Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancer Chemother Pharmacol ; 83(1): 191-199, 2019 01.
Article in English | MEDLINE | ID: mdl-30406840

ABSTRACT

PURPOSE: Olaratumab is a recombinant human IgG1 monoclonal antibody against PGDFRα. Olaratumab plus doxorubicin improved survivalversus doxorubicin in an open-label, randomised phase 2 soft tissue sarcoma (STS) trial. We characterised the olaratumab exposure-response relationship for progression-free survival (PFS), overall survival (OS), and safety. METHODS: PFS and OS data from the 133 patients enrolled in the phase 2 study were analysed using time-to-event modelling. The effect of olaratumab on PFS/OS was explored using the trough serum concentration after cycle 1 (Cmin1) and the average concentration throughout treatment (Cavg). The rate of treatment-emergent adverse events (TEAEs) was compared across olaratumab exposure quartiles. RESULTS: PFS and OS were described by models with an exponential hazard function and inhibitory EMAX functions to describe the effect of olaratumab, regardless of the PK endpoint. The olaratumab EC50s for PFS (ECmin150 = 82.0 µg/mL, ECavg50 = 179 µg/mL) and OS (ECmin150 = 66.1 µg/mL, ECavg50 = 134 µg/mL) corresponded to the median and 25th percentile of Cmin1/Cavg in the study, respectively. Maximum predicted improvement in the hazard ratio for OS and PFS was approximately 75% and 60%, respectively. There was no change in the rate of TEAEs with increasing olaratumab serum levels. CONCLUSIONS: PFS/OS benefits occurred without a rate change in TEAEs across quartiles. Maximum benefit in OS was achieved in the upper three quartiles and a potential of early disease progression in the lower quartile of olaratumab serum exposure. These results prompted a loading dose strategy in the ongoing phase 3 STS trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Sarcoma/mortality , Antibodies, Monoclonal/administration & dosage , Doxorubicin/administration & dosage , Follow-Up Studies , Humans , Prognosis , Sarcoma/drug therapy , Sarcoma/pathology , Survival Rate
2.
Clin Pharmacokinet ; 57(3): 355-365, 2018 03.
Article in English | MEDLINE | ID: mdl-28620891

ABSTRACT

BACKGROUND AND OBJECTIVES: Olaratumab is a recombinant human monoclonal antibody that binds to platelet-derived growth factor receptor-α (PDGFRα). In a randomized phase II study, olaratumab plus doxorubicin met its predefined primary endpoint for progression-free survival and achieved a highly significant improvement in overall survival versus doxorubicin alone in patients with advanced or metastatic soft tissue sarcoma (STS). In this study, we characterize the pharmacokinetics (PKs) of olaratumab in a cancer patient population. METHODS: Olaratumab was tested at 15 or 20 mg/kg in four phase II studies (in patients with nonsmall cell lung cancer, glioblastoma multiforme, STS, and gastrointestinal stromal tumors) as a single agent or in combination with chemotherapy. PK sampling was performed to measure olaratumab serum levels. PK data were analyzed by nonlinear mixed-effect modeling techniques using NONMEM®. RESULTS: The PKs of olaratumab were best described by a two-compartment PK model with linear clearance (CL). Patient body weight was found to have a significant effect on both CL and central volume of distribution (V 1), whereas tumor size significantly affected CL. A small subset of patients developed treatment-emergent anti-drug antibodies (TE-ADAs); however, TE-ADAs did not have any effect on CL or PK time course of olaratumab. There was no difference in the PKs of olaratumab between patients who received olaratumab as a single agent or in combination with chemotherapy. CONCLUSION: The PKs of olaratumab were best described by a model with linear disposition. Patient body weight and tumor size were found to be significant covariates. The PKs of olaratumab were not affected by immunogenicity or chemotherapeutic agents.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Models, Biological , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Body Weight , Disease-Free Survival , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Nonlinear Dynamics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Survival Rate , Time Factors , Young Adult
3.
J Hematol Oncol ; 10(1): 73, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28327200

ABSTRACT

BACKGROUND: Hepcidin plays a central role in iron homeostasis and erythropoiesis. Neutralizing hepcidin with a monoclonal antibody (mAb) may prevent ferroportin internalization, restore iron efflux from cells, and allow transferrin-mediated iron transport to the bone marrow. This multicenter, phase 1 study evaluated the safety, pharmacokinetics (PK), pharmacodynamics (PD), and efficacy of a fully humanized mAb (LY2787106) with high affinity for hepcidin in cancer patients with anemia. METHODS: Thirty-three patients with hepcidin levels ≥5 ng/mL received LY2787106 either every 3 weeks (19 patients, dose range 0.3-10 mg/kg) (part A) or weekly (14 patients, dose 10 mg/kg) (part B). LY2787106 PK/PD markers of iron and hematology biology were measured. RESULTS: LY2787106 clearance (32 mL/h) and volume of distribution (7.7 L) were independent of dose and time, leading to a dose-proportional increase in concentration with dose. Consistent dose-dependent increases in serum iron, and transferrin saturation were seen at the 3 and 10 mg/kg dose levels, typically peaking within 24 h after LY2787106 administration and returning to baseline by day 8. CONCLUSIONS: Our findings indicate that LY2787106 was well tolerated in cancer patients with anemia and that targeting the hepcidin-ferroportin pathway by neutralizing hepcidin resulted in transient iron mobilization, thus supporting the role of hepcidin in iron regulation. TRIAL REGISTRATION: ClinicalTrial.gov, NCT01340976.


Subject(s)
Anemia/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Hepcidins/immunology , Neoplasms/complications , Adult , Aged , Aged, 80 and over , Anemia/complications , Anemia/etiology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Female , Hemoglobins/analysis , Hemoglobins/drug effects , Humans , Iron/blood , Iron/metabolism , Male , Middle Aged , Time Factors , Treatment Outcome
4.
Cancer ; 117(20): 4732-9, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21456002

ABSTRACT

BACKGROUND: Tasisulam sodium (hereafter, tasisulam) is a novel anticancer agent that induces apoptosis through the intrinsic pathway and has antiangiogenic activity in preclinical models. Tasisulam demonstrated activity across a broad range of tumors, including melanoma. The primary objective of this phase 2 study was to determine the objective response rate (ORR) in patients who had received 1 previous systemic chemotherapy for unresectable/metastatic melanoma; secondary objectives were to evaluate the clinical response rate (CRR), progression-free survival (PFS), overall survival (OS), duration of response, safety, and pharmacokinetics. METHODS: Tasisulam was administered intravenously on Day 1 of 21-day cycles according to a lean body weight-based dosing algorithm targeting a peak plasma concentration (C(max)) of 420 µg/mL. RESULTS: In 68 enrolled patients, the median age was 59 years (range, 26-83 years). No patients had a complete response (CR), 8 patients had a partial response (PR), and 24 patients had stable disease (SD); the ORR (CR + PR) was 11.8%, and the CRR (CR + PR + SD) was 47.1%. The median PFS was 2.6 months, and the median OS was 9.6 months. The predominant treatment-related grade 3/4 toxicity was thrombocytopenia (20.6% of patients). Tasisulam exhibited a biexponential disposition with a predicted distribution half-life of 0.3 hours to 2.8 hours and a median terminal elimination half-life of 10 days (consistent with the turnover of albumin), suggesting that tasisulam is very tightly bound to albumin. CONCLUSIONS: Tasisulam administered at a targeted C(max) of 420 µg/mL on Day 1 of 21-day cycles demonstrated activity and tolerable toxicity as second-line treatment in malignant melanoma. These results led to a registration trial in metastatic melanoma.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzamides/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Algorithms , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Benzamides/administration & dosage , Benzamides/adverse effects , Benzamides/pharmacokinetics , Body Weight , Disease-Free Survival , Drug Administration Schedule , Fatigue/chemically induced , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/mortality , Middle Aged , Skin Neoplasms/mortality , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Thrombocytopenia/chemically induced , Treatment Outcome
5.
Cancer Chemother Pharmacol ; 68(5): 1233-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21431416

ABSTRACT

PURPOSE: This phase I study was carried out to determine the phase II recommended dose of tasisulam sodium (hereafter, tasisulam), a novel anticancer agent with a unique mechanism of action. METHODS: Tasisulam was administered intravenously, every 21 days, in patients with refractory solid tumors using a three-plus-three dose-escalation schema. RESULTS: Fifty-three patients were enrolled; the first 34 were treated with a flat dose of tasisulam of up to 2,400 mg, the dose level at which all three patients had dose-limiting toxicity (DLT). Controlling for C(max) proved important to reduce the risk of toxicity; therefore, we initially focused on identifying which parameters explained C(max) (end-of-infusion concentration) variability. Pharmacokinetic analysis indicated that C(max) negatively correlates with lean body weight (LBW). Thus, the dosing regimen was revised using a LBW-based algorithm targeting a specific C(max). A loading/chronic dose paradigm was then implemented as pharmacokinetic results revealed a long terminal half-life of tasisulam, likely because of its high-affinity albumin binding. C(max)-based dose escalation was stopped at the 420-µg/mL cohort, in which one of the 16 patients had DLT (transient hepatic transaminase elevation); grade 3/4 hematologic toxicity was noted in later cycles in three patients. Although response was not a primary objective, 33% of heavily pretreated patients with post-dose radiological assessments had stable disease. CONCLUSION: Implementation of a novel targeted C(max)-based dosing regimen allowed for the recommendation of a phase II tasisulam dose (loading dose of 420 µg/mL targeted C(max) with all subsequent doses administered at 65% of chronic dose given every 21 days) despite pharmacological challenges posed by high albumin binding.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Neoplasms/drug therapy , Sulfonamides/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Algorithms , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Benzamides/adverse effects , Benzamides/blood , Benzamides/pharmacokinetics , Chromatography, Liquid , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Middle Aged , Reproducibility of Results , Sulfonamides/adverse effects , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Tandem Mass Spectrometry , Young Adult
6.
J Immunol ; 178(4): 2028-37, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277106

ABSTRACT

Imatinib mesylate (IM) is effective at inducing complete cytogenetic remission in patients with chronic myelogenous leukemia. Because its influence on CD8 T cell responsiveness in vivo is unknown, we investigated the effects of IM by analyzing the response of OT-1 CD8 T cells to Listeria monocytogenes (LM) that express the cognate epitope OVA(257-264) (LM-OVA). In vitro, IM had no effect on Ag-specific expansion, cell division, cell cycle progression, or IFN-gamma expression in naive or memory OT-1 T cells. However, IM induced apoptosis of naive and memory OT-1 T cells at doses of >5 microM. At 15 microM IM, OT-1 T cells did not survive in in vitro cultures. The primary response of OT-1 T cells in vivo to LM-OVA infection was unaltered. In contrast, continuous IM treatment resulted in a diminished memory OT-1 response. The expression of IL-7Ralpha, a receptor required for memory cell survival, was lower (on OT-1 cells) in animals receiving IM. These results indicate that IM treatment affects the ability of the CD8 memory pool to respond to Ag and has the potential to increase susceptibility to infection.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Immunologic Memory/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Antigens, Bacterial/immunology , Apoptosis/immunology , Benzamides , CD8-Positive T-Lymphocytes/metabolism , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Imatinib Mesylate , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/immunology , Listeriosis/metabolism , Mice , Mice, Transgenic , Receptors, Interleukin-7/biosynthesis , Receptors, Interleukin-7/immunology
8.
Blood ; 107(12): 4917-25, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16522816

ABSTRACT

The role of signal transducers and activators of transcription 5 (STAT5) in chronic myelogenous leukemia (CML) is controversial. To clarify the role of STAT5 signaling in P210(BCR/ABL) leukemogenesis, P210 was introduced into primary murine STAT5A-deficient (STAT5A(-/-)) bone marrow (BM) cells, which, unlike STAT5A/5B double knockout BM cells, have no major intrinsic hematopoietic defects. Interestingly, only 21% of mice reconstituted with P210-transduced STAT5A(-/-) BM cells developed classic CML, compared with 80% to 100% of P210/STAT5A(+/+) and P210/STAT5A(+/-)-reconstituted animals. The remainder of P210/STAT5A(-/-) animals died from an acute B-cell lymphoblastic leukemia (ALL)-like disease (32%) or a CML/ALL mix (47%), reflecting impairment in the induction and maintenance of CML, which normally predominates in this mouse model. Of mice that ultimately developed CML, P210/STAT5A(-/-) animals had prolonged survival and increased myeloid immaturity. Importantly, reconstitution of wild-type mice with BM cells coexpressing P210 and dominant-negative STAT5 also profoundly reduced the incidence of CML, without impairing the induction of ALL. Altogether, these findings indicate that STAT5 and STAT5A play an important role in the pathogenesis of the CML-like disease in mice. A greater understanding of the STAT5 target genes involved in CML induction may lead to new therapeutic targets that influence CML progenitor cell biology.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Genes, abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , STAT5 Transcription Factor/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Transformation, Neoplastic/genetics , Drug Design , Enzyme Inhibitors/therapeutic use , Genes, abl/genetics , Hematopoiesis/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Knockout , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/deficiency
9.
Article in English | MEDLINE | ID: mdl-16304379

ABSTRACT

Despite recent improvements in the treatment of early-stage disease, the blastic phase of chronic myeloid leukemia (CML) remains a therapeutic challenge. For imatinib-naïve patients, imatinib provided encouraging hematologic and cytogenetic benefits; however, the vast majority of CML blast crisis cases today arise in patients already on imatinib-based therapy. Clonal evolution and duplication of the Philadelphia chromosome continue to be associated with blastic phase transformation, but recent studies have identified BCR/ABL kinase domain mutations in 30%-40% of blast crisis patients. This implies that BCR-ABL-targeted therapy might have influenced the molecular road map to blastic transformation. In this review, we will examine the effect of imatinib on primitive CML progenitors and how this might influence the pathophysiology of blast crisis. A rational framework for deciding how best to integrate stem cell transplantation, traditional chemotherapy, imatinib, and other BCR-ABL kinase inhibitors in the care of blast crisis patients will also be discussed.


Subject(s)
Antineoplastic Agents/adverse effects , Blast Crisis/chemically induced , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/adverse effects , Algorithms , Antineoplastic Agents/therapeutic use , Benzamides , Blast Crisis/drug therapy , Blast Crisis/genetics , Blast Crisis/physiopathology , Fusion Proteins, bcr-abl/metabolism , Genes, abl , Genomic Instability , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation , Protein-Tyrosine Kinases/metabolism
10.
Cancer Res ; 65(19): 8698-705, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204038

ABSTRACT

Ewing sarcoma is the second most common malignant pediatric bone tumor. Over 80% of Ewing sarcoma contain the oncogene EWS/FLI-1, which encodes the EWS/FLI-1 oncoprotein, a hybrid transcription factor comprised of NH2-terminal sequences from the RNA-binding protein EWS and the DNA-binding and COOH-terminal regions of the Ets transcription factor FLI-1. Although numerous genes are dysregulated by EWS/FLI-1, advances in Ewing sarcoma cancer biology have been hindered by the lack of an animal model because of EWS/FLI-1-mediated cytotoxicity. In this study, we have developed conditions for the isolation and propagation of murine primary bone-derived cells (mPBDC) that stably express EWS/FLI-1. Early-passage EWS/FLI-1 mPBDCs were immortalized in culture but inefficient at tumor induction, whereas later-passage cells formed sarcomatous tumors in immunocompetent syngeneic mice. Murine EWS/FLI-1 tumors contained morphologically primitive cells that lacked definitive lineage markers. Molecular characterization of murine EWS/FLI-1 tumors revealed that some but not all had acquired a novel, clonal in-frame p53 mutation associated with a constitutive loss of p21 expression. Despite indications that secondary events facilitated EWS/FLI-1 mPBDC tumorigenesis, cells remained highly dependent on EWS/FLI-1 for efficient transformation in clonogenic assays. This Ewing sarcoma animal model will be a useful tool for dissecting the molecular pathogenesis of Ewing sarcoma and provides rationale for the broader use of organ-specific progenitor cell populations for the study of human sarcoma.


Subject(s)
Bone Neoplasms/metabolism , Oncogene Proteins, Fusion/biosynthesis , Proto-Oncogene Protein c-fli-1/biosynthesis , RNA-Binding Protein EWS/biosynthesis , Sarcoma, Ewing/metabolism , Amino Acid Sequence , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Cycle/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Gene Expression , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
11.
Blood ; 105(10): 3995-4003, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15657179

ABSTRACT

Imatinib mesylate is highly effective in newly diagnosed chronic myeloid leukemia (CML), but BCR/ABL (breakpoint cluster region/abelson murine leukemia)-positive progenitors persist in most patients with CML treated with imatinib mesylate, indicating the need for novel therapeutic approaches. In this study, we have used the murine CML-like myeloproliferative disorder as a platform to characterize the pharmacokinetic, signal transduction, and antileukemic properties of PD166326, one of the most potent members of the pyridopyrimidine class of protein tyrosine kinase inhibitors. In mice with the CML-like disease, PD166326 rapidly inhibited Bcr/Abl kinase activity after a single oral dose and demonstrated marked antileukemic activity in vivo. Seventy percent of PD166326-treated mice achieved a white blood cell (WBC) count less than 20.0 x 10(9)/L (20,000/microL) at necropsy, compared with only 8% of imatinib mesylate-treated animals. Further, two thirds of PD166326-treated animals had complete resolution of splenomegaly, compared with none of the imatinib mesylate-treated animals. Consistent with its more potent antileukemic effect in vivo, PD166326 was also superior to imatinib mesylate in inhibiting the constitutive tyrosine phosphorylation of numerous leukemia-cell proteins, including the src family member Lyn. PD166326 also prolonged the survival of mice with imatinib mesylate-resistant CML induced by the Bcr/Abl mutants P210/H396P and P210/M351T. Altogether, these findings demonstrate the potential of more potent Bcr/Abl inhibitors to provide more effective antileukemic activity. Clinical development of PD166326 or a related analog may lead to more effective drugs for the treatment of de novo and imatinib mesylate-resistant CML.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Animals , Benzamides , Cell Line , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Molecular Structure , Mutation/genetics , Phosphorylation , Phosphotyrosine/metabolism , Piperazines/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Signal Transduction , Stem Cell Factor/metabolism , Survival Rate , Time Factors
12.
Hematol Oncol Clin North Am ; 18(3): 525-43, vii, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15271391

ABSTRACT

Of the current mouse chronic myelogenous leukemia (CML) models,the murine bone marrow (BM) transduction and transplantation model most efficiently mimics many of the central features of human CML. In this model, lethally irradiated mice are reconstituted with primary murine BM cells transduced with a P210BCR/ABL retrovirus. All recipient mice develop a fatal peripheral blood and BM granulocytosis and splenomegaly, a disease termed the murine CML-like myeloproliferative disorder. This model has been used to establish the causative role of Bcr/Abl in CML, identify those signaling pathways and regions of Bcr/Abl critical for leukemogenesis, and explore the limitations of targeted CML therapy. Future refinements in this CML mouse model will make it a more effective tool for studying imatinib-resistant CML, reproducing chronic- and blastic-phase human CML, and performing CML progenitor studies.


Subject(s)
Disease Models, Animal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Mice , Retroviridae/genetics , Transduction, Genetic
13.
Clin Cancer Res ; 10(10): 3528-34, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15161712

ABSTRACT

PURPOSE: Despite recent advances in cancer therapy, long-term survival in small cell lung cancer (SCLC) remains uncommon, underscoring the need for novel therapeutic approaches. Previous studies have identified constitutive expression of the receptor tyrosine kinase, c-Kit, and its ligand, stem cell factor, in a substantial proportion of SCLC specimens. The purpose of this study was to determine whether imatinib mesylate, an inhibitor of c-Kit, could achieve therapeutic concentrations in tumors and in brain (a frequent site of SCLC metastasis) and interfere with SCLC tumor growth in vivo. EXPERIMENTAL DESIGN: Human SCLC tumor cell lines with constitutive c-kit expression and tyrosine phosphorylation (NCI-H209, NCI-H526, and NCI-H1607) were used to establish SCLC tumor xenografts in NCr nude (nu/nu)-immunodeficient mice. SCLC tumor-bearing mice were randomly assigned to imatinib or control (water) administered twice a day by oral gavage. Imatinib concentrations in plasma, brain, and tumor were quantitated and correlated with tumor response. RESULTS: Therapeutic concentrations of imatinib were achieved in plasma and tumor xenografts but not in the brain. Imatinib blocked the constitutive activation of c-kit in SCLC tumor cell lines in vitro but had a negligible effect on SCLC xenograft growth in vivo. CONCLUSIONS: Orally administered imatinib rapidly reaches therapeutic concentrations in SCLC xenografts, suggesting the feasibility of combining imatinib with other novel or traditional chemotherapeutic agents in SCLC or other solid tumors. The c-Kit signaling pathway does not appear to play a critical role in SCLC proliferation and viability in vivo, however, suggesting that imatinib is unlikely to be effective as monotherapy for SCLC.


Subject(s)
Carcinoma, Small Cell/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzamides , Brain Neoplasms/secondary , Cell Line, Tumor , Humans , Imatinib Mesylate , Immunoblotting , Immunoprecipitation , Ligands , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Metastasis , Neoplasm Transplantation , Phosphorylation , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Stem Cell Factor/metabolism , Time Factors , Tyrosine/metabolism
15.
J Pediatr Hematol Oncol ; 25(6): 459-66, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12794524

ABSTRACT

BACKGROUND: Although chimeric EWS gene and Ets gene fusions are pathognomonic of Ewing sarcoma (ES) and primitive neuroectodermal tumors (PNET), the molecular pathogenesis of these pediatric malignancies is poorly understood. Recently, the human epidermal growth factor (HER)-2 receptor, which plays an important role in the biology of certain epithelial cancers, has been implicated in ES tumor cell line growth and chemosensitivity. MATERIALS: To investigate whether HER2 might be a rational target for ES/PNET therapy, five ES cell lines and 13 archival primary ES/PNET samples were examined by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) for evidence of HER2 overexpression. RESULTS: Although several ES cell lines demonstrated modest constitutive HER2 expression by immunoblot, none of the ES cell lines or primary tumor samples showed evidence of HER2 overexpression by IHC or HER2 gene amplification by FISH. Moreover, treatment of human ES cell lines with the HER2-targeted agent trastuzumab (Herceptin) had little effect on cell survival, proliferation, or growth in semi-solid medium. CONCLUSIONS: These results suggest that HER2 is not a biologically or therapeutically important pathway in ES/PNET.


Subject(s)
Bone Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Sarcoma, Ewing/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Blotting, Western , Bone Neoplasms/drug therapy , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Sarcoma, Ewing/drug therapy , Trastuzumab , Tumor Cells, Cultured
16.
Blood ; 101(12): 5010-3, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12595307

ABSTRACT

The chronic myelogenous leukemia (CML)-like myeloproliferative disorder observed in the BCR/ABL murine bone marrow transduction and transplantation model shares several features with the human disease, including a high response rate to the tyrosine kinase inhibitor imatinib mesylate (STI571). To study the impact of chronic imatinib mesylate treatment on the CML-like illness, mice were maintained on therapeutic doses of this drug and serially monitored. Unexpectedly, despite excellent systemic control of the CML-like illness, many of the mice developed progressive neurologic deficits after 2 to 4 months of imatinib mesylate therapy because of central nervous system (CNS) leukemia. Analysis of imatinib mesylate cerebral spinal fluid concentrations revealed levels 155- fold lower than in plasma. Thus, in the mouse, the limited ability of imatinib mesylate to cross the blood-brain barrier allowed the CNS to become a sanctuary for Bcr/Abl-induced leukemia. This model will be a useful tool for the future study of novel anti-CML drugs and in better defining the mechanisms for limited imatinib mesylate penetration into the CNS.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms/pathology , Genes, abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Spinal Cord Neoplasms/pathology , Animals , Benzamides , Bone Marrow , Central Nervous System Diseases , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Piperazines/cerebrospinal fluid , Piperazines/therapeutic use , Pyrimidines/cerebrospinal fluid , Pyrimidines/therapeutic use , Transfection
17.
Mol Cell Biol ; 23(2): 482-92, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509448

ABSTRACT

The chimeric fusion gene EWS/FLI-1 is detected in most cases of Ewing's sarcoma (ES), the second most common malignant bone tumor of childhood. Although 80% of ES tumors develop in skeletal sites, the remainder can arise in almost any soft tissue location. The lineage of the cell developing the EWS/FLI-1 gene fusion has not been fully characterized but is generally considered to be of either mesenchymal or neural crest origin. To study this oncogene in a conceptually relevant target cell, EWS/FLI-1 was introduced into the murine cell line C2C12, a myoblast cell line capable of differentiation into muscle, bone, or fat. In this cellular context, EWS/FLI-1 profoundly inhibited the myogenic differentiation program. The block in C2C12 myogenic differentiation required the nuclear localization and DNA-binding functions of EWS/FLI-1 and was mediated by transcriptional and posttranscriptional suppression of the myogenic transcription factors MyoD and myogenin. Interestingly, C2C12-EWS/FLI-1 cells constitutively expressed alkaline phosphatase, a bone lineage marker, and were alkaline phosphatase positive by histochemistry but showed no other evidence of bone lineage commitment. Consistent with recent findings in human ES tumor cell lines, C2C12-EWS/FLI-1 cells constitutively expressed cyclin D1 and demonstrated decreased expression of the cell cycle regulator p21(cip1), even under differentiation conditions and at confluent density. This C2C12-EWS/FLI-1 cell model may assist in the identification of novel differentially expressed genes relevant to ES and provide further insight into the cell(s) of origin developing ES-associated genetic fusions.


Subject(s)
Oncogene Proteins, Fusion/physiology , Sarcoma, Ewing/metabolism , Transcription Factors/physiology , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Cell Cycle , Cell Differentiation , Cell Line , Cell Lineage , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Flow Cytometry , Genetic Vectors , Immunohistochemistry , Mice , Microscopy, Fluorescence , Models, Biological , MyoD Protein/metabolism , Myogenin/metabolism , Oncogene Proteins, Fusion/metabolism , Oncogenes/genetics , Plasmids/metabolism , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL