Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Methods Mol Biol ; 1866: 163-171, 2019.
Article in English | MEDLINE | ID: mdl-30725415

ABSTRACT

This chapter reviews how total methionine (MET) restriction (MR) of a human brain tumor xenograft, effected by the combination of recombinant L-methionine-α-deamino-γ-lyase (rMETase) and a MET-free diet, greatly potentiates standard chemotherapy for brain tumors in mouse models. The growth of human brain tumor Daoy, SWB77, and D-54 xenografts in nude mice was arrested after the depletion of mouse plasma methionine (MET) with a combination of an MR diet and rMETase and homocysteine to rescue normal cells and tissues. MET was depleted to below 5 µm by this treatment. MR for 10-12 days inhibited tumor growth, but did not prevent tumor regrowth after treatment cessation. A single dose of N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU), which was ineffective alone, was administered at the end of the MR regimen, and caused a more than 80-day growth delay for Daoy and D-54 and a 20-day growth delay for SWB77. The total MR treatment regimens also increased the efficacy of temozolomide (TMZ) against the SWB77 xenograft when administered at the end of the MET regimen.


Subject(s)
Methionine/deficiency , Neoplasms/diet therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Choline , Homocysteine/metabolism , Humans , Liver/drug effects , Liver/pathology , Methionine/blood , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
2.
Neoplasia ; 19(10): 848-855, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28923537

ABSTRACT

This is the first prospective study of the effects of human gut microbiota and metabolites on immune checkpoint inhibitor (ICT) response in metastatic melanoma patients. Whereas many melanoma patients exhibit profound response to ICT, there are fewer options for patients failing ICT-particularly with BRAF-wild-type disease. In preclinical studies, specific gut microbiota promotes regression of melanoma in mice. We therefore conducted a study of the effects of pretreatment gut microbiota and metabolites on ICT Response Evaluation Criteria in Solid Tumors response in 39 metastatic melanoma patients treated with ipilimumab, nivolumab, ipilimumab plus nivolumab (IN), or pembrolizumab (P). IN yielded 67% responses and 8% stable disease; P achieved 23% responses and 23% stable disease. ICT responders for all types of therapies were enriched for Bacteroides caccae. Among IN responders, the gut microbiome was enriched for Faecalibacterium prausnitzii, Bacteroides thetaiotamicron, and Holdemania filiformis. Among P responders, the microbiome was enriched for Dorea formicogenerans. Unbiased shotgun metabolomics revealed high levels of anacardic acid in ICT responders. Based on these pilot studies, both additional confirmatory clinical studies and preclinical testing of these bacterial species and metabolites are warranted to confirm their ICT enhancing activity.


Subject(s)
Gastrointestinal Microbiome , Melanoma/etiology , Melanoma/metabolism , Metabolomics , Metagenome , Metagenomics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Chromatography, High Pressure Liquid , Cluster Analysis , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Metabolomics/methods , Metagenomics/methods , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Tandem Mass Spectrometry , Treatment Outcome
3.
Oncologist ; 22(4): 369-374, 2017 04.
Article in English | MEDLINE | ID: mdl-28314841

ABSTRACT

Multiple factors critical to the effectiveness of academic phase I cancer programs were assessed among 16 academic centers in the U.S. Successful cancer centers were defined as having broad phase I and I/II clinical trial portfolios, multiple investigator-initiated studies, and correlative science. The most significant elements were institutional philanthropic support, experienced clinical research managers, robust institutional basic research, institutional administrative efforts to reduce bureaucratic regulatory delays, phase I navigators to inform patients and physicians of new studies, and a large cancer center patient base. New programs may benefit from a separate stand-alone operation, but mature phase I programs work well when many of the activities are transferred to disease-oriented teams. The metrics may be useful as a rubric for new and established academic phase I programs. The Oncologist 2017;22:369-374.


Subject(s)
Academic Medical Centers , Neoplasms/epidemiology , Clinical Trials as Topic , Humans , Neoplasms/genetics , Program Development , United States
4.
Article in English | MEDLINE | ID: mdl-26316815

ABSTRACT

PURPOSE: This study interrogates the molecular status of individual cells in patients with triple-negative breast cancers and explores the molecular identification and characterization of these tumors to consider the exploitation of a potential-targeted therapeutic approach. PATIENTS AND METHODS: Hyperspectral immunologic cell by cell analysis was applied to touch imprint smears obtained from fresh tumors of breast cancer patients. RESULTS: Cell by cell analysis confirms significant intratumoral molecular heterogeneity in cancer markers with differences from polymerase chain reaction marker reporting. The individual cell heterogeneity was recognized in adjacent cells examined with panels of ten molecular markers in each single cell and included some markers that are considered to express "stem-cell" character. In addition, heterogeneity did not relate either to the size or stage of the primary tumor or to the site from within the cancer. CONCLUSION: There is a very significant molecular heterogeneity when "adjacent cells" are examined in triple-negative breast cancer, thereby making a successful targeted approach unlikely. In addition, it is not reasonable to consider that these changes will provide an answer to tumor dormancy.

5.
Mol Cancer Res ; 13(4): 784-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25573953

ABSTRACT

UNLABELLED: Protein S-palmitoylation is a widespread and dynamic posttranslational modification that regulates protein-membrane interactions, protein-protein interactions, and protein stability. A large family of palmitoyl acyl transferases, termed the DHHC family due to the presence of a common catalytic motif, catalyzes S-palmitoylation; the role of these enzymes in cancer is largely unexplored. In this study, an RNAi-based screen targeting all 23 members of the DHHC family was conducted to examine the effects on the growth in non-small cell lung cancer (NSCLC). Interestingly, siRNAs directed against DHHC5 broadly inhibited the growth of multiple NSCLC lines but not normal human bronchial epithelial cell (HBEC) lines. Silencing of DHHC5 by lentivirus-mediated expression of DHHC5 shRNAs dramatically reduced in vitro cell proliferation, colony formation, and cell invasion in a subset of cell lines that were examined in further detail. The phenotypes were restored by transfection of a wild-type DHHC5 plasmid but not by a plasmid expressing a catalytically inactive DHHC5. Tumor xenograft formation was severely inhibited by DHHC5 knockdown and rescued by DHHC5 expression, using both a conventional and tetracycline-inducible shRNA. These data indicate that DHHC5 has oncogenic capacity and contributes to tumor formation in NSCLC, thus representing a potential novel therapeutic target. IMPLICATIONS: Inhibitors of DHHC5 enzyme activity may inhibit non-small cell lung cancer growth.


Subject(s)
Acyltransferases/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA, Small Interfering/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , Neoplasm Transplantation
6.
Melanoma Res ; 24(6): 556-67, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25304236

ABSTRACT

Metastatic melanoma is a deadly form of cancer with few therapeutic options and the cause of more than 9480 deaths annually in the USA alone. Novel treatment options for this disease are urgently needed. Here we test the efficacy of a novel melanoma drug, the human recombinant Co-arginase (CoArgIPEG), against an aggressive A375 melanoma mouse model. CoArgIPEG is a modification of the naturally occurring human enzyme with improved stability, catalytic activity, and potentially lower immunogenicity compared with current amino acid-depleting drugs. Marked tumor growth reductions (mean P=0.0057) with apoptosis induction and proliferation inhibition are noted with CoArgIPEG treatment, both in the presence and in the absence of supplemental citrulline. Further, improved therapeutic efficacy has been noted against A375 xenografts relative to the naturally occurring human recombinant arginase enzyme at lower doses of CoArgIPEG. Unfortunately, after 1 month, half of the relapsing tumors showed argininosuccinate synthase induction, which correlated with Ser62-phosphorylated cMyc. Although argininosuccinate synthase induction could not be induced in vitro, a drug targeting pathway previously demonstrated to be associated with Ser62 cMyc phosphorylation - U0126 - in combination with CoArgIPEG demonstrated an in-vitro synergistic response (combination indices 0.13±0.10 and 0.14±0.10 with or without citrulline, respectively). Overall, favorable efficacy and potential synergy with other antimelanoma drugs support CoArgIPEG as a potent, novel cancer therapeutic.


Subject(s)
Antineoplastic Agents/therapeutic use , Arginase/therapeutic use , Melanoma/drug therapy , Recombinant Proteins/therapeutic use , Skin Neoplasms/drug therapy , Animals , Cobalt/chemistry , Cobalt/therapeutic use , Female , Humans , Hydrolases/chemistry , Hydrolases/therapeutic use , Jurkat Cells , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Sci ; 105(8): 1040-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24837299

ABSTRACT

Epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) are validated molecular targets in cancer therapy. Dual blockade has been explored and one such agent, lapatinib, is in clinical practice but with modest activity. Through chemical screening, we discovered a novel EGFR and HER2 inhibitor, S-222611, that selectively inhibited both kinases with IC50 s below 10 nmol/L. S-222611 also inhibited intracellular kinase activity and the growth of EGFR-expressing and HER2-expressing cancer cells. In addition, S-222611 showed potent antitumor activity over lapatinib in a variety of xenograft models. In evaluations with two patient-oriented models, the intrafemoral implantation model and the intracranial implantation model, S-222611 exhibited excellent activity and could be effective against bone and brain metastasis. Compared to neratinib and afatinib, irreversible EGFR/HER2 inhibitors, S-222611 showed equivalent or slightly weaker antitumor activity but a safer profile. These results indicated that S-222611 is a potent EGFR and HER2 inhibitor with substantially better antitumor activity than lapatinib at clinically relevant doses. Considering the safer profile than for irreversible inhibitors, S-222611 could be an important option in future cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays
8.
ACS Nano ; 7(10): 8816-23, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24016305

ABSTRACT

Sensitive and quantitative assessment of changes in circulating tumor cells (CTCs) can help in cancer prognosis and in the evaluation of therapeutics efficacy. However, extremely low occurrence of CTCs in the peripheral blood (approximately one CTC per billion blood cells) and potential changes in molecular biomarkers during the process of epithelial to mesenchymal transition create technical hurdles to the enrichment and enumeration of CTCs. Recently, efforts have been directed toward development of antibody-capture assays based on the expression of the common biomarker-the epithelial cell adhesion molecule (EpCAM) of epithelium-derived cancer cells. Despite some promising results, the assays relying on EpCAM capture have shown inconsistent sensitivity in clinical settings and often fail to detect CTCs in patients with metastatic cancer. We have addressed this problem by the development of an assay based on hybrid magnetic/plasmonic nanocarriers and a microfluidic channel. In this assay, cancer cells are specifically targeted by antibody-conjugated magnetic nanocarriers and are separated from normal blood cells by a magnetic force in a microfluidic chamber. Subsequently, immunofluorescence staining is used to differentiate CTCs from normal blood cells. We demonstrated in cell models of colon, breast, and skin cancers that this platform can be easily adapted to a variety of biomarkers, targeting both surface receptor molecules and intracellular biomarkers of epithelial-derived cancer cells. Experiments in whole blood showed capture efficiency greater than 90% when two cancer biomarkers are used for cell capture. Thus, the combination of immunotargeted magnetic nanocarriers with microfluidics provides an important platform that can improve the effectiveness of current CTC assays by overcoming the problem of heterogeneity of tumor cells in the circulation.


Subject(s)
Immunomagnetic Separation , Nanostructures , Neoplasms/pathology , Biomarkers, Tumor/metabolism , Humans , Microscopy, Electron, Transmission
9.
Neurology ; 81(3): 256-63, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23771486

ABSTRACT

OBJECTIVE: The study goal was to assess the benefits and potential limitations in the use of ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles in the MRI diagnosis of CNS inflammatory diseases and primary CNS lymphoma. METHODS: Twenty patients with presumptive or known CNS lesions underwent MRI study. Eighteen patients received both gadolinium-based contrast agents (GBCAs) and 1 of 2 USPIO contrast agents (ferumoxytol and ferumoxtran-10) 24 hours apart, which allowed direct comparative analysis. The remaining 2 patients had only USPIO-enhanced MRI because of a renal contraindication to GBCA. Conventional T1- and T2-weighted MRI were acquired before and after contrast administration in all patients, and perfusion MRI for relative cerebral blood volume (rCBV) assessment was obtained in all 9 patients receiving ferumoxytol. RESULTS: USPIO-enhanced MRI showed an equal number of enhancing brain lesions in 9 of 18 patients (50%), more enhancing lesions in 2 of 18 patients (11%), and fewer enhancing lesions in 3 of 18 patients (17%) compared with GBCA-enhanced MRI. Four of 18 patients (22%) showed no MRI enhancement. Dynamic susceptibility-weighted contrast-enhanced perfusion MRI using ferumoxytol showed low rCBV (ratio <1.0) in 3 cases of demyelination or inflammation, modestly elevated rCBV in 5 cases of CNS lymphoma or lymphoproliferative disorder (range: 1.3-4.1), and no measurable disease in one case. CONCLUSIONS: This study showed that USPIO-enhanced brain MRI can be useful in the diagnosis of CNS inflammatory disorders and lymphoma, and is also useful for patients with renal compromise at risk of nephrogenic systemic fibrosis who are unable to receive GBCA.


Subject(s)
Brain Diseases/diagnosis , Brain/pathology , Central Nervous System Neoplasms/diagnosis , Ferric Compounds , Lymphoma/diagnosis , Nanoparticles , Adult , Aged , Brain Diseases/pathology , Central Nervous System Neoplasms/pathology , Female , Humans , Inflammation/diagnosis , Inflammation/pathology , Lymphoma/pathology , Magnetic Resonance Imaging , Male , Middle Aged
10.
Blood ; 121(14): 2669-77, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23365458

ABSTRACT

Mutations in the transcription factor GATA2 underlie the syndrome of monocytopenia and B- and natural killer (NK)-cell lymphopenia associated with opportunistic infections and cancers. In addition, patients have recurrent and severe viral infections. NK cells play a critical role in mediating antiviral immunity. Human NK cells are thought to mature in a linear fashion, with the CD56(bright) stage preceding terminal maturation to the CD56(dim) stage, considered the most enabled for cytotoxicity. Here we report an NK cell functional defect in GATA2-deficient patients and extend this genetic lesion to what is considered to be the original NK cell-deficient patient. In most cases, GATA2 deficiency is accompanied by a severe reduction in peripheral blood NK cells and marked functional impairment. The NK cells detected in peripheral blood of some GATA2-deficient patients are exclusively of the CD56(dim) subset, which is recapitulated on in vitro NK cell differentiation. In vivo, interferon α treatment increased NK cell number and partially restored function but did not correct the paucity of CD56(bright) cells. Thus, GATA2 is required for the maturation of human NK cells and the maintenance of the CD56(bright) pool in the periphery. Defects in GATA2 are a novel cause of profound NK cell dysfunction.


Subject(s)
CD56 Antigen/immunology , Cell Differentiation/immunology , GATA2 Transcription Factor/genetics , Killer Cells, Natural/immunology , Lymphopenia/genetics , Antigens, CD34/metabolism , CD56 Antigen/metabolism , Cytotoxicity, Immunologic/immunology , GATA2 Transcription Factor/immunology , GATA2 Transcription Factor/metabolism , Humans , Immunophenotyping , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphopenia/immunology , Lymphopenia/metabolism , Stromal Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
Biomed Microdevices ; 15(4): 673-681, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23109037

ABSTRACT

Combining the power of immunomagnetic assay and microfluidic microchip operations, we successfully detected rare CTCs from clinical blood samples. The microfluidic system is operated in a flip-flop mode, where a computer-controlled rotational holder with an array of microfluidic chips inverts the microchannels. We have demonstrated both theoretically and experimentally that the direction of red blood cell (RBC) sedimentation with regards to the magnetic force required for cell separation is important for capture efficiency, throughput, and purity. The flip-flop operation reduces the stagnation of RBCs and non-specific binding on the capture surface by alternating the direction of the magnetic field with respect to gravity. The developed immunomagnetic microchip-based screening system exhibits high capture rates (more than 90%) for SkBr3, PC3, and Colo205 cell lines in spiked screening experiments and successfully isolates CTCs from patient blood samples. The proposed motion controlled microchip-based immunomagnetic system shows great promise as a clinical tool for cancer diagnosis and prognosis.


Subject(s)
Immunomagnetic Separation/instrumentation , Microfluidic Analytical Techniques/instrumentation , Motion , Nanotechnology/instrumentation , Neoplastic Cells, Circulating/pathology , Blood Sedimentation , Cell Line, Tumor , Erythrocytes/cytology , Humans , Magnetic Fields , Neoplasms/blood , Viscosity
12.
Transl Res ; 159(5): 366-75, 2012 May.
Article in English | MEDLINE | ID: mdl-22500509

ABSTRACT

We developed a hyperspectral microscopic imaging (HMI) platform that can precisely identify and quantify 10 molecular markers in individual cancer cells in a single pass. The exploitation of an improved separation of circulating tumor cells and the application of HMI provided an opportunity (1) to identify molecular changes in these cells, (2) to recognize the coexpression of these markers, (3) to pose an important opportunity for noninvasive diagnosis, and (4) to use targeted therapy. We balanced the intensity of 10 fluorochromes bound to 10 different antibodies, each specific to a particular tumor marker, so that the intensity of each fluorochrome can be discerned from overlapping emissions. Using 2 touch preps from each primary breast cancer, the average molecular marker intensities of 25 tumor cells gave a representative molecular signature for the tumor despite some cellular heterogeneity. The intensities determined by the HMI correlate well with the conventional 0-3+ analysis by experts in cellular pathology. Because additional multiplexes can be developed using the same fluorochromes but different antibodies, this analysis allows quantification of many molecular markers on a population of tumor cells. HMI can be automated completely, and eventually, it could allow the standardization of protein biomarkers and improve reproducibility among clinical pathology laboratories.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Fluorescent Dyes , Genes, erbB-2 , Humans , Microscopy
13.
Expert Opin Biol Ther ; 12(1): 53-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22171665

ABSTRACT

INTRODUCTION: Amino acid auxotrophy or the metabolic defect which renders cancer incapable of surviving under amino acid depleted conditions is being exploited and explored as a therapeutic against cancer. Early clinical data on asparagine- and arginine-depleting drugs have demonstrated low toxicity and efficacy in melanoma, hepatocellular carcinoma and acute lymphoblastic leukemia. Methionine auxotrophy is a novel niche currently under exploration for targeting certain cancers. AREAS COVERED: In this review we explore the discovery of methionine auxotrophy followed by in vitro, in vivo and patient data on targeting cancer with methionine depletion. We end with a small discussion on bioengineering, pegylation and red blood cell encapsulation as mechanisms for decreasing immunogenicity of methionine-depleting drugs. We hope to provide a platform for future pharmacology, toxicology and cytotoxicity studies with methionine depletion therapy and drugs. EXPERT OPINION: Although methionine auxotrophy seems as a viable target, extensive research addressing normal versus cancer cell toxicity needs to be conducted. Further research also needs to be conducted into the molecular mechanism associated with methionine depletion therapy. Finally, novel methods need to be developed to decrease the immunogenicity of methionine-depleting drugs, a current issue with protein therapeutics.


Subject(s)
Methionine/metabolism , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Bioengineering/methods , Cell Line, Tumor , Erythrocytes/cytology , Humans , Immune System , Medical Oncology/methods , Methionine/therapeutic use , Mice , Models, Biological , Neoplasm Transplantation , Neoplasms/diet therapy , Polyethylene Glycols/chemistry , Pseudomonas putida/metabolism , Rats
14.
Clin Appl Thromb Hemost ; 18(2): 215-7, 2012.
Article in English | MEDLINE | ID: mdl-22067544

ABSTRACT

UNLABELLED: Heparin-induced thrombocytopenia (HIT) antibodies are screened by an enzyme-linked immunosorbent assay (ELISA). Polyspecific ELISA detects anti-PF4/heparin IgG, IgA, and IgM. Recently, anti-PF4/heparin IgG ELISA has been shown to be more specific. However, the impact of using the IgG-ELISA on the incidence of isolated HIT (thrombocytopenia alone without clinically evident thrombosis) and the risk of developing subsequent thrombosis are still unknown. METHODS: A total of 492 consecutive patients with clinically suspected HIT at The University of Texas Southwestern Medical Center and affiliated hospitals were retrospectively reviewed from December 2008 to May 2010. RESULTS: 29 patients (6%) were diagnosed with HIT based on clinical findings and positive ELISA. 19 of the 29 patients (65%) had thrombosis at the time of diagnosis; whereas 10 of the 29 (35%) had only isolated HIT. The ten patients with isolated HIT had serial follow up for at least 3 months. 3 of 10 were treated with direct thrombin inhibitors and 5 of 10 were treated with Warfarin for at least 1 month upon discharge. None of them developed symptoms or signs of thrombosis during 3 months of follow up. CONCLUSION: The incidence of isolated HIT in this study was 35%, which is significantly lower than previously reported in the literature. It is possible that some patients previously thought to have HIT by the poly-specific ELISA assay had false positive results. The improved specificity of the IgG- ELISA appears to reduce the incidence of isolated HIT which may have lower risk of subsequent thrombosis.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Heparin/adverse effects , Immunoglobulin G/immunology , Platelet Factor 4/immunology , Purpura, Thrombocytopenic, Idiopathic/chemically induced , Thrombophilia/etiology , Thrombosis/epidemiology , Antibody Specificity , Anticoagulants/therapeutic use , False Positive Reactions , Follow-Up Studies , Heparin/immunology , Heparin/therapeutic use , Humans , Incidence , Purpura, Thrombocytopenic, Idiopathic/epidemiology , Reproducibility of Results , Retrospective Studies , Risk , Sensitivity and Specificity , Texas/epidemiology , Thrombophilia/drug therapy , Thrombosis/drug therapy , Thrombosis/etiology , Warfarin/therapeutic use
15.
Lab Chip ; 11(20): 3449-57, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21863182

ABSTRACT

Screening for circulating tumor cells (CTCs) in blood has been an object of interest for evidence of progressive disease, status of disease activity, recognition of clonal evolution of molecular changes and for possible early diagnosis of cancer. We describe a new method of microchip-based immunomagnetic CTC detection, in which the benefits of both immunomagnetic assay and the microfluidic device are combined. As the blood sample flows through the microchannel closely above arrayed magnets, cancer cells labeled with magnetic nanoparticles are separated from blood flow and deposited at the bottom wall of the glass coverslip, which allows direct observation of captured cells with a fluorescence microscope. A polydimethylsiloxane (PDMS)-based microchannel fixed on a glass coverslip was used to screen blood samples. The thin, flat dimensions of the microchannel, combined with the sharp magnetic field gradient in the vicinity of arrayed magnets with alternate polarities, lead to an effective capture of labeled cells. Compared to the commercially available CellSearch™ system, fewer (25%) magnetic particles are required to achieve a comparable capture rate, while the screening speed (at an optimal blood flow rate of 10 mL h(-1)) is more than five times faster than those reported previously with a microchannel-based assay. For the screening experiment, blood drawn from healthy subjects into CellSave™ tubes was spiked with cultured cancer cell lines of COLO205 and SKBR3. The blood was then kept at room temperature for 48 hours before the screening, emulating the actual clinical cases of blood screening. Customized Fe(3)O(4) magnetic nanoparticles (Veridex Ferrofluid™) conjugated to anti-epithelial cell adhesion molecule (EpCAM) antibodies were introduced into the blood samples to label cancer cells, and the blood was then run through the microchip device to capture the labelled cells. After capture, the cells were stained with fluorescent labelled anti-cytokeratin, DAPI and anti-CD45. Subsequent immunofluorescence images were taken for the captured cells, followed by comprehensive computer aided analysis based on fluorescence intensities and cell morphology. Rare cancer cells (from ∼1000 cells down to ∼5 cells per mL) with very low tumor cell to blood cell ratios (about 1 : 10(7) to 10(9), including red blood cells) were successfully detected. Cancer cell capture rates of 90% and 86% were demonstrated for COLO205 and SKBR3 cells, respectively.


Subject(s)
Cell Separation/instrumentation , Immunoassay/instrumentation , Lab-On-A-Chip Devices , Magnets , Neoplastic Cells, Circulating/pathology , Cell Count , Cell Line, Tumor , Humans
16.
Neurology ; 76(1): 87-93, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21205697

ABSTRACT

One rationale behind the use of agents that inhibit vascular endothelial growth factor in the therapy of primary CNS malignancies is based upon the concept that normalization of tumor vasculature with a decrease in tumor interstitial pressure will improve access of cytoreductive drugs and improve radiotherapy efficacy due to increased oxygen delivery. However, several studies have raised the concern that these agents may both rapidly restore the low permeability characteristics of the blood-brain barrier and counteract the beneficial effect of pseudoprogression. The result may be decreased therapeutic efficacy while increasing infiltration by co-opting normal vessels. In this discussion, we examine both histologic and radiographic tumor progression in the context of antiangiogenic agents. Issues dealing with the safety of bevacizumab (Avastin®, Genentech, South San Francisco, CA) and its potential to decrease efficacy of standard radiochemotherapy when used to treat patients with newly diagnosed malignant glioma are emphasized.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Glioma/drug therapy , Antibodies, Monoclonal, Humanized , Bevacizumab , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Clinical Trials as Topic , Combined Modality Therapy , Glioma/diagnosis , Humans , Magnetic Resonance Imaging
17.
Int J Cancer ; 128(12): 2823-32, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-20734393

ABSTRACT

Conventional chemotherapy is commonly used for advanced stages of bladder cancer with modest success and high morbidity. Identifying markers of resistance will allow clinicians to tailor treatment to a specific patient population. T24-tumorigenic cell line was grown orthotopically in nude mice and monitored using bioluminescence imaging and microcomputed tomography until they developed metastases. Stable sublines were then developed from primary bladder (T24-P), lung (T24-L) and bone (T24-B) tissues. Chromosomal analysis and DNA microarray were used to characterize these sublines. Real-time quantitative polymerase chain reaction and immunohistochemistry were used for validation. Epigenetic modifiers were used to study gene regulation. The cell viability was quantified with MTT assay. Chromosomal analysis revealed multiple alterations in metastatic cell lines compared to T24-P. DNA microarray analysis showed that taxol resistance-associated gene (TRAG) 3 was the most upregulated gene. From real-time quantitative polymerase chain reaction and immunohistochemistry, TRAG3 was significantly higher in T24-L and T24-B than T24-P. TRAG3 gene expression is likely controlled by DNA methylation but not histone acetylation. Interestingly, T24-B and T24-L cells were more resistant than T24-P to treatment with antimicrotubule agents such as docetaxel, paclitaxel and vinblastine. TRAG3 mRNA expression was higher in 20% of patients with ≤ pT2 (n = 10) and 60% of patients with ≥ pT3 (n = 20) compared to normal adjacent tissue (p = 0.05). In addition, the median TRAG3 expression was 6.7-fold higher in ≥ pT3 tumors compared to ≤ pT2 tumors. Knowing the status of TRAG3 expression could help clinicians tailor treatment to a particular patient population that could benefit from treatment, while allocating patients with resistant tumors to new experimental therapies.


Subject(s)
Carcinoma, Transitional Cell/genetics , Neoplasm Proteins/genetics , Up-Regulation , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Carcinoma, Transitional Cell/pathology , DNA Primers , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Urinary Bladder Neoplasms/pathology
18.
J Thorac Oncol ; 4(2): 161-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19179890

ABSTRACT

INTRODUCTION: Most epidermal growth factor receptor (EGFR) mutant non-small cell lung cancers (NSCLCs) are sensitive to EGFR tyrosine kinase inhibitors (TKIs) such as erlotinib or gefitinib, but many EGFR wild type NSCLCs are resistant to TKIs. In this study, we examined the effects of the histone deacetylase inhibitor, romidepsin, in combination with erlotinib, in NSCLC cell lines and xenografts. METHODS: For in vitro studies, nine NSCLC cell lines with varying mutation status and histology were treated with erlotinib and romidepsin alone or in combination. 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assays were performed to determine the concentration that inhibits 50% (IC50) value of each drug or the combination. For in vivo studies, NCI-H1299 xenografts were inoculated subcutaneously into athymic nude mice. Romidepsin and/or erlotinib were injected intraperitoneally after tumors developed and tumor sizes were measured. RESULTS: We found that romidepsin increased the sensitivity of erlotinib synergistically in all nine NSCLC cell lines including EGFR and KRAS wild type cell lines, KRAS mutant cell lines, and TKI resistant EGFR mutant cell lines. This effect was partially due to enhanced apoptosis. Furthermore, cotreatment of erlotinib and romidepsin inhibited NCI-H1299 xenograft growth in athymic nude mice. CONCLUSIONS: These observations support a role for the combination of a histone deacetylase inhibitor and a TKI in the treatment of NSCLCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Depsipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Erlotinib Hydrochloride , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
19.
Am J Clin Oncol ; 31(3): 219-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18525298

ABSTRACT

OBJECTIVES: Preclinical studies using sequences of topoisomerase I and II inhibitors suggested synergism; preliminary clinical studies, resulting in enhanced antitumor responses, confirm this in selected malignancies. This study determined the maximum-tolerated dose (MTD), toxicity, and pharmacokinetics of irinotecan (CPT-11), capecitabine, and epirubicin in patients with metastatic adenocarcinoma of lung, breast, or gastrointestinal tract. Correlation of topoisomerase IIbeta was also done. METHODS: Eligibility criteria included the following: documented adenocarcinoma of the lung, breast, or gastrointestinal tract, <3 prior chemotherapy regimens, Eastern Cooperative Oncology Group (ECOG) performance status 0 to 2, age > or =18 years, adequate organ function, and signed informed consent. Irinotecan was administered at 250 mg/m2 intravenously day 1 every 21-day cycle, but was reduced to 180 mg/m2 in cohort 2 due to toxicity. Capecitabine was administered at 750 mg/m2 twice daily days 2 to 14 in cohort 1 but only on days 2 to 7 from cohort 2 due to early neutropenia and to allow for prophylactic granulocyte colony-stimulating factor (GCSF) support. Epirubicin was administered at 40 mg/m2 in cohort 1, but reduced to 30 mg/m2 in cohort 2, then reescalated until the MTD was reached. RESULTS: Toxicity was assessed in 21 patients; response was assessed in 17 patients. The most common grade 3 to 4 toxicities included neutropenia (57.1%) and anemia (28.6%). The MTD of epirubicin was 50 mg/m2. In evaluable patients, there were 2 partial responses (11.8%) and 13 stable disease (76.5%); these correlated well with topoisomerase IIbeta. CONCLUSIONS: The recommended doses for phase II studies: irinotecan 180 mg/m2 day 1, epirubicin 50 mg/m2 day 2, and capecitabine 750 mg/m2 twice daily days 2 to 7 of each 21-day cycle. This combination is reasonably active and warrants evaluation in the phase II setting.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Epirubicin/administration & dosage , Gastrointestinal Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Topoisomerase II Inhibitors , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Administration, Oral , Adult , Aged , Alopecia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Capecitabine , DNA Topoisomerases, Type I/blood , DNA Topoisomerases, Type II/blood , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Drug Resistance, Neoplasm , Drug Synergism , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Gastrointestinal Diseases/chemically induced , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematologic Diseases/chemically induced , Humans , Infusions, Intravenous , Irinotecan , Male , Maximum Tolerated Dose , Middle Aged , Treatment Outcome
20.
Platelets ; 18(7): 491-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17852774

ABSTRACT

Platelet aggregation inhibition by clopidogrel may be suboptimal in 4-30% of patients. Traditionally, optical aggregometry is used to assess clopidogrel's anti-platelet effects by inhibition of ADP-induced aggregation in platelet rich plasma. Red blood cells are an important source of ADP and, thus, are known to modulate platelet function. Because the whole blood aggregation by impedance method assesses platelet function in a physiological milieu, we compared clopidogrel response by this method with the optical method in platelet rich plasma (PRP) and the Platelet Function Analyzer (PFA-100). Platelet function studies were performed in 17 healthy subjects at baseline and after 10 days of clopidogrel intake (75 mg/day). Optical and impedance aggregometry were performed after addition of ADP (10 and 20 microM) and collagen (1 and 2 microg/mL). For PFA-100 analysis, whole blood closure time was measured in collagen-coated cartridges with ADP and epinephrine. All subjects except one showed a decrease in ADP-induced aggregation using both aggregation methods. However, ADP-induced platelet aggregation was significantly inhibited when assessed in whole blood as compared to the optical method (71+/- 34% vs. 34.2+/- 23%, p = 0.0002); this suggests that whole blood aggregometry is more sensitive in the detection of clopidogrel effect in the presence of red cells, which are known to modulate platelet function. The PFA-100 ADP closure time was slightly prolonged above the reference interval in only 5/17 (29%) subjects, suggesting that this instrument is not able to detect clopidogrel effect. We conclude that whole blood aggregation appears to be more sensitive in detecting clopidogrel effect compared with the platelet rich plasma method; the PFA-100 was unable to detect clopidogrel effect in the majority of the subjects.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Function Tests/instrumentation , Ticlopidine/analogs & derivatives , Adult , Aged , Clopidogrel , Female , Hematologic Tests/methods , Humans , Male , Middle Aged , Optics and Photonics , Sensitivity and Specificity , Ticlopidine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...