Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
bioRxiv ; 2023 Jun 14.
Article En | MEDLINE | ID: mdl-37398342

Mechanistic modeling of cancers such as Medullary Thyroid Carcinoma (MTC) to emulate patient-specific phenotypes is challenging. The discovery of potential diagnostic markers and druggable targets in MTC urgently requires clinically relevant animal models. Here we established orthotopic mouse models of MTC driven by aberrantly active Cdk5 using cell-specific promoters. Each of the two models elicits distinct growth differences that recapitulate the less or more aggressive forms of human tumors. The comparative mutational and transcriptomic landscape of tumors revealed significant alterations in mitotic cell cycle processes coupled with the slow-growing tumor phenotype. Conversely, perturbation in metabolic pathways emerged as critical for aggressive tumor growth. Moreover, an overlapping mutational profile was identified between mouse and human tumors. Gene prioritization revealed putative downstream effectors of Cdk5 which may contribute to the slow and aggressive growth in the mouse MTC models. In addition, Cdk5/p25 phosphorylation sites identified as biomarkers for Cdk5-driven neuroendocrine tumors (NETs) were detected in both slow and rapid onset models and were also histologically present in human MTC. Thus, this study directly relates mouse and human MTC models and uncovers vulnerable pathways potentially responsible for differential tumor growth rates. Functional validation of our findings may lead to better prediction of patient-specific personalized combinational therapies.

2.
Front Oncol ; 13: 1168710, 2023.
Article En | MEDLINE | ID: mdl-37205196

Introduction: Immunotherapy is an effective treatment for a subset of cancer patients, and expanding the benefits of immunotherapy to all cancer patients will require predictive biomarkers of response and immune-related adverse events (irAEs). To support correlative studies in immunotherapy clinical trials, we are developing highly validated assays for quantifying immunomodulatory proteins in human biospecimens. Methods: Here, we developed a panel of novel monoclonal antibodies and incorporated them into a novel, multiplexed, immuno-multiple reaction monitoring mass spectrometry (MRM-MS)-based proteomic assay targeting 49 proteotypic peptides representing 43 immunomodulatory proteins. Results and discussion: The multiplex assay was validated in human tissue and plasma matrices, where the linearity of quantification was >3 orders of magnitude with median interday CVs of 8.7% (tissue) and 10.1% (plasma). Proof-of-principle demonstration of the assay was conducted in plasma samples collected in clinical trials from lymphoma patients receiving an immune checkpoint inhibitor. We provide the assays and novel monoclonal antibodies as a publicly available resource for the biomedical community.

3.
Front Immunol ; 12: 765898, 2021.
Article En | MEDLINE | ID: mdl-34858420

Immunotherapies are revolutionizing cancer care, producing durable responses and potentially cures in a subset of patients. However, response rates are low for most tumors, grade 3/4 toxicities are not uncommon, and our current understanding of tumor immunobiology is incomplete. While hundreds of immunomodulatory proteins in the tumor microenvironment shape the anti-tumor response, few of them can be reliably quantified. To address this need, we developed a multiplex panel of targeted proteomic assays targeting 52 peptides representing 46 proteins using peptide immunoaffinity enrichment coupled to multiple reaction monitoring-mass spectrometry. We validated the assays in tissue and plasma matrices, where performance figures of merit showed over 3 orders of dynamic range and median inter-day CVs of 5.2% (tissue) and 21% (plasma). A feasibility study in clinical biospecimens showed detection of 48/52 peptides in frozen tissue and 38/52 peptides in plasma. The assays are publicly available as a resource for the research community.


Chromatography, Liquid/methods , Mass Spectrometry/methods , Peptides/analysis , Proteome/analysis , Proteomics/methods , Specimen Handling/methods , Antibodies/analysis , Antibodies/immunology , Blotting, Western , Cell Line, Tumor , HeLa Cells , Humans , Jurkat Cells , MCF-7 Cells , Peptides/blood , Peptides/immunology , Proteome/genetics , Proteome/immunology , RNA-Seq/methods , Reproducibility of Results
4.
Cell Rep Methods ; 1(3)2021 07 26.
Article En | MEDLINE | ID: mdl-34671754

SUMMARY: A primary goal of the US National Cancer Institute's Ras initiative at the Frederick National Laboratory for Cancer Research is to develop methods to quantify RAS signaling to facilitate development of novel cancer therapeutics. We use targeted proteomics technologies to develop a community resource consisting of 256 validated multiple reaction monitoring (MRM)-based, multiplexed assays for quantifying protein expression and phosphorylation through the receptor tyrosine kinase, MAPK, and AKT signaling networks. As proof of concept, we quantify the response of melanoma (A375 and SK-MEL-2) and colorectal cancer (HCT-116 and HT-29) cell lines to BRAF inhibition by PLX-4720. These assays replace over 60 Western blots with quantitative mass spectrometry-based assays of high molecular specificity and quantitative precision, showing the value of these methods for pharmacodynamic measurements and mechanism of action studies. Methods, fit-for-purpose validation, and results are publicly available as a resource for the community at assays.cancer.gov. MOTIVATION: A lack of quantitative, multiplexable assays for phosphosignaling limits comprehensive investigation of aberrant signaling in cancer and evaluation of novel treatments. To alleviate this limitation, we sought to develop assays using targeted mass spectrometry for quantifying protein expression and phosphorylation through the receptor tyrosine kinase, MAPK, and AKT signaling networks. The resulting assays provide a resource for replacing over 60 Western blots in examining cancer signaling and tumor biology with high molecular specificity and quantitative rigor.


Melanoma , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Mass Spectrometry/methods , Receptor Protein-Tyrosine Kinases , Mitogen-Activated Protein Kinase Kinases , Tyrosine
5.
Cancers (Basel) ; 13(15)2021 Jul 30.
Article En | MEDLINE | ID: mdl-34359745

The ATM serine/threonine kinase (HGNC: ATM) is involved in initiation of repair of DNA double-stranded breaks, and ATM inhibitors are currently being tested as anti-cancer agents in clinical trials, where pharmacodynamic (PD) assays are crucial to help guide dose and scheduling and support mechanism of action studies. To identify and quantify PD biomarkers of ATM inhibition, we developed and analytically validated a 51-plex assay (DDR-2) quantifying protein expression and DNA damage-responsive phosphorylation. The median lower limit of quantification was 1.28 fmol, the linear range was over 3 orders of magnitude, the median inter-assay variability was 11% CV, and 86% of peptides were stable for storage prior to analysis. Use of the assay was demonstrated to quantify signaling following ionizing radiation-induced DNA damage in both immortalized lymphoblast cell lines and primary human peripheral blood mononuclear cells, identifying PD biomarkers for ATM inhibition to support preclinical and clinical studies.

6.
Sci Data ; 6(1): 160, 2019 08 29.
Article En | MEDLINE | ID: mdl-31467290

RAS genes are frequently mutated in cancer and have for decades eluded effective therapeutic attack. The National Cancer Institute's RAS Initiative has a focus on understanding pathways and discovering therapies for RAS-driven cancers. Part of these efforts is the generation of novel reagents to enable the quantification of RAS network proteins. Here we present a dataset describing the development, validation (following consensus principles developed by the broader research community), and distribution of 104 monoclonal antibodies (mAbs) enabling detection of 27 phosphopeptides and 69 unmodified peptides from 20 proteins in the RAS network. The dataset characterizes the utility of the antibodies in a variety of applications, including Western blotting, immunoprecipitation, protein array, immunohistochemistry, and targeted mass spectrometry. All antibodies and characterization data are publicly available through the CPTAC Antibody Portal, Panorama Public Repository, and/or PRIDE databases. These reagents will aid researchers in discerning pathways and measuring expression changes in the RAS signaling network.


Antibodies, Monoclonal/chemistry , Genes, ras , Signal Transduction , Cell Line , DNA Fingerprinting , Humans , Indicators and Reagents/chemistry , Microsatellite Repeats , Neoplasms/genetics
7.
PLoS One ; 13(6): e0199361, 2018.
Article En | MEDLINE | ID: mdl-29928062

The presence of cancer stem cells (CSCs) and the induction of epithelial-to-mesenchymal transition (EMT) in tumors are associated with tumor aggressiveness, metastasis, drug resistance, and poor prognosis, necessitating the development of reagents for unambiguous detection of CSC- and EMT-associated proteins in tumor specimens. To this end, we generated novel antibodies to EMT- and CSC-associated proteins, including Goosecoid, Sox9, Slug, Snail, and CD133. Importantly, unlike several widely used antibodies to CD133, the anti-CD133 antibodies we generated recognize epitopes distal to known glycosylation sites, enabling analyses that are not confounded by differences in CD133 glycosylation. For all target proteins, we selected antibodies that yielded the expected target protein molecular weights by Western analysis and the correct subcellular localization patterns by immunofluorescence microscopy assay (IFA); binding selectivity was verified by immunoprecipitation-mass spectrometry and by immunohistochemistry and IFA peptide blocking experiments. Finally, we applied these reagents to assess modulation of the respective markers of EMT and CSCs in xenograft tumor models by IFA. We observed that the constitutive presence of human hepatocyte growth factor (hHGF) in the tumor microenvironment of H596 non-small cell lung cancer tumors implanted in homozygous hHGF knock-in transgenic mice induced a more mesenchymal-like tumor state (relative to the epithelial-like state when implanted in control SCID mice), as evidenced by the elevated expression of EMT-associated transcription factors detected by our novel antibodies. Similarly, our new anti-CD133 antibody enabled detection and quantitation of drug-induced reductions in CD133-positive tumor cells following treatment of SUM149PT triple-negative breast cancer xenograft models with the CSC/focal adhesion kinase (FAK) inhibitor VS-6063. Thus, our novel antibodies to CSC- and EMT-associated factors exhibit sufficient sensitivity and selectivity for immunofluorescence microscopy studies of these processes in preclinical xenograft tumor specimens and the potential for application with clinical samples.


Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Neoplastic Stem Cells/pathology , Tumor Microenvironment/drug effects , AC133 Antigen/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Gene Knock-In Techniques , Hepatocyte Growth Factor/genetics , Humans , Indicators and Reagents , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Pyrazines/pharmacology , Pyrazines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Nat Methods ; 15(5): 330-338, 2018 05.
Article En | MEDLINE | ID: mdl-29638227

A key component of efforts to address the reproducibility crisis in biomedical research is the development of rigorously validated and renewable protein-affinity reagents. As part of the US National Institutes of Health (NIH) Protein Capture Reagents Program (PCRP), we have generated a collection of 1,406 highly validated immunoprecipitation- and/or immunoblotting-grade mouse monoclonal antibodies (mAbs) to 737 human transcription factors, using an integrated production and validation pipeline. We used HuProt human protein microarrays as a primary validation tool to identify mAbs with high specificity for their cognate targets. We further validated PCRP mAbs by means of multiple experimental applications, including immunoprecipitation, immunoblotting, chromatin immunoprecipitation followed by sequencing (ChIP-seq), and immunohistochemistry. We also conducted a meta-analysis that identified critical variables that contribute to the generation of high-quality mAbs. All validation data, protocols, and links to PCRP mAb suppliers are available at http://proteincapture.org.


Antibodies, Monoclonal/immunology , Protein Array Analysis/methods , Transcription Factors/metabolism , Animals , Cloning, Molecular , Databases, Factual , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Reproducibility of Results
9.
Int Immunopharmacol ; 29(2): 476-487, 2015 Dec.
Article En | MEDLINE | ID: mdl-26474694

Targeted inhibiting insulin-like growth factor 1 is an effective approach for cancer therapy. Insulin-like growth factor binding protein 7 (IGFBP7) is considered as a potential therapeutic protein. However, producing high quality of such non-IgG proteins in mammalian cells is still a challenge in biopharmaceutical development. Here, we report a rapid production process by using transient gene transfection in HEK 293E cells. A set of constructs combining several expression promoters, leader sequences, and 5' un-translated regions were generated and optimized, from which the best vector with expression level at ~50mg/L was selected for production at 2L cell culture scale. Comparison study in downstream purification methods led to development of a scalable, non-affinity chromatography strategy through Super Q, Fast Flow Q, and Heparin columns. The product was characterized in purity (99%), isoelectric point, molecule weight, glycosylation, and stability by using SEC-HPLC, SDS-PAGE, isoelectric focusing and mass spectrometry. The highly purified product shows IGF-1 binding activity and inhibits IGF-1-induced cell proliferation. This process not only provides a remarkable high expression at ~50mg/L and pure glycosylated mammalian rhIGFBP7, also highlights that transient gene expression technology is practical to be used for production and early development of recombinant non-IgG therapeutic proteins.


Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/pharmacology , Insulin-Like Growth Factor I/drug effects , 5' Untranslated Regions/genetics , Amino Acid Sequence , Cell Proliferation/drug effects , Gene Expression/drug effects , Genetic Vectors , Glycosylation , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Molecular Sequence Data , Recombinant Proteins/pharmacology , Transfection
10.
MAbs ; 6(5): 1190-200, 2014.
Article En | MEDLINE | ID: mdl-25517304

Conjugation of small molecule drugs to specific sites on the antibody molecule has been increasingly used for the generation of relatively homogenous preparations of antibody-drug conjugates (ADCs) with physicochemical properties similar or identical to those of the naked antibody. Previously a method for conjugation of small molecules to glycoproteins through existing glycans by using an engineered glycotransferase and a chemically reactive sugar as a handle was developed. Here, for the first time, we report the use of this method with some modifications to generate an ADC from a monoclonal antibody, m860, which we identified from a human naïve phage display Fab library by panning against the extracellular domain of human HER2. M860 bound to cell surface-associated HER2 with affinity comparable to that of Trastuzumab (Herceptin), but to a different epitope. The m860ADC was generated by enzymatically adding a reactive keto-galactose to m860 using an engineered glycotransferase and conjugating the reactive m860 to aminooxy auristatin F. It exhibited potent and specific cell-killing activity against HER2 positive cancer cells, including trastuzumab-resistant breast cancer cells. This unique ADC may have utility as a potential therapeutic for HER2 positive cancers alone or in combination with other drugs. Our results also validate the keto-galactose/engineered glycotransferase method for generation of functional ADCs, which could potentially also be used for preparation of ADCs targeting other disease markers.


Antibodies/chemistry , Carbohydrates/chemistry , Glycosyltransferases/metabolism , Immunoconjugates/chemistry , Pharmaceutical Preparations/chemistry , Aminobenzoates/chemistry , Antibodies/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Glycosyltransferases/genetics , Humans , Immunoconjugates/genetics , Immunoconjugates/pharmacology , MCF-7 Cells , Models, Molecular , Oligopeptides/chemistry , Protein Binding/immunology , Protein Engineering/methods , Protein Structure, Tertiary , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Methods Mol Biol ; 1002: 253-65, 2013.
Article En | MEDLINE | ID: mdl-23625409

Matrix-assisted laser desorption/ionization (MALDI) imaging is rapidly gaining importance in the -biomarkers field because it is able to detect several analytes at the same time and to assign to each one of them not only an m/z value but also spatial distribution. Here we present the detailed description of sample preparation for protein and peptide MALDI imaging assays. This chapter describes the microtomy performed in a cryostat to produce tissue slides mounted onto glass slides suitable for MALDI. Sample preparation will include matrix coating procedure with a sensor-controlled aerosol. Finally, we will show some examples of how data can be visualized to suit the purposes of the research.


Peptides/analysis , Proteins/analysis , Proteomics/methods , Specimen Handling/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Amino Acid Sequence , Humans , Microtomy , Peptides/chemistry , Proteins/chemistry
12.
J Biol Chem ; 288(17): 12305-12, 2013 Apr 26.
Article En | MEDLINE | ID: mdl-23486472

HA22 is a recombinant immunotoxin that kills CD22-expressing cells by ADP-ribosylating and inactivating elongation factor-2 (EF2). HA22 is composed of an Fv that binds to CD22 fused to a portion of Pseudomonas exotoxin A. HA22 is very active in drug-resistant hairy cell leukemia but is less active in children with acute lymphoblastic leukemia. To understand why some patients do not respond to HA22, we isolated an HA22-resistant lymphoma cell line and showed that resistance was due to the inability of HA22 to ADP-ribosylate and inactivate EF2. We analyzed the diphthamide synthesis genes and found that the WDR85 gene was deleted. We show that WDR85 knockdown conferred HA22 resistance to sensitive cells and that sensitivity was restored by introduction of a WDR85 cDNA into resistant cells. Analysis of EF2 in the mutant cells revealed a novel form of diphthamide with an additional methyl group that prevented ADP-ribosylation and inactivation of EF2. The abnormal methylation appeared to be catalyzed by DPH5. Inactivation of the WDR85 gene could be a mechanism of immunotoxin resistance in patients undergoing immunotoxin therapy.


Drug Resistance, Neoplasm , Gene Deletion , Histidine/analogs & derivatives , Immunotoxins/pharmacology , Lymphoma/metabolism , Neoplasm Proteins/metabolism , Peptide Elongation Factor 2/metabolism , Proteins , Carboxylic Ester Hydrolases , Cell Line, Tumor , Histidine/genetics , Histidine/metabolism , Humans , Lymphoma/drug therapy , Lymphoma/genetics , Methylation/drug effects , Methyltransferases/genetics , Methyltransferases/metabolism , Neoplasm Proteins/genetics , Peptide Elongation Factor 2/genetics
13.
Pathol Int ; 62(1): 28-35, 2012 Jan.
Article En | MEDLINE | ID: mdl-22192801

To date, most of the proteomic analyses on lung cancer tissue samples have been performed using surgical specimens, which are obtained after a diagnosis is made. To determine if a proteomic signature obtained from bronchoscopic biopsy samples could be found to assist with diagnosis, 50 lung cancer bronchoscopic biopsy samples and 13 adjacent normal lung tissue samples were analyzed using histology-directed, matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS). Lung tissue samples were cryosectioned, and sinapinic acid was robotically deposited on areas of each tissue section enriched in epithelial cells, either tumor or normal. Mass spectra were acquired using a MALDI-time of flight instrument. Small cell lung cancers (SCLCs) demonstrated clearly different protein profiles from normal lung tissue and from non-small cell lung cancers (NSCLCs). Calcyclin (m/z= 10,094.7) was identified to be underexpressed in small cell lung cancers, as compared with non-small cell lung cancers and normal lung tissue. An immunohistochemistry study using 152 NSCLCs and 21 SCLCs confirmed significantly reduced calcyclin stain in SCLCs. Thus, protein profiles obtained from bronchoscopic biopsy samples via MALDI MS distinguish cancerous epithelium from normal lung tissue and between NSCLCs and SCLCs.


Cell Cycle Proteins/metabolism , Lung Neoplasms/diagnosis , S100 Proteins/metabolism , Small Cell Lung Carcinoma/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/analysis , Proteomics/methods , S100 Calcium Binding Protein A6 , Sensitivity and Specificity , Small Cell Lung Carcinoma/pathology
14.
Lipids ; 46(5): 469-77, 2011 May.
Article En | MEDLINE | ID: mdl-21327726

Since its introduction as an ionization technique in mass spectrometry, matrix-assisted laser desorption ionization (MALDI) has been applied to a wide range of applications. Quantitative small molecule analysis by MALDI, however, is limited due to the presence of intense signals from the matrix coupled with non-homogeneous surfaces. The surface used in nano-structured laser desorption ionization (NALDI) eliminates the need for a matrix and the resulting interferences, and allows for quantitative analysis of small molecules. This study was designed to analyze and quantitate phospholipid components of liposomes. Here we have developed an assay to quantitate the DPPC and DC(8,9)PC in liposomes by NALDI following various treatments. To test our method we chose to analyze a liposome system composed of DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine) and DC(8,9)PC (1,2-bis(tricosa-10,12-diynoyl)-sn-glycero-3-phosphocholine), as DC(8,9)PC is known to undergo cross-linking upon treatment with UV (254 nm) and this reaction converts the monomer into a polymer. First, calibration curves for pure lipids (DPPC and DC(8,9)PC) were created using DMPC (1,2-dimyristoyl-sn-glycero-3-phosphocholine) as an internal standard. The calibration curve for both DPPC and DC(8,9)PC showed an R(2) of 0.992, obtained using the intensity ratio of analyte and internal standard. Next, DPPC:DC(8,9)PC liposomes were treated with UV radiation (254 nm). Following this treatment, lipids were extracted from the liposomes and analyzed. The analysis of the lipids before and after UV exposure confirmed a decrease in the signal of DC(8,9)PC of about 90%. In contrast, there was no reduction in DPPC signal.


Phospholipids/chemistry , Liposomes/chemistry , Mass Spectrometry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
J Proteome Res ; 9(8): 4123-30, 2010 Aug 06.
Article En | MEDLINE | ID: mdl-20557134

To date, proteomic analyses on gastrointestinal cancer tissue samples have been performed using surgical specimens only, which are obtained after a diagnosis is made. To determine if a proteomic signature obtained from endoscopic biopsy samples could be found to assist with diagnosis, frozen endoscopic biopsy samples collected from 63 gastric cancer patients and 43 healthy volunteers were analyzed using matrix-assisted laser desorption/ionization (MALDI) mass spectrometry. A statistical classification model was developed to distinguish tumor from normal tissues using half the samples and validated with the other half. A protein profile was discovered consisting of 73 signals that could classify 32 cancer and 22 normal samples in the validation set with high predictive values (positive and negative predictive values for cancer, 96.8% and 91.3%; sensitivity, 93.8%; specificity, 95.5%). Signals overexpressed in tumors were identified as alpha-defensin-1, alpha-defensin-2, calgranulin A, and calgranulin B. A protein profile was also found to distinguish pathologic stage Ia (pT1N0M0) samples (n = 10) from more advanced stage (Ib or higher) tumors (n = 48). Thus, protein profiles obtained from endoscopic biopsy samples may be useful in assisting with the diagnosis of gastric cancer and, possibly, in identifying early stage disease.


Biomarkers, Tumor/analysis , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Stomach Neoplasms/diagnosis , Stomach Neoplasms/metabolism , Biopsy , Case-Control Studies , Defensins/metabolism , Gastroscopy/methods , Humans , Leukocyte L1 Antigen Complex/metabolism , Models, Statistical , Sensitivity and Specificity , Stomach Neoplasms/pathology
16.
Clin Lab Med ; 29(1): 57-69, 2009 Mar.
Article En | MEDLINE | ID: mdl-19389551

The potential of using mass spectrometry profiling as a diagnostic tool has been demonstrated for a wide variety of diseases. Various cancers and cancer-related diseases have been the focus of much of this work because of both the paucity of good diagnostic markers and the knowledge that early diagnosis is the most powerful weapon in treating cancer. The implementation of mass spectrometry as a routine diagnostic tool has proved to be difficult, however, primarily because of the stringent controls that are required for the method to be reproducible. The method is evolving as a powerful guide to the discovery of biomarkers that could, in turn, be used either individually or in an array or panel of tests for early disease detection. Using proteomic patterns to guide biomarker discovery and the possibility of deployment in the clinical laboratory environment on current instrumentation or in a hybrid technology has the possibility of being the early diagnosis tool that is needed.


Biomarkers/blood , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Humans , Immunochemistry , Reproducibility of Results , Sensitivity and Specificity
17.
Clin Pharmacokinet ; 48(2): 131-41, 2009.
Article En | MEDLINE | ID: mdl-19271785

BACKGROUND AND OBJECTIVES: Gemcitabine (2,2-difluorodeoxycytidine [dFdC]) can be administered in a standard 30-minute infusion or in a fixed-dose-rate (FDR) infusion to maximize the rate of accumulation of triphosphate, its major intracellular metabolite. The standard 30-minute infusion requires dose adjustment in patients with organ dysfunction, especially in patients with elevated baseline serum bilirubin levels. On the other hand, the FDR infusion is burdened by increased haematological toxicity. The primary aim of this study was to evaluate the pharmacokinetics of dFdC and its metabolite difluorodeoxyuridine (dFdU) in patients with normal and impaired hepatic function. PATIENTS AND METHODS: In this prospective study, patients with pancreatic or biliary tract carcinoma and normal or impaired hepatic function tests were considered eligible for recruitment. Patients were recruited according to the following criteria: (i) serum bilirubin <1.6 mg/dL and AST and ALT <2 times the upper the limit of normal (ULN) [cohort I]; and (ii) serum bilirubin >1.6 mg/dL and/or AST/ALT >2 times the ULN (cohort II). An FDR infusion of gemcitabine 1000 mg/m2 was administered on days 1, 8 and 15 every 4 weeks. The pharmacokinetic analysis of gemcitabine and dFdU was performed with high-performance liquid chromatography-tandem mass spectrometry assay in cycles 1 and 2. RESULTS: Thirteen patients were enrolled, four in cohort I and nine in cohort II. All patients were assessable for toxicity and pharmacokinetic analysis. The grade and rate of toxicities were similar in both groups, and patients with elevation of bilirubin and/or transaminases did not require dose reduction of gemcitabine. Pharmacokinetic analysis revealed a reduction of the experimental area under the plasma concentration-time curve for gemcitabine and dFdU in patients with hepatic dysfunction when compared with patients with normal hepatic function. All other pharmacokinetic parameters were similar in the two cohorts. No statistical difference was demonstrated for all parameters evaluated between cycle 1 and cycle 2 in the two groups. CONCLUSION: Gemcitabine 1000 mg/m2 can be administered as an FDR infusion in patients with altered hepatic function without causing additional toxicity compared with patients with normal hepatic function.


Adenocarcinoma/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Biliary Tract Neoplasms/blood , Deoxycytidine/analogs & derivatives , Liver Diseases/blood , Pancreatic Neoplasms/blood , Adenocarcinoma/complications , Adenocarcinoma/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Area Under Curve , Biliary Tract Neoplasms/complications , Biliary Tract Neoplasms/drug therapy , Chromatography, High Pressure Liquid , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Floxuridine/analogs & derivatives , Floxuridine/pharmacokinetics , Humans , Infusions, Intravenous , Liver Diseases/complications , Liver Diseases/physiopathology , Liver Function Tests , Male , Middle Aged , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/drug therapy , Prospective Studies , Tandem Mass Spectrometry , Gemcitabine
18.
In Vivo ; 20(6A): 735-8, 2006.
Article En | MEDLINE | ID: mdl-17203757

BACKGROUND: A combination of doxorubicin and tumor necrosis factor alpha (TNFalpha) has been proven to be very effective in the perfusional treatment of advanced soft tissue limb sarcoma both in terms of tumor necrosis and limb conservative surgery rate. Unfortunately, in some patients a grade IV limb reaction has been recorded. The key solution might be the use of liposomal doxorubicin (Caelyx) because the carrier seems to release the drug preferentially in the tumor rather than in the healthy tissue. PATIENTS AND METHODS: Twenty patients were treated with Caelyx: 14 with Caelyx alone and 6 in combination with a low TNFalpha dose (1 mg). In the first series of 14 patients a dose escalation study was carried out starting from a dose of 10 mg/L of limb volume. Six patients were treated with Caelyx (16 mg) and TNFalpha (1 mg). RESULTS: The maximum tolerated dose (MTD) was 16 mg/L as in two patients treated with 18 mg/L a grade IV limb reaction was observed. Tumor response was satisfactory and conservative surgery was carried out in 13 patients. In 6 patients treated with Caelyx and TNFalpha, only a grade I limb reaction was recorded, thus, confirming that TNFalpha did not increase toxicity, at least at a dose of 1 mg. The Caelyx-TNFalpha combination did increase treatment efficacy. Tumor necrosis > or = 70% was observed in 4 out of 6 patients, one with 100% necrosis (pathological complete response). All the patients underwent conservative surgery. CONCLUSION: The Caelyx-TNFalpha combination was proven to increase the efficacy of Caelyx alone, with a very low toxicity. These preliminary results have to be tested in a larger patient population.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Adult , Aged , Chemotherapy, Cancer, Regional Perfusion , Combined Modality Therapy , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Male , Middle Aged , Sarcoma/pathology , Sarcoma/surgery , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/surgery , Treatment Outcome , Tumor Necrosis Factor-alpha/administration & dosage
...