Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Ann Oncol ; 28(6): 1346-1351, 2017 06 01.
Article En | MEDLINE | ID: mdl-28383677

Background: Therapeutic options for patients with chemoresistant germ cell tumors (GCTs) are limited. Pazopanib is a selective tyrosine kinase inhibitor with distinct antiangiogenic activity. We aimed to evaluate pazopanib activity in patients with refractory GCT. Patients and methods: In the open-label, single-arm, phase 2 Pazotest study (NCT01743482), patient eligibility included failure of ≥2 platinum-based regimens, and allowed prior high-dose chemotherapy administration. Patients were given pazopanib 800 mg/day until disease progression (PD) or onset of unacceptable toxicity. Measurements of serum tumor markers (STM), computed tomography and FDG-PET were carried out at baseline, after 4 weeks of pazopanib treatment, and every 8 weeks thereafter. PD was defined as increasing levels of STM, increasing size of non-teratomatous masses, or appearance of new lesions. The study primary endpoint was progression-free survival (PFS, H0: 3-month PFS ≤ 10%, H1: ≥25%, α = 5%, ß = 20%). Results: Forty-three patients were enrolled from May 2013 to July 2016. The number of prior chemotherapy regimens was: 2 (11.6%), 3 (51.2%), >3 (37.2%). Grade 3 adverse events were observed in six patients (13.9%). Overall, 70.3% of patients had reduced levels of STM after 4 weeks. There were 2 partial responses (4.7%), 19 cases of stable disease, and 16 cases of PD (6 not evaluable by RECIST). The median follow-up duration was 29.6 months. The 3-month PFS probability was 12.8% [95% confidence interval (CI): 5.7%-28.9%]. The 24-month OS probability was 14.2% (95% CI: 6.0%-33.7%). In patients with a >50% decline in STM, the 24-month OS probability was 24.1% (95% CI: 8.3%-69.6%). The small sample size was the major limitation. Conclusions: Despite pazopanib showed potent but short-lived activity in refractory GCT, long-term survival was obtained in a proportion of treated patients. According to the kinetics of pazopanib activity, this drug may be investigated in less pre-treated patients as an optimal bridging therapy preceding and/or combined with salvage chemotherapy.


Angiogenesis Inhibitors/therapeutic use , Neoplasms, Germ Cell and Embryonal/drug therapy , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Testicular Neoplasms/drug therapy , Adult , Disease Progression , Humans , Indazoles , Male , Treatment Outcome
2.
Invest New Drugs ; 32(3): 555-60, 2014 Jun.
Article En | MEDLINE | ID: mdl-24566706

Despite a compelling preclinical rationale for the use of anti-angiogenic drugs in urothelial cancer (UC), short-living responses have been observed in clinical trials. PF-03446962 is a novel monoclonal antibody against Activin Receptor-Like Kinase-1 (ALK1), a type I subclass of the TGFß receptor, with dose-dependent anti-angiogenic activity. An open label, single-group, phase 2 trial of PF-03446962 was conducted in salvage setting. Patients failing at least one chemotherapy regimen were eligible. Design provided PF-03446962 10 mg/Kg intravenously fortnightly until disease progression (PD) or unacceptable toxicity. Two-month progression-free survival (PFS) was the primary endpoint. The trial was registered with ClinicalTrials.gov, number NCT01620970. Fourteen patients were enrolled from October 2012 to July 2013. Median age was 64 years (interquartile range [IQR]: 58.2-69.5), 9 patients had a Bellmunt score of 1-2, median number of prior drugs was 3. One stable disease and 13 PD were recorded and the study met the futility stopping rule of interim analysis. Median PFS was 1.8 months (95 %CI, 1.4-2.0). After a median follow up of 7.4 months (IQR 4.5-10.9), 8 patients are alive. Median overall survival (OS) was 8 months (95 %CI, 2.9-not estimable). Most common toxicities were thrombocytopenia (G1-2 in 5 cases, persistent G3 in one, with 3 dose delays and 1 dose interruption), fatigue and abdominal pain (G1-2 in 4 cases each). Impairment of quality of life (ESAS score) was observed as well as an increase from baseline to +2 month median levels of vascular endothelial growth factor (VEGF) and interleukin-8. PF-03446962 had no activity as single drug in refractory UC and we do not recommend further investigation outside of the combination with agents targeting the VEGF receptor axis.


Activin Receptors, Type II/antagonists & inhibitors , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Urologic Neoplasms/drug therapy , Activin Receptors, Type II/immunology , Aged , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Female , Humans , Male , Middle Aged
3.
Curr Med Chem ; 21(14): 1654-66, 2014.
Article En | MEDLINE | ID: mdl-24180279

A large number of indolyl-4-azaindolyl thiazoles, nortopsentin analogues, were conveniently synthesized. The antiproliferative activity of the new derivatives was examined against four human tumor cell lines with different histologic origin. Seven derivatives consistently reduced the growth of the experimental models independently of TP53 gene status and exhibited the highest activity against the malignant peritoneal mesothelioma (STO) cell line. The most active compound of this series acts as a CDK1 inhibitor, and was found to cause cell cycle arrest at G2/M phase, to induce apoptosis by preventing the phosphorylation of survivin in Thr(34) and to increase the cytotoxic activity of paclitaxel in STO cells.


Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Pyridines/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line , Humans , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyridines/chemical synthesis , Structure-Activity Relationship
4.
Br J Cancer ; 110(1): 26-33, 2014 Jan 07.
Article En | MEDLINE | ID: mdl-24231947

BACKGROUND: Pazopanib achieved the end point of clinical activity in pretreated patients with urothelial cancer in a single-group, phase 2 trial. The objective was to identify biological predictors of clinical benefit to pazopanib in these patients. METHODS: EDTA blood samples were collected at baseline (T0) and after 4 weeks (T1) of treatment, together with radiological imaging in all 41 patients to analyse plasma circulating angiogenic factor levels by multiplex ELISA plates. Changes from T0 to T1 in marker levels were matched with response with the covariance analysis. Univariable and multivariable analyses evaluated the association with overall survival (OS), adjusted for prespecified clinical variables. Net reclassification improvement (NRI) tested the performance of the recognised Cox model. RESULTS: Increasing IL8(T1) level associated with lower response probability at covariance analysis (P=0.010). Both IL8(T0) (P=0.019) and IL8(T1) (P=0.004) associated with OS and the prognostic model, including clinical variables and IL8(T1) best-predicted OS after backward selection. The NRI for this model was 39%.When analysed as a time-varying covariate, IL8(T1) level<80 pg ml(-1) portended significantly greater response (∼80%) and 6-month OS (∼60%) probability than level ≥ 80. CONCLUSION: IL8-level changes during pazopanib allowed for a prognostic improvement and were associated with response probability.


Angiogenesis Inducing Agents/blood , Cytokines/blood , Interleukin-8/blood , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Urologic Neoplasms/blood , Urologic Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Transitional Cell/blood , Carcinoma, Transitional Cell/drug therapy , Enzyme-Linked Immunosorbent Assay , Humans , Indazoles , Multimodal Imaging , Positron-Emission Tomography , Prognosis , Proportional Hazards Models , Tomography, X-Ray Computed
5.
J Pharmacol Exp Ther ; 348(3): 360-71, 2014 Mar.
Article En | MEDLINE | ID: mdl-24345465

Since response to platinum-based therapy in non-small-cell lung cancer (NSCLC) is poor, the present study was designed to rationally identify novel drug combinations in cell models including the A549 cell line and the cisplatin-resistant subline A549/Pt, characterized by reduced sensitivity to cisplatin-induced apoptosis and by upregulation of efflux transporters of the ATP binding cassette (ABC) superfamily. Given the molecular features of these cells, we focused on compounds triggering apoptosis through different mechanisms, such as the mitochondria-targeting drug arsenic trioxide and the phenanthridine analog sanguinarine, which induce apoptosis through the extrinsic pathway. Sanguinarine, not recognized by ABC transporters, could overcome cisplatin resistance and, when used in combination with arsenic trioxide, was synergistic in A549 and A549/Pt cells. The arsenic trioxide/sanguinarine cotreatment upregulated genes implicated in apoptosis activation through the extrinsic pathway. Drug combination experiments indicated that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment improved arsenic trioxide/sanguinarine efficacy, a feature associated with a striking apoptosis induction, particularly in the cisplatin-resistant variant. Thus, a synergistic interaction between sanguinarine and arsenic trioxide could be obtained independent of relative cell sensitivity to arsenic trioxide, and an enhanced apoptosis induction could be achieved in combination with TRAIL through modulation of the extrinsic apoptotic pathway. Antitumor activity studies supported the interest of drug combinations including TRAIL in NSCLC, indicating that drug-resistant NSCLC cells can efficiently be killed by the combination of proapoptotic agents. Our results suggest that the molecular changes occurring in treated cells may be exploited to rationally hit surviving cells.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Benzophenanthridines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Isoquinolines/pharmacology , Lung Neoplasms/drug therapy , Oxides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Arsenic Trioxide , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Drug Resistance, Neoplasm , Drug Synergism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
6.
Br J Cancer ; 100(5): 739-46, 2009 Mar 10.
Article En | MEDLINE | ID: mdl-19223905

We analysed the effects of small interfering RNA (siRNA)-mediated silencing of Apollon, a member of the inhibitors of apoptosis protein family, on the proliferative potential and ability of human breast cancer cell lines to undergo apoptosis. In wild-type p53 ZR75.1 cells, Apollon knockdown resulted in a marked, time-dependent decline of cell growth and an increased rate of apoptosis, which was associated with p53 stabilisation and activation of the mitochondrial-dependent apoptotic pathway. Pre-incubation of cells with a p53-specific siRNA resulted in a partial rescue of cell growth inhibition, as well as in a marked reduction of the apoptotic response, indicating p53 as a major player in cell growth impairment consequent on Apollon silencing. Apollon knockdown induced consistently less pronounced anti-proliferative and pro-apoptotic effects in mutant p53 MDA-MB-231 cells than in ZR75.1 cells. Furthermore, the activation of caspase-3 seemed to be essential for the induction of apoptosis after Apollon knockdown, as the Apollon-specific siRNA had no effect on the viability of caspase-3-deficient, wild-type p53 MCF-7 cells or the ZR75.1 cells after RNA interference-mediated caspase-3 silencing. Our results indicate that p53 stabilisation and caspase-3 activation concur to determine the apoptotic response mediated by Apollon knockdown in breast cancer cells, and suggest Apollon to be a potential new therapeutic target for this malignancy.


Apoptosis/drug effects , Breast Neoplasms/therapy , Caspase 3/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Female , Gene Silencing/physiology , Humans , Inhibitor of Apoptosis Proteins/genetics , Models, Biological , Protein Stability/drug effects , RNA, Small Interfering/therapeutic use , Tumor Cells, Cultured
7.
Mini Rev Med Chem ; 3(1): 51-60, 2003 Feb.
Article En | MEDLINE | ID: mdl-12570854

The ability of peptide nucleic acids and hammerhead ribozymes, which target different subunits of human telomerase, to efficiently inhibit the enzyme's catalytic activity has been clearly demonstrated in several in vitro studies carried out in human immortalized and cancer cell lines. However, the actual efficacy of these molecules still needs to be validated in in vivo human tumor models, and such validation appears to be largely dependent on the development of reliable systems for their intracellular delivery.


Enzyme Inhibitors/pharmacology , Peptide Nucleic Acids/pharmacology , RNA, Catalytic/pharmacology , Telomerase/antagonists & inhibitors , Amino Acid Sequence , Base Sequence , DNA Primers , Humans
8.
Cell Mol Life Sci ; 59(8): 1406-12, 2002 Aug.
Article En | MEDLINE | ID: mdl-12363043

Stable transfection of human ovarian carcinoma cells with survivin cDNA caused a four- to sixfold increase in cell resistance to taxotere and taxol (two-sided Student's t test, p < 0.05), with a concomitant reduction in the apoptotic response to taxol, but did not affect cell sensitivity to cisplatin or oxaliplatin. Such findings were indirectly supported by similar observations obtained with clinical tumours. In fact, high levels of survivin protein expression (>30% positive cells), detected by immunohistochemistry in 90/124 (73%) advanced ovarian carcinomas, were significantly associated with clinical resistance to a taxol/platinum-based regimen but unrelated to tumour shrinkage following cisplatin-including combinations (non-taxol based). In the 95 patients receiving a taxol/platinum-based regimen, survivin overexpression correlated with a lower clinical or pathologic complete remission rate than absent/low protein expression (43 vs 75%, p = 0.0058 by logistic regression adjusted for tumour stage, histological grade and p53 expression). Conversely, in the 29 cases treated with cisplatin-containing regimens (not taxol based), survivin expression was unrelated to tumour response. Cellular studies and clinical data suggest a direct link between survivin expression and tumour cell susceptibility to taxol.


Antineoplastic Agents, Phytogenic/pharmacology , Chromosomal Proteins, Non-Histone/genetics , Drug Resistance, Neoplasm/genetics , Microtubule-Associated Proteins , Ovarian Neoplasms/genetics , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , Apoptosis/physiology , Chromosomal Proteins, Non-Histone/physiology , Cisplatin/pharmacology , Docetaxel , Female , Humans , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Mutation , Neoplasm Proteins , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Oxaliplatin , Phosphorylation/drug effects , Phosphotransferases/drug effects , Survivin , Transfection , Tumor Cells, Cultured
9.
Curr Med Chem Anticancer Agents ; 2(5): 605-12, 2002 Sep.
Article En | MEDLINE | ID: mdl-12678727

Human telomerase is a ribonucleoprotein enzyme complex that enables cells to maintain telomere length, allowing indefinite replicative capacity. The notion that telomerase is reactivated in 80-90% of human cancers has led to the proposal of telomerase as a promising therapeutic target for novel anticancer interventions. Due to its inherent accessibility to nucleic acids, telomerase appears an ideal target for strategies based on the use of antisense oligonucleotides and ribozymes that target its RNA template. In this review a summary of the different antisense- and ribozyme-based approaches used thus far to inhibit telomerase activity in human cancer cells is provided. All these strategies significantly inhibited the enzyme's catalytic activity in in vitro and in vivo tumor models. However, while in some studies tumor cell growth arrest was observed as a consequence of telomere shortening after prolonged telomerase inhibition, other studies have shown that antisense- and ribozyme-based treatments targeting telomerase induced rapid loss (i.e. within a few days) of tumor cell viability with concomitant apoptosis. In the latter case it is unlikely that cell death was related to telomere erosion since the cells would not have undergone enough divisions to significantly shorten their telomeres. A possible explanation is that telomerase inhibitors may induce apoptosis in cancer cells directly by interfering with the capping function of the enzyme. Overall, the available results indicate antisense oligonucleotides and ribozymes as good tools to inhibit telomerase and suggest that abrogation of telomerase activity may affect tumor cell proliferation also through pathways that are not dependent on telomere erosion.


Antineoplastic Agents/pharmacology , Oligonucleotides, Antisense/pharmacology , RNA, Catalytic/pharmacology , Telomerase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/therapeutic use , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/pharmacology , Peptide Nucleic Acids/therapeutic use , RNA, Catalytic/chemistry , RNA, Catalytic/therapeutic use , Structure-Activity Relationship
10.
Br J Cancer ; 84(10): 1387-90, 2001 May 18.
Article En | MEDLINE | ID: mdl-11355952

A collateral sensitivity or a very modest cross-resistance to BBR 3464 was found in 2 ovarian cancer cell lines with experimentally induced resistance to cisplatin. Loss of mismatch repair proteins (hMLH1, hPMS2) or overexpression of nucleotide excision repair proteins (ERCC1) was not detrimental for the cellular sensitivity to BBR 3464. Moreover, interesting differences in the kinetics of formation and removal of DNA lesions at the single-gene (N- ras) level were observed between BBR 3464 and CDDP.


Adenosine Triphosphatases , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Cisplatin/toxicity , DNA Repair Enzymes , DNA Repair/genetics , DNA-Binding Proteins , Drug Resistance, Neoplasm , Endonucleases , Organoplatinum Compounds/toxicity , Adaptor Proteins, Signal Transducing , Base Pair Mismatch , Carrier Proteins , DNA Repair/drug effects , Female , Humans , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Nuclear Proteins , Ovarian Neoplasms , Polymerase Chain Reaction , Proteins/genetics , Tumor Cells, Cultured
...