Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
JCO Clin Cancer Inform ; 6: e2100173, 2022 03.
Article in English | MEDLINE | ID: mdl-35467964

ABSTRACT

PURPOSE: Overall survival (OS) is the gold standard end point for establishing clinical benefits in phase III oncology trials. However, these trials are associated with low success rates, largely driven by failure to meet the primary end point. Surrogate end points such as progression-free survival (PFS) are increasingly being used as indicators of biologic drug activity and to inform early go/no-go decisions in oncology drug development. We developed OSPred, a digital health aid that combines actual clinical data and machine intelligence approaches to visualize correlation trends between early (PFS-based) and late (OS) end points and provide support for shared decision making in the drug development pipeline. METHODS: OSPred is based on a trial-level data set of 81 reports (35 anticancer drugs with various mechanisms of action; 156 observations) in non-small-cell lung cancer (NSCLC). OSPred was developed using R Shiny, with packages ggplot2, metafor, boot, dplyr, and mvtnorm, to analyze and visualize correlation results and predict OS hazard ratio (HR OS) on the basis of user-inputted PFS-based data, namely, HR PFS, or the odds ratio of PFS at 4 (OR PFS4) or 6 (OR PFS6) months. RESULTS: The three main features of the tool are as follows: prediction of HR OS on the basis of user-inputted early end point values; visualization of comparisons of the user's investigational drug with other drugs in the NSCLC setting, including by specific MoA; and creation of a probability density chart, providing point prediction and CIs for HR OS. A working version of the tool for download is linked. CONCLUSION: The OSPred tool offers interactive visualization of clinical trial end point correlations with reference to a large pool of historical NSCLC studies. Its focused capability has the potential to digitally transform and accelerate data-driven decision making as part of the drug development process.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Clinical Trials, Phase III as Topic , Endpoint Determination , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Progression-Free Survival , Proportional Hazards Models
2.
Front Oncol ; 11: 672916, 2021.
Article in English | MEDLINE | ID: mdl-34381708

ABSTRACT

Early endpoints, such as progression-free survival (PFS), are increasingly used as surrogates for overall survival (OS) to accelerate approval of novel oncology agents. Compiling trial-level data from randomized controlled trials (RCTs) could help to develop a predictive framework to ascertain correlation trends between treatment effects for early and late endpoints. Through trial-level correlation and random-effects meta-regression analysis, we assessed the relationship between hazard ratio (HR) OS and (1) HR PFS and (2) odds ratio (OR) PFS at 4 and 6 months, stratified according to the mechanism of action of the investigational product. Using multiple source databases, we compiled a data set including 81 phase II-IV RCTs (35 drugs and 156 observations) of patients with non-small-cell lung cancer. Low-to-moderate correlations were generally observed between treatment effects for early endpoints (based on PFS) and HR OS across trials of agents with different mechanisms of action. Moderate correlations were seen between treatment effects for HR PFS and HR OS across all trials, and in the programmed cell death-1/programmed cell death ligand-1 and epidermal growth factor receptor trial subsets. Although these results constitute an important step, caution is advised, as there are some limitations to our evaluation, and an additional patient-level analysis would be needed to establish true surrogacy.

4.
Bone ; 50(4): 885-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245630

ABSTRACT

Saracatinib (AZD0530) is an orally active once-daily Src kinase inhibitor which modulates key signaling pathways in cancer cells. In a Phase I study in patients with advanced solid malignancies resistant to standard treatment we assessed the effect of saracatinib on bone turnover. Fifty-one patients were randomized into three parallel groups to receive saracatinib 50, 125 or 175 mg/day. After a single dose followed by a 7-day washout, patients received once-daily doses for 21 days. Bone turnover markers were measured in serum and urine samples collected before dosing on days 1, 2, 3, 17 and 28. Samples were available at baseline and more than one other time point for 44 patients. Bone resorption markers were significantly decreased by saracatinib. Serum cross-linked C-terminal telopeptide of type I collagen (sCTX) changed in the 50, 125 and 175 mg/day groups by -36% (95% CI -58, -4), -64% (95% CI -75, -48) and -75% (95% CI -83, -61), respectively, at day 28. Urinary cross-linked N-terminal telopeptide of type I collagen/creatinine ratio (uNTX/Cr) changed in the 50, 125 and 175 mg/day groups by; -13% (95% CI -33, 13), -48% (95% CI -59, -34) and -50% (95% CI -62, -35), respectively, at day 28. The significant decreases in bone resorption markers indicate that suppression of Src kinase inhibits osteoclast activity in patients with advanced cancer. This result suggests that saracatinib may have therapeutic benefit in metastatic bone disease.


Subject(s)
Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Bone Remodeling/drug effects , Neoplasms/drug therapy , Neoplasms/physiopathology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , src-Family Kinases/antagonists & inhibitors , Adult , Aged , Benzodioxoles/adverse effects , Biomarkers, Tumor/metabolism , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/adverse effects , Young Adult , src-Family Kinases/metabolism
5.
Clin Cancer Res ; 16(19): 4876-83, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20805299

ABSTRACT

PURPOSE: This dose-escalation study evaluated the safety, tolerability, and pharmacokinetics (PK) of the oral Src inhibitor saracatinib (AZD0530) in patients with advanced solid malignancies. Tumor biopsy samples were taken to investigate the effect of saracatinib on Src activity in tumors. EXPERIMENTAL DESIGN: Part A of the study followed a multiple-ascending dose design to establish the maximum tolerated dose (MTD) of saracatinib. Part B was a randomized, parallel-group, cohort-expansion phase to further assess tolerated doses. Safety, tolerability, and Src activity (immunohistochemistry and lysate-based methodologies) were assessed after 21 days of once-daily oral dosing. PK was assessed after single and multiple dosing. RESULTS: In part A, 30 patients received once-daily saracatinib at doses of 60 to 250 mg; the MTD was established as 175 mg. In part B, 51 patients were randomized to receive 50 mg (n = 16), 125 mg (n = 16), or 175 mg (n = 19) of saracatinib. The most common grade ≥3 events considered to be treatment related were anemia, diarrhea, and asthenia. Tumor Src activity was reduced following saracatinib treatment. The area under the concentration-time curve and C(max) of saracatinib increased with increasing dose. Saracatinib accumulated 4- to 5-fold on once-daily dosing to reach steady-state exposure after 10 to 17 days of dosing. The half-life was ∼40 hours. CONCLUSIONS: Saracatinib was well tolerated in patients with advanced solid malignancies. A reduction in tumor Src activity was observed. PK data show that saracatinib is suitable for once-daily oral dosing. Based on this study, the recommended dose for the phase II studies was chosen to be 175 mg/d.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Benzodioxoles/pharmacokinetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Quinazolines/pharmacokinetics , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Benzodioxoles/administration & dosage , Benzodioxoles/adverse effects , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Enzyme Activation/drug effects , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/diagnosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/adverse effects , Treatment Outcome , Young Adult , src-Family Kinases/metabolism
6.
Anticancer Res ; 30(7): 2935-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683035

ABSTRACT

BACKGROUND: Saracatinib (AZD0530), a potent Src inhibitor, is a subject of current evaluation as an anticancer therapy. Increased plasma creatinine levels have previously been observed after saracatinib administration in healthy subjects and this study was undertaken to characterize the underlying mechanism of this increase. SUBJECTS AND METHODS: 56 healthy male subjects were assigned to either single- (n=28; randomised to placebo or saracatinib 500 mg) or multiple-dose oral treatment (n=28; randomised to placebo or saracatinib 125 mg for 14 days). Renal function variables assessed included inulin clearance and tubular secretion of creatinine. RESULTS: Saracatinib led to a reduction in mean creatinine fractional excretion ratio, which was due to a reduction in tubular secretion of creatinine. Increased plasma creatinine was not associated with decreased glomerular filtration rate or increased creatinine production. CONCLUSION: The observed increase in plasma creatinine after saracatinib administration was due to reduced tubular secretion of creatinine, but was not considered to be clinically relevant in the context of this study.


Subject(s)
Benzodioxoles/adverse effects , Kidney/drug effects , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , src-Family Kinases/antagonists & inhibitors , Adolescent , Adult , Benzodioxoles/administration & dosage , Creatinine/blood , Creatinine/urine , Double-Blind Method , Humans , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Young Adult
7.
J Cancer Res Clin Oncol ; 135(3): 467-76, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18787840

ABSTRACT

PURPOSE: Two large, randomized, placebo-controlled trials (IRESSA NSCLC Trial Assessing Combination Therapy; INTACT 1 and 2) in non-small-cell lung cancer (NSCLC) failed to show survival benefit for gefitinib (IRESSA) in combination with first-line platinum-based chemotherapy. Epidermal growth factor receptor (EGFR) staining was assessed retrospectively in relation to survival response to gefitinib in combination with chemotherapy. METHODS: Tumor biopsies obtained prior to start of therapy were assessed by immunohistochemistry for EGFR using the Dako EGFR pharmDx assay (Dako, Denmark). Analyses were stratified by trial and performed independently for patients randomized to placebo and gefitinib as well as for both treatment groups combined. A restricted backwards elimination Cox regression analysis was conducted to identify independent EGFR factors that were statistically significant (P < 0.10), and these were also tested for treatment interaction to assess if they served as predictive factors. RESULTS: Analyses found two statistically significant EGFR-based prognostic factors representing growth pattern and percent membrane staining in patients treated with gefitinib (P = 0.0023), placebo (P = 0.0128), and both combined (P < 0.0001). The prognostic effect was independent of other known prognostic factors. There was no predictive effect of either the growth pattern or membrane staining variable. CONCLUSIONS: While some previous studies indicate that higher EGFR expression correlates with poor survival, our analyses provide statistically significant evidence that the combination of EGFR expression and growth pattern is a strong prognostic indicator for improved survival within this setting. The effects of membrane staining and growth pattern are still significant when adjusting for mutation.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Biopsy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , ErbB Receptors/metabolism , Gefitinib , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplasm Staging , Placebos , Platinum Compounds/administration & dosage , Prognosis , Survival Analysis
8.
BJU Int ; 100(4): 765-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17822457

ABSTRACT

OBJECTIVE: To investigate, in a phase II trial, the use of the epidermal growth factor receptor (EGFR) inhibitor gefitinib as monotherapy in patients with non-metastatic hormone refractory prostate cancer (HRPC), as current treatment options for this disease are limited, and agents which target the EGFR should be assessed because EGFR is highly expressed in prostate cancer and associated with a poor prognosis. PATIENTS AND METHODS: Patients with histologically or cytologically confirmed cancer of the prostate with no evidence of metastatic disease were enrolled into this open-label, multicentre study of monotherapy with gefitinib 500 mg/day. The primary endpoint of the study was biochemical response, defined as a >/=50% decrease in serum prostate-specific antigen (PSA) level. RESULTS: Fifty-eight men were enrolled across 10 centres in the USA; none of the 40 evaluable patients had a PSA response. Gefitinib was generally well tolerated, with diarrhoea being the most common treatment- related adverse event, in 71% of patients. There was treatment-related grade 3 diarrhoea in 5% of patients, with no grade 4 adverse events or deaths during the course of the study. CONCLUSIONS: Gefitinib has no single-agent activity in non-metastatic HRPC, as assessed by decreases in serum PSA level. This phase II study also confirmed the well-established favourable tolerability profile of gefitinib monotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Quinazolines/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Gefitinib , Humans , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Quinazolines/adverse effects , Treatment Outcome
9.
J Clin Oncol ; 23(31): 8081-92, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16204011

ABSTRACT

PURPOSE: Most cases of non-small-cell lung cancer (NSCLC) with dramatic responses to gefitinib have specific activating mutations in the epidermal growth factor receptor (EGFR), but the predictive value of these mutations has not been defined in large clinical trials. The goal of this study was to determine the contribution of molecular alterations in EGFR to response and survival within the phase II (IDEAL) and phase III (INTACT) trials of gefitinib. PATIENTS AND METHODS: We analyzed the frequency of EGFR mutations in lung cancer specimens from both the IDEAL and INTACT trials and compared it with EGFR gene amplification, another genetic abnormality in NSCLC. RESULTS: EGFR mutations correlated with previously identified clinical features of gefitinib response, including adenocarcinoma histology, absence of smoking history, female sex, and Asian ethnicity. No such association was seen in patients whose tumors had EGFR amplification, suggesting that these molecular markers identify different biologic subsets of NSCLC. In the IDEAL trials, responses to gefitinib were seen in six of 13 tumors (46%) with an EGFR mutation, two of seven tumors (29%) with amplification, and five of 56 tumors (9%) with neither mutation nor amplification (P = .001 for either EGFR mutation or amplification v neither abnormality). Analysis of the INTACT trials did not show a statistically significant difference in response to gefitinib plus chemotherapy according to EGFR genotype. CONCLUSION: EGFR mutations and, to a lesser extent, amplification appear to identify distinct subsets of NSCLC with an increased response to gefitinib. The combination of gefitinib with chemotherapy does not improve survival in patients with these molecular markers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Gene Amplification , Lung Neoplasms/genetics , Mutation , Quinazolines/therapeutic use , Adult , Aged , Antineoplastic Agents , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Gefitinib , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...