to IL-6, exerts regulatory effects on normal B cell functions, but its functional significance in haematological malignancies has not been defined. The purpose of this study was to evaluate the biological effects and signalling pathways that are induced by BSF-3 in malignant plasma cells. Recombinant human BSF-3 was found to have growth stimulatory activity on plasmacytoma cell lines and primary tumour cells. In addition, BSF-3 was able to protect from Dexamethasone (Dex)-induced apoptosis. BSF-3 stimulated cell growth could not be inhibited by neutralizing anti-IL-6 or anti-IL-6 receptor antibodies, but was abrogated by anti-gp130 antibodies. In INA-6.Tu11 cells, a subline of the IL-6-dependent human plasma cell line INA-6 expressing gp130 and the receptor for leukaemia inhibitory factor (LIF), stimulation with BSF-3 induced tyrosine phosphorylation of signal transducer and activator of transcription 3 (STAT3). AG490, an inhibitor of Janus kinases, decreased BSF-3 induced cell growth in a dose-dependent manner. This correlated with a reduction of STAT3 phosphorylation levels, while p44/42 mitogen-activated protein kinase (MAPK) phosphorylation was not affected. In conclusion, BSF-3 is a novel myeloma growth and survival factor with a potential role in the pathophysiology of the disease.


Subject(s)
Multiple Myeloma/metabolism , Signal Transduction , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division , Cytokine Receptor gp130 , Cytokines/metabolism , Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Membrane Glycoproteins/immunology , Multiple Myeloma/pathology , Phosphorylation , Recombinant Proteins/pharmacology , STAT3 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured , Tyrphostins/pharmacology
19.
Endocrinology ; 144(4): 1202-10, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12639901

ABSTRACT

Novel neurotrophin-1/B cell-stimulating factor-3 (NNT-1/BSF-3) is a recently cloned gp130 cytokine, acting through the tripartite ciliary neurotrophic factor receptor (CNTFR) alpha/leukemia inhibitory factor receptor (LIFR)/gp130 receptor complex. The aim of the current study was to investigate the role of NNT-1/BSF-3 in corticotroph cell function and further characterize NNT-1/BSF-3 signaling pathways. Using RT-PCR, expression of ciliary neurotrophic factor receptor alpha, leukemia inhibitory factor receptor, and gp130 could be demonstrated in mRNA derived from murine corticotroph AtT-20 cells and murine pituitary tissue. Incubation of AtT-20 cells with 10 ng/ml recombinant human NNT-1/BSF-3 rapidly induced tyrosine-phosphorylation of signal transducer and activator of transcription (STAT)3 and STAT1 at 5 and 10 min. Proopiomelanocortin promoter activity and suppressor of cytokine signaling (SOCS)-3 promoter activity were significantly stimulated by NNT-1/BSF-3 4.0 +/- 0.3- and 5.9 +/- 0.2-fold, respectively. In comparison with untreated control, NNT-1/BSF-3 significantly stimulated ACTH secretion at 24 and 48 h 1.7 +/- 0.2-fold and 1.5 +/- 0.1-fold above baseline. In comparison with mock-transfected cells, stable overexpression of SOCS-3 in AtT-20 cells abolished NNT-1/BSF-3-induced STAT1 and STAT3 phosphorylation and almost completely inhibited STAT-dependent proopiomelanocortin promoter and SOCS-3 promoter activities. In addition, NNT-1/BSF-3-induced ACTH secretion at 48 h was significantly attenuated by SOCS-3 overexpression. In summary, we have shown that NNT-1/BSF-3 is a modulator of corticotroph cell function, which is negatively regulated by SOCS-3. Our data indicate that the activation of the Jak-STAT cascade is essential for corticotroph NNT-1/BSF-3 signaling. Further studies will have to investigate the possible in vivo role of NNT-1/BSF-3 as a neuroimmunoendocrine modulator of hypothalamus-pituitary-adrenal axis stress response.


Subject(s)
Cytokines/metabolism , Pituitary Gland/cytology , Proteins/metabolism , Repressor Proteins , Signal Transduction/physiology , Transcription Factors , Adrenocorticotropic Hormone/metabolism , Animals , Antigens, CD/genetics , Cells, Cultured , Cytokine Receptor gp130 , Cytokines/genetics , DNA-Binding Proteins/metabolism , Gene Expression/physiology , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit , Membrane Glycoproteins/genetics , Neuroimmunomodulation/physiology , Phosphorylation , Pituitary Gland/physiology , Pro-Opiomelanocortin/genetics , Proteins/genetics , RNA, Messenger/analysis , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Cytokine/genetics , Receptors, OSM-LIF , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism
20.
J Immunol ; 170(4): 2100-5, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574381

ABSTRACT

IL-18 is an important cytokine in autoimmune and inflammatory diseases through the induction of IFN-gamma, TNF-alpha, and IL-1. We report herein that collagen-induced arthritis (CIA) in mice is inhibited by treatment with murine IL-18 binding protein (mIL-18BP). CIA was induced in DBA/1J mice by the injection of bovine type II collagen (CII) in IFA with added Mycobacterium tuberculosis on days 0 and 21. The mice were then treated for 3 wk with PBS or with two doses of mIL-18BP (0.5 and 3 mg/kg) as a fusion protein with the Fc portion of murine IgG1. Both the clinical disease activity scores and the histological scores of joint damage were reduced 50% in mice treated with either dose of mIL-18BP. Proliferation of CII-stimulated spleen and lymph node cells as well as the change in serum levels of IgG1 and IgG2a Ab to collagen between days 21 and 42 were decreased in mice treated with mIL-18BP. The production of IFN-gamma, TNF-alpha, and IL-1beta in cultured spleen cells was reduced by in vivo treatment with low dose, but not high dose, mIL-18BP. FACS analysis showed a slight decrease in NK cells and an increase in CD4(+) T cells in spleens of mice treated with mIL-18BP. The steady state mRNA levels of IFN-gamma, TNF-alpha, and IL-1beta in isolated joints were all decreased in mice treated with both doses of mIL-18BP. The mechanisms of mIL-18BP inhibition of CIA include reductions in cell-mediated and humoral immunity to collagen as well as decreases in production of proinflammatory cytokines in the spleen and joints.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Collagen/administration & dosage , Collagen/immunology , Glycoproteins/physiology , Interleukin-18/metabolism , Animals , Arthritis, Experimental/pathology , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cattle , Cell Count , Collagen/antagonists & inhibitors , Glycoproteins/therapeutic use , Immunoglobulin G/biosynthesis , Intercellular Signaling Peptides and Proteins , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Subsets/cytology , Macrophages/cytology , Mice , Mice, Inbred DBA , RNA, Messenger/analysis , RNA, Messenger/antagonists & inhibitors , Severity of Illness Index , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...