Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Endocrinol Metab ; 99(6): E936-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24628546

ABSTRACT

BACKGROUND: Vascular endothelial growth factor-targeted kinase inhibitors have emerged as highly promising therapies for radioiodine-refractory metastatic differentiated thyroid cancer. Unfortunately, drug resistance uniformly develops, limiting their therapeutic efficacies and thereby constituting a major clinical problem. APPROACH AND METHODS: To study acquired drug resistance and elucidate underlying mechanisms in this setting, BHP2-7 human differentiated thyroid cancer cells were subjected to prolonged continuous in vitro selection with 18 µM pazopanib, a clinically relevant concentration; acquisition of pazopanib resistance was serially assessed, with the resulting resistant cells thereafter subcloned and characterized to assess potential mechanisms of acquired pazopanib resistance. RESULTS: Stable 2- to 4-fold in vitro pazopanib resistance emerged in response to pazopanib selection associated with similar in vitro growth characteristics but with markedly more aggressive in vivo xenograft growth. Selected cells were cross-resistant to sunitinib and to a lesser extent sorafenib but not to MAPK kinase (MEK1/2) inhibition by GSK1120212. Genotyping demonstrated acquisition of a novel activating KRAS codon 13 GGC to GTT (glycine to valine) mutation, consistent with the observed resistance to upstream vascular endothelial growth factor receptor inhibition yet sensitivity to downstream MAPK kinase (MEK1/2) inhibition. CONCLUSIONS: Selection of thyroid cancer cells with clinically utilized therapeutics can lead to acquired drug resistance and altered in vivo xenograft behavior that can recapitulate analogous drug resistance observed in patients. This approach has the potential to lead to insights into acquired treatment-related drug resistance in thyroid cancers that can be subjected to subsequent validation in serially collected patient samples and that has the potential to yield preemptive and responsive approaches to dealing with this important clinical problem.


Subject(s)
Carcinoma/drug therapy , Carcinoma/pathology , Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sulfonamides/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Animals , Carcinoma/genetics , Carcinoma, Papillary , Cell Culture Techniques , Cell Line, Tumor , DNA Mutational Analysis , Humans , Indazoles , Mice , Molecular Targeted Therapy , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Xenograft Model Antitumor Assays , ras Proteins/genetics
2.
Sci Transl Med ; 5(166): 166ra3, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23283368

ABSTRACT

Anaplastic thyroid cancer (ATC) has perhaps the worst prognosis of any cancer, with a median survival of only about 5 months regardless of stage. Pazopanib monotherapy has promising clinical activity in differentiated thyroid cancers (generally attributed to vascular endothelial growth factor receptor inhibition), yet has less effective single-agent activity in ATC. We now report that combining pazopanib with microtubule inhibitors such as paclitaxel produced heightened and synergistic antitumor effects in ATC cells and xenografts that were associated with potentiated mitotic catastrophe. We hypothesized that combined effects may reflect enhanced paclitaxel-induced cytotoxicity mediated by cell cycle regulatory kinase inhibition by pazopanib. Indeed, pazopanib potently inhibited aurora A, with pazopanib/paclitaxel synergy recapitulated by aurora A short hairpin RNA knockdown or by specific aurora A pharmacological inhibition. Pazopanib/paclitaxel synergy was reversed by aurora A knockdown. Moreover, aurora A (but not B or C) message and protein levels were significantly increased in patient ATCs, and durable benefit resulted from pilot clinical translation of pazopanib/paclitaxel therapy in a patient with metastatic ATC. Collectively, these results suggest that the pazopanib/paclitaxel combination is a promising candidate therapeutic approach in ATC and that aurora A may represent a potentially viable therapeutic molecular target in ATC.


Subject(s)
Drug Synergism , Paclitaxel/pharmacology , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Thyroid Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols , Aurora Kinase A , Aurora Kinases , Cell Cycle , Cell Line, Tumor , Cell Separation , Dose-Response Relationship, Drug , Female , Humans , Indazoles , Mice , Mice, Nude , Mitosis , Neoplasm Metastasis , Neoplasm Transplantation , Protein Serine-Threonine Kinases/pharmacology , RNA, Small Interfering/metabolism , Thyroid Carcinoma, Anaplastic , Time Factors , Tubulin Modulators/therapeutic use
3.
J Clin Endocrinol Metab ; 97(9): 3179-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22774206

ABSTRACT

CONTEXT/OBJECTIVES: Pazopanib, an inhibitor of kinases including vascular endothelial growth factor receptor, demonstrated impressive activity in progressive metastatic differentiated thyroid cancer, prompting its evaluation in anaplastic thyroid cancer (ATC). DESIGN/SETTING/PATIENTS/INTERVENTIONS/OUTCOME MEASURES: Preclinical studies, followed by a multicenter single arm phase 2 trial of continuously administered 800 mg pazopanib daily by mouth (designed to provide 90% chance of detecting a response rate of >20% at the 0.10 significance level when the true response rate is >5%), were undertaken. The primary trial end point was Response Evaluation Criteria in Solid Tumors (RECIST) response. RESULTS: Pazopanib displayed activity in the KTC2 ATC xenograft model, prompting clinical evaluation. Sixteen trial patients were enrolled; 15 were treated: 66.7% were female, median age was 66 yr (range 45-77 yr), and 11 of 15 had progressed through prior systemic therapy. Enrollment was halted, triggered by a stopping rule requiring more than one confirmed RECIST response among the first 14 of 33 potential patients. Four patients required one to two dose reductions; severe toxicities (National Cancer Institute Common Toxicity Criteria-Adverse Events version 3.0 grades >3) were hypertension (13%) and pharyngolaryngeal pain (13%). Treatment was discontinued because of the following: disease progression (12 patients), death due to a possibly treatment-related tumor hemorrhage (one patient), and intolerability (radiation recall tracheitis and uncontrolled hypertension, one patient each). Although transient disease regression was observed in several patients, there were no confirmed RECIST responses. Median time to progression was 62 d; median survival time was 111 d. Two patients are alive with disease 9.9 and 35 months after the registration; 13 died of disease. CONCLUSIONS: Despite preclinical in vivo activity in ATC, pazopanib has minimal single-agent clinical activity in advanced ATC.


Subject(s)
Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Thyroid Neoplasms/drug therapy , Aged , Animals , Disease Progression , Disease-Free Survival , Endpoint Determination , Female , Humans , Indazoles , Male , Mice , Mice, Nude , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Survival Analysis , Thyroid Carcinoma, Anaplastic , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...