Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
ESMO Open ; 9(6): 103488, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838500

ABSTRACT

BACKGROUND: Fibroblast growth factor receptor 2 (FGFR2) fusions and rearrangements are clinically actionable genomic alterations in cholangiocarcinoma (CCA). Pemigatinib is a selective, potent, oral inhibitor of FGFR1-3 and demonstrated efficacy in patients with previously treated, advanced/metastatic CCA with FGFR2 alterations in FIGHT-202 (NCT02924376). We report final outcomes from the extended follow-up period. PATIENTS AND METHODS: The multicenter, open-label, single-arm, phase II FIGHT-202 study enrolled patients ≥18 years old with previously treated advanced/metastatic CCA with FGFR2 fusions or rearrangements (cohort A), other FGF/FGFR alterations (cohort B), or no FGF/FGFR alterations (cohort C). Patients received once-daily oral pemigatinib 13.5 mg in 21-day cycles (2 weeks on, 1 week off) until disease progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR) in cohort A assessed as per RECIST v1.1 by an independent review committee; secondary endpoints included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS: FIGHT-202 enrolled 147 patients (cohort A, 108; cohort B, 20; cohort C, 17; unconfirmed FGF/FGFR alterations, 2). By final analysis, 145 (98.6%) had discontinued treatment due to progressive disease (71.4%), withdrawal by patient (8.2%), or adverse events (AEs; 6.8%). Median follow-up was 45.4 months. The ORR in cohort A was 37.0% (95% confidence interval 27.9% to 46.9%); complete and partial responses were observed in 3 and 37 patients, respectively. Median DOR was 9.1 (6.0-14.5) months; median PFS and OS were 7.0 (6.1-10.5) months and 17.5 (14.4-22.9) months, respectively. The most common treatment-emergent AEs (TEAEs) were hyperphosphatemia (58.5%), alopecia (49.7%), and diarrhea (47.6%). Overall, 15 (10.2%) patients experienced TEAEs leading to pemigatinib discontinuation; intestinal obstruction and acute kidney injury (n = 2 each) occurred most frequently. CONCLUSIONS: Pemigatinib demonstrated durable response and prolonged OS with manageable AEs in patients with previously treated, advanced/metastatic CCA with FGFR2 alterations in the extended follow-up period of FIGHT-202.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Pyrimidines , Humans , Cholangiocarcinoma/drug therapy , Male , Female , Middle Aged , Aged , Adult , Bile Duct Neoplasms/drug therapy , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 2 , Aged, 80 and over , Morpholines , Pyrroles
2.
Clin Oncol (R Coll Radiol) ; 36(2): 87-97, 2024 02.
Article in English | MEDLINE | ID: mdl-38129199

ABSTRACT

AIMS: The combination of 5-fluorouracil/leucovorin (5-FU/LV) plus oxaliplatin (FOLFOX) is widely acknowledged as the standard regimen for second-line treatment in patients with advanced biliary tract cancer. Nanoliposomal irinotecan (nal-IRI) has demonstrated its activity in patients with advanced pancreatic cancer. Recent studies have investigated the activity of nal-IRI in combination with 5-FU/LV for biliary tract cancer. However, the results have been contradictory. We conducted a meta-analysis to assess survival outcomes and response rates in randomised trials investigating the activity of nal-IRI in previously treated biliary tract cancer patients. MATERIALS AND METHODS: We systematically collected potentially relevant findings from PubMed/Medline, the Cochrane library and EMBASE. Abstracts presented at major international oncological meetings were also reviewed. We extracted hazard ratios and 95% confidence intervals for progression-free survival and overall survival, as well as odds ratios and 95% confidence intervals for objective response rate. The outcomes of the accessible randomised studies evaluating the activity of nal-IRI plus 5-FU/LV were analysed. RESULTS: The combination therapy exhibited a statistically significant decrease in the risk of progression (hazard ratio 0.70; 95% confidence interval 0.50-0.97) when compared with 5-FU/LV alone. Additionally, the dual regimen yielded longer overall survival and a higher objective response rate. CONCLUSION: Our meta-analysis showed that nal-IRI plus 5-FU/LV had a superior activity in comparison with 5-FU/LV. Further investigations are required to elucidate the role of nal-IRI in this setting and to identify subgroups of patients who could derive the greatest benefit from its administration.


Subject(s)
Biliary Tract Neoplasms , Pancreatic Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biliary Tract Neoplasms/drug therapy , Fluorouracil/therapeutic use , Irinotecan , Leucovorin/adverse effects , Liposomes/therapeutic use , Pancreatic Neoplasms/drug therapy
3.
ESMO Open ; 7(3): 100505, 2022 06.
Article in English | MEDLINE | ID: mdl-35696744

ABSTRACT

The incidence of cholangiocarcinoma (CCA) has steadily increased during the past 20 years, and mortality is increasing. The majority of patients with CCA have advanced or metastatic disease at diagnosis, and treatment options for unresectable disease are limited, resulting in poor prognosis. However, recent identification of targetable genomic alterations has expanded treatment options for eligible patients. Given the importance of early and accurate diagnosis in optimizing patient outcomes, this review discusses best practices in CCA diagnosis, with a focus on categorizing molecular genetics and available targeted therapies. Imaging and staging of CCAs are discussed, as well as recommended biopsy collection techniques, and molecular and genomic profiling methodologies, which have become increasingly important as molecular biomarker data accumulate. Approved agents targeting actionable genomic alterations specifically in patients with CCA include ivosidenib for tumors harboring IDH1 mutations, and infigratinib and pemigatinib for those with FGFR2 fusions. Other agents currently under development in this indication have shown promising results, which are presented here.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Humans , Molecular Biology
4.
ESMO Open ; 6(1): 100032, 2021 02.
Article in English | MEDLINE | ID: mdl-33399070

ABSTRACT

OBJECTIVE: Germline BRCA1-2 pathogenic variants (gBRCApv) increase the risk of pancreatic cancer and predict for response to platinating agents and poly(ADP-ribose) polymerase inhibitors. Data on worldwide gBRCApv incidence among pancreatic ductal adenocarcinoma (PDAC) patients are sparse and describe a remarkable geographic heterogeneity. The aim of this study is to analyze the epidemiology of gBRCApv in Italian patients. MATERIALS AND METHODS: Patients of any age with pancreatic adenocarcinoma, screened within 3 months from diagnosis for gBRCApv in Italian oncologic centers systematically performing tests without any selection. For the purposes of our analysis, breast, ovarian, pancreas, and prostate cancer in a patient's family history was considered as potentially BRCA-associated. Patients or disease characteristics were examined using the χ2 test or Fisher's exact test for qualitative variables and the Student's t-test or Mann-Whitney test for continuous variables, as appropriate. RESULTS: Between June 2015 and May 2020, 939 patients were tested by 14 Italian centers; 492 (52%) males, median age 62 years (range 28-87), 569 (61%) metastatic, 273 (29%) with a family history of potentially BRCA-associated cancers. gBRCA1-2pv were found in 76 patients (8.1%; 9.1% in metastatic; 6.4% in non-metastatic). The gBRCA2/gBRCA1 ratio was 5.4 : 1. Patients with gBRCApv were younger compared with wild-type (59 versus 62 years, P = 0.01). The gBRCApv rate was 17.1% among patients <40 years old, 10.4% among patients 41-50 years old, 9.2% among patients 51-60 years old, 6.7% among patients aged 61-70 years, and 6.2% among patients >70 years old (none out of 94 patients >73 years old). gBRCApv frequency in 845 patients <74 years old was 9%. Patients with/without a family history of potentially BRCA-associated tumors had 14%/6% mutations. CONCLUSION: Based on our findings of a gBRCApv incidence higher than expected in a real-life series of Italian patients with incident PDAC, we recommend screening all PDAC patients <74 years old, regardless of family history and stage, due to the therapeutic implications and cancer risk prevention in patients' relatives.


Subject(s)
Adenocarcinoma , BRCA1 Protein , BRCA2 Protein , Pancreatic Neoplasms , Adenocarcinoma/epidemiology , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Female , Germ-Line Mutation , Humans , Italy/epidemiology , Male , Middle Aged , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/genetics
5.
Int J Obes (Lond) ; 42(3): 334-343, 2018 03.
Article in English | MEDLINE | ID: mdl-29151594

ABSTRACT

BACKGROUND: Solid epidemiological evidences connect obesity with incidence, stage and survival in pancreatic cancer. However, the underlying mechanistic basis linking adipocytes to pancreatic cancer progression remain largely elusive. We hypothesized that factors secreted by adipocytes could be responsible for epithelial-to-mesenchymal transition (EMT) induction and, in turn, a more aggressive phenotype in models of pancreatic preneoplastic lesions. METHODS: We studied the role of factors secreted by two adipogenic model systems from primary human bone marrow stromal cells (hBMSCs) in an in vitro experimental cell transformation model system of human pancreatic ductal epithelial (HPDE) cell stably expressing activated KRAS (HPDE/KRAS),Results:We measured a significant induction of EMT and aggressiveness in HPDE and HPDE/KRAS cell lines when cultured with medium conditioned by fully differentiated adipocytes (ADIPOCM) if compared with the same cells cultured with medium conditioned by hBMSC (hBMSCCM) from two different healthy donors. Several genes coding for soluble modulators of the non-canonical WNT signaling pathway, including FRZB, SFRP2, RSPO1, WNT5A and 5B were significantly overexpressed in fully differentiated adipocytes than in their respective in hBMSC. ADIPOCM induced the overexpression and the nuclear translocation of the Frizzled family member receptor tyrosine kinase-like orphan receptor (Ror) 2 in HPDE and HPDE/KRAS cells. Vantictumab, an anti-Frizzled monoclonal antibody, reduced ROR2 nuclear translocation and in turn the EMT and aggressiveness in HPDE and HPDE/KRAS cells. CONCLUSIONS: We demonstrated that adipocytes could induce EMT and aggressiveness in models of pancreatic preneoplastic lesions by orchestrating a complex paracrine signaling of soluble modulators of the non-canonical WNT signaling pathway that determine, in turn, the activation and nuclear translocation of ROR2. This signaling pathway could represent a novel target for pancreatic cancer chemoprevention. Most importantly, these factors could serve as novel biomarkers to select a risk population among obese subjects for screening and, thus, early diagnosis of pancreatic cancer.


Subject(s)
Adipocytes/cytology , Pancreatic Neoplasms/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Epithelial-Mesenchymal Transition/genetics , Humans , Mesenchymal Stem Cells , Models, Biological , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction/genetics , Wnt Proteins/genetics
6.
Int J Cancer ; 139(12): 2859-2864, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27578417

ABSTRACT

Mechanisms of acquired resistance to trastuzumab-based treatment in gastric cancer are largely unknown. In this study, we analyzed 22 pairs of tumor samples taken at baseline and post-progression in patients receiving chemotherapy and trastuzumab for advanced HER2-positive [immunohistochemistry (IHC) 3+ or 2+ with in-situ hybridization (ISH) amplification] gastric or gastroesophageal cancers. Strict clinical criteria for defining acquired trastuzumab resistance were adopted. Loss of HER2 positivity and loss of HER2 over-expression were defined as post-trastuzumab IHC score <3+ and absence of ISH amplification, and IHC "downscoring" from 2+/3+ to 0/1+, respectively. HER2 IHC was always performed, while ISH was missing in 3 post-progression samples. Patients with initial HER2 IHC score 3+ and 2+ were 14 (64%) and 8 (36%), respectively. Loss of HER2 positivity and HER2 over-expression was observed in 32 and 32% samples, respectively. The chance of HER2 loss was not associated with any of the baseline clinicopathological variables. The only exception was in patients with initial IHC score 2+ versus 3+, for both endpoints of HER2 positivity (80 vs. 14%; p = 0.008) and HER2 over-expression (63 vs. 14%; p = 0.025). As already shown in breast cancer, loss of HER2 may be observed also in gastric cancers patients treated with trastuzumab-based chemotherapy in the clinical practice. This phenomenon may be one of the biological reasons explaining the failure of anti-HER2 second-line strategies in initially HER2-positive disease.


Subject(s)
Esophageal Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Stomach Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Female , Humans , Immunohistochemistry , Male , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Staging , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Trastuzumab/therapeutic use , Treatment Outcome
7.
Br J Cancer ; 113(6): 878-85, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26291056

ABSTRACT

BACKGROUND: About 20% of resectable oesophageal carcinoma is resistant to preoperative chemoradiotherapy. Here we hypothesised that the expression of the antiapoptotic gene Baculoviral inhibitor of apoptosis repeat containing (BIRC)3 induced by the transforming growth factor ß activated kinase 1 (TAK1) might be responsible for the resistance to the proapoptotic effect of chemoradiotherapy in oesophageal carcinoma. METHODS: TAK1 kinase activity was inhibited in FLO-1 and KYAE-1 oesophageal adenocarcinoma cells using (5Z)-7-oxozeaenol. The BIRC3 mRNA expression was measured by qRT-PCR in 65 pretreatment frozen biopsies from patients receiving preoperatively docetaxel, cisplatin, 5-fluorouracil, and concurrent radiotherapy. Receiver operator characteristic (ROC) analyses were performed to determine the performance of BIRC3 expression levels in distinguishing patients with sensitive or resistant carcinoma. RESULTS: In vitro, (5Z)-7-oxozeaenol significantly reduced BIRC3 expression in FLO-1 and KYAE-1 cells. Exposure to chemotherapeutic agents or radiotherapy plus (5Z)-7-oxozeaenol resulted in a strong synergistic antiapoptotic effect. In patients, median expression of BIRC3 was significantly (P<0.0001) higher in adenocarcinoma than in the more sensitive squamous cell carcinoma subtype. The BIRC3 expression significantly discriminated patients with sensitive or resistant adenocarcinoma (AUC-ROC=0.7773 and 0.8074 by size-based pathological response or Mandard's tumour regression grade classifications, respectively). CONCLUSIONS: The BIRC3 expression might be a valid biomarker for predicting patients with oesophageal adenocarcinoma that could most likely benefit from preoperative chemoradiotherapy.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy/methods , Esophageal Neoplasms/therapy , Inhibitor of Apoptosis Proteins/metabolism , MAP Kinase Kinase Kinases/physiology , Neoplasm Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Zearalenone/analogs & derivatives , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Baculoviral IAP Repeat-Containing 3 Protein , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cisplatin/administration & dosage , Docetaxel , Down-Regulation , Drug Resistance, Neoplasm , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagogastric Junction , Female , Fluorouracil/administration & dosage , Humans , In Vitro Techniques , Inhibitor of Apoptosis Proteins/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Middle Aged , Neoplasm Proteins/genetics , RNA, Messenger/metabolism , ROC Curve , Radiation Tolerance , Taxoids/administration & dosage , Ubiquitin-Protein Ligases/genetics , Zearalenone/pharmacology
8.
Behav Brain Res ; 210(1): 99-106, 2010 Jun 26.
Article in English | MEDLINE | ID: mdl-20156489

ABSTRACT

The psychostimulant methylphenidate and the non-stimulant atomoxetine are two approved drugs for attention-deficit hyperactivity disorder (ADHD) therapy. The aim of this study was to investigate the long-term effects of prepuberal subchronic methylphenidate and atomoxetine on adult behaviour and the forebrain neurotransmitter and metabolite content of Naples High-Excitability (NHE) rats, a genetic model for the mesocortical variant of ADHD. Male NHE rats were given a daily intraperitoneal injection (1.0mg/kg) of methylphenidate, atomoxetine or vehicle from postnatal day 29 to 42. At postnatal day 70-75, rats were exposed to spatial novelty in the Làt and radial (Olton) mazes. Behavioural analysis for indices of horizontal, vertical, non-selective (NSA) and selective spatial attention (SSA) indicated that only methylphenidate significantly reduced horizontal activity to a different extent, i.e., 39 and 16% respectively. Moreover methylphenidate increased NSA as assessed by higher leaning duration. The high-performance liquid chromatography (HPLC) tissue content assessment of dopamine, norepinephrine, serotonin and relative metabolites in the prefrontal cortex (PFC), cortical motor area (MC), dorsal striatum (DS), ventral striatum (VS), hippocampus and mesencephalon indicated that methylphenidate decreased (i) dopamine, DOPAC, norepinephrine, MHPG, 5-HT and 5-HIAA in the PFC, (ii) dopamine, DOPAC, HVA, serotonin, 5-HIAA in the DS, (iii) dopamine, DOPAC, HVA and MHPG (but increased norepinephrine) in the VS and (iv) norepinephrine, MHPG, serotonin and 5-HIAA in the hippocampus. Atomoxetine increased dopamine and decreased MHPG in the PFC. Like methylphenidate, atomoxetine decreased dopamine, DOPAC, HVA, serotonin and 5-HIAA in the DS, but decreased MHPG in the VS. These results suggest that methylphenidate determined long-term effects on behavioural and neurochemical parameters, whereas atomoxetine affected only the latter.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Attention Deficit Disorder with Hyperactivity/drug therapy , Central Nervous System Stimulants/pharmacology , Methylphenidate/pharmacology , Propylamines/pharmacology , Prosencephalon/drug effects , Adrenergic Uptake Inhibitors/administration & dosage , Aging , Animals , Atomoxetine Hydrochloride , Attention/drug effects , Attention Deficit Disorder with Hyperactivity/metabolism , Central Nervous System Stimulants/administration & dosage , Disease Models, Animal , Dopamine/metabolism , Male , Maze Learning/drug effects , Methylphenidate/administration & dosage , Norepinephrine/metabolism , Propylamines/administration & dosage , Prosencephalon/metabolism , Random Allocation , Rats , Serotonin/metabolism , Time Factors
9.
Behav Brain Res ; 198(1): 37-44, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-19041899

ABSTRACT

The excitatory amino acids (EAA) L-glutamate (L-Glu), L-aspartate (L-Asp) and D-aspartate (D-Asp) are thought to play a neurotransmitter/neuromodulator role in neuronal communications. Recently, a high level of EAA L-Glu, D- and L-Asp isomers has been found in the forebrain of Naples high-excitability (NHE) rat line that models the mesocortical variant of Attention-Deficit Hyperactivity Disorder (ADHD). The aim of this study was to assess the functions of D-Asp using two forms, i.e. free D-Asp or D-Asp diethyl ester (DEE) as prodrug, on brain and behaviour. Thus, prepuberal rats were given, for two weeks daily, an i.p. injection of D-Asp or DEE or vehicle. Then rats were exposed to two spatial novelties i.e. Làt and radial Olton maze. Behaviour was monitored for indices of activity, non-selective attention (NSA), selective spatial attention (SSA) and emotional reactivity. L-Glu and D- and L-Asp were detected by HPLC in cognitive and non-cognitive brain areas such as prefrontal cortex, striatum, hippocampus and hypothalamus. Results indicate that subchronic D-Asp or DEE (i) reduced EAA levels in the NHE and increased it in the random-bred controls (NRB) rats, (ii) in the Làt-maze D-Asp increased horizontal activity in NHE but DEE decreased it in NRB rats, (iii) in the Olton maze D-Asp and DEE decreased vertical activity in NHE and NRB rats respectively, (iv) D-Asp impaired attention only in NRB decreasing number of arms visited before first repetition. Therefore, data demonstrate differential effects of prepuberal subchronic D-Asp and DEE that may be related to different basal EAA levels in NHE and NRB rats.


Subject(s)
Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Attention Deficit Disorder with Hyperactivity/metabolism , Behavior, Animal , Excitatory Amino Acids/metabolism , Prodrugs/pharmacology , Prosencephalon/metabolism , Animals , Aspartic Acid/administration & dosage , Attention/drug effects , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/physiopathology , Behavior, Animal/drug effects , Chromatography, High Pressure Liquid , Corpus Striatum/metabolism , Disease Models, Animal , Emotions/drug effects , Emotions/physiology , Exploratory Behavior/drug effects , Glutamic Acid/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Injections, Subcutaneous , Maze Learning/drug effects , Motor Activity/drug effects , Prefrontal Cortex/metabolism , Prodrugs/administration & dosage , Prodrugs/metabolism , Prosencephalon/drug effects , Rats , Rats, Inbred Strains , Space Perception/drug effects , Treatment Outcome
10.
Neuroscience ; 152(1): 234-44, 2008 Mar 03.
Article in English | MEDLINE | ID: mdl-18164552

ABSTRACT

Pathological conditions, such as Parkinson's disease and attention deficit hyperactivity disorder, have been linked to alterations of specific dopamine (DA) pathways. However, since exogenous DA does not cross the blood-brain barrier, DA levels can be modulated e.g. by DA precursors or DA reuptake blockers. Hereby histochemical, analytical and behavioral evidence shows that a galactosylated form of DA (GAL-DA) carries DA into the brain, thus modulating activity and nonselective attention in rats. To this aim adult male rats of the Naples high-excitability (NHE) and random bred controls (NRB) lines were given a single i.p. injection of GAL-DA (10 or 100 mg/kg). Three hours later the behavior was videotaped and analyzed for horizontal activity, orienting frequency and scanning duration. The dose of 100 mglkg of GAL-DA reduced by 25% the horizontal activity in NHE rats, mainly in the first part of the testing period. No effect was observed on orienting frequency or on scanning duration. However, GAL-DA 100 mg/kg was associated with longer rearing episodes in the second part of the testing period in NHE rats. In parallel experiments histochemistry with a galactose-specific lectin showed 10% increase in galactose residues into the striatum between 0.5 and 3.0 h. To quantify the level of GAL-DA, its metabolite DA-succinate and DA in the prefrontal cortex, neostriatum, and cerebellum, rats were killed 2.0 h after the injection of prodrug. Mass high performance liquid chromatography (HPLC) was used for analysis of GAL-DA and DA succinate whereas electrochemical HPLC for DA. Both HPLC techniques demonstrate that GAL-DA carries and releases DA into the brain. Specifically 100 mg/kg of GAL-DA increased DA level in the striatum in the NHE rats only. Moreover, DA in the mesencephalon (MES) was correlated positively with striatal and prefrontal cortex DA in NHE rats. In contrast DA in the MES was negatively correlated with striatal DA in NRB. GAL-DA disrupted these correlations in both rat lines. Thus, this new DA prodrug may modify DA neurotransmission and might have a potential clinical application.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Dopamine/metabolism , Dopamine/pharmacology , Galactose/metabolism , Prodrugs/metabolism , Animals , Brain/metabolism , Brain Chemistry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Galactose/pharmacology , Image Processing, Computer-Assisted , Male , Rats
11.
J Biomed Mater Res A ; 87(4): 843-9, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18200553

ABSTRACT

Controlled and local drug-delivery systems for anti-inflammatory agents are drawing increasing attention for possible pharmaceutical and biomedical applications, because of their extended therapeutic effect and reduced side effects. A single-step sol-gel process was used to precipitate silica microspheres containing Ketoprofen, Indomethacin, Ketorolac tris salt, or Triamcinolone acetonide, for controlled drug delivery applications. The amorphous nature of the gels was ascertained by X-ray diffraction analysis. Release kinetics in a simulated body fluid (SBF) has been subsequently investigated. The amount of drug released has been detected by UV-vis spectroscopy. The pure anti-inflammatory agent exhibited linear release with time, while sol-gel silica-entrapped drugs were released with a logarithmic time dependence, starting with an initial burst effect followed by a gradual decrease. Finally, SEM micrography and EDS analysis showed the formation of a hydroxyapatite layer on the surface of the samples soaked in SBF. All the materials showed good release and therefore could be used as drug-delivery systems.


Subject(s)
Anti-Inflammatory Agents , Drug Carriers , Drug Delivery Systems , Gels , Microspheres , Silicon Dioxide , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Body Fluids/metabolism , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Durapatite/chemistry , Gels/chemistry , Gels/pharmacokinetics , Humans , Materials Testing , Microscopy, Electron, Scanning , Molecular Weight , Particle Size , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , X-Ray Diffraction
12.
Behav Brain Res ; 187(2): 449-54, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18063140

ABSTRACT

Different strategies can be used to carry dopamine into the brain such as L-Dopa precursors or galactosilated form of DA (GAL-DA). The aim of this study was to investigate whether GAL-DA would reduce hyperactivity and increase non-selective attention (NSA) in a mouse model of attention deficit hyperactivity disorder (ADHD), as, i.e. C57BL/6 as did in NHE rats. Here we report that GAL-DA increases NSA in a spatial novelty in C57BL/6 mice. They received a single i.p. injection of GAL-DA (10 mg/kg or 100 mg/kg) or equimolar galactose vehicle. Another mouse strain the Swiss albino was introduced as inbred control group. Three hours after last injection mice were tested in a Làt-maze for 30-min. Behaviour was analyzed for horizontal (traveled distance) and vertical activity (orienting frequency and scanning durations) which shares cognitive and non-cognitive nature, respectively. Ten milligram per kilograms of GAL-DA, increases scanning duration in C57BL/6 mice. Thus a low dose of GAL-DA increases NSA without reducing hyperactivity in this mouse model of ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/metabolism , Attention/physiology , Dopamine/metabolism , Galactose/metabolism , Maze Learning/physiology , Motor Activity/physiology , Analysis of Variance , Animals , Attention Deficit Disorder with Hyperactivity/drug therapy , Disease Models, Animal , Dopamine/administration & dosage , Dose-Response Relationship, Drug , Drug Carriers , Female , Galactose/administration & dosage , Galactose/analogs & derivatives , Male , Mice , Mice, Inbred C57BL , Species Specificity , Statistics, Nonparametric
13.
J Mater Sci Mater Med ; 17(5): 413-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16688581

ABSTRACT

Local drug delivery of antimicrobics by sustained release delivery system can be used to treat periodontal disease. Advantages of these systems may include maintaining high levels of antibiotic in the gingival crevicular fluid for a sustained period of time and ease of use with high patient acceptance. The materials used are TiO(2) and TiO(2)4SiO(2), mixed with sodium ampicillin, a broad-spectrum antibiotic, have been synthesized by sol-gel method. The amorphous nature of the gels was ascertained by X-ray diffraction analysis. Release kinetics in a simulated body fluid (SBF) have been subsequently investigated. The amount of sodium ampicillin released has been detected by UV-VIS spectroscopy and SEM. The release kinetics seems to occur in more than one stage. HPLC analysis has also been taken to ensure the integrity of ampicillin after the synthetic treatment. Finally, SEM micrographs and EDS analysis showed the formation of a hydroxyapatite layer on the surface of the samples soaked in SBF. Both the materials showed good release and could be used as drug delivery bioactive systems. High antimicrobial effects of samples against Escherichia coli and Streptococcus mutants were found.


Subject(s)
Silicon Dioxide/chemistry , Titanium/chemistry , Ampicillin/chemistry , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Durapatite/chemistry , Escherichia coli/metabolism , Gingiva/metabolism , In Vitro Techniques , Kinetics , Materials Testing/methods , Microscopy, Electron, Scanning , Phase Transition , Streptococcus/metabolism , Surface Properties , X-Ray Diffraction
14.
Endocr Relat Cancer ; 11(1): 51-68, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15027885

ABSTRACT

The currently available treatment of cancer patients is based on the use of cytotoxic drugs and/or of ionizing radiations, which have potent antitumor activity, but also cause clinically relevant side effects, since they affect cellular targets that are common to both cancer cells and normal proliferating cells. In the past 20 years, the discoveries on the molecular mechanisms of cancer development and progression have prompted the search for agents which are more selective for cancer cell molecular targets. The possibility of combining conventional cytotoxic drugs with novel agents that specifically interfere with key pathways controlling cancer cell survival, proliferation, invasion and/or metastatic spreading has generated a wide interest. This could be a promising therapeutic approach for several reasons. First, since the cellular targets for these agents and their mechanism(s) of action are different from those of cytotoxic drugs, it is possible for their combination with chemotherapy without cross-resistance. Second, alterations in the expression and/or the activity of genes that regulate mitogenic signals not only can directly cause perturbation of cell growth, but also may affect the sensitivity of cancer cells to conventional chemotherapy and radiotherapy. In this review, we will discuss the biologic bases of the combination of molecular targeted drugs with conventional medical cancer treatments and the available results of the first series of clinical trials in cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Signal Transduction/drug effects , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Combined Modality Therapy , ErbB Receptors/antagonists & inhibitors , Farnesyltranstransferase , Genes, bcl-2/drug effects , Humans , Neoplasms/therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Protein Kinase C/antagonists & inhibitors , Thionucleotides/pharmacology , ras Proteins/antagonists & inhibitors
15.
Clin Cancer Res ; 7(12): 4156-63, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751516

ABSTRACT

PURPOSE: Protein kinase A type I (PKAI) and the epidermal growth factor receptor (EGFR) play a role in neoplastic transformation and interact with each other in transducing mitogenic signals. We developed different PKAI and EGFR inhibitors, demonstrating their cooperation with cytotoxic drugs and the therapeutic potential of the combined blockade of PKAI and EGFR. In this study, we investigated the effect of orally active PKAI and EGFR inhibitors in combination with a novel taxane. EXPERIMENTAL DESIGN: We combined a hybrid PKAI antisense oligonucleotide sequence (AS-PKAI), the EGFR inhibitor ZD1839 (Iressa), and the taxane IDN5109, studying their effect on human cancer growth, apoptosis, and angiogenesis and measuring vascular endothelial growth factor (VEGF) expression and vessel formation in vitro and after oral administration in nude mice. RESULTS: We demonstrated cooperative growth inhibitory and proapoptotic effects and inhibition of VEGF expression with any combination of two drugs and a marked synergistic effect when all three agents were combined. Oral administration of AS-PKAI, ZD1839, and IDN5109 in combination to nude mice caused a remarkable antitumor effect with no histological evidence of tumors in 50% of mice 5 weeks after treatment withdrawal, accompanied by complete suppression of vessel formation and VEGF expression. CONCLUSION: This is the first demonstration of the cooperative antitumor and antiangiogenic activity of three novel agents that block multiple signaling pathways after oral administration. Because all agents are under clinical evaluation in cancer patients, our results provide a rationale to translate this feasible therapeutic strategy in a clinical setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Cyclic AMP-Dependent Protein Kinases/genetics , Oligodeoxyribonucleotides, Antisense/therapeutic use , Ovarian Neoplasms/drug therapy , Quinazolines/therapeutic use , Administration, Oral , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Endothelial Growth Factors/genetics , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Lymphokines/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Oligodeoxyribonucleotides, Antisense/administration & dosage , Ovarian Neoplasms/pathology , Quinazolines/administration & dosage , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...