Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Commun ; 10(1): 2030, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31048689

ABSTRACT

Acquired resistance to MEK1/2 inhibitors (MEKi) arises through amplification of BRAFV600E or KRASG13D to reinstate ERK1/2 signalling. Here we show that BRAFV600E amplification and MEKi resistance are reversible following drug withdrawal. Cells with BRAFV600E amplification are addicted to MEKi to maintain a precise level of ERK1/2 signalling that is optimal for cell proliferation and survival, and tumour growth in vivo. Robust ERK1/2 activation following MEKi withdrawal drives a p57KIP2-dependent G1 cell cycle arrest and senescence or expression of NOXA and cell death, selecting against those cells with amplified BRAFV600E. p57KIP2 expression is required for loss of BRAFV600E amplification and reversal of MEKi resistance. Thus, BRAFV600E amplification confers a selective disadvantage during drug withdrawal, validating intermittent dosing to forestall resistance. In contrast, resistance driven by KRASG13D amplification is not reversible; rather ERK1/2 hyperactivation drives ZEB1-dependent epithelial-to-mesenchymal transition and chemoresistance, arguing strongly against the use of drug holidays in cases of KRASG13D amplification.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Amplification/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Withholding Treatment , Zinc Finger E-box-Binding Homeobox 1/metabolism
2.
Mol Cancer Ther ; 18(5): 909-919, 2019 05.
Article in English | MEDLINE | ID: mdl-30872381

ABSTRACT

Barasertib (AZD1152), a pro-drug of the highly potent and selective Aurora B kinase inhibitor AZD2811, showed promising clinical activity in relapsed/refractory diffuse large B-cell lymphoma (DLBCL) patients administered as a 4-day infusion. To improve potential therapeutic benefit of Aurora B kinase inhibition, a nanoparticle formulation of AZD2811 has been developed to address limitations of repeated intravenous infusion. One of the challenges with the use of nanoparticles for chronic treatment of tumors is optimizing dose and schedule required to enable repeat administration to sustain tumor growth inhibition. AZD2811 gives potent cell growth inhibition across a range of DLBCL cells lines in vitro In vivo, repeat administration of the AZD2811 nanoparticle gave antitumor activity at half the dose intensity of AZD1152. Compared with AZD1152, a single dose of AZD2811 nanoparticle gave less reduction in pHH3, but increased apoptosis and reduction of cells in G1 and G2-M, albeit at later time points, suggesting that duration and depth of target inhibition influence the nature of the tumor cell response to drug. Further exploration of the influence of dose and schedule on efficacy revealed that AZD2811 nanoparticle can be used flexibly with repeat administration of 25 mg/kg administered up to 7 days apart being sufficient to maintain equivalent tumor control. Timing of repeat administration could be varied with 50 mg/kg every 2 weeks controlling tumor control as effectively as 25 mg/kg every week. AZD2811 nanoparticle can be administered with very different doses and schedules to inhibit DLBCL tumor growth, although maximal tumor growth inhibition was achieved with the highest dose intensities.


Subject(s)
Acetanilides/pharmacology , Aurora Kinase B/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Acetanilides/chemistry , Animals , Aurora Kinase B/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Nanoparticles/chemistry , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry , Xenograft Model Antitumor Assays
3.
Cancer Discov ; 9(2): 230-247, 2019 02.
Article in English | MEDLINE | ID: mdl-30373918

ABSTRACT

Small cell lung cancer (SCLC) accounts for 15% of lung cancers and is almost always linked to inactivating RB1 and TP53 mutations. SCLC frequently responds, albeit briefly, to chemotherapy. The canonical function of the RB1 gene product RB1 is to repress the E2F transcription factor family. RB1 also plays both E2F-dependent and E2F-independent mitotic roles. We performed a synthetic lethal CRISPR/Cas9 screen in an RB1 -/- SCLC cell line that conditionally expresses RB1 to identify dependencies that are caused by RB1 loss and discovered that RB1 -/- SCLC cell lines are hyperdependent on multiple proteins linked to chromosomal segregation, including Aurora B kinase. Moreover, we show that an Aurora B kinase inhibitor is efficacious in multiple preclinical SCLC models at concentrations that are well tolerated in mice. These results suggest that RB1 loss is a predictive biomarker for sensitivity to Aurora B kinase inhibitors in SCLC and perhaps other RB1 -/- cancers. SIGNIFICANCE: SCLC is rarely associated with actionable protooncogene mutations. We did a CRISPR/Cas9-based screen that showed that RB1 -/- SCLC are hyperdependent on AURKB, likely because both genes control mitotic fidelity, and confirmed that Aurora B kinase inhibitors are efficacious against RB1 -/- SCLC tumors in mice at nontoxic doses.See related commentary by Dick and Li, p. 169.This article is highlighted in the In This Issue feature, p. 151.


Subject(s)
Aurora Kinase B/metabolism , Cell Proliferation , Genes, Tumor Suppressor , Lung Neoplasms/pathology , Mutation , Retinoblastoma Binding Proteins/metabolism , Small Cell Lung Carcinoma/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Aurora Kinase B/genetics , CRISPR-Cas Systems , Chromosome Segregation , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Retinoblastoma Binding Proteins/antagonists & inhibitors , Retinoblastoma Binding Proteins/genetics , Signal Transduction , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 16(6): 1031-1040, 2017 06.
Article in English | MEDLINE | ID: mdl-28292940

ABSTRACT

Barasertib (AZD1152), a highly potent and selective aurora kinase B inhibitor, gave promising clinical activity in elderly acute myeloid leukemia (AML) patients. However, clinical utility was limited by the requirement for a 7-day infusion. Here we assessed the potential of a nanoparticle formulation of the selective Aurora kinase B inhibitor AZD2811 (formerly known as AZD1152-hQPA) in preclinical models of AML. When administered to HL-60 tumor xenografts at a single dose between 25 and 98.7 mg/kg, AZD2811 nanoparticle treatment delivered profound inhibition of tumor growth, exceeding the activity of AZD1152. The improved antitumor activity was associated with increased phospho-histone H3 inhibition, polyploidy, and tumor cell apoptosis. Moreover, AZD2811 nanoparticles increased antitumor activity when combined with cytosine arabinoside. By modifying dose of AZD2811 nanoparticle, therapeutic benefit in a range of preclinical models was further optimized. At high-dose, antitumor activity was seen in a range of models including the MOLM-13 disseminated model. At these higher doses, a transient reduction in bone marrow cellularity was observed demonstrating the potential for the formulation to target residual disease in the bone marrow, a key consideration when treating AML. Collectively, these data establish that AZD2811 nanoparticles have activity in preclinical models of AML. Targeting Aurora B kinase with AZD2811 nanoparticles is a novel approach to deliver a cell-cycle inhibitor in AML, and have potential to improve on the clinical activity seen with cell-cycle agents in this disease. Mol Cancer Ther; 16(6); 1031-40. ©2017 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Aurora Kinase B/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Nanoparticles , Organophosphates/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Cytarabine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Mice , Organophosphates/pharmacokinetics , Polyploidy , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Rats , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Oncotarget ; 7(52): 86313-86325, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27861144

ABSTRACT

Metabolic plasticity is an emerging hallmark of cancer, and increased glycolysis is often observed in transformed cells. Small molecule inhibitors that target driver oncogenes can potentially inhibit the glycolytic pathway. Osimertinib (AZD9291) is a novel EGFR tyrosine kinase inhibitor (TKI) that is potent and selective for sensitising (EGFRm) and T790M resistance mutations. Clinical studies have shown osimertinib to be efficacious in patients with EGFRm/ T790M advanced NSCLC who have progressed after EGFR-TKI treatment. However experience with targeted therapies suggests that acquired resistance may emerge. Thus there is a need to characterize resistance mechanisms and to devise ways to prevent, delay or overcome osimertinib resistance. We show here that osimertinib suppresses glycolysis in parental EGFR-mutant lung adenocarcinoma lines, but has not in osimertinib-resistant cell lines. Critically, we show osimertinib treatment induces a strict dependence on mitochondrial oxidative phosphorylation (OxPhos), as OxPhos inhibitors significantly delay the long-term development of osimertinib resistance in osimertinib-sensitive lines. Accordingly, growth conditions which promote a less glycolytic phenotype confer a degree of osimertinib resistance. Our data support a model in which the combination of osimertinib and OxPhos inhibitors can delay or prevent resistance in osimertinib-naïve tumour cells, and represents a novel strategy that warrants further pre-clinical investigation.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Oxidative Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Acrylamides , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aniline Compounds , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Glycolysis/drug effects , Humans , Lung Neoplasms/pathology
6.
Mol Cancer Ther ; 15(10): 2378-2387, 2016 10.
Article in English | MEDLINE | ID: mdl-27439477

ABSTRACT

Osimertinib (AZD9291) is a potent, selective, irreversible inhibitor of EGFR-sensitizing (exon 19 and L858R) and T790M-resistant mutation. In vivo, in the mouse, it is metabolized to an active des-methyl metabolite, AZ5104. To understand the therapeutic potential in patients, this study aimed to assess the relationship between osimertinib pharmacokinetics, the pharmacokinetics of the active metabolite, the pharmacodynamics of phosphorylated EGFR reduction, and efficacy in mouse xenograft models of EGFR-driven cancers, including two NSCLC lines. Osimertinib was dosed in xenografted models of EGFR-driven cancers. In one set of experiments, changes in phosphorylated EGFR were measured to confirm target engagement. In a second set of efficacy studies, the resulting changes in tumor volume over time after repeat dosing of osimertinib were observed. To account for the contributions of both molecules, a mathematical modeling approach was taken to integrate the resulting datasets. The model was able to describe the pharmacokinetics, pharmacodynamics, and efficacy in A431, PC9, and NCI-H1975 xenografts, with the differences in sensitivity described by the varying potency against wild-type, sensitizing, and T790M-mutant EGFR and the phosphorylated EGFR reduction required to reduce tumor volume. It was inferred that recovery of pEGFR is slower after chronic dosing due to reduced resynthesis. It was predicted and further demonstrated that although inhibition is irreversible, the resynthesis of EGFR is such that infrequent intermittent dosing is not as efficacious as once daily dosing. Mol Cancer Ther; 15(10); 2378-87. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacokinetics , ErbB Receptors/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Acrylamides , Algorithms , Aniline Compounds , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/chemistry , Humans , Mice , Models, Biological , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Xenograft Model Antitumor Assays
7.
ACS Med Chem Lett ; 7(5): 514-9, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190603

ABSTRACT

A novel series of covalent inhibitors of EGFR (epidermal growth factor receptor) kinase was discovered through a combination of subset screening and structure-based design. These compounds preferentially inhibit mutant forms of EGFR (activating mutant and T790M mutant) over wild-type EGFR in cellular assays measuring EGFR autophosphorylation and proliferation, suggesting an improved therapeutic index in non-small cell lung cancer patients would be achievable relative to established EGFR inhibitors. We describe our design approaches, resulting in the identification of the lead compound 5, and our efforts to develop an understanding of the structure-activity relationships within this series. In addition, strategies to overcome challenges around metabolic stability and aqueous solubility are discussed. Despite limitations in its physical properties, 5 is orally bioavailable in mice and demonstrates pronounced antitumor activity in in vivo models of mutant EGFR-driven cancers.

8.
J Control Release ; 229: 106-119, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27001894

ABSTRACT

The present studies were aimed at formulating AZD2811-loaded polylactic acid-polyethylene glycol (PLA-PEG) nanoparticles with adjustable release rates without altering the chemical structures of the polymer or active pharmaceutical ingredient (API). This was accomplished through the use of a hydrophobic ion pairing approach. A series of AZD2811-containing nanoparticles with a variety of hydrophobic counterions including oleic acid, 1-hydroxy-2-naphthoic acid, cholic acid, deoxycholic acid, dioctylsulfosuccinic acid, and pamoic acid is described. The hydrophobicity of AZD2811 was increased through formation of ion pairs with these hydrophobic counterions, producing nanoparticles with exceptionally high drug loading-up to five fold higher encapsulation efficiency and drug loading compared to nanoparticles made without hydrophobic ion pairs. Furthermore, the rate at which the drug was released from the nanoparticles could be controlled by employing counterions with various hydrophobicities and structures, resulting in release half-lives ranging from about 2 to 120h using the same polymer, nanoparticle size, and nanoemulsion process. Process recipe variables affecting drug load and release rate were identified, including pH and molarity of quench buffer. Ion pair formation between AZD2811 and pamoic acid as a model counterion was investigated using solubility enhancement as well as nuclear magnetic resonance spectroscopy to demonstrate solution-state interactions. Further evidence for an ion pairing mechanism of controlled release was provided through the measurement of API and counterion release profiles using high-performance liquid chromatography, which had stoichiometric relationships. Finally, Raman spectra of an AZD2811-pamoate salt compared well with those of the formulated nanoparticles, while single components (AZD2811, pamoic acid) alone did not. A library of AZD2811 batches was created for analytical and preclinical characterization. Dramatically improved preclinical efficacy and tolerability data were generated for the pamoic acid lead formulation, which has been selected for evaluation in a Phase 1 clinical trial (ClinicalTrials.gov Identifier NCT 02579226). This work clearly demonstrates the importance of assessing a wide range of drug release rates during formulation screening as a critical step for new drug product development, and how utilizing hydrophobic ion pairing enabled this promising nanoparticle formulation to proceed into clinical development.


Subject(s)
Acetanilides/administration & dosage , Antineoplastic Agents , Drug Delivery Systems , Nanoparticles , Organophosphates , Prodrugs , Quinazolines/administration & dosage , Acetanilides/chemistry , Acetanilides/pharmacokinetics , Acetanilides/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Line, Tumor , Cholic Acid/chemistry , Deoxycholic Acid/chemistry , Dioctyl Sulfosuccinic Acid/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Naphthols/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Organophosphates/administration & dosage , Organophosphates/chemistry , Organophosphates/pharmacokinetics , Organophosphates/therapeutic use , Polyethylene Glycols/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Rats, Nude , Rats, Wistar , Tumor Burden/drug effects
9.
Sci Transl Med ; 8(325): 325ra17, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26865565

ABSTRACT

Efforts to apply nanotechnology in cancer have focused almost exclusively on the delivery of cytotoxic drugs to improve therapeutic index. There has been little consideration of molecularly targeted agents, in particular kinase inhibitors, which can also present considerable therapeutic index limitations. We describe the development of Accurin polymeric nanoparticles that encapsulate the clinical candidate AZD2811, an Aurora B kinase inhibitor, using an ion pairing approach. Accurins increase biodistribution to tumor sites and provide extended release of encapsulated drug payloads. AZD2811 nanoparticles containing pharmaceutically acceptable organic acids as ion pairing agents displayed continuous drug release for more than 1 week in vitro and a corresponding extended pharmacodynamic reduction of tumor phosphorylated histone H3 levels in vivo for up to 96 hours after a single administration. A specific AZD2811 nanoparticle formulation profile showed accumulation and retention in tumors with minimal impact on bone marrow pathology, and resulted in lower toxicity and increased efficacy in multiple tumor models at half the dose intensity of AZD1152, a water-soluble prodrug of AZD2811. These studies demonstrate that AZD2811 can be formulated in nanoparticles using ion pairing agents to give improved efficacy and tolerability in preclinical models with less frequent dosing. Accurins specifically, and nanotechnology in general, can increase the therapeutic index of molecularly targeted agents, including kinase inhibitors targeting cell cycle and oncogenic signal transduction pathways, which have to date proved toxic in humans.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Nanoparticles/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Animals , Aurora Kinases/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Line, Tumor , Drug Liberation , Female , Humans , Male , Mass Spectrometry , Mice , Mice, SCID , Organophosphates/chemistry , Organophosphates/pharmacokinetics , Organophosphates/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Rats, Nude , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Dementia (London) ; 15(5): 958-75, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25187482

ABSTRACT

BACKGROUND: End of life decisions for people with advanced dementia are reported as often being difficult for families as they attempt to make appropriate and justified decisions. AIM: To explore the experiences of advance care planning amongst family caregivers of people with advanced dementia. DESIGN: Qualitative research including a series of single cases (close family relatives). METHODS: A purposive sample of 12 family caregivers within a specialist dementia unit was interviewed about their experiences of advance care planning between August 2009 and February 2010. RESULTS/FINDINGS: Family caregivers need encouragement to ask the right questions during advance care planning to discuss the appropriateness of nursing and medical interventions at the end of life. CONCLUSIONS: Advance care planning can be facilitated with the family caregiver in the context of everyday practice within the nursing home environment for older people with dementia.


Subject(s)
Advance Care Planning , Caregivers/psychology , Dementia/psychology , Terminal Care/psychology , Adult , Aged , Aged, 80 and over , Decision Making , Female , Humans , Male , Middle Aged , Qualitative Research
11.
Toxicol Pathol ; 44(1): 98-111, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26534939

ABSTRACT

The growth plate, ovary, adrenal gland, and rodent incisor tooth are sentinel organs for antiangiogenic effects since they respond reliably, quantitatively, and sensitively to inhibition of the vascular endothelial growth factor receptor (VEGFR). Here we report that treatment of rats with platelet-derived growth factor receptor beta (PDGFRß) inhibitors that target pericytes results in severe ovarian hemorrhage with degeneration and eventual rupture of the corpus luteum. Evaluation of the growth plate, adrenal gland, and incisor tooth that are typical target organs for antiangiogenic treatment in the rodent revealed no abnormalities. Histologically, the changes in the ovary were characterized by sinusoidal dilatation, increased vessel fragility, and hemorrhage into the corpus luteum. Immunocytochemical staining of vessels with alpha smooth muscle actin and CD31 that recognize pericytes and vascular endothelium, respectively, demonstrated that this effect was due to selective pericyte deficiency within corpora lutea. Further experiments in which rats were treated concurrently with both PDGFRß and VEGFR inhibitors ablated the hemorrhagic response, resulting instead in corpus luteum necrosis. These changes are consistent with the notion that selective pericyte loss in the primitive capillary network resulted in increased vessel fragility and hemorrhage, whereas concomitant VEGFR inhibition resulted in vessel regression and reduced vascular perfusion that restricted development of the hemorrhagic vessels. These results also highlight the utility of the rodent ovary to respond differentially to VEGFR and PDGFR inhibitors, which may provide useful information during routine safety assessment for determining target organ toxicity.


Subject(s)
Corpus Luteum/drug effects , Hemorrhage/chemically induced , Ovary/drug effects , Pericytes/drug effects , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Actins/metabolism , Animals , Corpus Luteum/physiopathology , Female , Histocytochemistry , Ovary/pathology , Ovary/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
12.
J Med Chem ; 57(20): 8249-67, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25271963

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitors have been used clinically in the treatment of non-small-cell lung cancer (NSCLC) patients harboring sensitizing (or activating) mutations for a number of years. Despite encouraging clinical efficacy with these agents, in many patients resistance develops leading to disease progression. In most cases, this resistance is in the form of the T790M mutation. In addition, EGFR wild type receptor inhibition inherent with these agents can lead to dose limiting toxicities of rash and diarrhea. We describe herein the evolution of an early, mutant selective lead to the clinical candidate AZD9291, an irreversible inhibitor of both EGFR sensitizing (EGFRm+) and T790M resistance mutations with selectivity over the wild type form of the receptor. Following observations of significant tumor inhibition in preclinical models, the clinical candidate was administered clinically to patients with T790M positive EGFR-TKI resistant NSCLC and early efficacy has been observed, accompanied by an encouraging safety profile.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/genetics , Chemistry Techniques, Synthetic , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Female , Humans , Inhibitory Concentration 50 , Lung Neoplasms/genetics , Male , Mice , Middle Aged , Mutation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats, Inbred Strains , Xenograft Model Antitumor Assays
13.
Nurse Res ; 22(1): 27-31, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25251817

ABSTRACT

AIM: To reflect on the author's personal and professional journey when undertaking semi-structured interviews on sensitive topics with potentially vulnerable people. BACKGROUND: When discussing care at the end of life, researchers must accept that some participants may become distressed or emotional, depending on their previous experiences. Interviews that involve sensitive topics require careful planning. DATA SOURCES: The semi-structured interviews were conducted as part of the author's PhD study examining the experiences of advance care planning among family caregivers of people with advanced dementia. REVIEW METHODS: A reflection on my personal and professional journey when undertaking semi-structured interviews on sensitive topics with potentially vulnerable people. DISCUSSION: The frustration and tragedy of dementia, as experienced by the family caregivers, were powerful and required the author to exert self-control to avoid being overly sympathetic and offering words of reassurance, agreement and comfort. CONCLUSION: This blurring of roles between researcher and nurse has implications for all nurse researchers who undertake qualitative interviews, particularly when an intense emotional response is likely. IMPLICATIONS FOR RESEARCH/PRACTICE: Nurse researchers should plan and prepare for potential blurring of roles during emotional interviews and should never automatically assume that they are sufficiently prepared as a result of their previous experience and nurse training.


Subject(s)
Interviews as Topic , Nursing Research , Research Personnel , Empathy , Humans , Workforce
14.
Cancer Discov ; 4(9): 1046-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24893891

ABSTRACT

UNLABELLED: First-generation EGFR tyrosine kinase inhibitors (EGFR TKI) provide significant clinical benefit in patients with advanced EGFR-mutant (EGFRm(+)) non-small cell lung cancer (NSCLC). Patients ultimately develop disease progression, often driven by acquisition of a second T790M EGFR TKI resistance mutation. AZD9291 is a novel oral, potent, and selective third-generation irreversible inhibitor of both EGFRm(+) sensitizing and T790M resistance mutants that spares wild-type EGFR. This mono-anilino-pyrimidine compound is structurally distinct from other third-generation EGFR TKIs and offers a pharmacologically differentiated profile from earlier generation EGFR TKIs. Preclinically, the drug potently inhibits signaling pathways and cellular growth in both EGFRm(+) and EGFRm(+)/T790M(+) mutant cell lines in vitro, with lower activity against wild-type EGFR lines, translating into profound and sustained tumor regression in EGFR-mutant tumor xenograft and transgenic models. The treatment of 2 patients with advanced EGFRm(+) T790M(+) NSCLC is described as proof of principle. SIGNIFICANCE: We report the development of a novel structurally distinct third-generation EGFR TKI, AZD9291, that irreversibly and selectively targets both sensitizing and resistant T790M(+) mutant EGFR while harboring less activity toward wild-type EGFR. AZD9291 is showing promising responses in a phase I trial even at the first-dose level, with first published clinical proof-of-principle validation being presented.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Acrylamides/chemistry , Acrylamides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , ErbB Receptors/chemistry , Female , Genes, erbB-2 , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
J Med Chem ; 56(17): 7025-48, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23930994

ABSTRACT

A novel series of small-molecule inhibitors has been developed to target the double mutant form of the epidermal growth factor receptor (EGFR) tyrosine kinase, which is resistant to treatment with gefitinib and erlotinib. Our reported compounds also show selectivity over wild-type EGFR. Guided by molecular modeling, this series was evolved to target a cysteine residue in the ATP binding site via covalent bond formation and demonstrates high levels of activity in cellular models of the double mutant form of EGFR. In addition, these compounds show significant activity against the activating mutations, which gefitinib and erlotinib target and inhibition of which gives rise to their observed clinical efficacy. A glutathione (GSH)-based assay was used to measure thiol reactivity toward the electrophilic functionality of the inhibitor series, enabling both the identification of a suitable reactivity window for their potency and the development of a reactivity quantitative structure-property relationship (QSPR) to support design.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Models, Molecular , Mutation , Structure-Activity Relationship
16.
Int J Cancer ; 129(4): 847-58, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21328342

ABSTRACT

Neutrophils are important innate immune cells that are involved in microbial clearance at sites of infection and in wound healing. The microenvironment of tumors often resembles that of chronic inflammation and increased numbers of neutrophils have been observed in several tumors and, in some cases, these positively correlate with poor prognosis. Neutrophil recruitment into tumors appears to be dependent on chemokines that bind to CXCR1 and CXCR2 expressed by neutrophils. In our study, we used lung adenocarcinoma A549 multicellular tumor spheroids and A549 tumor xenografts along with a CXCR2-specific small molecule inhibitor (AZ10397767) to investigate the recruitment and function of human neutrophils in tumors. We found that A549 spheroids constitutively secrete high levels of CXCL chemokines and that neutrophil recruitment into A549 tumors in vitro and in vivo is largely dependent on CXCR2 activation. AZ10397767 significantly reduced the numbers of infiltrating neutrophils into both in vitro and in vivo tumor models, which was associated with slower growing tumors. Neutrophil infiltration into A549 tumor spheroids increased their size compared to noninfiltrated spheroids and neutrophil-derived factors increased the proliferation of A549 tumor cells and induced endothelial cell tubule formation in vitro. In contrast, we saw no reduction in microvascular density in AZ10397767-treated A549 tumors or in tumors grown in CXCR2(-/-) mice, suggesting that angiogenesis in these tumors is CXCR2-independent. Our data show that neutrophils can contribute to lung tumor growth and that CXCR2 antagonists may be a useful therapeutic agent in the treatment of lung carcinomas.


Subject(s)
Adenocarcinoma/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Neutrophil Infiltration/drug effects , Quinazolines/therapeutic use , Receptors, Interleukin-8B/antagonists & inhibitors , Triazoles/therapeutic use , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Animals , Blotting, Western , Cell Proliferation , Chemotaxis , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques , In Vitro Techniques , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , RNA, Messenger/genetics , Receptors, Interleukin-8B/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 16(14): 3548-61, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20606037

ABSTRACT

PURPOSE: Vascular endothelial growth factor (VEGF) signaling is key to tumor angiogenesis and is an important target in the development of anticancer drugs. However, VEGF receptor (VEGFR) expression in human cancers, particularly the relative expression of VEGFR-2 and VEGFR-3 in tumor vasculature versus tumor cells, is poorly defined. EXPERIMENTAL DESIGN: VEGFR-2- and VEGFR-3-specific antibodies were identified and used in the immunohistochemical analysis of human primary cancers and normal tissue. The relative vascular localization of both receptors in colorectal and breast cancers was determined by coimmunofluorescence with vascular markers. RESULTS: VEGFR-2 and VEGFR-3 were expressed on vascular endothelium but not on malignant cells in 13 common human solid tumor types (n > 400, bladder, breast, colorectal, head and neck, liver, lung, skin, ovarian, pancreatic, prostate, renal, stomach, and thyroid). The signal intensity of both receptors was significantly greater in vessels associated with malignant colorectal, lung, and breast than adjacent nontumor tissue. In colorectal cancers, VEGFR-2 was expressed on both intratumoral blood and lymphatic vessels, whereas VEGFR-3 was found predominantly on lymphatic vessels. In breast cancers, both receptors were localized to and upregulated on blood vessels. CONCLUSIONS: VEGFR-2 and VEGFR-3 are primarily localized to, and significantly upregulated on, tumor vasculature (blood and/or lymphatic) supporting the majority of solid cancers. The primary clinical mechanism of action of VEGF signaling inhibitors is likely to be through the targeting of tumor vessels rather than tumor cells. The upregulation of VEGFR-3 on tumor blood vessels indicates a potential additional antiangiogenic effect for dual VEGFR-2/VEGFR-3-targeted therapy.


Subject(s)
Endothelium, Vascular/metabolism , Neoplasms, Experimental/metabolism , Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Endothelium, Vascular/pathology , Humans , Immunohistochemistry , Mice , Mice, Nude , Mice, SCID , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms/pathology , Neoplasms, Experimental/pathology
18.
J Natl Cancer Inst ; 99(22): 1724-8, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18000220

ABSTRACT

Antivascular agents that act by destabilizing microtubules, such as ZD6126 (N-acetylcolchinol-O-phosphate), are associated with adverse cardiovascular effects, including transient hypertension, cardiac ischemia, myocardial infarction, and increases in circulating levels of markers of cardiac damage (e.g., troponins). We investigated mechanisms underlying these effects of ZD6126 in rats by continuously monitoring their heart rate and blood pressure and by assessing heart histopathology and plasma troponin T levels. ZD6126 induced acute transient hemodynamic changes (hypertension and delayed tachycardia), which were associated with statistically significant increases in circulating troponin T levels (median level 3 hours after treatment with vehicle or 12.5 mg/kg ZD6126: <9 pg/mL and 563 pg/mL, respectively; P <.001 [two-sided Wilcoxon rank sum test]) and in the incidence of left ventricular myocardial fiber necrosis (incidence 24 hours after treatment with vehicle or 12.5 mg/kg ZD6126: 0/10 rats and 9/10 rats, respectively; P <.001 [two-sided Wilcoxon rank sum test]). Pretreatment of rats with atenolol and nifedipine ameliorated the acute hemodynamic changes and prevented ZD6126-induced increases in both troponin T and myocardial necrosis but did not prevent ZD6126-induced tumor necrosis in an Hras5 tumor xenograft model in nude rats. Our findings suggest that ZD6126-induced acute hemodynamic changes are a prerequisite for cardiac damage in rats.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Antihypertensive Agents/pharmacology , Antineoplastic Agents/toxicity , Atenolol/pharmacology , Hemodynamics/drug effects , Hypertension/prevention & control , Nifedipine/pharmacology , Organophosphorus Compounds/toxicity , Tachycardia/prevention & control , Adrenergic beta-Antagonists/pharmacology , Animals , Anti-Arrhythmia Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Atenolol/therapeutic use , Biomarkers/blood , Calcium Channel Blockers/pharmacology , Disease Models, Animal , Drug Administration Schedule , Female , Heart/drug effects , Hypertension/chemically induced , Myocardium/pathology , Neoplasms/drug therapy , Nifedipine/therapeutic use , Organophosphorus Compounds/administration & dosage , Random Allocation , Rats , Tachycardia/chemically induced , Transplantation, Heterologous , Troponin T/blood , Vasodilator Agents/pharmacology
19.
Neoplasia ; 9(5): 382-91, 2007 May.
Article in English | MEDLINE | ID: mdl-17534443

ABSTRACT

Magnetic resonance imaging (MRI) can measure the effects of therapies targeting the tumor vasculature and has demonstrated that vascular-damaging agents (VDA) induce acute vascular shutdown in tumors in human and animal models. However, at subtherapeutic doses, blood flow may recover before the induction of significant levels of necrosis. We present the relationship between changes in MRI biomarkers and tumor necrosis. Multiple MRI measurements were taken at 4.7 T in athymic rats (n = 24) bearing 1.94 +/- 0.2-cm3 subcutaneous Hras5 tumors (ATCC 41000) before and 24 hours after clinically relevant doses of the VDA, ZD6126 (0-10 mg/kg, i.v.). We measured effective transverse relaxation rate (R2*), initial area under the gadolinium concentration-time curve (IAUGC(60/150)), equivalent enhancing fractions (EHF(60/150)), time constant (K(trans)), proportion of hypoperfused voxels as estimated from fit failures in K(trans) analysis, and signal intensity (SI) in T2-weighted MRI (T(2)W). ZD6126 treatment induced > 90% dose-dependent tumor necrosis at 10 mg/kg; correspondingly, SI changes were evident from T2W MRI. Although R2* did not correlate, other MRI biomarkers significantly correlated with necrosis at doses of > or = 5 mg/kg ZD6126. These data on Hras5 tumors suggest that the quantification of hypoperfused voxels might provide a useful biomarker of tumor necrosis.


Subject(s)
Magnetic Resonance Imaging , Neoplasms, Experimental/drug therapy , Organophosphorus Compounds/therapeutic use , Animals , Biomarkers , Dose-Response Relationship, Drug , Genes, ras , Male , Mice , NIH 3T3 Cells , Necrosis , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Rats , Rats, Nude , Transplantation, Heterologous
20.
Patient Educ Couns ; 62(3): 330-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16860522

ABSTRACT

OBJECTIVE: For women who are experiencing intimate partner violence (IPV), making changes toward safety is often a gradual process. When providing counseling and support, health care providers may benefit from better understanding of where women are in their readiness to change. Our objective was to apply the transtheoretical model's stages of change to the experiences of women who experienced IPV and map their experiences of change as they moved toward increased safety. METHODS: A multi-disciplinary team designed a qualitative interview process with 20 women who had current or past histories of IPV in order to explore their experiences. RESULTS: The women in our study (1) moved through stages of readiness generally in a nonlinear fashion, with varying rates of progression between safe and nonsafe situations, (2) were able to identify a "turning-point" in their situations, (3) attempted multiple "action" steps and (4) were influenced by internal and external factors. CONCLUSIONS: Our study suggests that focusing on the transtheoretical model to develop stage-based interventions for IPV may not be the most appropriate given the nonsequential movement between stages and influence of external factors. PRACTICE IMPLICATIONS: The "change mapping" technique can be used as an educational and counseling tool with patients, as well as a training tool for health care providers.


Subject(s)
Adaptation, Psychological , Battered Women/psychology , Health Behavior , Models, Psychological , Patient Acceptance of Health Care/psychology , Spouse Abuse/psychology , Adult , Battered Women/education , Counseling/organization & administration , Educational Status , Female , Health Knowledge, Attitudes, Practice , Humans , Middle Aged , Motivation , Narration , Patient Education as Topic/organization & administration , Pennsylvania , Qualitative Research , Safety Management , Self Care/methods , Self Care/psychology , Socioeconomic Factors , Spouse Abuse/prevention & control , Surveys and Questionnaires , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...