Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell Rep ; 24(12): 3237-3250, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30232005

ABSTRACT

Differentiation events contribute to phenotypic cellular heterogeneity within tumors and influence disease progression and response to therapy. Here, we dissect mechanisms controlling intratumoral heterogeneity within triple-negative basal-like breast cancers. Tumor cells expressing the cytokeratin K14 possess a differentiation state that is associated with that of normal luminal progenitors, and K14-negative cells are in a state closer to that of mature luminal cells. We show that cells can transition between these states through asymmetric divisions, which produce one K14+ and one K14- daughter cell, and that these asymmetric divisions contribute to the generation of cellular heterogeneity. We identified several regulators that control the proportion of K14+ cells in the population. EZH2 and Notch increase the numbers of K14+ cells and their rates of symmetric divisions, and FOXA1 has an opposing effect. Our findings demonstrate that asymmetric divisions generate differentiation transitions and heterogeneity, and identify pathways that control breast cancer cellular composition.


Subject(s)
Asymmetric Cell Division , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Keratins/genetics , Keratins/metabolism , Mice , Receptors, Notch/genetics , Receptors, Notch/metabolism , Triple Negative Breast Neoplasms/metabolism
3.
Hum Cell ; 30(3): 226-236, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28251557

ABSTRACT

High grade serous ovarian cancer (HGSOC) patients have a high recurrence rate after surgery and adjuvant chemotherapy due to inherent or acquired drug resistance. Cell lines derived from HGSOC tumors that are resistant to chemotherapeutic agents represent useful pre-clinical models for drug discovery. Here, we describe establishment of a human ovarian carcinoma cell line, which we term WHIRC01, from a patient-derived mouse xenograft established from a chemorefractory HGSOC patient who did not respond to carboplatin and paclitaxel therapy. This newly derived cell line is platinum- and paclitaxel-resistant with cisplatin, carboplatin, and paclitaxel half-maximal lethal doses of 15, 130, and 20 µM, respectively. Molecular characterization of this cell line was performed using targeted DNA exome sequencing, transcriptomics (RNA-seq), and mass spectrometry-based proteomic analyses. Results from exomic sequencing revealed mutations in TP53 consistent with HGSOC. Transcriptomic and proteomic analyses of WHIRC01 showed high level of alpha-enolase and vimentin, which are associated with cell migration and epithelial-mesenchymal transition. WHIRC01 represents a chemorefractory human HGSOC cell line model with a comprehensive molecular profile to aid future investigations of drug resistance mechanisms and screening of chemotherapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Carcinoma/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Animals , Carcinoma/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Discovery , Exome/genetics , Female , Gene Expression Profiling , Heterografts , Humans , Mice , Mutation , Neoplasm Staging , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Proteomics , Tumor Suppressor Protein p53/genetics
4.
Clin Lung Cancer ; 18(4): e223-e232, 2017 07.
Article in English | MEDLINE | ID: mdl-28082048

ABSTRACT

BACKGROUND: RET (rearranged during transfection) fusions have been reported in 1% to 2% of lung adenocarcinoma (LADC) cases. In contrast, KIF5B-RET and CCDC6-RET fusion genes have been identified in 70% to 90% and 10% to 25% of tumors, respectively. The natural history and management of RET-rearranged LADC are still being delineated. MATERIALS AND METHODS: We present a series of 14 patients with RET-rearranged LADC. The response to therapy was assessed by the clinical response and an avatar model in 2 cases. Patients underwent chemotherapy, targeted therapy, and immunotherapy. RESULTS: A total of 14 patients (8 women; 10 never smokers; 4 light smokers; mean age, 57 years) were included. KIF5B-RET and CCDC6-RET variants were diagnosed in 10 and 4 cases, respectively. Eight patients had an early disseminated manifestation, seven with KIF5B-RET rearranged tumor. The features of this subset included bilateral miliary lung metastases, bone metastases, and unusual early visceral abdominal involvement. One such patient demonstrated an early and durable complete response to cabozantinib for 7 months. Another 2 patients treated with cabozantinib experienced a partial response, with rapid significant clinical improvement. Four patients with tumors harboring CCDC6-RET and KIF5B-RET fusions showed pronounced and durable responses to platinum-based chemotherapy that lasted for 8 to 15 months. Two patients' tumors showed programmed cell death ligand 1-positive staining but did not respond to pembrolizumab. The median overall survival was 22.8 months. CONCLUSION: RET-rearranged LADC in our series tended to occur as bilateral disease with early visceral involvement, especially with KIF5B fusion. Treatment with cabozantinib achieved responses, including 1 complete response. However, further studies are required in this group of patients.


Subject(s)
Adenocarcinoma/drug therapy , Anilides/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Platinum Compounds/therapeutic use , Pyridines/therapeutic use , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adult , Aged , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-ret/genetics , Survival Analysis , Transcription Factors/genetics , Treatment Outcome
5.
Br J Cancer ; 116(4): 515-523, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28118322

ABSTRACT

BACKGROUND: Screening of patients for cancer-driving mutations is now used for cancer prognosis, remission scoring and treatment selection. Although recently emerged targeted next-generation sequencing-based approaches offer promising diagnostic capabilities, there are still limitations. There is a pressing clinical need for a well-validated, rapid, cost-effective mutation profiling system in patient specimens. Given their speed and cost-effectiveness, quantitative PCR mutation detection techniques are well suited for the clinical environment. The qBiomarker mutation PCR array has high sensitivity and shorter turnaround times compared with other methods. However, a direct comparison with existing viable alternatives are required to assess its true potential and limitations. METHODS: In this study, we evaluated a panel of 117 patient-derived tumour xenografts by the qBiomarker array and compared with other methods for mutation detection, including Ion AmpliSeq sequencing, whole-exome sequencing and droplet digital PCR. RESULTS: Our broad analysis demonstrates that the qBiomarker's performance is on par with that of other labour-intensive and expensive methods of cancer mutation detection of frequently altered cancer-associated genes, and provides a foundation for supporting its consideration as an option for molecular diagnostics. CONCLUSIONS: This large-scale direct comparison and validation of currently available mutation detection approaches is extremely relevant for the current scenario of precision medicine and will lead to informed choice of screening methodologies, especially in lower budget conditions or time frame limitations.


Subject(s)
DNA Mutational Analysis/methods , Heterografts , Neoplasms/genetics , Animals , Heterografts/metabolism , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Neoplasm Transplantation , Neoplasms/pathology , Polymerase Chain Reaction/methods , Reproducibility of Results , Tumor Cells, Cultured
6.
Nat Commun ; 7: 13701, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27922010

ABSTRACT

Lung cancer is the leading cause of cancer deaths, and effective treatments are urgently needed. Loss-of-function mutations in the DNA damage response kinase ATM are common in lung adenocarcinoma but directly targeting these with drugs remains challenging. Here we report that ATM loss-of-function is synthetic lethal with drugs inhibiting the central growth factor kinases MEK1/2, including the FDA-approved drug trametinib. Lung cancer cells resistant to MEK inhibition become highly sensitive upon loss of ATM both in vitro and in vivo. Mechanistically, ATM mediates crosstalk between the prosurvival MEK/ERK and AKT/mTOR pathways. ATM loss also enhances the sensitivity of KRAS- or BRAF-mutant lung cancer cells to MEK inhibition. Thus, ATM mutational status in lung cancer is a mechanistic biomarker for MEK inhibitor response, which may improve patient stratification and extend the applicability of these drugs beyond RAS and BRAF mutant tumours.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cell Proliferation/drug effects , Lung Neoplasms/prevention & control , Mutation , Protein Kinase Inhibitors/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , RNA Interference , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
7.
Clin Cancer Res ; 22(24): 6153-6163, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27384421

ABSTRACT

PURPOSE: Even when diagnosed prior to metastasis, pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with almost 90% lethality, emphasizing the need for new therapies optimally targeting the tumors of individual patients. EXPERIMENTAL DESIGN: We first developed a panel of new physiologic models for study of PDAC, expanding surgical PDAC tumor samples in culture using short-term culture and conditional reprogramming with the Rho kinase inhibitor Y-27632, and creating matched patient-derived xenografts (PDX). These were evaluated for sensitivity to a large panel of clinical agents, and promising leads further evaluated mechanistically. RESULTS: Only a small minority of tested agents was cytotoxic in minimally passaged PDAC cultures in vitro Drugs interfering with protein turnover and transcription were among most cytotoxic. Among transcriptional repressors, triptolide, a covalent inhibitor of ERCC3, was most consistently effective in vitro and in vivo causing prolonged complete regression in multiple PDX models resistant to standard PDAC therapies. Importantly, triptolide showed superior activity in MYC-amplified PDX models and elicited rapid and profound depletion of the oncoprotein MYC, a transcriptional regulator. Expression of ERCC3 and MYC was interdependent in PDACs, and acquired resistance to triptolide depended on elevated ERCC3 and MYC expression. The Cancer Genome Atlas analysis indicates ERCC3 expression predicts poor prognosis, particularly in CDKN2A-null, highly proliferative tumors. CONCLUSIONS: This provides initial preclinical evidence for an essential role of MYC-ERCC3 interactions in PDAC, and suggests a new mechanistic approach for disruption of critical survival signaling in MYC-dependent cancers. Clin Cancer Res; 22(24); 6153-63. ©2016 AACR.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Amides/pharmacology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Heterografts/metabolism , Humans , Mice , Mice, SCID , NIH 3T3 Cells , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phenanthrenes/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Transcription, Genetic/drug effects , rho-Associated Kinases/metabolism , Pancreatic Neoplasms
8.
Oncotarget ; 7(29): 46263-46272, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27323832

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) represents a block in differentiation of malignant myoblasts. Genomic events implicated in the pathogenesis of ARMS involve PAX3-FKHR (FOXO1) or PAX7-FKHR (FOXO1) translocation with corresponding fusion transcripts and fusion proteins. Commonalities in ARMS include uncontrollable proliferation and failure to differentiate. The genomic-molecular correlates contributing to the etiopathogenesis of ARMS incorporate PAX3-FKHR (FOXO1) fusion protein stimulation of the IGF-1R, c-Met and GSK3-ß pathways. With sequential morphoproteomic profiling on such a case in conjunction with personalized tumor graft testing, we provide an expanded definition of the biology of PAX3-FKHR (FOXO1) ARMS that integrates genomics, proteomics and pharmacogenomics. Moreover, therapies that target the genomic and molecular biology and lead to tumoral regression and/or tumoral growth inhibition in a xenograft model of ARMS are identified. SIGNIFICANCE: This case study could serve as a model for clinical trials using relatively low toxicity agents in both initial and maintenance therapies to induce remission and reduce the risk of recurrent disease in PAX3-FKHR (FOXO1) subtype of ARMS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Precision Medicine/methods , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Animals , Cell Proliferation/drug effects , Child , Hand/pathology , Humans , Male , Mice , Oncogene Proteins, Fusion/genetics , Proteomics , Xenograft Model Antitumor Assays
9.
Cancer Biol Ther ; 16(8): 1184-93, 2015.
Article in English | MEDLINE | ID: mdl-26046946

ABSTRACT

Triple negative breast cancer has an extremely poor prognosis when chemotherapy is no longer effective. To overcome drug resistance, novel drug delivery systems based on nanoparticles have had remarkable success. We produced a novel nanoparticle component 'MDC' from milk-derived colloid. In order to evaluate the anti-cancer effect of MDC, we conducted in vitro and in vivo experiments on cancer cell lines and a primary tumor derived breast xenograft. Doxorubicin (Dox) conjugated to MDC (MDC-Dox) showed higher cancer cell growth inhibition than MDC alone especially in cell lines with high EGFR expression. In a mouse melanoma model, MDC-Dox significantly suppressed tumor growth when compared with free Dox. Moreover, in a primary tumor derived breast xenograft, one of the mice treated with MDC-Dox showed partial regression, while mice treated with free Dox failed to show any suppression of tumor growth. We have shown that a novel nanoparticle compound made of simple milk-derived colloid has the capability for drug conjugation, and serves as a tumor-specific carrier of anti-cancer drugs. Further research on its safety and ability to carry various anti-cancer drugs into multiple drug-resistant primary breast models is warranted.


Subject(s)
Colloids/chemistry , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Milk/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor/drug effects , Colloids/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Delivery Systems/methods , Female , Humans , Immunocompromised Host , Melanoma/drug therapy , Melanoma/pathology , Mice, Inbred C57BL , Nanoparticles/chemistry , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
J Proteome Res ; 14(4): 1900-10, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25748058

ABSTRACT

A majority of high-grade (HG) serous ovarian cancer (SOC) patients develop resistant disease despite high initial response rates to platinum/paclitaxel-based chemotherapy. We identified shed/secreted proteins in preclinical models of paclitaxel-resistant human HGSOC models and correlated these candidate proteins with patient outcomes using public data from HGSOC patients. Proteomic analyses of a HGSOC cell line secretome was compared to those from a syngeneic paclitaxel-resistant variant and from a line established from an intrinsically chemorefractory HGSOC patient. Associations between the identified candidate proteins and patient outcome were assessed in a discovery cohort of 545 patients and two validation cohorts totaling 795 independent SOC patients. Among the 81 differentially abundant proteins identified (q < 0.05) from paclitaxel-sensitive vs -resistant HGSOC cell secretomes, AKAP12 was verified to be elevated in all models of paclitaxel-resistant HGSOC. Furthermore, elevated AKAP12 transcript expression was associated with worse progression-free and overall survival. Associations with outcome were observed in three independent cohorts and remained significant after adjusted multivariate modeling. We further provide evidence to support that differential gene methylation status is associated with elevated expression of AKAP12 in taxol-resistant ovarian cancer cells and ovarian cancer patient subsets. Elevated expression and shedding/secretion of AKAP12 is characteristic of paclitaxel-resistant HGSOC cells, and elevated AKAP12 transcript expression is a poor prognostic and predictive marker for progression-free and overall survival in SOC patients.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Methylation/genetics , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/physiology , Ovarian Neoplasms/diagnosis , Paclitaxel/metabolism , Cohort Studies , Female , Humans , Patient Outcome Assessment , Prognosis , Proteomics/methods
11.
Hum Genome Var ; 2: 15009, 2015.
Article in English | MEDLINE | ID: mdl-27081524

ABSTRACT

Cetuximab, a monoclonal antibody against epidermal growth factor receptor (EGFR), was shown to be active in colorectal cancer. Although some patients who harbor K-ras wild-type tumors benefit from cetuximab treatment, 40 to 60% of patients with wild-type K-ras tumors do not respond to cetuximab. Currently, there is no universal marker or method of clinical utility that could guide the treatment of cetuximab in colorectal cancer. Here, we demonstrate a method to predict response to cetuximab in patients with colorectal cancer using OncoFinder pathway activation strength (PAS), based on the transcriptomic data of the tumors. We first evaluated our OncoFinder pathway activation strength model in a set of transcriptomic data obtained from patient-derived xenograft (PDx) models established from colorectal cancer biopsies. Then, the approach and models were validated using a clinical trial data set. PAS could efficiently predict patients' response to cetuximab, and thus holds promise as a selection criterion for cetuximab treatment in metastatic colorectal cancer.

12.
Cancer Biol Ther ; 15(9): 1208-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24921944

ABSTRACT

Stem cell factor receptor (c-Kit) exerts multiple biological effects on target cells upon binding its ligand stem cell factor (SCF). Aberrant activation of c-Kit results in dysregulated signaling and is implicated in the pathogenesis of numerous cancers. The development of more specific and effective c-Kit therapies is warranted given its essential role in tumorigenesis. In this study, we describe the biological properties of CK6, a fully human IgG1 monoclonal antibody against the extracellular region of human c-Kit. CK6 specifically binds c-Kit receptor with high affinity (EC 50 = 0.06 nM) and strongly blocks its interaction with SCF (IC 50 = 0.41 nM) in solid phase assays. Flow cytometry shows CK6 binding to c-Kit on the cell surface of human small cell lung carcinoma (SCLC), melanoma, and leukemia tumor cell lines. Furthermore, exposure to CK6 inhibits SCF stimulation of c-Kit tyrosine kinase activity and downstream signaling pathways such as mitogen-activated protein kinase (MAPK) and protein kinase B (AKT), in addition to reducing tumor cell line growth in vitro. CK6 treatment significantly decreases human xenograft tumor growth in NCI-H526 SCLC (T/C% = 57) and Malme-3M melanoma (T/C% = 58) models in vivo. The combination of CK6 with standard of care chemotherapy agents, cisplatin and etoposide for SCLC or dacarbazine for melanoma, more potently reduces tumor growth (SCLC T/C% = 24, melanoma T/C% = 38) compared with CK6 or chemotherapy alone. In summary, our results demonstrate that CK6 is a c-Kit antagonist antibody with tumor growth neutralizing properties and are highly suggestive of potential therapeutic application in treating human malignancies harboring c-Kit receptor.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoglobulin G/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Proto-Oncogene Proteins c-kit/metabolism , Small Cell Lung Carcinoma/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Etoposide/administration & dosage , Female , Heterografts , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
13.
Cancer Res ; 74(14): 3995-4005, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24830724

ABSTRACT

Although specific mutations in the tyrosine kinase domain of epidermal growth factor receptor (EGFR) identify tumors that are responsive to EGFR tyrosine kinase inhibitors (TKI), these genetic alterations are present in only a minority of patients. Patients with tumors expressing wild-type EGFR lack reliable predictive markers of their clinical response to EGFR TKIs. Although epithelial-mesenchymal transition (EMT) has been inversely correlated with the response of cancers to EGFR-targeted therapy, the precise molecular mechanisms underlying this association have not been defined and no specific EMT-associated biomarker of clinical benefit has been identified. Here, we show that during transforming growth factor ß (TGFß)-mediated EMT, inhibition of the microRNAs 200 (miR200) family results in upregulated expression of the mitogen-inducible gene 6 (MIG6), a negative regulator of EGFR. The MIG6-mediated reduction of EGFR occurs concomitantly with a TGFß-induced EMT-associated kinase switch of tumor cells to an AKT-activated EGFR-independent state. In a panel of 25 cancer cell lines of different tissue origins, we find that the ratio of the expression levels of MIG6 and miR200c is highly correlated with EMT and resistance to erlotinib. Analyses of primary tumor xenografts of patient-derived lung and pancreatic cancers carrying wild-type EGFR showed that the tumor MIG6(mRNA)/miR200 ratio was inversely correlated with response to erlotinib in vivo. Our data demonstrate that the TGFß-miR200-MIG6 network orchestrates the EMT-associated kinase switch that induces resistance to EGFR inhibitors, and identify a low ratio of MIG6 to miR200 as a promising predictive biomarker of the response of tumors to EGFR TKIs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , ErbB Receptors/antagonists & inhibitors , MicroRNAs/genetics , Protein Kinase Inhibitors/pharmacology , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Organ Specificity/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
14.
Mol Cancer Ther ; 13(6): 1611-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24688052

ABSTRACT

KRAS gene mutation is linked to poor prognosis and resistance to therapeutics in non-small cell lung cancer (NSCLC). In this study, we have explored the possibility of exploiting inherent differences in KRAS-mutant cell metabolism for treatment. This study identified a greater dependency on folate metabolism pathways in KRAS mutant compared with KRAS wild-type NSCLC cell lines. Microarray gene expression and biologic pathway analysis identified higher expression of folate metabolism- and purine synthesis-related pathways in KRAS-mutant NSCLC cells compared with wild-type counterparts. Moreover, pathway analysis and knockdown studies suggest a role for MYC transcriptional activity in the expression of these pathways in KRAS-mutant NSCLC cells. Furthermore, KRAS knockdown and overexpression studies demonstrated the ability of KRAS to regulate expression of genes that comprise folate metabolism pathways. Proliferation studies demonstrated higher responsiveness to methotrexate, pemetrexed, and other antifolates in KRAS-mutant NSCLC cells. Surprisingly, KRAS gene expression is downregulated in KRAS wild-type and KRAS-mutant cells by antifolates, which may also contribute to higher efficacy of antifolates in KRAS-mutant NSCLC cells. In vivo analysis of multiple tumorgraft models in nude mice identified a KRAS-mutant tumor among the pemetrexed-responsive tumors and also demonstrated an association between expression of the folate pathway gene, methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), and antifolate activity. Collectively, we identify altered regulation of folate metabolism in KRAS-mutant NSCLC cells that may account for higher antifolate activity in this subtype of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Folic Acid/metabolism , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mutation , Proto-Oncogene Proteins p21(ras) , Xenograft Model Antitumor Assays
15.
Cancer ; 120(13): 2006-15, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24705963

ABSTRACT

BACKGROUND: Patients with advanced, metastatic sarcoma have a poor prognosis, and the overall benefit from the few standard-of-care therapeutics available is small. The rarity of this tumor, combined with the wide range of subtypes, leads to difficulties in conducting clinical trials. The authors previously reported the outcome of patients with a variety of common solid tumors who received treatment with drug regimens that were first tested in patient-derived xenografts using a proprietary method ("TumorGrafts"). METHODS: Tumors resected from 29 patients with sarcoma were implanted into immunodeficient mice to identify drug targets and drugs for clinical use. The results of drug sensitivity testing in the TumorGrafts were used to personalize cancer treatment. RESULTS: Of 29 implanted tumors, 22 (76%) successfully engrafted, permitting the identification of treatment regimens for these patients. Although 6 patients died before the completion of TumorGraft testing, a correlation between TumorGraft results and clinical outcome was observed in 13 of 16 (81%) of the remaining individuals. No patients progressed during the TumorGraft-predicted therapy. CONCLUSIONS: The current data support the use of the personalized TumorGraft model as an investigational platform for therapeutic decision-making that can guide treatment for rare tumors such as sarcomas. A randomized phase 3 trial versus physician's choice is warranted.


Subject(s)
Heterografts , Precision Medicine/methods , Sarcoma/surgery , Transplantation, Heterologous , Aged , Animals , Child , Chondrosarcoma/surgery , Female , Humans , Leiomyosarcoma/surgery , Liposarcoma/surgery , Male , Mice , Mice, SCID , Middle Aged , Myxoma/surgery , Rhabdomyosarcoma/surgery , Sarcoma/genetics , Sarcoma/pathology , Sarcoma/secondary , Sarcoma, Ewing/surgery , Sarcoma, Synovial/surgery , Treatment Outcome
16.
Clin Cancer Res ; 20(9): 2476-84, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24634382

ABSTRACT

BACKGROUND: Current technology permits an unbiased massive analysis of somatic genetic alterations from tumor DNA as well as the generation of individualized mouse xenografts (Avatar models). This work aimed to evaluate our experience integrating these two strategies to personalize the treatment of patients with cancer. METHODS: We performed whole-exome sequencing analysis of 25 patients with advanced solid tumors to identify putatively actionable tumor-specific genomic alterations. Avatar models were used as an in vivo platform to test proposed treatment strategies. RESULTS: Successful exome sequencing analyses have been obtained for 23 patients. Tumor-specific mutations and copy-number variations were identified. All samples profiled contained relevant genomic alterations. Tumor was implanted to create an Avatar model from 14 patients and 10 succeeded. Occasionally, actionable alterations such as mutations in NF1, PI3KA, and DDR2 failed to provide any benefit when a targeted drug was tested in the Avatar and, accordingly, treatment of the patients with these drugs was not effective. To date, 13 patients have received a personalized treatment and 6 achieved durable partial remissions. Prior testing of candidate treatments in Avatar models correlated with clinical response and helped to select empirical treatments in some patients with no actionable mutations. CONCLUSION: The use of full genomic analysis for cancer care is encouraging but presents important challenges that will need to be solved for broad clinical application. Avatar models are a promising investigational platform for therapeutic decision making. While limitations still exist, this strategy should be further tested.


Subject(s)
High-Throughput Nucleotide Sequencing , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine , Adult , Aged , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Computational Biology , DNA Copy Number Variations , Disease Models, Animal , Exome , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/chemistry , Female , Genomics , Humans , Male , Mice , Middle Aged , Models, Molecular , Mutation , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors , Protein Conformation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry , Retrospective Studies , Treatment Outcome , Tumor Burden , Xenograft Model Antitumor Assays
17.
Anticancer Res ; 31(6): 2149-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21737635

ABSTRACT

BACKGROUND: Clinically relevant targets for developmental drug efficacy in animal models of cancer are critical yet understudied parameters. MATERIALS AND METHODS: Cetuximab, a chimeric antibody to epidermal growth factor receptor (EGFR), was administered to athymic mice bearing subcutaneous tumors established with 13 human colorectal cancer cell lines of varying biomarker status, defined by DNA sequencing and RT-PCR. RESULTS: If tumor growth inhibition is taken as a target, as is commonly done, then in contrast to the clinical situation where KRAS mutation strongly predicts for a lack of clinically meaningful benefit in colorectal cancer patients, cetuximab alone and in combination with irinotecan-based chemotherapy were efficacious in a similar proportion of KRAS wild-type and mutant models. It was only when tumor regression was utilized to define relevant efficacy that cetuximab monotherapy was efficacious in KRAS wild-type, but not mutant models. Adding cytotoxic therapy to cetuximab treatment increased tumor regression frequency in both genotypes to the point that once again the response was similar for KRAS wild-type and mutant models. CONCLUSION: Our data support shifting the threshold for claiming clinically relevant targeted therapy efficacy in subcutaneous xenograft models towards tumor regression, rather than tumor growth inhibition, focusing on the evaluation of tumor cells that are addicted to the pathways being targeted.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, ras , Mutation , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/metabolism , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cetuximab , Colorectal Neoplasms/metabolism , Gene Dosage , Genes, erbB-1 , Humans , Irinotecan , Mice , Mice, Nude , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Proto-Oncogene Proteins B-raf/genetics , Xenograft Model Antitumor Assays
18.
IDrugs ; 11(1): 46-56, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18175263

ABSTRACT

An improved understanding of acute myelogenous leukemia (AML) over the past two decades has led to a characterization of associated recurring cytogenetic abnormalities. AML is often driven by the overexpression or constitutive activation of receptor tyrosine kinases such as Fms-like tyrosine kinase 3 (FLT3), which serves as a good therapeutic target. Millennium Pharmaceuticals Inc's tandutinib (MLN-518) is an orally active inhibitor of FLT3 kinase and family members PDGFR beta and c-Kit. Tandutinib inhibited FLT3 phosphorylation, downstream signaling and malignant growth in vitro and in animal models. The drug exhibited limited activity as a single agent in phase I and II clinical trials in patients with AML and myelodysplastic syndrome, but displayed promising antileukemic activity (90% complete remissions) in a phase I/II trial in patients with newly diagnosed AML when administered in combination with cytarabine and daunorubicin. Phase II clinical trials for tandutinib are ongoing in patients with AML or renal cell carcinoma.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Piperazines/therapeutic use , Quinazolines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Administration, Oral , Animals , Clinical Trials as Topic , Humans , Piperazines/administration & dosage , Quinazolines/administration & dosage
19.
Comb Chem High Throughput Screen ; 11(1): 62-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18220543

ABSTRACT

Recently, significant progress has been made towards understanding the pathogenesis of cancer from the molecular standpoint. To this end, a growing number of approaches are being exploited for the identification and validation of new therapeutic targets suitable for potent and specific intervention. The type 1 insulin-like growth factor receptor (IGF-1R) system has recently become the focus of major attention in the arena of cancer research. The involvement of the receptor and its downstream signaling cascades in the carcinogenesis process makes this system an excellent target for potential cancer therapy. Indeed, advances in the understanding of the molecular mechanisms behind IGF-1R activation have led to the discovery of agents designed selectively for targeting IGF-1R. The potential application of these inhibitors is currently under intense clinical investigation. This review describes the biology of IGF-1R particularly from a cancer perspective. The attempts to develop effective IGF-1R antagonists are discussed comprehensively with special emphasis on antibodies and small tyrosine kinase inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Blocking , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Line, Tumor , Humans , Neoplasms/metabolism , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/metabolism
20.
Cancer Res ; 66(5): 2544-52, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510571

ABSTRACT

Signal transducers and activators of transcription 3 (STAT3) is a transcription factor that is aberrantly activated in many cancer cells. Constitutively activated STAT3 is oncogenic, presumably as a consequence of the genes that it differentially regulates. Activated STAT3 correlated with elevated cyclin D1 protein in primary breast tumors and breast cancer-derived cell lines. Cyclin D1 mRNA levels were increased in primary rat-, mouse-, and human-derived cell lines expressing either the oncogenic variant of STAT3 (STAT3-C) or vSrc, which constitutively phosphorylates STAT3. Mutagenesis of STAT3 binding sites within the cyclin D1 promoter and chromatin immunoprecipitation studies showed an association between STAT3 and the transcriptional regulation of the human cyclin D1 gene. Introduction of STAT3-C and vSrc into immortalized cyclin D1(-/-) and cyclin D1(-/+) fibroblasts led to anchorage-independent growth of only cyclin D1(-/+) cells. Furthermore, knockdown of cyclin D1 in breast carcinoma cells led to a reduction in anchorage-independent growth. Phosphorylation of the retinoblastoma (Rb) protein [a target of the cyclin D1/cyclin-dependent kinase 4/6 (cdk4/6) holoenzyme] was delayed in the cyclin D1(-/-) cells relative to cyclin D1(-/+) cells. The E7 oncogene, whose activity includes degradation of Rb and dissociation of Rb from E2F, did not confer anchorage-independent growth to the cyclin D1(-/-) cells but, in conjunction with vSrc, resulted in robust growth in soft agar. These results suggest both a cdk-dependent and cdk-independent role for cyclin D1 in modulating transformation by different oncogenes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/biosynthesis , STAT3 Transcription Factor/metabolism , Animals , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , Cyclin D1/genetics , G1 Phase/genetics , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells , Papillomavirus E7 Proteins/genetics , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , STAT3 Transcription Factor/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...