Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Invest New Drugs ; 36(5): 886-894, 2018 10.
Article in English | MEDLINE | ID: mdl-29766337

ABSTRACT

Background The MET tyrosine kinase and its ligand, hepatocyte growth factor (HGF) also known as scatter factor, are associated with tumourigenesis and metastasis by promotion of scattering, proliferation, angiogenesis, motility and invasion. ASLAN-002 is a potent inhibitor of MET as well as related kinases. A phase I dose escalation study was conducted to determine the safety and pharmacokinetics of ASLAN-002 in patients with advanced cancer. Methods Patients with advanced or metastatic solid tumours, who had progressed on standard therapy or for whom standard therapy was not known, were administered ASLAN-002 orally. The starting dose was 100 mg once daily (QD) with subsequent cohorts to receive doses of 200 mg QD, 300 mg QD, 450 mg QD, 600 mg QD, 300 mg twice daily (BID), 450 mg BID, and 600 mg BID. Results Forty patients were included across 7 dose cohorts. Cohort 8 (600 mg BID) was not opened due to the lack of appreciable pharmacokinetic (PK) differences between 300 mg BID and 450 mg BID and higher incidences of grade 3 or 4 adverse events (AE) in Cohort 7 (450 mg BID). Fifteen patients (37.5%) experienced a grade 3 or 4 AE. The most commonly reported AEs were nausea (55%), fatigue (47.5%) and constipation (30%). One dose limiting toxicity (DLT) of atrial fibrillation was observed with 450 mg BID. Conclusions ASLAN-002 is well tolerated at 300 mg BID and is the recommended dose for future phase II studies (RP2D). Clinical Trials Registry Number: NCT01721148 .


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Fatigue/chemically induced , Fatigue/metabolism , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Nausea/metabolism , Neoplasms/metabolism
2.
Invest New Drugs ; 26(2): 159-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18219445

ABSTRACT

BACKGROUND: ZD6126 is a novel vascular-targeting agent that disrupts the endothelial tubulin cytoskeleton causing selective occlusion of tumor vasculature and extensive tumor necrosis. This Phase I clinical study was conducted to evaluate the dose and administration schedule of ZD6126. METHODS: Adult patients with solid tumors refractory to existing treatments received a 10-min, single-dose intravenous infusion of ZD6126 every 14 or 21 days. Subsequent dose escalation was performed, based on the incidence of adverse events (AEs) within the first cycle of drug administration. Blood samples were obtained for pharmacokinetic analysis, and the effects of ZD6126 on tumor vasculature were visualized using DCE-MRI technology. RESULTS: Forty-four patients received ZD6126 (5-112 mg/m2 in the 21-day schedule, n=35; 40-80 mg/m2 in the 14-day schedule, n=9). Common AEs were similar in both groups and included abdominal pain, nausea and vomiting, which appeared to be dose related. The incidence of abdominal pain at 112 mg/m2 in the 21-day study prevented further dose escalation. Pharmacokinetic studies confirmed that ZD6126 is rapidly hydrolyzed to ZD6126 phenol. There was no difference in the pharmacokinetics of ZD6126 phenol upon repeat administration or between the two dosing regimens. DCE-MRI evaluation has demonstrated the antivascular effects of ZD6126. CONCLUSIONS: This study identified that ZD6126 administered every 2 or 3 weeks at 80 mg/m2 was well tolerated, with mild but manageable gastrointestinal AEs. In approximately 11% (5 out of 44) of patients, ZD6126 was associated with cardiac events categorized as dose limiting toxicities (one patient with asymptomatic decreased left ventricular ejection fraction (LVEF), two with increased troponin concentrations, one with myocardial ischemia, and one with ECG signs of myocardial ischemia).


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Organophosphorus Compounds/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Female , Humans , Infusions, Intravenous , Magnetic Resonance Imaging , Male , Middle Aged , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/pharmacokinetics , Tubulin/drug effects
3.
Anticancer Drugs ; 18(5): 569-79, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17414626

ABSTRACT

ZD6474 is a novel, orally available inhibitor of vascular endothelial growth factor receptor kinase insert domain receptor/flk-1 tyrosine kinase activity with additional activity against the epidermal growth factor receptor-1 tyrosine kinase. The aim of this study was to evaluate ZD6474, alone and in combination with gemcitabine, in an orthotopic model of metastatic pancreatic cancer. Nude mice (nine to 10/group) were injected orthotopically with 1x10(6) L3.6pl human pancreatic cancer cells. Eight days later, treatment was initiated with vehicle only, gemcitabine (100 mg/kg intraperitoneal twice weekly), ZD6474 (50 mg/kg oral once daily) or a combination of the two treatments. Animals were killed on day 24 posttreatment initiation. The phosphorylation status level of vascular endothelial growth factor receptor-2 and epidermal growth factor receptor as well as the phosphorylation level of AKT and extracellular signal-regulated kinase-1/2 in different human pancreatic carcinoma cells and in human umbilical vein endothelial cells was analyzed by Western blotting. Compared with controls (1231 mg), the mean weight of treated tumors was reduced to 836, 541 and 308 mg in the gemcitabine, ZD6474 and combination groups, respectively. Lymph node metastasis was significantly reduced in both the ZD6474 alone and combined treatment groups, with 3/10 and 1/5 animals developing metastases, compared with 10/10 and 9/9 in the control and gemcitabine groups (P<0.003 and <0.0003, respectively). Microvessel density and cell proliferation were significantly reduced in the ZD6474 and combined treatment groups (P<0.02). Immunohistochemistry of tumor samples following treatment with ZD6474 resulted in a reduction of the activated and phosphorylated epidermal growth factor receptor, whereas total epidermal growth factor receptor levels were comparable with control tumors. On the basis of Western blot analysis, ZD6474 provides inhibition of tumor angiogenesis through an anti-vascular endothelial growth factor receptor-2 mechanism and inhibition of cancer cell growth through an anti-epidermal growth factor receptor mechanism. ZD6474 decreased primary pancreatic tumor growth and reduced lymph node and liver metastases compared with controls or gemcitabine alone. Tumor growth was inhibited further in animals receiving ZD6474 and gemcitabine in combination.


Subject(s)
Angiogenesis Inhibitors , Antineoplastic Agents , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Piperidines/pharmacology , Quinazolines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Skin/blood supply , Skin/pathology , Transplantation, Heterologous , Gemcitabine
4.
Contemp Clin Trials ; 28(2): 138-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16843736

ABSTRACT

This paper examines the design of phase II trials in oncology and recommends departing from the traditional uncontrolled trial design. Entrance into phase II clinical evaluation represents a key milestone in the development of any new cancer therapy. As novel molecular-targeted therapies are introduced, whose primary action is to slow the growth of tumors, it will be important to ensure that the clinical trial design will effectively capture any clinical benefit of these agents. The objective of a phase II trial should, in addition to identifying active therapies, be extended to identifying those that are likely to be successful in pivotal trials. It is therefore necessary to quantify the likelihood of either incorrectly halting the development of an active agent or continuing development of an ineffective agent. We believe only randomized studies with comparative intent and including a concurrent active control, can reliably assess these risks corresponding to significance and power. Given that the objective of phase II studies is to identify promising treatments, it is important not be constrained by conventional levels of significance. This paper will review the various approaches to phase II trial design in oncology and provide a framework for fully powered randomized trials of a moderate size. For example, a randomized trial of just 100 patients could lead to the termination of development of 90% of inactive agents whereas at least 80% of agents with a meaningful and realistic increase in progression-free survival would be identified for confirmatory study. We believe randomized studies with progression-free survival endpoints are the most powerful and economical method of determining the clinical activity of new cytostatic agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Trials, Phase II as Topic/methods , Randomized Controlled Trials as Topic/methods , Research Design/standards , Algorithms , Antineoplastic Agents/pharmacology , Clinical Trials, Phase II as Topic/statistics & numerical data , Computer Simulation , Disease-Free Survival , Humans , Models, Statistical , Neoplasms/drug therapy , Randomized Controlled Trials as Topic/statistics & numerical data
5.
Contemp Clin Trials ; 28(2): 146-52, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16807129

ABSTRACT

The traditional development paradigm for phase II trials in oncology has been challenged in recent years by the introduction of cytostatic therapies. These agents slow the growth of tumors rather than cause high rates of shrinkage, this argues for the use of endpoints that measure growth inhibition such as progression free survival. We have previously argued the need for randomized trials in this setting. Here we discuss methodological solutions to enhance the development decision at the end of phase II in the context of progression endpoints employed in randomized trials. There are well recognized issues associated with progression endpoints relating to bias in the timing and interpretation of assessments. In this paper we present design and analysis solutions that will minimize bias by using methods that are either partially or completely time independent. We also discuss other design features to maximize the information yielded in a phase II setting. We advocate the creation of progression endpoints that utilize all available progression data rather than early fixed time-point analyses and show that little is to be gained by assessing progression status any more frequently than would be required in routine clinical practice. Such design and analysis measures will optimize the development decision made at the end of phase II clinical evaluation.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Trials, Phase II as Topic/methods , Neoplasms/drug therapy , Randomized Controlled Trials as Topic/methods , Research Design , Algorithms , Antineoplastic Agents/pharmacology , Bias , Clinical Trials, Phase II as Topic/statistics & numerical data , Disease Progression , Disease-Free Survival , Humans , Models, Statistical , Randomized Controlled Trials as Topic/statistics & numerical data
6.
Neoplasia ; 7(10): 957-66, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16242078

ABSTRACT

ZD6126 is a novel vascular-targeting agent that acts by disrupting the tubulin cytoskeleton of an immature tumor endothelium, leading to an occlusion of tumor blood vessels and a subsequent tumor necrosis. We wanted to evaluate ZD6126 in primary and metastatic tumor models of human pancreatic cancer. Nude mice were injected orthotopically with L3.6pl pancreatic cancer cells. In single and multiple dosing experiments, mice received ZD6126, gemcitabine, a combination of both agents, or no treatment. For the induction of metastatic diseases, additional groups of mice were injected with L3.6pl cells into the spleen. Twenty-four hours after a single-dose treatment, ZD6126 therapy led to an extensive central tumor necrosis, which was not seen after gemcitabine treatment. Multiple dosing of ZD6126 resulted in a significant growth inhibition of primary tumors and a marked reduction of spontaneous liver and lymph node metastases. Experimental metastatic diseases could be significantly controlled by a combination of ZD6126 and gemcitabine, as shown by a reduction of the number and size of established liver metastases. As shown by additional in vitro and in vivo experiments, possible mechanisms involve antivascular activities and subsequent antiproliferative and proapoptotic effects of ZD6126 on tumor cells, whereas direct activities against tumor cells seem unlikely. These data highlight the antitumor and antimetastatic effects of ZD6126 in human pancreatic cancer and reveal benefits of adding ZD6126 to standard gemcitabine therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Organophosphorus Compounds/pharmacology , Pancreatic Neoplasms/pathology , Tubulin/chemistry , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Coloring Agents/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Liver/pathology , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Chemical , Necrosis , Neoplasm Metastasis , Neovascularization, Pathologic , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Umbilical Veins/cytology , Gemcitabine
7.
Clin Cancer Res ; 11(9): 3369-76, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867237

ABSTRACT

PURPOSE: To determine the efficacy and safety of ZD6474, an orally available inhibitor of vascular endothelial growth factor receptor-2 (VEGFR-2) tyrosine kinase with additional activity against the epidermal growth factor receptor (EGFR) tyrosine kinase, in patients with previously treated metastatic breast cancer. PATIENTS AND METHODS: Eligible patients had histologically confirmed metastatic breast cancer and had received prior treatment with an anthracycline and taxane; measurable disease was required. Patients were enrolled sequentially into one of two dose cohorts, 100 or 300 mg orally once daily; 28 days defined one cycle. The primary end point was objective response rate; pharmacokinetics and serial pharmacodynamic studies were obtained. RESULTS: Forty-six patients were enrolled between May 2002 and April 2003, and 44 were evaluable for response. Diarrhea was the most commonly reported toxicity and seemed dose related (grade >/=2: 4.5% and 37.5% in the 100 and 300 mg cohorts, respectively). Rash was reported by 26% of patients but was never worse than grade 2. Seven patients in the 300 mg cohort had asymptomatic grade 1 prolongation of the QTc interval. Hypertension requiring treatment was not reported. There were no objective responses; one patient in the 300 mg cohort had stable disease >/=24 weeks. All patients in the 300 mg cohort and 90% of patients in the 100 mg cohort achieved steady-state concentrations exceeding the IC(50) for VEGF inhibition in preclinical models. CONCLUSION: ZD6474 monotherapy was generally well tolerated but had limited monotherapy activity in patients with refractory metastatic breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Piperidines/therapeutic use , Quinazolines/therapeutic use , Adult , Aged , Aged, 80 and over , Anthracyclines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Bridged-Ring Compounds/administration & dosage , Diarrhea/chemically induced , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , Female , Humans , Middle Aged , Nausea/chemically induced , Neoplasm Metastasis , Piperidines/adverse effects , Piperidines/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Taxoids/administration & dosage , Treatment Outcome , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
8.
Clin Cancer Res ; 10(23): 8028-36, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585638

ABSTRACT

PURPOSE: The Src family comprises a family of nonreceptor intracellular tyrosine kinases that mediate a variety of cellular pathways. Src kinases are overexpressed in a variety of human tumors, including cancer of the colon, breast, and pancreas, and they are an integral part of tumor cell signaling pathways associated with migration, proliferation, adhesion, and angiogenesis. EXPERIMENTAL DESIGN: We investigated whether the blockade of Src kinase by daily oral administration of the novel Src tyrosine kinase inhibitor AZM475271 [kindly provided by AstraZeneca (Macclesfield, United Kingdom)], alone or in combination with intraperitoneal gemcitabine, can inhibit growth and metastasis of orthotopically implanted human pancreatic carcinoma cells in nude mice. RESULTS: Treatment with AZM475271 alone reduced the primary pancreatic tumor volume by approximately 40%, whereas AZM475271 plus gemcitabine reduced tumor volume by 90%. Furthermore, treatment with AZM475271 and gemcitabine significantly reduced metastasis: none of eight animals who received the combination treatment had lymph node or liver metastases, compared with five of five and three of five animals, respectively, in the control group (P = 0.001). Src inhibition by AZM475271 (alone or with gemcitabine) was associated with significantly reduced tumor cell proliferation, decreased tumor microvessel density, and increased apoptosis in vivo. Moreover, these effects were all significantly increased when gemcitabine was combined with AZM475271 compared with gemcitabine alone. CONCLUSIONS: Src inhibition by AZM475271, either alone or in combination with gemcitabine, demonstrated significant antitumor and antimetastatic activity in an orthotopic nude mouse model for human pancreatic cancer. The combination of AZM475271 with gemcitabine sensitized tumor cells to the cytotoxic effect of gemcitabine.


Subject(s)
Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Enzyme Inhibitors/therapeutic use , Pancreatic Neoplasms/drug therapy , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Drug Therapy, Combination , Flow Cytometry , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Male , Mice , Mice, Nude , NIH 3T3 Cells , Pancreatic Neoplasms/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , src-Family Kinases/metabolism , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...