Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Endocr Relat Cancer ; 23(5): 411-8, 2016 05.
Article in English | MEDLINE | ID: mdl-27080472

ABSTRACT

Neuroendocrine tumors (NETs) are highly vascular neoplasms overexpressing vascular endothelial growth factor (VEGF) as well as VEGF receptors (VEGFR). Axitinib is a potent, selective inhibitor of VEGFR-1, -2 and -3, currently approved for the treatment of advanced renal cell carcinoma. We performed an open-label, two-stage design, phase II trial of axitinib 5mg twice daily in patients with progressive unresectable/metastatic low-to-intermediate grade carcinoid tumors. The primary end points were progression-free survival (PFS) and 12-month PFS rate. The secondary end points included time to treatment failure (TTF), overall survival (OS), overall radiographic response rate (ORR), biochemical response rate and safety. A total of 30 patients were enrolled and assessable for toxicity; 22 patients were assessable for response. After a median follow-up of 29months, we observed a median PFS of 26.7months (95% CI, 11.4-35.1), with a 12-month PFS rate of 74.5% (±10.2). The median OS was 45.3 months (95% CI, 24.4-45.3), and the median TTF was 9.6months (95% CI, 5.5-12). The best radiographic response was partial response (PR) in 1/30 (3%) and stable disease (SD) in 21/30 patients (70%); 8/30 patients (27%) were unevaluable due to early withdrawal due to toxicity. Hypertension was the most common toxicity that developed in 27 patients (90%). Grade 3/4 hypertension was recorded in 19 patients (63%), leading to treatment discontinuation in six patients (20%). Although axitinib appears to have an inhibitory effect on tumor growth in patients with advanced, progressive carcinoid tumors, the high rate of grade 3/4 hypertension may represent a potential impediment to its use in unselected patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Imidazoles/therapeutic use , Indazoles/therapeutic use , Neuroendocrine Tumors/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Axitinib , Colonic Neoplasms/drug therapy , Female , Humans , Hypertension/chemically induced , Imidazoles/adverse effects , Indazoles/adverse effects , Intestinal Neoplasms/drug therapy , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Rectal Neoplasms/drug therapy , Thymus Neoplasms/drug therapy , Treatment Outcome
3.
Oncogene ; 33(12): 1590-600, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-23542178

ABSTRACT

The mammalian target of rapamycin (mTOR) regulates cell growth by integrating nutrient and growth factor signaling and is strongly implicated in cancer. But mTOR is not an oncogene, and which tumors will be resistant or sensitive to new adenosine triphosphate (ATP) competitive mTOR inhibitors now in clinical trials remains unknown. We screened a panel of over 600 human cancer cell lines to identify markers of resistance and sensitivity to the mTOR inhibitor PP242. RAS and phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) mutations were the most significant genetic markers for resistance and sensitivity to PP242, respectively; colon origin was the most significant marker for resistance based on tissue type. Among colon cancer cell lines, those with KRAS mutations were most resistant to PP242, whereas those without KRAS mutations most sensitive. Surprisingly, cell lines with co-mutation of PIK3CA and KRAS had intermediate sensitivity. Immunoblot analysis of the signaling targets downstream of mTOR revealed that the degree of cellular growth inhibition induced by PP242 was correlated with inhibition of phosphorylation of the translational repressor eIF4E-binding protein 1 (4E-BP1), but not ribosomal protein S6 (rpS6). In a tumor growth inhibition trial of PP242 in patient-derived colon cancer xenografts, resistance to PP242-induced inhibition of 4E-BP1 phosphorylation and xenograft growth was again observed in KRAS mutant tumors without PIK3CA co-mutation, compared with KRAS wild-type controls. We show that, in the absence of PIK3CA co-mutation, KRAS mutations are associated with resistance to PP242 and that this is specifically linked to changes in the level of phosphorylation of 4E-BP1.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Binding, Competitive/drug effects , Carrier Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Phosphoproteins/metabolism , Purines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Colonic Neoplasms/pathology , Eukaryotic Initiation Factors , Female , Humans , MAP Kinase Signaling System/drug effects , Mice , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Ribosomal Protein S6/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , ras Proteins/genetics
4.
Br J Cancer ; 109(7): 1725-34, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24022191

ABSTRACT

BACKGROUND: This phase 1 clinical trial was conducted to determine the safety, maximum-tolerated dose (MTD), and pharmacokinetics of imatinib, bevacizumab, and metronomic cyclophosphamide in patients with advanced colorectal cancer (CRC). METHODS: Patients with refractory stage IV CRC were treated with bevacizumab 5 mg kg(-1) i.v. every 2 weeks (fixed dose) plus oral cyclophosphamide q.d. and imatinib q.d. or b.i.d. in 28-day cycles with 3+3 dose escalation. Response was assessed every two cycles. Pharmacokinetics of imatinib and cyclophosphamide and circulating tumour, endothelial, and immune cell subsets were measured. RESULTS: Thirty-five patients were enrolled. Maximum-tolerated doses were cyclophosphamide 50 mg q.d., imatinib 400 mg q.d., and bevacizumab 5 mg kg(-1) i.v. every 2 weeks. Dose-limiting toxicities (DLTs) included nausea/vomiting, neutropaenia, hyponatraemia, fistula, and haematuria. The DLT window required expansion to 42 days (1.5 cycles) to capture delayed toxicities. Imatinib exposure increased insignificantly after adding cyclophosphamide. Seven patients (20%) experienced stable disease for >6 months. Circulating tumour, endothelial, or immune cells were not associated with progression-free survival. CONCLUSION: The combination of metronomic cyclophosphamide, imatinib, and bevacizumab is safe and tolerable without significant drug interactions. A subset of patients experienced prolonged stable disease independent of dose level.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Colorectal Neoplasms/drug therapy , Cyclophosphamide/therapeutic use , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/adverse effects , Benzamides/pharmacokinetics , Bevacizumab , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacokinetics , Drug Administration Schedule , Female , Humans , Imatinib Mesylate , Male , Maximum Tolerated Dose , Middle Aged , Neoplastic Cells, Circulating/drug effects , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Treatment Outcome
5.
Ann Oncol ; 24(7): 1900-1907, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23519998

ABSTRACT

BACKGROUND: Based upon preclinical evidence for improved antitumor activity in combination, this phase I study investigated the maximum-tolerated dose (MTD), safety, activity, pharmacokinetics (PK), and biomarkers of the mammalian target of rapamycin inhibitor, temsirolimus, combined with sorafenib in hepatocellular carcinoma (HCC). PATIENTS AND METHODS: Patients with incurable HCC and Child Pugh score ≤B7 were treated with sorafenib plus temsirolimus by 3 + 3 design. The dose-limiting toxicity (DLT) interval was 28 days. The response was assessed every two cycles. PK of temsirolimus was measured in a cohort at MTD. RESULTS: Twenty-five patients were enrolled. The MTD was temsirolimus 10 mg weekly plus sorafenib 200 mg twice daily. Among 18 patients at MTD, DLT included grade 3 hand-foot skin reaction (HFSR) and grade 3 thrombocytopenia. Grade 3 or 4 related adverse events at MTD included hypophosphatemia (33%), infection (22%), thrombocytopenia (17%), HFSR (11%), and fatigue (11%). With sorafenib, temsirolimus clearance was more rapid (P < 0.05). Two patients (8%) had a confirmed partial response (PR); 15 (60%) had stable disease (SD). Alpha-fetoprotein (AFP) declined ≥50% in 60% assessable patients. CONCLUSION: The MTD of sorafenib plus temsirolimus in HCC was lower than in other tumor types. HCC-specific phase I studies are necessary. The observed efficacy warrants further study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , alpha-Fetoproteins/metabolism , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplastic Cells, Circulating , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Protein Precursors/blood , Prothrombin , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sorafenib , Treatment Outcome
6.
Br J Cancer ; 93(2): 195-9, 2005 Jul 25.
Article in English | MEDLINE | ID: mdl-15999098

ABSTRACT

The use of serial serum measurements of the carbohydrate antigen 19-9 (CA19-9) to guide treatment decisions and serve as a surrogate end point in clinical trial design requires further validation. We investigated whether CA19-9 decline represents an accurate surrogate for survival and time to treatment failure (TTF) in a cohort of 76 patients with advanced pancreatic cancer receiving fixed-dose rate gemcitabine in three separate studies. Statistically significant correlations between percentage CA19-9 decline and both overall survival and TTF were found, with median survival ranging from 12.0 months for patients with the greatest degree of biomarker decline (> 75%) compared with 4.3 months in those whose CA19-9 did not decline during therapy (P < 0.001). Using specific thresholds, patients with > or = 25% decline in CA19-9 during treatment had significantly better outcomes than those who did not (median survival and TTF of 9.6 and 4.6 months vs 4.4 and 1.5 months; P < 0.001). Similar results were seen using both 50 and 75% as cutoff points. We conclude that serial CA19-9 measurements correlate well with clinical outcomes in this patient population, and that decline in this biomarker should be entertained for possible use as a surrogate end point in clinical trials for the selection of new treatments in this disease.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Pancreatic Neoplasms/drug therapy , Endpoint Determination , Humans , Pancreatic Neoplasms/pathology , Predictive Value of Tests , Prognosis , Retrospective Studies , Sensitivity and Specificity , Survival Analysis , Treatment Outcome , Gemcitabine
7.
Semin Oncol ; 28(5): 521-31, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11685745

ABSTRACT

Hepatocellular carcinoma (HCC) is a disease that is extremely difficult to manage and is markedly increasing in incidence. Malignant transformation generally occurs in the setting of liver dysfunction related to a number of different diseases, including viral hepatitis, alcoholic liver disease, and aflatoxin exposure. Short of surgical or ablative approaches, no standard therapy exists for HCC and the prognosis is poor. Perhaps our best hope is that further elucidation of the specific molecular features underlying the disease will translate into innovative, and potentially disease-specific strategies to manage this difficult cancer. Exposure to aflatoxin is associated with a specific mutation in the tumor-suppressor gene p53. The exact molecular events underlying hepatocarcinogenesis in the setting of viral infection have yet to be elucidated, although there is evidence to suggest that virus-encoded proteins contribute to malignant transformation. Both hepatitis B X antigen and hepatitis C core protein appear to interact with a variety of cellular proteins leading to alterations in signal transduction and transcriptional activity. These events presumably cooperate to facilitate malignant progression by promoting extended hepatocyte survival, evasion of the immune response, and acquisition of mutations through genomic instability.


Subject(s)
Carcinoma, Hepatocellular/etiology , Cell Transformation, Neoplastic , Liver Neoplasms/etiology , Animals , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/prevention & control , Cell Transformation, Viral , Chromosome Aberrations , Humans , Liver Neoplasms/epidemiology , Liver Neoplasms/prevention & control , Models, Animal , Mutation
8.
Curr Opin Oncol ; 12(4): 357-61, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888422

ABSTRACT

Hepatocellular carcinoma (HCC) is a disease that is extremely difficult to manage and is increasing markedly in incidence. This presents both an opportunity and a challenge. At-risk patients can be identified and early detection of HCC is feasible. New surgical techniques and postoperative therapies, including hepatic intra-arterial radiation, may improve the outlook for some patients with resectable cancer. Unfortunately, the vast majority of patients with HCC will have unresectable cancers. Regional treatments may shrink or necrose tumors, but no clear benefit to such therapies has been demonstrated. Recent evidence suggests combination chemotherapy may help some patients, although this needs validation. Perhaps the best hope is that the further elucidation of the genetic and molecular features of HCC will lend us insight into innovative strategies to manage this difficult cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Combined Modality Therapy , Disease Models, Animal , Drug Therapy , Hormone Replacement Therapy , Humans , Immunotherapy , Liver Neoplasms/epidemiology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Neoplasm Recurrence, Local
SELECTION OF CITATIONS
SEARCH DETAIL
...