Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Lancet Oncol ; 22(3): 370-380, 2021 03.
Article in English | MEDLINE | ID: mdl-33587894

ABSTRACT

BACKGROUND: Melanoma lacks validated blood-based biomarkers for monitoring and predicting treatment efficacy. Cell-free circulating tumour DNA (ctDNA) is a promising biomarker; however, various detection methods have been used, and, to date, no large studies have examined the association between serial changes in ctDNA and survival after BRAF, MEK, or BRAF plus MEK inhibitor therapy. We aimed to evaluate whether baseline ctDNA concentrations and kinetics could predict survival outcomes. METHODS: In this clinical validation study, we used analytically validated droplet digital PCR assays to measure BRAFV600-mutant ctDNA in pretreatment and on-treatment plasma samples from patients aged 18 years or older enrolled in two clinical trials. COMBI-d (NCT01584648) was a double-blind, randomised phase 3 study of dabrafenib plus trametinib versus dabrafenib plus placebo in previously untreated patients with BRAFV600 mutation-positive unresectable or metastatic melanoma. Patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. COMBI-MB (NCT02039947) was an open-label, non-randomised, phase 2 study evaluating dabrafenib plus trametinib in patients with BRAFV600 mutation-positive metastatic melanoma and brain metastases. Patients in cohort A of COMBI-MB had asymptomatic brain metastases, no previous local brain-directed therapy, and an ECOG performance status of 0 or 1. Biomarker analysis was a prespecified exploratory endpoint in both trials and performed in the intention-to-treat populations in COMBI-d and COMBI-MB. We investigated the association between mutant copy number (baseline or week 4 or zero conversion status) and efficacy endpoints (progression-free survival, overall survival, and best overall response). We used Cox models, Kaplan-Meier plots, and log-rank tests to explore the association of pretreatment ctDNA concentrations with progression-free survival and overall survival. The effect of additional prognostic variables such as lactate dehydrogenase was also investigated in addition to the mutant copy number. FINDINGS: In COMBI-d, pretreatment plasma samples were available from 345 (82%) of 423 patients and on-treatment (week 4) plasma samples were available from 224 (53%) of 423 patients. In cohort A of COMBI-MB, pretreatment and on-treatment samples were available from 38 (50%) of 76 patients with intracranial and extracranial metastatic melanoma. ctDNA was detected in pretreatment samples from 320 (93%) of 345 patients (COMBI-d) and 34 (89%) of 38 patients (COMBI-MB). When assessed as a continuous variable, elevated baseline BRAFV600 mutation-positive ctDNA concentration was associated with worse overall survival outcome (hazard ratio [HR] 1·13 [95% CI 1·09-1·18], p<0·0001 by univariate analysis), independent of treatment group and baseline lactate dehydrogenase concentrations (1·08 [1·03-1·13], p=0·0020), in COMBI-d. A ctDNA cutoff point of 64 copies per mL of plasma stratified patients enrolled in COMBI-d as high risk or low risk with respect to survival outcomes (HR 1·74 [95% CI 1·37-2·21], p<0·0001 for progression-free survival; 2·23 [1·73-2·87], p<0·0001 for overall survival) and was validated in the COMBI-MB cohort (3·20 [1·39-7·34], p=0·0047 for progression-free survival; 2·94 [1·18-7·32], p=0·016 for overall survival). In COMBI-d, undetectable ctDNA at week 4 was significantly associated with extended progression-free and overall survival, particularly in patients with elevated lactate dehydrogenase concentrations (HR 1·99 [95% CI 1·08-3·64], p=0·027 for progression-free survival; 2·38 [1·24-4·54], p=0·0089 for overall survival). INTERPRETATION: Pretreatment and on-treatment BRAFV600-mutant ctDNA measurements could serve as independent, predictive biomarkers of clinical outcome with targeted therapy. FUNDING: Novartis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/secondary , Circulating Tumor DNA/genetics , Melanoma/pathology , Aged , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Circulating Tumor DNA/analysis , Double-Blind Method , Female , Follow-Up Studies , Humans , Imidazoles/administration & dosage , Male , Melanoma/drug therapy , Melanoma/genetics , Middle Aged , Oximes/administration & dosage , Prognosis , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Survival Rate
2.
Lancet Oncol ; 21(3): 358-372, 2020 03.
Article in English | MEDLINE | ID: mdl-32007138

ABSTRACT

BACKGROUND: Adjuvant dabrafenib plus trametinib reduced the risk of relapse versus placebo in patients with resected, BRAFV600-mutant, stage III melanoma in the phase 3 COMBI-AD trial. This prespecified exploratory biomarker analysis aimed to evaluate potential prognostic or predictive factors and mechanisms of resistance to adjuvant targeted therapy. METHODS: COMBI-AD is a randomised, double-blind, placebo-controlled, phase 3 trial comparing dabrafenib 150 mg orally twice daily plus trametinib 2 mg orally once daily versus two matched placebos. Study participants were at least 18 years of age and underwent complete resection of stage IIIA (lymph node metastases >1 mm), IIIB, or IIIC cutaneous melanoma, per American Joint Committee on Cancer 7th edition criteria, with a BRAFV600E or BRAFV600K mutation. Patients were randomly assigned (1:1) to the two treatment groups by an interactive voice response system, stratified by mutation type and disease stage. Patients, physicians, and the investigators who analysed data were masked to treatment allocation. The primary outcome was relapse-free survival, defined as the time from randomisation to disease recurrence or death from any cause. Biomarker assessment was a prespecified exploratory outcome of the trial. We assessed intrinsic tumour genomic features by use of next-generation DNA sequencing and characteristics of the tumour microenvironment by use of a NanoString RNA assay, which might provide prognostic and predictive information. This trial is registered with ClinicalTrials.gov, number NCT01682083, and is ongoing but no longer recruiting participants. FINDINGS: Between Jan 31, 2013, and Dec 11, 2014, 870 patients were enrolled in the trial. Median follow-up at data cutoff (April 30, 2018) was 44 months (IQR 38-49) in the dabrafenib plus trametinib group and 42 months (21-49) in the placebo group. Intrinsic tumour genomic features were assessed in 368 patients (DNA sequencing set) and tumour microenvironment characteristics were assessed in 507 patients (NanoString biomarker set). MAPK pathway genomic alterations at baseline did not affect treatment benefit or clinical outcome. An IFNγ gene expression signature higher than the median was prognostic for prolonged relapse-free survival in both treatment groups. Tumour mutational burden was independently prognostic for relapse-free survival in the placebo group (high TMB, top third; hazard ratio [HR] 0·56, 95% CI 0·37-0·85, p=0·0056), but not in the dabrafenib plus trametinib group (0·83, 95% CI 0·53-1·32, p=0·44). Patients with tumour mutational burden in the lower two terciles seem to derive a substantial long-term relapse-free survival benefit from targeted therapy (HR [versus placebo] 0·49, 95% CI 0·35-0·68, p<0·0001). However, patients with high tumour mutational burden seem to have a less pronounced benefit with targeted therapy (HR [versus placebo] 0·75, 95% CI 0·44-1·26, p=0·27), especially if they had an IFNγ signature lower than the median (HR 0·88 [95% CI 0·40-1·93], p=0·74). INTERPRETATION: Tumour mutational burden alone or in combination with IFNγ gene expression signature or other markers for an adaptive immune response might be of relevance for identifying patients with stage III melanoma who might derive clinical benefit from targeted therapy. Further validation in prospective clinical trials is warranted. FUNDING: Novartis Pharmaceuticals.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Mutation , Neoplasm Recurrence, Local/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Double-Blind Method , Female , Follow-Up Studies , Humans , Imidazoles/administration & dosage , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Oximes/administration & dosage , Prognosis , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Salvage Therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Survival Rate , Young Adult
3.
Breast Cancer Res ; 19(1): 18, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183331

ABSTRACT

BACKGROUND: Overexpression of fibroblast growth factor receptor 1 (FGFR1), found in ≤8% of hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer cases, is correlated with decreased overall survival and resistance to endocrine therapy (ET). Dovitinib, a potent FGFR inhibitor, has demonstrated antitumor activity in heavily pretreated patients with FGFR pathway-amplified breast cancer. METHODS: In this randomized, placebo-controlled phase II trial, we evaluated whether the addition of dovitinib to fulvestrant would improve outcomes in postmenopausal patients with HR+, HER2- advanced breast cancer that had progressed during or after prior ET. Patients were stratified by FGF pathway amplification and presence of visceral disease, and they were randomized 1:1 to receive fulvestrant plus dovitinib or placebo. The primary endpoint was progression-free survival (PFS). RESULTS: From 15 May 2012 to 26 November 2014, 97 patients from 36 centers were enrolled. The frequency of FGF pathway amplification was lower than anticipated, and the study was terminated early owing to slow accrual of patients with FGF pathway amplification. The median PFS (95% CI) was 5.5 (3.8-14.0) months vs 5.5 (3.5-10.7) months in the dovitinib vs placebo arms, respectively (HR, 0.68; did not meet predefined efficacy criteria). For the FGF pathway-amplified subgroup (n = 31), the median PFS (95% CI) was 10.9 (3.5-16.5) months vs 5.5 (3.5-16.4) months in the dovitinib vs placebo arms, respectively (HR, 0.64; met the predefined superiority criteria). Frequently reported adverse events in the dovitinib (diarrhea, nausea, vomiting, asthenia, and headache) and placebo (diarrhea, fatigue, nausea, and asthenia) arms were mostly low grade. CONCLUSIONS: The safety profile of dovitinib plus fulvestrant was consistent with the known safety profile of single-agent dovitinib. Dovitinib in combination with fulvestrant showed promising clinical activity in the FGF pathway-amplified subgroup. However, the data reported herein should be interpreted with caution, given that fewer PFS events occurred in the FGF pathway-amplified patients than was expected and that an effect of dovitinib regardless of FGR pathway amplification status cannot be excluded, because the population was smaller than expected. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01528345 . Registered 31 January 2012.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles/administration & dosage , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease Progression , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Fulvestrant , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Postmenopause , Quinolones/administration & dosage , Retreatment , Survival Analysis , Treatment Outcome
4.
Blood ; 123(14): 2157-60, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24458439

ABSTRACT

The JAK1/JAK2 inhibitor ruxolitinib produced significant reductions in splenomegaly and symptomatic burden and improved survival in patients with myelofibrosis (MF), irrespective of their JAK2 mutation status, in 2 phase III studies against placebo (COMFORT-I) and best available therapy (COMFORT-II). We performed a comprehensive mutation analysis to evaluate the impact of 14 MF-associated mutations on clinical outcomes in 166 patients included in COMFORT-II. We found that responses in splenomegaly and symptoms, as well as the risk of developing ruxolitinib-associated anemia and thrombocytopenia, occurred at similar frequencies across different mutation profiles. Ruxolitinib improved survival independent of mutation profile and reduced the risk of death in patients harboring a set of prognostically detrimental mutations (ASXL1, EZH2, SRSF2, IDH1/2) with an hazard ratio of 0.57 (95% confidence interval: 0.30-1.08) vs best available therapy. These data indicate that clinical efficacy and survival improvement may occur across different molecular subsets of patients with MF treated with ruxolitinib.


Subject(s)
Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , DNA Mutational Analysis , Enhancer of Zeste Homolog 2 Protein , Humans , Isocitrate Dehydrogenase/genetics , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Mutation , Nitriles , Nuclear Proteins/genetics , Polycomb Repressive Complex 2/genetics , Primary Myelofibrosis/mortality , Prognosis , Pyrimidines , Repressor Proteins/genetics , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors , Treatment Outcome
5.
Lancet Oncol ; 15(3): 286-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24556040

ABSTRACT

BACKGROUND: An unmet medical need exists for patients with metastatic renal cell carcinoma who have progressed on VEGF-targeted and mTOR-inhibitor therapies. Fibroblast growth factor (FGF) pathway activation has been proposed as a mechanism of escape from VEGF-targeted therapies. Dovitinib is an oral tyrosine-kinase inhibitor that inhibits VEGF and FGF receptors. We therefore compared dovitinib with sorafenib as third-line targeted therapies in patients with metastatic renal cell carcinoma. METHODS: In this multicentre phase 3 study, patients with clear cell metastatic renal cell carcinoma who received one previous VEGF-targeted therapy and one previous mTOR inhibitor were randomly assigned through an interactive voice and web response system to receive open-label dovitinib (500 mg orally according to a 5-days-on and 2-days-off schedule) or sorafenib (400 mg orally twice daily) in a 1:1 ratio. Randomisation was stratified by risk group and region. The primary endpoint was progression-free survival (PFS) assessed by masked central review. Efficacy was assessed in all patients who were randomly assigned and safety was assessed in patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01223027. FINDINGS: 284 patients were randomly assigned to the dovitinib group and 286 to the sorafenib group. Median follow-up was 11·3 months (IQR 7·9-14·6). Median PFS was 3·7 months (95% CI 3·5-3·9) in the dovitinib group and 3·6 months (3·5-3·7) in the sorafenib group (hazard ratio 0·86, 95% CI 0·72-1·04; one-sided p=0·063). 280 patients in the dovitinib group and 284 in the sorafenib group received at least one dose of study drug. Common grade 3 or 4 adverse events included hypertriglyceridaemia (38 [14%]), fatigue (28 [10%]), hypertension (22 [8%]), and diarrhoea (20 [7%]) in the dovitinib group, and hypertension (47 [17%]), fatigue (24 [8%]), dyspnoea (21 [7%]), and palmar-plantar erythrodysaesthesia (18 [6%]) in the sorafenib group. The most common serious adverse event was dyspnoea (16 [6%] and 15 [5%] in the dovitinib and sorafenib groups, respectively). INTERPRETATION: Dovitinib showed activity, but this was no better than that of sorafenib in patients with renal cell carcinoma who had progressed on previous VEGF-targeted therapies and mTOR inhibitors. This trial provides reference outcome data for future studies of targeted inhibitors in the third-line setting. FUNDING: Novartis Pharmaceuticals Corporation.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Quinolones/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Benzimidazoles/adverse effects , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Female , Humans , Kidney Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Quinolones/adverse effects , Sorafenib , Vascular Endothelial Growth Factor A/antagonists & inhibitors
6.
Cancer Immunol Res ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990554

ABSTRACT

Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Here we report clinical and translational results from a phase Ib trial testing whether IL-1ß blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD-1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n=10) were treated with canakinumab, a high-affinity monoclonal human anti-interleukin-1ß (IL-1ß), the PD-1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSCs) by flow cytometry for patients in the trial, but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD-1 and IL-1ß blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.

7.
Comput Methods Programs Biomed ; 242: 107771, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37717523

ABSTRACT

Repetitive Transcranial Magnetic Stimulation (rTMS) is an evidence-based treatment for depression. However, the patterns of response to this treatment modality are inconsistent. Whilst many people see a significant reduction in the severity of their depression following rTMS treatment, some patients do not. To support and improve patient outcomes, recent work is exploring the possibility of using Machine Learning to predict rTMS treatment outcomes. Our proposed model is the first to combine functional magnetic resonance imaging (fMRI) connectivity with deep learning techniques to predict treatment outcomes before treatment starts. Furthermore, with the use of Explainable AI (XAI) techniques, we identify potential biomarkers that may discriminate between rTMS responders and non-responders. Our experiments utilize 200 runs of repeated bootstrap sampling on two rTMS datasets. We compare performances between our proposed feedforward deep neural network against existing methods, and compare the average accuracy, balanced accuracy and F1-score on a held-out test set. The results of these experiments show that our model outperforms existing methods with an average accuracy of 0.9423, balanced accuracy of 0.9423, and F1-score of 0.9461 in a sample of 61 patients. We found that functional connectivity measures between the Subgenual Anterior Cingulate Cortex and Centeral Opercular Cortex are a key determinant of rTMS treatment response. This knowledge provides psychiatrists with further information to explore the potential mechanisms of responses to rTMS treatment. Our developed prototype is ready to be deployed across large datasets in multiple centres and different countries.


Subject(s)
Depression , Transcranial Magnetic Stimulation , Humans , Transcranial Magnetic Stimulation/methods , Depression/therapy , Prefrontal Cortex , Magnetic Resonance Imaging/methods , Biomarkers
8.
Brain Inform ; 10(1): 10, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37093301

ABSTRACT

Informatics paradigms for brain and mental health research have seen significant advances in recent years. These developments can largely be attributed to the emergence of new technologies such as machine learning, deep learning, and artificial intelligence. Data-driven methods have the potential to support mental health care by providing more precise and personalised approaches to detection, diagnosis, and treatment of depression. In particular, precision psychiatry is an emerging field that utilises advanced computational techniques to achieve a more individualised approach to mental health care. This survey provides an overview of the ways in which artificial intelligence is currently being used to support precision psychiatry. Advanced algorithms are being used to support all phases of the treatment cycle. These systems have the potential to identify individuals suffering from mental health conditions, allowing them to receive the care they need and tailor treatments to individual patients who are mostly to benefit. Additionally, unsupervised learning techniques are breaking down existing discrete diagnostic categories and highlighting the vast disease heterogeneity observed within depression diagnoses. Artificial intelligence also provides the opportunity to shift towards evidence-based treatment prescription, moving away from existing methods based on group averages. However, our analysis suggests there are several limitations currently inhibiting the progress of data-driven paradigms in care. Significantly, none of the surveyed articles demonstrate empirically improved patient outcomes over existing methods. Furthermore, greater consideration needs to be given to uncertainty quantification, model validation, constructing interdisciplinary teams of researchers, improved access to diverse data and standardised definitions within the field. Empirical validation of computer algorithms via randomised control trials which demonstrate measurable improvement to patient outcomes are the next step in progressing models to clinical implementation.

9.
Int J Cancer ; 130(12): 2997-3005, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-21796626

ABSTRACT

Aurora kinases are oncogenic serine/threonine kinases that play key roles in regulating the mitotic phase of the eukaryotic cell cycle. Auroras are overexpressed in numerous tumors including B-cell non-Hodgkin's lymphomas and are validated oncology targets. AT9283, a pan-aurora inhibitor inhibited growth and survival of multiple solid tumors in vitro and in vivo. In this study, we demonstrated that AT9283 had potent activity against Aurora B in a variety of aggressive B-(non-Hodgkin lymphoma) B-NHL cell lines. Cells treated with AT9283 exhibited endoreduplication confirming the mechanism of action of an Aurora B inhibitor. Also, treatment of B-NHL cell lines with AT9283 induced apoptosis in a dose and time dependent manner and inhibited cell proliferation with an IC(50) < 1 µM. It is well known that inhibition of auroras (A or B) synergistically enhances the effects of microtubule targeting agents such as taxanes and vinca alkaloids to induce antiproliferation and apoptosis. We evaluated whether AT9283 in combination with docetaxel is more efficient in inducing apoptosis than AT9283 or docetaxel alone. At very low doses (5 nM) apoptosis was doubled in the combination (23%) compared to AT9283 or docetaxel alone (10%). A mouse xenograft model of mantle cell lymphoma demonstrated that AT9283 at 15 mg/kg and docetaxel (10 mg/kg) alone had modest anti-tumor activity. However, AT9283 at 20 mg/kg and AT9283 (15 or 20 mg/kg) plus docetaxel (10 mg/kg) demonstrated a statistically significant tumor growth inhibition and enhanced survival. Together, our results suggest that AT9283 plus docetaxel may represent a novel therapeutic strategy in B-cell NHL and warrant early phase clinical trial evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Lymphoma, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Taxoids/pharmacology , Urea/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Humans , Lymphoma, B-Cell/pathology , Lymphoma, Mantle-Cell/drug therapy , Mice , Mice, SCID , Urea/pharmacology , Xenograft Model Antitumor Assays
10.
Blood ; 116(12): 2089-95, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20548094

ABSTRACT

Despite promising clinical results from imatinib mesylate and second-generation ABL tyrosine kinase inhibitors (TKIs) for most BCR-ABL(+) leukemia, BCR-ABL harboring the mutation of threonine 315 to isoleucine (BCR-ABL/T315I) is not targeted by any of these agents. We describe the in vitro and in vivo effects of AT9283 (1-cyclopropyl-3[5-morpholin-4yl methyl-1H-benzomidazol-2-yl]-urea), a potent inhibitor of several protein kinases, including Aurora A, Aurora B, Janus kinase 2 (JAK2), JAK3, and ABL on diverse imatinib-resistant BCR-ABL(+) cells. AT9283 showed potent antiproliferative activity on cells transformed by wild-type BCR-ABL and BCR-ABL/T315I. AT9283 inhibited proliferation in a panel of BaF3 and human BCR-ABL(+) cell lines both sensitive and resistant to imatinib because of a variety of mechanisms. In BCR-ABL(+) cells, we confirmed inhibition of substrates of both BCR-ABL (signal transducer and activator of transcription-5) and Aurora B (histone H3) at physiologically achievable concentrations. The in vivo effects of AT9283 were examined in several mouse models engrafted either subcutaneously or intravenously with BaF3/BCR-ABL, human BCR-ABL(+) cell lines, or primary patient samples expressing BCR-ABL/T315I or glutamic acid 255 to lysine, another imatinib-resistant mutation. These data together support further clinical investigation of AT9283 in patients with imatinib- and second-generation ABL TKI-resistant BCR-ABL(+) cells, including T315I.


Subject(s)
Benzimidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Urea/analogs & derivatives , Animals , Antineoplastic Agents , Benzamides , Benzimidazoles/therapeutic use , Cell Proliferation/drug effects , Drug Delivery Systems , Drug Resistance, Neoplasm , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Urea/pharmacology , Urea/therapeutic use
11.
Biomedicines ; 10(5)2022 May 13.
Article in English | MEDLINE | ID: mdl-35625872

ABSTRACT

Advanced gastrointestinal stromal tumors (GIST) are typically treated with tyrosine kinase inhibitors, and imatinib is the most commonly used standard of care in first line treatments. The use of this and other tyrosine kinase inhibitors is associated with objective tumor responses and prolongation of progression-free and overall survival, but the treatment of metastatic disease is non-curative due to the selection or acquisition of secondary mutations and the activation of alternative kinase signaling pathways, leading to resistance and disease progression after an initial response. The present preclinical study evaluated the potential use of the fibroblast growth factor receptor inhibitors infigratinib and dovitinib alone or in combination with the mitogen-activated protein kinase inhibitor binimetinib in mouse models of GIST with different sensitivity or resistance to imatinib. Patient- and cell-line-derived GIST xenografts were established by bilateral, subcutaneous transplantation of human GIST tissue in female adult nu/nu NMRI mice. The mice were treated with dovitinib, infigratinib, or binimetinib, either alone or in combination with imatinib. The safety of treated animals was assessed by well-being inspection and body weight measurement. Antitumor effects were assessed by caliper-based tumor measurement. H&E staining and immunohistochemistry were used for assessing anti-mitotic and pro-apoptotic activity of the experimental treatments. Western blotting was used for assessing effects of the agents on kinase signaling pathways. Anti-angiogenic activity was assessed by measuring tumor vessel density. Dovitinib was found to have antitumor efficacy in GIST xenografts characterized by different imatinib resistance patterns. Dovitinib had better efficacy than imatinib (both at standard and increased dose) and was found to be well tolerated. Dovitinib had better efficacy in a KIT exon 9 mutant model, highlighting a role of patient selection in clinical GIST trials with the agent. In a model with KIT exon 11 and 17 mutations, dovitinib induced tumor necrosis, most likely due to anti-angiogenic effects. Additive effects combining dovitinib with binimetinib were limited.

13.
Br J Haematol ; 150(1): 46-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20507304

ABSTRACT

Constitutive activation of Janus kinase (Jak) 2 is the most prevalent pathogenic event observed in the myeloproliferative disorders (MPD), suggesting that inhibitors of Jak2 may prove valuable in their management. Inhibition of the Aurora kinases has also proven to be an effective therapeutic strategy in a number of haematological malignancies. AT9283 is a multi-targeted kinase inhibitor with potent activity against Jak2 and Aurora kinases A and B, and is currently being evaluated in clinical trials. To investigate the therapeutic potential of AT9283 in the MPD we studied its activity in a number of Jak2-dependent systems. AT9283 potently inhibited proliferation and Jak2-related signalling in Jak2-dependent cell lines as well as inhibiting the formation of erythroid colonies from haematopoietic progenitors isolated from MPD patients with Jak2 mutations. The compound also demonstrated significant therapeutic potential in vivo in an ETV6-JAK2 (TEL-JAK2) murine leukaemia model. Inhibition of both Jak2 and Aurora B was observed in the model systems used, indicating a dual mechanism of action. Our results suggest that AT9283 may be a valuable therapy in patients with MPD and that the dual inhibition of Jak2 and the Aurora kinases may potentially offer combinatorial efficacy in the treatment of these diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Janus Kinase 2/antagonists & inhibitors , Myeloproliferative Disorders/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Urea/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Aurora Kinase B , Aurora Kinases , Benzimidazoles/therapeutic use , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Erythroid Precursor Cells/drug effects , Humans , Janus Kinase 2/genetics , Janus Kinase 2/physiology , Leukemia, Experimental/drug therapy , Leukemia, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Signal Transduction/drug effects , Tumor Cells, Cultured , Urea/pharmacology , Urea/therapeutic use , Xenograft Model Antitumor Assays
14.
Mol Cancer Ther ; 8(2): 324-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19174555

ABSTRACT

Cyclin-dependent kinases (CDK), and their regulatory cyclin partners, play a central role in eukaryotic cell growth, division, and death. This key role in cell cycle progression, as well as their deregulation in several human cancers, makes them attractive therapeutic targets in oncology. A series of CDK inhibitors was developed using Astex's fragment-based medicinal chemistry approach, linked to high-throughput X-ray crystallography. A compound from this series, designated AT7519, is currently in early-phase clinical development. We describe here the biological characterization of AT7519, a potent inhibitor of several CDK family members. AT7519 showed potent antiproliferative activity (40-940 nmol/L) in a panel of human tumor cell lines, and the mechanism of action was shown here to be consistent with the inhibition of CDK1 and CDK2 in solid tumor cell lines. AT7519 caused cell cycle arrest followed by apoptosis in human tumor cells and inhibited tumor growth in human tumor xenograft models. Tumor regression was observed following twice daily dosing of AT7519 in the HCT116 and HT29 colon cancer xenograft models. We show that these biological effects are linked to inhibition of CDKs in vivo and that AT7519 induces tumor cell apoptosis in these xenograft models. AT7519 has an attractive biological profile for development as a clinical candidate, and the tolerability and efficacy in animal models compare favorably with other CDK inhibitors in clinical development. Studies described here formed the biological rationale for investigating the potential therapeutic benefit of AT7519 in cancer patients.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Piperidines/pharmacology , Pyrazoles/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Nude , Piperidines/blood , Piperidines/chemistry , Piperidines/pharmacokinetics , Pyrazoles/blood , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/pharmacology , Time Factors , Xenograft Model Antitumor Assays
15.
Rev Sci Instrum ; 91(3): 035108, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-32259989

ABSTRACT

We report the fabrication, characterization, and use of rubidium vapor dispensers based on highly oriented pyrolytic graphite (HOPG) intercalated with metallic rubidium. Compared to commercial chromate salt dispensers, these intercalated HOPG (IHOPG) dispensers hold an order of magnitude more rubidium in a similar volume, require less than one-fourth the heating power, and emit less than one-half as many impurities. Appropriate processing permits exposure of the IHOPG to atmosphere for over ninety minutes without any adverse effects. Intercalation of cesium, potassium, and lithium into HOPG has also been demonstrated in the literature, which suggests that IHOPG dispensers may also be made for those metals.

16.
Cell Signal ; 20(5): 836-43, 2008 May.
Article in English | MEDLINE | ID: mdl-18280112

ABSTRACT

Extracellular signal-regulated kinase-1 and -2 (ERK1/2) are activated by dual threonine and tyrosine phosphorylation of a TEY motif. The highly related kinase ERK5 is also activated by phosphorylation at a TEY motif. Inactivation of ERK1/2 is achieved by distinct members of the dual-specificity protein phosphatase (DUSP) family, which are responsible for the specific, regulated de-phosphorylation of the TEY motif. These include both nuclear (DUSP5) and cytoplasmic (DUSP6) enzymes. DUSP6, a candidate tumour suppressor gene, is thought to be highly specific for inactivation of ERK1/2 but several reports have suggested that it may also inactivate ERK5. Here we have compared the ability of DUSP6 to regulate the ERK1/2 and ERK5 protein kinases. We find that DUSP6 binds to ERK1/2 in both yeast and human cells but fails to bind to ERK5. Recombinant ERK2 can induce catalytic activation of DUSP6 whereas ERK5 cannot. Ectopic expression of DUSP6 can de-phosphorylate a co-expressed ERK2 construct but does not de-phosphorylate ERK5. Finally, expression of DUSP6 blocks the MEK1-driven activation of GAL4-ELK1, an ERK1/2-regulated transcription factor, but fails to block the MEK5-driven activation of GAL4-MEF2D, an ERK5-regulated transcription factor. These results demonstrate that even upon over-expression DUSP6 fails to inactivate ERK5, confirming that it is indeed an ERK1/2-specific DUSP.


Subject(s)
Dual Specificity Phosphatase 6/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Cell Line , Dual Specificity Phosphatase 6/genetics , Humans , MADS Domain Proteins/metabolism , MEF2 Transcription Factors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Myogenic Regulatory Factors/metabolism , Phosphorylation , Protein Binding , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Two-Hybrid System Techniques , ets-Domain Protein Elk-1/metabolism
17.
J Med Imaging Radiat Sci ; 49(2): 145-152, 2018 Jun.
Article in English | MEDLINE | ID: mdl-32074032

ABSTRACT

INTRODUCTION: Radiation oncology is a vital tool in resisting the cancer epidemic affecting millions worldwide. This study queried the perspectives of radiation oncology professionals and sought their solutions in regard to current practice, future practice, outsourcing, and common values. METHODS: A mass consumer survey was distributed globally that collected quantitative and qualitative data from 245 radiation oncology professionals based in 47 countries. RESULTS: Participants scored the sector highly on the quality of current practice. The United States was identified as the perceived global leader in the practice of radiation oncology. The sector was considered moderately open to reform with suggestions of better training, greater resources, and incorporation of data informatics. A preference for paradigms involving private enterprise emerged. Appropriate outsourcing tasks and companies were identified and industry leaders evaluated. Remarkable accord was observed in values and priorities across professional groups and examined subsets. CONCLUSIONS: There is an opportunity to realize value through the application of successful global paradigms. Our research generated a unique performance evaluation of the sector by identifying the current situation and areas open to reform. The required shift in the role of government from provider to regulator implied a mandate for a reorientation of policy settings with greater emphasis on free enterprise solutions that maximize value and ultimately advance patient care.

18.
J Med Phys ; 42(3): 163-170, 2017.
Article in English | MEDLINE | ID: mdl-28974863

ABSTRACT

Dedicated rapid access palliative radiation therapy improves patients' access to care, allowing more timely treatment which would positively impact on quality of life. The TomoTherapy (Accuray, Sunnyvale, CA) system provides megavoltage (MV) fan-beam computed tomography (FBCT) as the image guidance technique, and a module called "statRT" that allows the use of these MV FBCT images for direct planning. The possibility of using this imaging modality for palliative radiotherapy treatment planning is assessed against accepted planning CT standards by performing tests following AAPM TG 66 and an end-to-end measurement. Results have shown that MV FBCT images acquired by TomoTherapy are of sufficient quality for the purpose of target delineation and dose calculation for palliative treatments. Large image noise and extended scan acquisition time are the two main drawbacks, so this imaging modality should only be used for palliative treatments at areas with well-known, easily distinguishable, and relatively immobile targets such as spine and whole brain.

19.
J Med Radiat Sci ; 64(4): 281-289, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28580762

ABSTRACT

INTRODUCTION: TomoTherapy (Accuray, Sunnyvale, CA) has recently introduced a static form of tomotherapy: TomoDirect™ (TD). This study aimed to evaluate TD against a contemporary intensity modulated radiation therapy (IMRT) alternative through comparison of target and organ at risk (OAR) doses in breast cancer cases. A secondary objective was to evaluate planning efficiency by measuring optimisation times. METHODS: Treatment plans of 27 whole-breast radiation therapy (WBRT) patients optimised with a tangential hybrid IMRT technique were replanned using TD. Parameters included a dynamic field width of 2.5 cm, a pitch of 0.251 and a modulation factor of 2.000; 50 Gy in 25 fractions was prescribed and planning time recorded. The planning metrics used in analysis were ICRU based, with the mean PTV minimum (D99 ) used as the point of comparison. RESULTS: Both modalities met ICRU50 target heterogeneity objectives (TD D99 = 48.0 Gy vs. IMRT = 48.1 Gy, P = 0.26; TD D1 = 53.5 Gy vs. IMRT = 53.0 Gy, P = 0.02; Homogeneity index TD = 0.11 vs. IMRT = 0.10, P = 0.03), with TD plans generating higher median doses (TD D50 = 51.1 Gy vs. IMRT = 50.9 Gy, P = 0.03). No significant difference was found in prescription dose coverage (TD V50 = 85.5% vs. IMRT = 82.0%, P = 0.09). TD plans produced a statistically significant reduction in V5 ipsilateral lung doses (TD V5 = 23.2% vs. IMRT = 27.2%, P = 0.04), while other queried OARs remained comparable (TD ipsilateral lung V20 = 13.2% vs. IMRT = 14.6%, P = 0.30; TD heart V5 = 2.7% vs. IMRT = 2.8%, P = 0.47; TD heart V10 = 1.7% vs. IMRT = 1.8%, P = 0.44). TD reduced planning time considerably (TD = 9.8 m vs. IMRT = 27.6 m, P < 0.01), saving an average planning time of 17.8 min per patient. CONCLUSIONS: TD represents a suitable WBRT treatment approach both in terms of plan quality metrics and planning efficiency.


Subject(s)
Breast Neoplasms/radiotherapy , Radiotherapy, Intensity-Modulated/methods , Female , Humans , Organs at Risk/radiation effects , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted/methods , Radiotherapy Planning, Computer-Assisted/standards , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy, Intensity-Modulated/standards , Random Allocation
20.
Mol Cancer Ther ; 16(6): 1010-1020, 2017 06.
Article in English | MEDLINE | ID: mdl-28341788

ABSTRACT

Fibroblast growth factor (FGF) signaling plays critical roles in key biological processes ranging from embryogenesis to wound healing and has strong links to several hallmarks of cancer. Genetic alterations in FGF receptor (FGFR) family members are associated with increased tumor growth, metastasis, angiogenesis, and decreased survival. JNJ-42756493, erdafitinib, is an orally active small molecule with potent tyrosine kinase inhibitory activity against all four FGFR family members and selectivity versus other highly related kinases. JNJ-42756493 shows rapid uptake into the lysosomal compartment of cells in culture, which is associated with prolonged inhibition of FGFR signaling, possibly due to sustained release of the inhibitor. In xenografts from human tumor cell lines or patient-derived tumor tissue with activating FGFR alterations, JNJ-42756493 administration results in potent and dose-dependent antitumor activity accompanied by pharmacodynamic modulation of phospho-FGFR and phospho-ERK in tumors. The results of the current study provide a strong rationale for the clinical investigation of JNJ-42756493 in patients with tumors harboring FGFR pathway alterations. Mol Cancer Ther; 16(6); 1010-20. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Lysosomes/metabolism , Male , Mice , Molecular Targeted Therapy , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Quinoxalines/administration & dosage , Quinoxalines/pharmacokinetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL