Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
3.
J Mol Diagn ; 25(10): 709-728, 2023 10.
Article in English | MEDLINE | ID: mdl-37517472

ABSTRACT

DNA methylation array profiling for classifying pediatric central nervous system (CNS) tumors is a valuable adjunct to histopathology. However, unbiased prospective and interlaboratory validation studies have been lacking. The AIM BRAIN diagnostic trial involving 11 pediatric cancer centers in Australia and New Zealand was designed to test the feasibility of routine clinical testing and ran in parallel with the Molecular Neuropathology 2.0 (MNP2.0) study at Deutsches Krebsforschungszentrum (German Cancer Research Center). CNS tumors from 269 pediatric patients were prospectively tested on Illumina EPIC arrays, including 104 cases co-enrolled on MNP2.0. Using MNP classifier versions 11b4 and 12.5, we report classifications with a probability score ≥0.90 in 176 of 265 (66.4%) and 213 of 269 (79.2%) cases, respectively. Significant diagnostic information was obtained in 130 of 176 (74%) for 11b4, and 12 of 174 (7%) classifications were discordant with histopathology. Cases prospectively co-enrolled on MNP2.0 gave concordant classifications (99%) and score thresholds (93%), demonstrating excellent test reproducibility and sensitivity. Overall, DNA methylation profiling is a robust single workflow technique with an acceptable diagnostic yield that is considerably enhanced by the extensive subgroup and copy number profile information generated by the platform. The platform has excellent test reproducibility and sensitivity and contributes significantly to CNS tumor diagnosis.


Subject(s)
Central Nervous System Neoplasms , DNA Methylation , Child , Humans , Australia , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , DNA Methylation/genetics , New Zealand , Prospective Studies , Reproducibility of Results
4.
Neurooncol Adv ; 5(1): vdad057, 2023.
Article in English | MEDLINE | ID: mdl-37287693

ABSTRACT

Background: ZFTA-RELA (formerly known as c11orf-RELA) fused supratentorial ependymoma (ZFTAfus ST-EPN) has been recognized as a novel entity in the 2016 WHO classification of CNS tumors and further defined in the recent 2021 edition. ZFTAfus ST-EPN was reported to portend poorer prognosis when compared to its counterpart, YAP1 ST-EPN in some previously published series. The aim of this study was to determine the treatment outcome of molecularly confirmed and conventionally treated ZFTAfus ST-EPN patients treated in multiple institutions. Methods: We conducted a retrospective analysis of all pediatric patients with molecularly confirmed ZFTAfus ST-EPN patients treated in multiple institutions in 5 different countries (Australia, Canada, Germany, Switzerland, and Czechia). Survival outcomes were analyzed and correlated with clinical characteristics and treatment approaches. Results: A total of 108 patients were collated from multiple institutions in 5 different countries across three continents. We found across the entire cohort that the 5- and 10-year PFS were 65% and 63%, respectively. The 5- and 10-year OS of this cohort of patients were 87% and 73%. The rates of gross total resection (GTR) were high with 84 out of 108 (77.8%) patients achieving GTR. The vast majority of patients also received post-operative radiotherapy, 98 out of 108 (90.7%). Chemotherapy did not appear to provide any survival benefit in our patient cohort. Conclusion: This is the largest study to date of contemporaneously treated molecularly confirmed ZFTAfus ST-EPN patients which identified markedly improved survival outcomes compared to previously published series. This study also re-emphasizes the importance of maximal surgical resection in achieving optimal outcomes in pediatric patients with supratentorial ependymoma.

5.
Nat Med ; 29(4): 917-926, 2023 04.
Article in English | MEDLINE | ID: mdl-36928815

ABSTRACT

The large diversity of central nervous system (CNS) tumor types in children and adolescents results in disparate patient outcomes and renders accurate diagnosis challenging. In this study, we prospectively integrated DNA methylation profiling and targeted gene panel sequencing with blinded neuropathological reference diagnostics for a population-based cohort of more than 1,200 newly diagnosed pediatric patients with CNS tumors, to assess their utility in routine neuropathology. We show that the multi-omic integration increased diagnostic accuracy in a substantial proportion of patients through annotation to a refining DNA methylation class (50%), detection of diagnostic or therapeutically relevant genetic alterations (47%) or identification of cancer predisposition syndromes (10%). Discrepant results by neuropathological WHO-based and DNA methylation-based classification (30%) were enriched in histological high-grade gliomas, implicating relevance for current clinical patient management in 5% of all patients. Follow-up (median 2.5 years) suggests improved survival for patients with histological high-grade gliomas displaying lower-grade molecular profiles. These results provide preliminary evidence of the utility of integrating multi-omics in neuropathology for pediatric neuro-oncology.


Subject(s)
Brain Neoplasms , Glioma , Adolescent , Humans , Child , Multiomics , Glioma/diagnosis , Glioma/genetics , Neuropathology , DNA Methylation/genetics , Mutation , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics
7.
FASEB J ; 29(4): 1426-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25550458

ABSTRACT

Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of PI3K, are among the most common mutations found in human cancer and have also recently been implicated in a range of overgrowth syndromes in humans. We have used a novel inducible "exon-switch" approach to knock in the constitutively active Pik3ca(H1047R) mutation into the endogenous Pik3ca gene of the mouse. Ubiquitous expression of the Pik3ca(H1047R) mutation throughout the body resulted in a dramatic increase in body weight within 3 weeks of induction (mutant 150 ± 5%; wild-type 117 ± 3%, mean ± sem), which was associated with increased organ size rather than adiposity. Severe metabolic effects, including a reduction in blood glucose levels to 59 ± 4% of baseline (11 days postinduction) and undetectable insulin levels, were also observed. Pik3ca(H1047R) mutant mice died earlier (median survival 46.5 d post-mutation induction) than wild-type control mice (100% survival > 250 days). Although deletion of Akt2 increased median survival by 44%, neither organ overgrowth, nor hypoglycemia were rescued, indicating that both the growth and metabolic functions of constitutive PI3K activity can be Akt2 independent. This mouse model demonstrates the critical role of PI3K in the regulation of both organ size and glucose metabolism at the whole animal level.


Subject(s)
Hypoglycemia/enzymology , Hypoglycemia/genetics , Insulin/blood , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Substitution , Animals , Class I Phosphatidylinositol 3-Kinases , Female , Gene Expression , Gene Knock-In Techniques , Glucose/metabolism , Humans , Hypoglycemia/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Organ Size/genetics , Organ Size/physiology , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Weight Gain
8.
Cancer Discov ; 4(4): 423-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469106

ABSTRACT

UNLABELLED: Deregulated glucose metabolism fulfills the energetic and biosynthetic requirements for tumor growth driven by oncogenes. Because inhibition of oncogenic BRAF causes profound reductions in glucose uptake and a strong clinical benefit in BRAF-mutant melanoma, we examined the role of energy metabolism in responses to BRAF inhibition. We observed pronounced and consistent decreases in glycolytic activity in BRAF-mutant melanoma cells. Moreover, we identified a network of BRAF-regulated transcription factors that control glycolysis in melanoma cells. Remarkably, this network of transcription factors, including hypoxia-inducible factor-1α, MYC, and MONDOA (MLXIP), drives glycolysis downstream of BRAF(V600), is critical for responses to BRAF inhibition, and is modulated by BRAF inhibition in clinical melanoma specimens. Furthermore, we show that concurrent inhibition of BRAF and glycolysis induces cell death in BRAF inhibitor (BRAFi)-resistant melanoma cells. Thus, we provide a proof-of-principle for treatment of melanoma with combinations of BRAFis and glycolysis inhibitors. SIGNIFICANCE: BRAF is suppress glycolysis and provide strong clinical benefi t in BRAF V600 melanoma. We show that BRAF inhibition suppresses glycolysis via a network of transcription factors that are critical for complete BRAFi responses. Furthermore, we provide evidence for the clinical potential of therapies that combine BRAFis with glycolysis inhibitors.


Subject(s)
Glycolysis/drug effects , Melanoma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , HEK293 Cells , Humans , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Piperazines/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Vemurafenib
9.
Biochem J ; 458(2): 251-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24320611

ABSTRACT

PIK3CA, the gene encoding the p110α catalytic subunit of PI3K (phosphoinositide 3-kinase), is mutated in approximately 20% of sporadic CRCs (colorectal cancers), but the role of these mutations in the pathogenesis of CRC remains unclear. In the present study we used a novel mouse model to investigate the role of the Pik3caH1047R mutation, the most common PIK3CA mutation in CRC, during the development and progression of intestinal cancer. Our results demonstrate that Pik3caH1047R, when expressed at physiological levels, is insufficient to initiate intestinal tumorigenesis; however, in the context of Apc (adenomatous polyposis coli) loss, which is observed in 80% of CRCs and by itself results in benign intestinal adenomas, the Pik3caH1047R mutation promotes the development of highly aggressive and invasive adenocarcinomas in both the small and large intestines. The results of the present study show that an activating Pik3ca mutation can act in tandem with Apc loss to drive the progression of gastrointestinal cancer and thus this disease may be susceptible to therapeutic targeting using PI3K pathway inhibitors.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli Protein/deficiency , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , Mutation/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Disease Progression , Female , Gene Knock-In Techniques , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/biosynthesis
10.
Eur J Cancer ; 49(18): 3936-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24011934

ABSTRACT

BACKGROUND: Ovarian cancer is the major cause of death from gynaecological malignancy with a 5year survival of only ∼30% due to resistance to platinum and paclitaxel-based first line therapy. Dysregulation of the phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) and RAS/extracellular signal-regulated kinase (ERK) pathways is common in ovarian cancer, providing potential new targets for 2nd line therapy. METHODS: We determined the inhibition of proliferation of an extensive panel of ovarian cancer cell lines, encompassing all the major histotypes, by the dual PI3K/mTOR inhibitor PF-04691502 and a MEK inhibitor, PD-0325901. In addition, we analysed global gene expression, mutation status of key PI3K/mTOR and RAS/ERK pathway members and pathway activation to identify predictors of drug response. RESULTS: PF-04691502 inhibits proliferation of the majority of cell lines with potencies that correlate with the extent of pathway inhibition. Resistant cell lines were characterised by activation of the RAS/ERK pathway as indicated by differential gene expression profiles and pathway activity analysis. PD-0325901 suppressed growth of a subset of cell lines that were characterised by high basal RAS/ERK signalling. Strikingly, using PF-04691502 and PD-0325901 in combination resulted in synergistic growth inhibition in 5/6 of PF-04691502 resistant cell lines and two cell lines resistant to both single agents showed robust synergistic growth arrest. Xenograft studies confirm the utility of combination therapy to synergistically inhibit tumour growth of PF-04691502-resistant tumours in vivo. CONCLUSIONS: These studies identify dual targeted inhibitors of PI3K/mTOR in combination with inhibitors of RAS/ERK signalling as a potentially effective new approach to treating ovarian cancer.


Subject(s)
Benzamides/pharmacology , Cell Proliferation/drug effects , Diphenylamine/analogs & derivatives , Ovarian Neoplasms/drug therapy , Pyridones/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/administration & dosage , Cell Line, Tumor , Diphenylamine/administration & dosage , Diphenylamine/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
11.
J Clin Invest ; 123(3): 1371-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23454771

ABSTRACT

The BRAF mutant, BRAF(V600E), is expressed in nearly half of melanomas, and oral BRAF inhibitors induce substantial tumor regression in patients with BRAF(V600E) metastatic melanoma. The inhibitors are believed to work primarily by inhibiting BRAF(V600E)-induced oncogenic MAPK signaling; however, some patients treated with BRAF inhibitors exhibit increased tumor immune infiltration, suggesting that a combination of BRAF inhibitors and immunotherapy may be beneficial. We used two relatively resistant variants of Braf(V600E)-driven mouse melanoma (SM1 and SM1WT1) and melanoma-prone mice to determine the role of host immunity in type I BRAF inhibitor PLX4720 antitumor activity. We found that PLX4720 treatment downregulated tumor Ccl2 gene expression and decreased tumor CCL2 expression in both Braf(V600E) mouse melanoma transplants and in de novo melanomas in a manner that was coincident with reduced tumor growth. While PLX4720 did not directly increase tumor immunogenicity, analysis of SM1 tumor-infiltrating leukocytes in PLX4720-treated mice demonstrated a robust increase in CD8(+) T/FoxP3(+)CD4(+) T cell ratio and NK cells. Combination therapy with PLX4720 and anti-CCL2 or agonistic anti-CD137 antibodies demonstrated significant antitumor activity in mouse transplant and de novo tumorigenesis models. These data elucidate a role for host CCR2 in the mechanism of action of type I BRAF inhibitors and support the therapeutic potential of combining BRAF inhibitors with immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Immunity, Cellular , Immunotherapy , Indoles/administration & dosage , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Receptors, CCR2/metabolism , Sulfonamides/administration & dosage , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Xenograft Model Antitumor Assays
12.
Cancer Discov ; 3(1): 82-95, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23242809

ABSTRACT

UNLABELLED: MYC deregulation is common in human cancer. IG-MYC translocations that are modeled in Eµ-Myc mice occur in almost all cases of Burkitt lymphoma as well as in other B-cell lymphoproliferative disorders. Deregulated expression of MYC results in increased mTOR complex 1 (mTORC1) signaling. As tumors with mTORC1 activation are sensitive to mTORC1 inhibition, we used everolimus, a potent and specific mTORC1 inhibitor, to test the requirement for mTORC1 in the initiation and maintenance of Eµ-Myc lymphoma. Everolimus selectively cleared premalignant B cells from the bone marrow and spleen, restored a normal pattern of B-cell differentiation, and strongly protected against lymphoma development. Established Eµ-Myc lymphoma also regressed after everolimus therapy. Therapeutic response correlated with a cellular senescence phenotype and induction of p53 activity. Therefore, mTORC1-dependent evasion of senescence is critical for cellular transformation and tumor maintenance by MYC in B lymphocytes. SIGNIFICANCE: This work provides novel insights into the requirements for MYC-induced oncogenesis by showing that mTORC1 activity is necessary to bypass senescence during transformation of B lymphocytes. Furthermore, tumor eradication through senescence elicited by targeted inhibition of mTORC1 identifies a previously uncharacterized mechanism responsible for significant anticancer activity of rapamycin analogues and serves as proof-of-concept that senescence can be harnessed for therapeutic benefit


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma/drug therapy , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Sirolimus/analogs & derivatives , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation/drug effects , Cellular Senescence , Everolimus , Lymphoma/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
13.
Crit Rev Oncog ; 17(1): 69-95, 2012.
Article in English | MEDLINE | ID: mdl-22471665

ABSTRACT

The phosphatidylinositol 3 kinase (PI3K) pathway is one of the major pathways modulating cell growth, proliferation, metabolism, survival, and angiogenesis. Hyperactivation of this pathway is one of the most frequent occurrences in human cancer and is thus an obvious target for treatment of this disease. Currently there are 26 novel compounds targeting the PI3K pathway being assessed in more than 150 cancer-related clinical trials. Although this pathway is involved in many vital biologic functions, data emanating from these clinical trials indicate that these drugs are well tolerated. This review outlines the interaction of the PI3K pathway with other signaling cascades, highlights mechanisms involved in hyperactivation, discusses current therapeutics in cancer-related clinical trials that target this pathway, and, based on preclinical data, discusses possible leads on patient selection and combinational therapy, including targeting multiple components of the associated signaling network.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Humans , Models, Biological , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/physiology
14.
J Clin Invest ; 122(2): 553-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22214849

ABSTRACT

Mutations in the gene encoding the p110α subunit of PI3K (PIK3CA) that result in enhanced PI3K activity are frequently observed in human cancers. To better understand the role of mutant PIK3CA in the initiation or progression of tumorigenesis, we generated mice in which a PIK3CA mutation commonly detected in human cancers (the H1047R mutation) could be conditionally knocked into the endogenous Pik3ca locus. Activation of this mutation in the mouse ovary revealed that alone, Pik3caH1047R induced premalignant hyperplasia of the ovarian surface epithelium but no tumors. Concomitantly, we analyzed several human ovarian cancers and found PIK3CA mutations coexistent with KRAS and/or PTEN mutations, raising the possibility that a secondary defect in a co-regulator of PI3K activity may be required for mutant PIK3CA to promote transformation. Consistent with this notion, we found that Pik3caH1047R mutation plus Pten deletion in the mouse ovary led to the development of ovarian serous adenocarcinomas and granulosa cell tumors. Both mutational events were required for early, robust Akt activation. Pharmacological inhibition of PI3K/mTOR in these mice delayed tumor growth and prolonged survival. These results demonstrate that the Pik3caH1047R mutation with loss of Pten is enough to promote ovarian cell transformation and that we have developed a model system for studying possible therapies.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation , Ovarian Neoplasms/genetics , PTEN Phosphohydrolase/deficiency , Phosphatidylinositol 3-Kinases/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovarian Neoplasms/pathology , Ovary/anatomy & histology , Ovary/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Survival Rate
15.
Mol Cancer Ther ; 10(8): 1440-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632463

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is commonly dysregulated in human cancer, making it an attractive target for novel anticancer therapeutics. We have used a mouse model of ovarian cancer generated by Kras(G12D) activation and Pten deletion in the ovarian surface epithelium for the preclinical assessment of a novel PI3K/mTOR inhibitor PF-04691502. To enable higher throughput studies, we developed an orthotopic primary transplant model from these mice and evaluated therapeutic response to PF-04691502 using small-animal ultrasound and FDG-PET imaging. PF-04691502 inhibited tumor growth at 7 days by 72% ± 9. FDG-PET imaging revealed that PF-04691502 reduced glucose metabolism dramatically, suggesting FDG-PET may be exploited as an imaging biomarker of target inhibition by PF-04691502. Tissue biomarkers of PI3K/mTOR pathway activity, p-AKT (S473), and p-RPS6 (S240/244), were also dramatically inhibited following PF-04691502 treatment. However, as a single agent, PF-04691502 did not induce tumor regression and the long-term efficacy was limited, with tumor proliferation continuing in the presence of drug treatment. We hypothesized that tumor progression was because of concomitant activation of the mitogen-activated protein kinase pathway downstream of Kras(G12D) expression promoting cell survival and that the therapeutic effect of PF-04691502 would be enhanced by combinatory inhibition of MEK using PD-0325901. This combination induced striking tumor regression, apoptosis associated with upregulation of Bim and downregulation of Mcl-1, and greatly improved duration of survival. These data suggest that contemporaneous MEK inhibition enhances the cytotoxicity associated with abrogation of PI3K/mTOR signaling, converting tumor growth inhibition to tumor regression in a mouse model of ovarian cancer driven by PTEN loss and mutant K-Ras.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , ras Proteins/genetics , Animals , Carbohydrate Metabolism/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Glucose/metabolism , Humans , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Burden/drug effects
16.
Mol Imaging Biol ; 13(3): 518-525, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20632117

ABSTRACT

PURPOSE: The aim of the study was to improve abdominal tumor detection by use of a dual radiologic contrast protocol. PROCEDURES: eXia160® (Benitio international) was mixed with 2-deoxy-2-[¹8F]fluoro-D: -glucose or 3'-[¹8F]fluoro-3'-deoxythymidine for intravenous (IV) injections. Omnipaque® 300 (GE healthcare) was used for intraperitoneal (IP) injections. Positron emission tomography/computed tomography (PET/CT) scans were acquired on a Siemens Biograph® equipped with point spread function reconstruction. The optimal concentration and injection schedule of IP contrast agent was studied in 12 mice. The impact of IP contrast media on PET quantitative accuracy was investigated by phantom studies and by imaging six mice before and after IP injection of Omnipaque®. The impact of a dual contrast media protocol on tumor delineation and quantitation was evaluated in 15 tumor-bearing mice using ex vivo counting as the reference. RESULTS: The optimal sequence was a mixture of tracer plus IV contrast agent followed by 1 mL of IP contrast agent (20 mg iodine/mL) administered 10 min before PET/CT acquisition. Phantom studies showed that the use of a 20-mg iodine/mL concentration of Omnipaque® led to a 4.8% overestimation of radioactivity concentration, as compared to saline. This was confirmed by animal studies that demonstrated a 4.3% overestimation. Tumor detection was excellent and correlation between PET/CT quantitative data and ex vivo counting was good (r² = 0.91, slope = 0.7). CONCLUSIONS: A dual radiologic contrast protocol is useful in PET/CT scanning of mice bearing abdominal tumors. Contrast agents used in this manner lead to a small but acceptable overestimation of quantitative PET data.


Subject(s)
Abdominal Neoplasms/diagnostic imaging , Contrast Media , Dideoxynucleosides , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Tomography, X-Ray Computed , Animals , Ascites/pathology , Contrast Media/administration & dosage , Dideoxynucleosides/administration & dosage , Fluorodeoxyglucose F18/administration & dosage , Injections, Intravenous , Mice
17.
J Nucl Med ; 51(10): 1559-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20847160

ABSTRACT

UNLABELLED: Targeting the mammalian target of rapamycin (mTOR) pathway is a potential means of overcoming cisplatin resistance in ovarian cancer patients. Because mTOR inhibition affects cell proliferation, we aimed to study whether 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) PET could be useful for monitoring early response to treatment with mTOR inhibitors in an animal model of cisplatin-resistant ovarian tumor. METHODS: BALB/c nude mice bearing subcutaneous human SKOV3 ovarian cancer xenografts were treated with either the mTOR inhibitor everolimus (5 mg/kg) or vehicle, and (18)F-FLT PET was performed at baseline, day 2, and day 7 of treatment. (18)F-FLT uptake was evaluated by calculation of mean standardized uptake value (SUVmean) corrected for partial-volume effect. Ex vivo immunohistochemistry studies were performed on separate cohorts of mice treated as above and sacrificed at the same time points as for the PET studies. The ex vivo analysis included bromodeoxyuridine incorporation as a marker of cell proliferation, and phosphorylation of ribosomal protein S6 as a downstream marker of mTOR activation. RESULTS: During the treatment period, no significant change in tumor (18)F-FLT uptake was observed in the vehicle group, whereas in everolimus-treated mice, (18)F-FLT SUVmean decreased by 33% (P = 0.003) at day 2 and 66% (P < 0.001) at day 7, compared with baseline. Notably, the reduction of (18)F-FLT uptake observed at day 2 in the everolimus group preceded changes in tumor volume, and a significant difference in (18)F-FLT uptake was observed between vehicle and drug-treated tumors at both day 2 (P = 0.0008) and day 7 (P = 0.01). In ex vivo studies, everolimus treatment resulted in a 98% reduction in phosphorylated ribosomal protein S6 immunostaining at day 2 (P = 0.02) and 91% reduction at day 7 (P = 0.003), compared with the vehicle group. Bromodeoxyuridine incorporation was reduced by 65% at day 2 (not significant) and by 41% at day 7 (P = 0.02) in drug versus vehicle groups. CONCLUSION: Reduction in (18)F-FLT uptake correlates well with the level of mTOR inhibition by everolimus in the SKOV3 ovarian tumor model. These data suggest that early treatment monitoring by (18)F-FLT PET may be of use in future preclinical or clinical trials evaluating treatment of cisplatin-resistant ovarian tumors by mTOR inhibitors.


Subject(s)
Dideoxynucleosides , Disease Models, Animal , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Positron-Emission Tomography/methods , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Everolimus , Female , Humans , Mice , Mice, Nude , Radiopharmaceuticals , Reproducibility of Results , Sensitivity and Specificity , Sirolimus/administration & dosage , Treatment Outcome
18.
Eur J Nucl Med Mol Imaging ; 37(5): 991-1001, 2010 May.
Article in English | MEDLINE | ID: mdl-20107792

ABSTRACT

PURPOSE: The objective of the study was to evaluate state-of-the-art clinical PET/CT technology in performing static and dynamic imaging of several mice simultaneously. METHODS: A mouse-sized phantom was imaged mimicking simultaneous imaging of three mice with computation of recovery coefficients (RCs) and spillover ratios (SORs). Fifteen mice harbouring abdominal or subcutaneous tumours were imaged on clinical PET/CT with point spread function (PSF) reconstruction after injection of [18F]fluorodeoxyglucose or [18F]fluorothymidine. Three of these mice were imaged alone and simultaneously at radial positions -5, 0 and 5 cm. The remaining 12 tumour-bearing mice were imaged in groups of 3 to establish the quantitative accuracy of PET data using ex vivo gamma counting as the reference. Finally, a dynamic scan was performed in three mice simultaneously after the injection of (68)Ga-ethylenediaminetetraacetic acid (EDTA). RESULTS: For typical lesion sizes of 7-8 mm phantom experiments indicated RCs of 0.42 and 0.76 for ordered subsets expectation maximization (OSEM) and PSF reconstruction, respectively. For PSF reconstruction, SOR(air) and SOR(water) were 5.3 and 7.5%, respectively. A strong correlation (r (2) = 0.97, p < 0.0001) between quantitative data obtained in mice imaged alone and simultaneously in a group of three was found following PSF reconstruction. The correlation between ex vivo counting and PET/CT data was better with PSF reconstruction (r (2) = 0.98; slope = 0.89, p < 0.0001) than without (r (2) = 0.96; slope = 0.62, p < 0.001). Valid time-activity curves of the blood pool, kidneys and bladder could be derived from (68)Ga-EDTA dynamic acquisition. CONCLUSION: New generation clinical PET/CT can be used for simultaneous imaging of multiple small animals in experiments requiring high throughput and where a dedicated small animal PET system is not available.


Subject(s)
Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Animals , Edetic Acid , Mice , Phantoms, Imaging , Time Factors
19.
Blood ; 108(3): 886-95, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16861343

ABSTRACT

The E2F proteins are major regulators of the transcriptional program required to coordinate cell cycle progression and exit. In particular, E2f4 has been proposed to be the principal family member responsible for the regulation of cell cycle exit chiefly through its transcriptional repressive properties. We have previously shown that E2f4(-/-) mice display a marked macrocytic anemia implicating E2f4 in the regulation of erythropoiesis. However, these studies could not distinguish whether E2f4 was required for differentiation, survival, or proliferation control. Here, we describe a novel function for E2f4 in the promotion of erythroid proliferation. We show that loss of E2f4 results in an impaired expansion of the fetal erythroid compartment in vivo that is associated with impaired cell cycle progression and decreased erythroid proliferation. Consistent with these observations, cDNA microarray analysis reveals cell cycle control genes as one of the major class of genes down-regulated in E2f4(-/-) FLs, and we provide evidence that E2f4 may directly regulate the transcriptional expression of a number of these genes. We conclude that the macrocytic anemia of E2f4(-/-) mice results primarily from impaired cellular proliferation and that the major role of E2f4 in fetal erythropoiesis is to promote cell cycle progression and cellular proliferation.


Subject(s)
Cell Proliferation , E2F4 Transcription Factor/physiology , Erythroid Cells/cytology , Erythropoiesis , Anemia, Macrocytic/etiology , Animals , Cell Cycle , E2F4 Transcription Factor/deficiency , Embryo, Mammalian , Gene Expression Profiling , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...