Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Fetal Pediatr Pathol ; 42(2): 241-252, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36062956

ABSTRACT

Background: Ewing sarcoma (ES) can be confirmed by identifying the EWSR1-FLI1 fusion transcript. This study is to investigate whether immunostaining (IHC) of PRKCB-a protein directly regulated by EWSR1-FLI1 is a surrogate maker for diagnosing ES in routine practice. Methods: Microarray gene expression analyses were conducted. RKCB IHC was applied to 69 ES confirmed by morphology and molecular methods, and 41 non-Ewing small round cell tumors. EWSR1 rearrangement, EWSR1-FLI1 fusion or t(11;22)(q24;q12) were identified by fluorescence in situ hybridization, reverse transcriptase polymerase chain reaction, or cytogenetic analysis, respectively. Results: Gene array analyses showed significant overexpression of the PRKCB in ES. PRKCB IHC was positive in 19 cases of ES with EWSR1-FLI1 fusion, 3 cases with cytogenetic 11:22 translocation and 59 cases with EWSR1 rearrangement while negative in only one EWSR1 rearranged case. PRKCB IHC is sensitive (98%) and specific (96%) in detecting EWSR1 rearranged ES. Conclusions: PRKCB is a reliable antibody for diagnosing ES in routine practice.


Subject(s)
Sarcoma, Ewing , Sarcoma , Humans , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/genetics , Immunohistochemistry , In Situ Hybridization, Fluorescence , RNA-Binding Protein EWS/genetics , Biomarkers , Oncogene Proteins, Fusion/genetics , Protein Kinase C beta/genetics , Protein Kinase C beta/metabolism
2.
Cancer Immunol Immunother ; 67(12): 1853-1862, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30209589

ABSTRACT

The GM.CD40L vaccine, which recruits and activates dendritic cells, migrates to lymph nodes, activating T cells and leading to systemic tumor cell killing. When combined with the CCL21 chemokine, which recruits T cells and enhances T-cell responses, additive effects have been demonstrated in non-small cell lung cancer mouse models. Here, we compared GM.CD40L versus GM.CD40L plus CCL21 (GM.CD40L.CCL21) in lung adenocarcinoma patients with ≥ 1 line of treatment. In this phase I/II randomized trial (NCT01433172), patients received intradermal vaccines every 14 days (3 doses) and then monthly (3 doses). A two-stage minimax design was used. During phase I, no dose-limiting toxicities were shown in three patients who received GM.CD40L.CCL21. During phase II, of evaluable patients, 5/33 patients (15.2%) randomized for GM.DCD40L (p = .023) and 3/32 patients (9.4%) randomized for GM.DCD40L.CCL21 (p = .20) showed 6-month progression-free survival. Median overall survival was 9.3 versus 9.5 months with GM.DCD40L versus GM.DCD40L.CCL21 (95% CI 0.70-2.25; p = .44). For GM.CD40L versus GM.CD40L.CCL21, the most common treatment-related adverse events (TRAEs) were grade 1/2 injection site reaction (51.4% versus 61.1%) and grade 1/2 fatigue (35.1% versus 47.2%). Grade 1 immune-mediated TRAEs were isolated to skin. No patients showed evidence of pseudo-progression or immune-related TRAEs of grade 1 or greater of pneumonitis, endocrinopathy, or colitis, and none discontinued treatment due to toxicity. Although we found no significant associations between vaccine immunogenicity and outcomes, in limited biopsies, one patient treated with GMCD40L.CCL21 displayed abundant tumor-infiltrating lymphocytes. This possible effectiveness warrants further investigation of GM.CD40L in combination approaches.


Subject(s)
Adenocarcinoma/therapy , CD40 Ligand/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , Chemokine CCL21/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunotherapy , Adenocarcinoma/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/immunology , Female , Follow-Up Studies , Humans , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Male , Middle Aged , Prognosis , Survival Rate
3.
PLoS One ; 12(1): e0169797, 2017.
Article in English | MEDLINE | ID: mdl-28085924

ABSTRACT

The tumor microenvironment is composed of cellular and stromal components such as tumor cells, mesenchymal cells, immune cells, cancer associated fibroblasts and the supporting extracellular matrix. The tumor microenvironment provides crucial support for growth and progression of tumor cells and affects tumor response to therapeutic interventions. To better understand tumor biology and to develop effective cancer therapeutic agents it is important to develop preclinical platforms that can faithfully recapitulate the tumor microenvironment and the complex interaction between the tumor and its surrounding stromal elements. Drug studies performed in vitro with conventional two-dimensional cancer cell line models do not optimally represent clinical drug response as they lack true tumor heterogeneity and are often performed in static culture conditions lacking stromal tumor components that significantly influence the metabolic activity and proliferation of cells. Recent microfluidic approaches aim to overcome such obstacles with the use of cell lines derived in artificial three-dimensional supportive gels or micro-chambers. However, absence of a true tumor microenvironment and full interstitial flow, leads to less than optimal evaluation of tumor response to drug treatment. Here we report a continuous perfusion microfluidic device coupled with microscopy and image analysis for the assessment of drug effects on intact fresh tumor tissue. We have demonstrated that fine needle aspirate biopsies obtained from patient-derived xenograft models of adenocarcinoma of the lung can successfully be analyzed for their response to ex vivo drug treatment within this biopsy trapping microfluidic device, wherein a protein kinase C inhibitor, staurosporine, was used to assess tumor cell death as a proof of principle. This approach has the potential to study tumor tissue within its intact microenvironment to better understand tumor response to drug treatments and eventually to choose the most effective drug and drug combination for individual patients in a cost effective and timely manner.


Subject(s)
Antibodies, Monoclonal/pharmacology , Doxorubicin/pharmacology , Microfluidic Analytical Techniques/instrumentation , Neoplasms/pathology , Staurosporine/pharmacology , Tumor Microenvironment/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Biopsy , Cell Culture Techniques , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, Hairless , Mice, SCID , Neoplasms/drug therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Sci Rep ; 5: 16991, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26601688

ABSTRACT

Systemic therapy has improved osteosarcoma event-free and overall survival, but 30-50% of patients originally diagnosed will have progressive or recurrent disease, which is difficult to cure. Osteosarcoma has a complex karyotype, with loss of p53 in the vast majority of cases and an absence of recurrent, targetable pathways. In this study, we explored 54 agents that are clinically approved for other oncologic indications, agents in active clinical development, and others with promising preclinical data in osteosarcoma at clinically achievable concentrations in 5 osteosarcoma cell lines. We found significant single-agent activity of multiple agents and tested 10 drugs in all permutations of two-drug combinations to define synergistic combinations by Chou and Talalay analysis. We then evaluated order of addition to choose the combinations that may be best to translate to the clinic. We conclude that the repurposing of chemotherapeutics in osteosarcoma by using an in vitro system may define novel drug combinations with significant in vivo activity. In particular, combinations of proteasome inhibitors with histone deacetylase inhibitors and ixabepilone and MK1775 demonstrated excellent activity in our assays.


Subject(s)
Bone Neoplasms/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Osteosarcoma/drug therapy , Proteasome Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Bone Neoplasms/pathology , Cell Survival/drug effects , Drug Synergism , Drug Therapy, Combination , Epothilones/therapeutic use , Epothilones/toxicity , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Osteosarcoma/pathology , Proteasome Inhibitors/pharmacology , Pyrazoles/therapeutic use , Pyrazoles/toxicity , Pyrimidines/therapeutic use , Pyrimidines/toxicity , Pyrimidinones , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Cancer Control ; 22(2): 193-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26068764

ABSTRACT

BACKGROUND: Worldwide, lung cancer is the most common cause of mortality. Toxins from tobacco smoke are known to increase the risk of lung cancer; however, up to 15% of lung cancer-related deaths in men and up to 50% of lung cancer-related deaths in women occur in people who do not smoke. Despite the fact that chemotherapy generally provides a survival benefit for non-small-cell lung cancer, not every patient will respond to therapy and many experience therapy-related adverse events. Thus, predictive markers are used to determine which patients are more likely to respond to a given regimen. METHODS: We reviewed the current medical literature in English relating to predictive markers that may be positive, such as the presence of an activating EGFR mutation. RESULTS: The advances in using EGFR as a molecular predictive marker were summarized. This biomarker influences therapeutic response in patients with lung adenocarcinoma. Clinical evidence supporting its value is also reviewed. CONCLUSIONS: The use of EGFR as a predictive factor in lung adenocarcinoma may help target therapy to individual tumors to achieve the best likelihood for long-term survival and to avoid adverse events from medications unlikely to be effective.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Amino Acid Substitution , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/pathology , Chromosome Deletion , Drug Resistance, Neoplasm/genetics , Exons/genetics , Humans , Lung Neoplasms/pathology , Mutation , Prognosis
6.
Cancer Control ; 22(2): 226-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26068769

ABSTRACT

BACKGROUND: Predictive assays for cancer treatment are not new technology, but they have failed to meet the criteria necessary for standardized use in clinical decision-making. METHODS: The authors summarize the use of predictive assays and the challenges and values associated with these assays in the clinical setting. RESULTS: Predictive assays commercially available in the clinical setting are not standardized, have significant obstacles to overcome, and cannot be relied upon by health care professionals due to the limited value these assays provide to the decision-making process for the treatment of patients. CONCLUSIONS: A method that more closely recapitulates the human tumor microenvironment and accurately predicts response with high reproducibility would be beneficial to patient outcomes and quality of life.


Subject(s)
Neoplasms/pathology , Precision Medicine/methods , Biomarkers, Tumor , Cell Culture Techniques , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Immunohistochemistry , Reproducibility of Results , Xenograft Model Antitumor Assays
7.
Sci Signal ; 8(359): ra4, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25587191

ABSTRACT

Strategies to measure functional signaling-associated protein complexes have the potential to augment current molecular biomarker assays, such as genotyping and expression profiling, used to annotate diseases. Aberrant activation of epidermal growth factor receptor (EGFR) signaling contributes to diverse cancers. We used a proximity ligation assay (PLA) to detect EGFR in a complex with growth factor receptor-bound protein 2 (GRB2), the major signaling adaptor for EGFR. We used multiple lung cancer cell lines to develop and characterize EGFR:GRB2 PLA and correlated this assay with established biochemical measures of EGFR signaling. In a panel of patient-derived xenografts in mice, the intensity of EGFR:GRB2 PLA correlated with the reduction in tumor size in response to the EGFR inhibitor cetuximab. In tumor biopsies from three cohorts of lung cancer patients, positive EGFR:GRB2 PLA was observed in patients with and without EGFR mutations, and the intensity of EGFR:GRB2 PLA was predictive of overall survival in an EGFR inhibitor-treated cohort. Thus, we established the feasibility of using PLA to measure EGFR signaling-associated protein complexes in patient-based materials, suggesting the potential for similar assays for a broader array of receptor tyrosine kinases and other key signaling molecules.


Subject(s)
Biomarkers, Tumor/metabolism , ErbB Receptors/metabolism , Immunoassay/methods , Lung Neoplasms/diagnosis , Multiprotein Complexes/metabolism , Protein Interaction Mapping/methods , Signal Transduction/physiology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line, Tumor , Cetuximab , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , GRB2 Adaptor Protein/metabolism , Heterografts/drug effects , Heterografts/pathology , Humans , Lung Neoplasms/metabolism , Mice , Multiprotein Complexes/physiology
8.
J Thorac Oncol ; 9(7): 974-982, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24922005

ABSTRACT

INTRODUCTION: Interleukin-6 (IL-6) can activate downstream signaling pathways in lung cancer cells, such as the STAT3 pathway, and is reported to be produced by tumor cells with activating EGFR mutations. We examined IL-6/STAT3 in lung cancer tumor tissues and the effects of siltuximab, a neutralizing antibody to human IL-6, in mouse models of lung cancer. METHODS: IL-6 and STAT3 activation levels were compared with tumor histology and presence of KRAS mutations in snap-frozen, non-small-cell lung cancer tumors. The effects of siltuximab alone or in combination with erlotinib were examined in mouse xenograft models constructed using three cell line xenograft models and one primary explant mouse model. We examined the influence of cancer-associated fibroblasts (CAFs) on tumor growth and siltuximab effects. RESULTS: IL-6 levels were higher in tumors of squamous cell versus adenocarcinoma histology and were not associated with presence of KRAS mutations. Tyrosine phosphorylation status of STAT3 did not correlate with tumor IL-6 levels. Serine phosphorylation of STAT3 was correlated with KRAS mutation status. Both tumor and stromal cells contributed to total IL-6 within tumors. Siltuximab had minimal effect as a single agent in xenografts with tumor cells alone; however, in models coadministered with CAFs, siltuximab had more potent effects on tumor inhibition. We observed no effects of combined erlotinib and siltuximab. CONCLUSIONS: IL-6 is elevated in subsets of human NSCLCs, especially with squamous cell histology. Tumors supported by stromal production of IL-6 seem to be the most vulnerable to tumor growth inhibition by siltuximab.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Disease Models, Animal , Erlotinib Hydrochloride , Female , Fibroblasts , Humans , Interleukin-6/analysis , Interleukin-6/immunology , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Mice , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Quinazolines/administration & dosage , STAT3 Transcription Factor/metabolism , Serine/metabolism , Tyrosine/metabolism , ras Proteins/genetics
10.
Clin Cancer Res ; 20(6): 1644-55, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24429877

ABSTRACT

PURPOSE: Panobinostat, a histone deacetylase (HDAC) inhibitor, enhances antiproliferative activity in non-small cell lung cancer (NSCLC) cell lines when combined with erlotinib. We evaluated this combination in patients with advanced NSCLC and head and neck cancer. EXPERIMENTAL DESIGN: Eligible patients were enrolled in a 3+3 dose-escalation design to determine the maximum tolerated dose (MTD) of twice weekly panobinostat plus daily erlotinib at four planned dose levels (DL). Pharmacokinetics, blood, fat pad biopsies (FPB) for histone acetylation, and paired pre and posttherapy tumor biopsies for checkpoint kinase 1 (CHK1) expression were assessed. RESULTS: Of 42 enrolled patients, 33 were evaluable for efficacy. Dose-limiting toxicities were prolonged-QTc and nausea at DL3. Adverse events included fatigue and nausea (grades 1-3), and rash and anorexia (grades 1-2). Disease control rates were 54% for NSCLC (n = 26) and 43% for head and neck cancer (n = 7). Of 7 patients with NSCLC with EGF receptor (EGFR) mutations, 3 had partial response, 3 had stable disease, and 1 progressed. For EGFR-mutant versus EGFR wild-type patients, progression-free survival (PFS) was 4.7 versus 1.9 months (P = 0.43) and overall survival was 41 (estimated) versus 5.2 months (P = 0.39). Erlotinib pharmacokinetics was not significantly affected. Correlative studies confirmed panobinostat's pharmacodynamic effect in blood, FPB, and tumor samples. Low CHK1 expression levels correlated with PFS (P = 0.006) and response (P = 0.02). CONCLUSIONS: We determined MTD at 30 mg (panobinostat) and 100 mg (erlotinib). Further studies are needed to further explore the benefits of HDAC inhibitors in patients with EGFR-mutant NSCLC, investigate FPB as a potential surrogate source for biomarker investigations, and validate CHK1's predictive role.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Head and Neck Neoplasms/drug therapy , Hydroxamic Acids/administration & dosage , Indoles/administration & dosage , Lung Neoplasms/drug therapy , Quinazolines/administration & dosage , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Erlotinib Hydrochloride , Female , Head and Neck Neoplasms/mortality , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Indoles/adverse effects , Indoles/pharmacokinetics , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Maximum Tolerated Dose , Middle Aged , Panobinostat , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Treatment Outcome
11.
PLoS One ; 8(12): e82875, 2013.
Article in English | MEDLINE | ID: mdl-24358232

ABSTRACT

Combinations of targeted drugs have been employed to treat sarcomas, however, response rates have not improved notably, therefore emphasizing the need for novel treatments. In addition, imaging approaches to assess therapeutic response is lacking, as currently measurable indices, such as volume and/or diameter, do not accurately correlate with changes in tumor biology. In this study, quantitative and profound analyses of magnetic resonance imaging (MRI) were developed to evaluate these as imaging biomarkers for MK1775 and Gem in an osteosarcoma xenotransplant model at early time-points following treatment. Notably, we showed that Gem and Gem+MK1775 groups had significantly inhibited tumor growth by day 4, which was presaged by elevations in mean ADC by 24 hours post treatment. Significant differences were also observed at later time points for the Gem+MK1775 combination and MK1775 therapy. ADC distribution and entropy (randomness of ADC values) were also elevated by 24 hours following therapy. Immunohistochemistry demonstrated that these treatment-related increases in ADC correlated with apoptosis and observed cell condensations (dense- and exploded bodies). These findings underline the role of ADC as a quantitative imaging biomarker for therapy-induced response and show promising clinical relevance in the sarcoma patient population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/diagnosis , Bone Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Diffusion Magnetic Resonance Imaging , Osteosarcoma/diagnosis , Osteosarcoma/drug therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Animals , Bone Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Deoxycytidine/administration & dosage , Female , Humans , Mice , Mice, SCID , Middle Aged , Molecular Targeted Therapy , Osteosarcoma/pathology , Prognosis , Pyrimidinones , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine
12.
J Proteome Res ; 12(9): 4005-17, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23901793

ABSTRACT

While targeted therapy based on the idea of attenuating the activity of a preselected, therapeutically relevant protein has become one of the major trends in modern cancer therapy, no truly specific targeted drug has been developed and most clinical agents have displayed a degree of polypharmacology. Therefore, the specificity of anticancer therapeutics has emerged as a highly important but severely underestimated issue. Chemical proteomics is a powerful technique combining postgenomic drug-affinity chromatography with high-end mass spectrometry analysis and bioinformatic data processing to assemble a target profile of a desired therapeutic molecule. Due to high demands on the starting material, however, chemical proteomic studies have been mostly limited to cancer cell lines. Herein, we report a down-scaling of the technique to enable the analysis of very low abundance samples, as those obtained from needle biopsies. By a systematic investigation of several important parameters in pull-downs with the multikinase inhibitor bosutinib, the standard experimental protocol was optimized to 100 µg protein input. At this level, more than 30 well-known targets were detected per single pull-down replicate with high reproducibility. Moreover, as presented by the comprehensive target profile obtained from miniaturized pull-downs with another clinical drug, dasatinib, the optimized protocol seems to be extendable to other drugs of interest. Sixty distinct human and murine targets were finally identified for bosutinib and dasatinib in chemical proteomic experiments utilizing core needle biopsy samples from xenotransplants derived from patient tumor tissue. Altogether, the developed methodology proves robust and generic and holds many promises for the field of personalized health care.


Subject(s)
Aniline Compounds/chemistry , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Nitriles/chemistry , Protein-Tyrosine Kinases/isolation & purification , Pyrimidines/chemistry , Quinolines/chemistry , Thiazoles/chemistry , Animals , Biopsy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Chromatography, Affinity , Dasatinib , Humans , K562 Cells , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Annotation , Molecular Targeted Therapy , Neoplasm Transplantation , Protein Interaction Maps , Protein Kinase Inhibitors/chemistry , Proteomics
13.
PLoS One ; 8(3): e57523, 2013.
Article in English | MEDLINE | ID: mdl-23520471

ABSTRACT

Sarcomas are rare and heterogeneous mesenchymal tumors affecting both pediatric and adult populations with more than 70 recognized histologies. Doxorubicin and ifosfamide have been the main course of therapy for treatment of sarcomas; however, the response rate to these therapies is about 10-20% in metastatic setting. Toxicity with the drug combination is high, response rates remain low, and improvement in overall survival, especially in the metastatic disease, remains negligible and new agents are needed. Wee1 is a critical component of the G2/M cell cycle checkpoint control and mediates cell cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by MK1775 has been reported to enhance the cytotoxic effect of DNA damaging agents in different types of carcinomas. In this study we investigated the therapeutic efficacy of MK1775 in various sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo both alone and in combination with gemcitabine, which is frequently used in the treatment of sarcomas. Our data demonstrate that MK1775 treatment as a single agent at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. Additionally, MK1775 significantly enhances the cytotoxic effect of gemcitabine in sarcoma cells lines with different p53 mutational status. In patient-derived bone and soft tissue sarcoma samples we showed that MK1775 alone and in combination with gemcitabine causes significant apoptotic cell death. Magnetic resonance imaging (MRI) and histopathologic studies showed that MK1775 induces significant cell death and terminal differentiation in a patient-derived xenograft mouse model of osteosarcoma in vivo. Our results together with the high safety profile of MK1775 strongly suggest that this drug can be used as a potential therapeutic agent in the treatment of both adult as well as pediatric sarcoma patients.


Subject(s)
Antimetabolites, Antineoplastic , Cell Cycle Proteins/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Femoral Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Osteosarcoma/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles , Pyrimidines , Adolescent , Adult , Animals , Antimetabolites, Antineoplastic/agonists , Antimetabolites, Antineoplastic/pharmacology , Cell Death , Cell Differentiation , Cell Line, Tumor , Child , Child, Preschool , Deoxycytidine/agonists , Deoxycytidine/pharmacology , Drug Synergism , Female , Femoral Neoplasms/pathology , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Osteosarcoma/pathology , Pyrazoles/agonists , Pyrazoles/pharmacology , Pyrimidines/agonists , Pyrimidines/pharmacology , Pyrimidinones , Transplantation, Heterologous , Xenograft Model Antitumor Assays , Gemcitabine
14.
Diagn Cytopathol ; 41(2): 159-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23335454

ABSTRACT

Squamous cell carcinoma with rhabdoid features (SCCRF) is a very rare and unusual cutaneous tumor. Here, we report an extraordinary case diagnosed by fine needle aspiration biopsy, in a 66-year-old man, status post multiple organ transplantation. The patient presented with a large ulcerating fungating mass in his forearm that had all the light microscopic and immunohistochemical features of a SCCRF. Previously six cases of SCCRF phenotype diagnosed by surgical pathology have been reported. This is the first case diagnosed cytologically. A review of the literature with emphasis on the differential diagnoses of such unusual rhabdoid-like tumors in fine-needle aspiration biopsy and the potential molecular mechanism for rhabdoid phenotype in transplant patients are discussed.


Subject(s)
Carcinoma, Squamous Cell/pathology , Skin Neoplasms/pathology , Soft Tissue Neoplasms/pathology , Aged , Biopsy, Fine-Needle , Carcinoma, Squamous Cell/diagnosis , Cell Nucleus/pathology , Forearm , Humans , Male , Organ Transplantation , Phenotype , Rhabdoid Tumor/pathology , Skin Neoplasms/diagnosis , Soft Tissue Neoplasms/diagnosis
15.
Cancer ; 119(5): 1023-32, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23065656

ABSTRACT

BACKGROUND: Chemoprevention for lung cancer with nutraceutical or anti-inflammatory agents has had mixed clinical benefit. Novel targeted agents hold the promise of greater efficacy and selectivity. The authors of this report evaluated enzastaurin, a selective protein kinase C-ß (PKC-ß) inhibitor with antiproliferative and proapoptotic properties, in former smokers. METHODS: The primary objective of this study was to compare the average fraction of Ki-67-stained cells (the Ki-67 labeling index [LI]) in bronchial biopsy specimens that were collected before and after treatment. Participants were randomized (2:1) to receive either 6 months of daily oral enzastaurin (500 mg) or placebo. Stratification was based on morphology, history of lung cancer, and airway obstruction. RESULTS: In pretrial investigations, the rationale for PKC-ß inhibition and pathway interrogation was established in premalignant lesions and early stage lung cancer. In an intent-to-treat analysis, of 40 randomized participants, there was no significant difference in the pretreatment/post-treatment change in the Ki-67 LI between the enzastaurin group and the placebo group (P = .53). Six participants discontinued enzastaurin, including 4 participants who had adverse events, including abdominal distension, deep vein thrombosis, hyponatremia, and rash, and 2 participants who decided to discontinue. One participant in the placebo group was discontinued on the study because of noncompliance. Two participants had ≥1 serious adverse event (bradycardia, deep vein thrombosis, and hypotension). CONCLUSIONS: To the authors' knowledge, this represents the first chemoprevention trial with a non-US Food and Drug Administration-approved, oral, small-molecule-targeted agent. Although the primary endpoint was not met, enzastaurin was tolerable for 6 months by 75% of participants, and there was a suggestion of response in a subset analysis that was restricted to those who had metaplastic or dysplastic lesions.


Subject(s)
Indoles/pharmacology , Lung Neoplasms/prevention & control , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Smoking/adverse effects , Aged , Female , Humans , Indoles/adverse effects , Male , Middle Aged , Precancerous Conditions/drug therapy , Protein Kinase C beta , Risk
16.
Int J Clin Exp Pathol ; 5(9): 982-90, 2012.
Article in English | MEDLINE | ID: mdl-23119117

ABSTRACT

Mixed germ cell tumors are rare neoplasms that are known to occur in the anterior mediastinum. Characterized by two or more types of germ cell components, these tumors comprise upwards of 25% of mediastinal germ cell tumors. Even rarer are those harboring somatic-type malignancies such as carcinoma, sarcoma, and hematopoietic malignancies. To date, however, there are no known cases of melanoma arising in a malignant mixed germ cell tumor of the anterior mediastinum. We describe the first case of malignant melanoma with spindle and epithelioid components arising from respiratory epithelium in a mediastinal malignant mixed germ cell tumor of a 32-year-old male. In addition, we also provide evidence supporting the theory of neuroendocrine cells as the origin of melanoma arising in the respiratory epithelium. This case emphasizes the need to carefully evaluate all germ cell tumors, not only for a myriad of benign embryological components, but also for malignancies arising in these components, as they might change the prognosis and patient's course of treatment. This microscopic approach should bring to light the diversity of mixed germ cell tumors in addition to somatic malignancies with corresponding biologic potentials.


Subject(s)
Mediastinal Neoplasms/pathology , Melanoma/pathology , Neoplasms, Complex and Mixed/pathology , Respiratory Mucosa/pathology , Teratoma/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biopsy , Chemotherapy, Adjuvant , Humans , Immunohistochemistry , Male , Mediastinal Neoplasms/chemistry , Mediastinal Neoplasms/therapy , Melanoma/chemistry , Melanoma/therapy , Neoadjuvant Therapy , Neoplasms, Complex and Mixed/chemistry , Neoplasms, Complex and Mixed/therapy , Respiratory Mucosa/chemistry , Teratoma/chemistry , Teratoma/therapy , Tomography, X-Ray Computed
17.
Cancer Res ; 72(21): 5483-93, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22942258

ABSTRACT

Autophagy attenuates the efficacy of conventional chemotherapy but its effects on immunotherapy have been little studied. Here, we report that chemotherapy renders tumor cells more susceptible to lysis by CTL in vivo. Moreover, bystander tumor cells that did not express antigen were killed by CTL. This effect was mediated by transient but dramatic upregulation of the mannose-6-phosphate receptor (MPR) on the tumor cell surface. Antitumor effects of combined treatment related to the kinetics of MPR upregulation and abrogation of this event abolished the combined effect of immunotherapy and chemotherapy. MPR accumulation on the tumor cell surface during chemotherapy was observed in different mouse tumor models and in patients with multiple myeloma. Notably, this effect was the result of redistribution of the receptor caused by chemotherapy-inducible autophagy. Together, our findings reveal one molecular mechanism through which the antitumor effects of conventional cancer chemotherapy and immunotherapy are realized.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cytotoxicity, Immunologic/physiology , Immunotherapy/methods , Neoplasms/therapy , Animals , Blotting, Western , Combined Modality Therapy , Drug Therapy , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Neoplasms/immunology , Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation
18.
Mol Cancer ; 11: 73, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23009336

ABSTRACT

BACKGROUND: Cancer stem cells are thought to be responsible for the initiation and progression of cancers. In non-small cell lung cancers (NSCLCs), Hoechst 33342 dye effluxing side population (SP) cells are shown to have stem cell like properties. The oncogenic capacity of cancer stem-like cells is in part due to their ability to self-renew; however the mechanistic correlation between oncogenic pathways and self-renewal of cancer stem-like cells has remained elusive. Here we characterized the SP cells at the molecular level and evaluated its ability to generate tumors at the orthotopic site in the lung microenvironment. Further, we investigated if the self-renewal of SP cells is dependent on EGFR mediated signaling. RESULTS: SP cells were detected and isolated from multiple NSCLC cell lines (H1650, H1975, A549), as well as primary human tumor explants grown in nude mice. SP cells demonstrated stem-like properties including ability to self-renew and grow as spheres; they were able to generate primary and metastatic tumors upon orthotopic implantation into the lung of SCID mice. In vitro study revealed elevated expression of stem cell associated markers like Oct4, Sox2 and Nanog as well as demonstrated intrinsic epithelial to mesenchymal transition features in SP cells. Further, we show that abrogation of EGFR, Src and Akt signaling through pharmacological or genetic inhibitors suppresses the self-renewal growth and expansion of SP-cells and resulted in specific downregulation of Sox2 protein expression. siRNA mediated depletion of Sox2 significantly blocked the SP phenotype as well as its self-renewal capacity; whereas other transcription factors like Oct4 and Nanog played a relatively lesser role in regulating self-renewal. Interestingly, Sox2 was elevated in metastatic foci of human NSCLC samples. CONCLUSIONS: Our findings suggest that Sox2 is a novel target of EGFR-Src-Akt signaling in NSCLCs that modulates self-renewal and expansion of stem-like cells from NSCLC. Therefore, the outcome of the EGFR-Src-Akt targeted therapy may rely upon the expression and function of Sox2 within the NSCLC-CSCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , SOXB1 Transcription Factors/genetics , src-Family Kinases/metabolism , Animals , Biomarkers/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Progression , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Side-Population Cells/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Cancer Res ; 72(10): 2501-11, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22461510

ABSTRACT

Driver tyrosine kinase mutations are rare in sarcomas, and patterns of tyrosine phosphorylation are poorly understood. To better understand the signaling pathways active in sarcoma, we examined global tyrosine phosphorylation in sarcoma cell lines and human tumor samples. Anti-phosphotyrosine antibodies were used to purify tyrosine phosphorylated peptides, which were then identified by liquid chromatography and tandem mass spectrometry. The findings were validated with RNA interference, rescue, and small-molecule tyrosine kinase inhibitors. We identified 1,936 unique tyrosine phosphorylated peptides, corresponding to 844 unique phosphotyrosine proteins. In sarcoma cells alone, peptides corresponding to 39 tyrosine kinases were found. Four of 10 cell lines showed dependence on tyrosine kinases for growth and/or survival, including platelet-derived growth factor receptor (PDGFR)α, MET, insulin receptor/insulin-like growth factor receptor signaling, and SRC family kinase signaling. Rhabdomyosarcoma samples showed overexpression of PDGFRα in 13% of examined cases, and sarcomas showed abundant tyrosine phosphorylation and expression of a number of tyrosine phosphorylated tyrosine kinases, including DDR2, EphB4, TYR2, AXL, SRC, LYN, and FAK. Together, our findings suggest that integrating global phosphoproteomics with functional analyses with kinase inhibitors can identify drivers of sarcoma growth and survival.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proteomics/methods , Sarcoma/enzymology , Cell Line, Tumor , Humans , Phosphorylation , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction
20.
Mol Cancer Ther ; 11(1): 174-82, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22084170

ABSTRACT

Wee1 is a critical component of the G(2)-M cell-cycle checkpoint control and mediates cell-cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by a selective small molecule inhibitor MK1775 can abrogate G(2)-M checkpoint, resulting in premature mitotic entry and cell death. MK1775 has recently been tested in preclinical and clinical studies of human carcinoma to enhance the cytotoxic effect of DNA-damaging agents. However, its role in mesenchymal tumors, especially as a single agent, has not been explored. Here, we studied the cytotoxic effect of MK1775 in various sarcoma cell lines and patient-derived tumor explants ex vivo. Our data show that MK1775 treatment at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. In MK1775-treated cells, CDC2 activity was enhanced, as determined by decreased inhibitory phosphorylation of tyrosine-15 residue and increased expression of phosphorylated histone H3, a marker of mitotic entry. The cytotoxic effect of Wee1 inhibition on sarcoma cells seems to be independent of p53 status as all sarcoma cell lines with different p53 mutation were highly sensitive to MK1775 treatment. Finally, in patient-derived sarcoma samples, we showed that MK1775 as a single agent causes significant apoptotic cell death, suggesting that Wee1 inhibition may represent a novel approach in the treatment of sarcomas.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Mitosis/drug effects , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sarcoma/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B/metabolism , Cyclin-Dependent Kinases , Histones/metabolism , Humans , Phosphorylation/drug effects , Pyrimidinones , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...