Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
PLoS One ; 11(11): e0165856, 2016.
Article in English | MEDLINE | ID: mdl-27846280

ABSTRACT

In the age of personalized medicine stratifying tumors into molecularly defined subtypes associated with distinctive clinical behaviors and predictable responses to therapies holds tremendous value. Towards this end, we developed a custom microfluidics-based bladder cancer gene expression panel for characterization of archival clinical samples. In silico analysis indicated that the content of our panel was capable of accurately segregating bladder cancers from several public datasets into the clinically relevant basal and luminal subtypes. On a technical level, our bladder cancer panel yielded robust and reproducible results when analyzing formalin-fixed, paraffin-embedded (FFPE) tissues. We applied our panel in the analysis of a novel set of 204 FFPE samples that included non-muscle invasive bladder cancers (NMIBCs), muscle invasive disease (MIBCs), and bladder cancer metastases (METs). We found NMIBCs to be mostly luminal-like, MIBCs to include both luminal- and basal-like types, and METs to be predominantly of a basal-like transcriptional profile. Mutational analysis confirmed the expected enrichment of FGFR3 mutations in luminal samples, and, consistently, FGFR3 IHC showed high protein expression levels of the receptor in these tumors. Our bladder cancer panel enables basal/luminal characterization of FFPE tissues and with further development could be used for stratification of bladder cancer samples in the clinic.


Subject(s)
Biological Specimen Banks , Gene Expression Regulation, Neoplastic , Microfluidics/methods , Transcription, Genetic , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Computer Simulation , Female , Formaldehyde , Genes, Neoplasm , Humans , Male , Middle Aged , Paraffin Embedding , Reproducibility of Results , Tissue Fixation , Urinary Bladder Neoplasms/pathology
2.
NPJ Breast Cancer ; 2: 16022, 2016.
Article in English | MEDLINE | ID: mdl-28721382

ABSTRACT

Breast cancer is a heterogeneous disease and patients are managed clinically based on ER, PR, HER2 expression, and key risk factors. We sought to characterize the molecular landscape of high-risk breast cancer patients enrolled onto an adjuvant chemotherapy study to understand how disease subsets and tumor immune status impact survival. DNA and RNA were extracted from 861 breast cancer samples from patients enrolled onto the United States Oncology trial 01062. Samples were characterized using multiplex gene expression, copy number, and qPCR mutation assays. HR+ patients with a PIK3CA mutant tumor had a favorable disease-free survival (DFS; HR 0.66, P=0.05), however, the prognostic effect was specific to luminal A patients (Luminal A: HR 0.67, P=0.1; Luminal B: HR 1.01, P=0.98). Molecular subtyping of triple-negative breast cancers (TNBCs) suggested that the mesenchymal subtype had the worst DFS, whereas the immunomodulatory subtype had the best DFS. Profiling of immunologic genes revealed that TNBC tumors (n=280) displaying an activated T-cell signature had a longer DFS following adjuvant chemotherapy (HR 0.59, P=0.04), while a distinct set of immune genes was associated with DFS in HR+ cancers. Utilizing a discovery approach, we identified genes associated with a high risk of recurrence in HR+ patients, which were validated in an independent data set. Molecular classification based on PAM50 and TNBC subtyping stratified clinical high-risk patients into distinct prognostic subsets. Patients with high expression of immune-related genes showed superior DFS in both HR+ and TNBC. These results may inform patient management and drug development in early breast cancer.

3.
Breast Cancer Res Treat ; 148(2): 315-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25338319

ABSTRACT

Breast cancers are categorized into three subtypes based on protein expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2/ERBB2). Patients enroll onto experimental clinical trials based on ER, PR, and HER2 status and, as receptor status is prognostic and defines treatment regimens, central receptor confirmation is critical for interpreting results from these trials. Patients enrolling onto experimental clinical trials in the metastatic setting often have limited available archival tissue that might better be used for comprehensive molecular profiling rather than slide-intensive reconfirmation of receptor status. We developed a Random Forests-based algorithm using a training set of 158 samples with centrally confirmed IHC status, and subsequently validated this algorithm on multiple test sets with known, locally determined IHC status. We observed a strong correlation between target mRNA expression and IHC assays for HER2 and ER, achieving an overall accuracy of 97 and 96%, respectively. For determining PR status, which had the highest discordance between central and local IHC, incorporation of expression of co-regulated genes in a multivariate approach added predictive value, outperforming the single, target gene approach by a 10% margin in overall accuracy. Our results suggest that multiplexed qRT-PCR profiling of ESR1, PGR, and ERBB2 mRNA, along with several other subtype associated genes, can effectively confirm breast cancer subtype, thereby conserving tumor sections and enabling additional biomarker data to be obtained from patients enrolled onto experimental clinical trials.


Subject(s)
Algorithms , Biomarkers, Tumor/genetics , Breast Neoplasms/classification , Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , RNA, Neoplasm/genetics , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Clinical Trials, Phase III as Topic , Female , Follow-Up Studies , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Limit of Detection , Multicenter Studies as Topic , Multivariate Analysis , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , ROC Curve , Randomized Controlled Trials as Topic , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
4.
PLoS One ; 9(3): e90761, 2014.
Article in English | MEDLINE | ID: mdl-24658394

ABSTRACT

Molecular profiling of tumor tissue to detect alterations, such as oncogenic mutations, plays a vital role in determining treatment options in oncology. Hence, there is an increasing need for a robust and high-throughput technology to detect oncogenic hotspot mutations. Although commercial assays are available to detect genetic alterations in single genes, only a limited amount of tissue is often available from patients, requiring multiplexing to allow for simultaneous detection of mutations in many genes using low DNA input. Even though next-generation sequencing (NGS) platforms provide powerful tools for this purpose, they face challenges such as high cost, large DNA input requirement, complex data analysis, and long turnaround times, limiting their use in clinical settings. We report the development of the next generation mutation multi-analyte panel (MUT-MAP), a high-throughput microfluidic, panel for detecting 120 somatic mutations across eleven genes of therapeutic interest (AKT1, BRAF, EGFR, FGFR3, FLT3, HRAS, KIT, KRAS, MET, NRAS, and PIK3CA) using allele-specific PCR (AS-PCR) and Taqman technology. This mutation panel requires as little as 2 ng of high quality DNA from fresh frozen or 100 ng of DNA from formalin-fixed paraffin-embedded (FFPE) tissues. Mutation calls, including an automated data analysis process, have been implemented to run 88 samples per day. Validation of this platform using plasmids showed robust signal and low cross-reactivity in all of the newly added assays and mutation calls in cell line samples were found to be consistent with the Catalogue of Somatic Mutations in Cancer (COSMIC) database allowing for direct comparison of our platform to Sanger sequencing. High correlation with NGS when compared to the SuraSeq500 panel run on the Ion Torrent platform in a FFPE dilution experiment showed assay sensitivity down to 0.45%. This multiplexed mutation panel is a valuable tool for high-throughput biomarker discovery in personalized medicine and cancer drug development.


Subject(s)
DNA Mutational Analysis , Microfluidics/methods , Class I Phosphatidylinositol 3-Kinases , ErbB Receptors/genetics , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Reproducibility of Results , fms-Like Tyrosine Kinase 3/genetics , ras Proteins/genetics
5.
PLoS One ; 9(2): e88401, 2014.
Article in English | MEDLINE | ID: mdl-24520381

ABSTRACT

Patients with newly diagnosed, early stage estrogen receptor positive (ER+) breast cancer often show disease free survival in excess of five years following surgery and systemic adjuvant therapy. An important question is whether diagnostic tumor tissue from the primary lesion offers an accurate molecular portrait of the cancer post recurrence and thus may be used for predictive diagnostic purposes for patients with relapsed, metastatic disease. As the class I phosphatidylinositol 3' kinase (PI3K) pathway is frequently activated in ER+ breast cancer and has been linked to acquired resistance to hormonal therapy, we hypothesized pathway status could evolve over time and treatment. Biomarker analyses were conducted on matched, asynchronous primary and metastatic tumors from 77 patients with ER+ breast cancer. We examined whether PIK3CA and AKT1 alterations or PTEN and Ki67 levels showed differences between primary and metastatic samples. We also sought to look more broadly at gene expression markers reflective of proliferation, molecular subtype, and key receptors and signaling pathways using an mRNA analysis platform developed on the Fluidigm BioMark™ microfluidics system to measure the relative expression of 90 breast cancer related genes in formalin-fixed paraffin-embedded (FFPE) tissue. Application of this panel of biomarker assays to matched tumor pairs showed a high concordance between primary and metastatic tissue, with generally few changes in mutation status, proliferative markers, or gene expression between matched samples. The collection of assays described here has been optimized for FFPE tissue and may have utility in exploratory analyses to identify patient subsets responsive to targeted therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Metabolome , Receptors, Estrogen/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Microfluidics , Mutation/genetics , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Signal Transduction , Tumor Cells, Cultured
6.
Methods Mol Biol ; 1050: 207-22, 2014.
Article in English | MEDLINE | ID: mdl-24297362

ABSTRACT

The ability to directly manipulate the human genome to correct a disease-related mutation, introduce a sequence change that would lead to site-specific gene knockout, or increase gene expression is a very powerful tool with tremendous clinical value. Triplex formation by synthetic DNA-binding molecules such as peptide nucleic acids (PNAs) has been studied for over 20 years and much of the work in the last 10 years has shown its great promise in its use to direct site-specific gene modification for the use in gene therapy. In this chapter, detailed protocols are described for the design and use of triplex-forming PNAs to bind and mediate gene modification at specific chromosomal targets. Target site identification, PNA and donor oligonucleotide design, in vitro characterization of binding, optimization with reporter systems, as well as various methods to assess gene modification and isolate modified cells are described.


Subject(s)
DNA Repair/genetics , Genetic Techniques , Genome, Human/genetics , Peptide Nucleic Acids/metabolism , Recombination, Genetic , Alleles , Animals , Base Sequence , Cell Line , Cell-Penetrating Peptides/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Genes, Reporter/genetics , Genetic Loci/genetics , Humans , Lactic Acid/chemistry , Molecular Sequence Data , Nanoparticles/chemistry , Nucleic Acid Conformation , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymerase Chain Reaction , Transfection
7.
Mol Ther Nucleic Acids ; 2: e135, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24253260

ABSTRACT

Biodegradable poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) encapsulating triplex-forming peptide nucleic acids (PNAs) and donor DNAs for recombination-mediated editing of the CCR5 gene were synthesized for delivery into human peripheral blood mononuclear cells (PBMCs). NPs containing the CCR5-targeting molecules efficiently entered PBMCs with low cytotoxicity. Deep sequencing revealed that a single treatment with the formulation resulted in a targeting frequency of 0.97% in the CCR5 gene and a low off-target frequency of 0.004% in the CCR2 gene, a 216-fold difference. NP-treated PBMCs efficiently engrafted immunodeficient NOD-scid IL-2rγ(-/-) mice, and the targeted CCR5 modification was detected in splenic lymphocytes 4 weeks posttransplantation. After infection with an R5-tropic strain of HIV-1, humanized mice with CCR5-NP-treated PBMCs displayed significantly higher levels of CD4(+) T cells and significantly reduced plasma viral RNA loads compared with control mice engrafted with mock-treated PBMCs. This work demonstrates the feasibility of PLGA-NP-encapsulated PNA-based gene-editing molecules for the targeted modification of CCR5 in human PBMCs as a platform for conferring HIV-1 resistance.Molecular Therapy-Nucleic Acids (2013) 2, e135; doi:10.1038/mtna.2013.59; published online 19 November 2013.

8.
Chem Biol ; 18(9): 1189-98, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21944757

ABSTRACT

Peptide nucleic acids (PNAs) bind duplex DNA in a sequence-specific manner, creating triplex structures that can provoke DNA repair and produce genome modification. CCR5 encodes a chemokine receptor required for HIV-1 entry into human cells, and individuals carrying mutations in this gene are resistant to HIV-1 infection. Transfection of human cells with PNAs targeted to the CCR5 gene, plus donor DNAs designed to introduce stop codons mimicking the naturally occurring CCR5-delta32 mutation, produced 2.46% targeted gene modification. CCR5 modification was confirmed at the DNA, RNA, and protein levels and was shown to confer resistance to infection with HIV-1. Targeting of CCR5 was achieved in human CD34(+) hematopoietic stem cells (HSCs) with subsequent engraftment into mice and persistence of the gene modification more than four months posttransplantation. This work suggests a therapeutic strategy for CCR5 knockout in HSCs from HIV-1-infected individuals, rendering cells resistant to HIV-1 and preserving immune system function.


Subject(s)
Hematopoietic Stem Cells/metabolism , Peptide Nucleic Acids/pharmacology , Receptors, CCR5/metabolism , Animals , Antigens, CD34/metabolism , Base Sequence , Binding Sites , CCR5 Receptor Antagonists , Cell Line , Codon, Terminator , DNA Repair , Gene Targeting/methods , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/virology , Humans , Mice , Mutation , Peptide Nucleic Acids/chemistry , Receptors, CCR5/genetics
9.
J Control Release ; 155(2): 312-6, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-21620910

ABSTRACT

Triplex-forming peptide nucleic acids (PNAs) can be used to coordinate the recombination of short 50-60bp "donor DNA" fragments into genomic DNA, resulting in site-specific correction of genetic mutations or the introduction of advantageous genetic modifications. Site-specific gene editing in hematopoietic stem and progenitor cells (HSPCs) could result in the treatment or cure of inherited disorders of the blood such as ß-thalassemia or sickle cell anemia. Gene editing in HSPCs and differentiated T cells could also help combat HIV infection by modifying the HIV co-receptor CCR5, which is necessary for R5-tropic HIV entry. However, translation of genome modification technologies to clinical practice is limited by challenges in intracellular delivery, especially in difficult-to-transfect hematolymphoid cells. Here, we review the use of engineered biodegradable polymer nanoparticles for site-specific genome editing in human hematopoietic cells, which represent a promising approach for ex vivo and in vivo gene therapy.


Subject(s)
Drug Carriers/chemistry , Peptide Nucleic Acids/administration & dosage , Polymers/chemistry , Targeted Gene Repair , Animals , Biocompatible Materials/chemistry , DNA/administration & dosage , DNA/chemistry , DNA/drug effects , Gene Transfer Techniques , Genome , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Nanoparticles/chemistry , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/genetics
10.
Mol Ther ; 19(1): 172-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20859257

ABSTRACT

Triplex-forming peptide nucleic acids (PNAs) are powerful gene therapy agents that can enhance recombination of short donor DNAs with genomic DNA, leading to targeted and specific correction of disease-causing genetic mutations. Therapeutic use of PNAs is severely limited, however, by challenges in intracellular delivery, particularly in clinically relevant targets such as hematopoietic stem and progenitor cells. Here, we demonstrate efficient and nontoxic PNA-mediated recombination in human CD34(+) cells using poly(lactic-co-glycolic acid) (PLGA) nanoparticles for intracellular oligonucleotide delivery. Treatment of progenitor cells with nanoparticles loaded with PNAs and DNAs targeting the ß-globin locus led to levels of site-specific modification in the range of 0.5-1% in a single treatment, without detectable loss in cell viability, resulting in a 60-fold increase in modified and viable cells as compared to nucleofection. As well, the differentiation capacity of the progenitor cells treated with nanoparticles did not change relative to untreated progenitor cells, indicating that nanoparticles are safe and minimally disruptive delivery vectors for PNAs and DNAs to mediate gene modification in human primary cells. This is the first demonstration of the use of biodegradable nanoparticles to deliver genome-editing agents to human primary cells, and provides a strong rationale for systemic delivery of complex nucleic acid mixtures designed for gene correction.


Subject(s)
Antigens, CD34/biosynthesis , Hematopoietic Stem Cells/physiology , Nanoparticles/administration & dosage , Peptide Nucleic Acids/administration & dosage , Recombination, Genetic , Targeted Gene Repair , Cell Survival/drug effects , Cells, Cultured , DNA/genetics , Gene Targeting/methods , Gene Transfer Techniques , Genome , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Lactic Acid/pharmacology , Nanoparticles/chemistry , Oligonucleotides/pharmacology , Particle Size , Peptide Nucleic Acids/genetics , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, CCR5/genetics , beta-Globins/genetics
11.
Methods Mol Biol ; 435: 175-90, 2008.
Article in English | MEDLINE | ID: mdl-18370076

ABSTRACT

Gene targeting with DNA-binding molecules such as triplex-forming oligonucleotides or peptide nucleic acids can be utilized to direct mutagenesis or induce recombination site-specifically. In this chapter, several detailed protocols are described for the design and use of triplex-forming molecules to bind and mediate gene modification at specific chromosomal targets. Target site identification, binding molecule design, as well as various methods to test binding and assess gene modification are described.


Subject(s)
Gene Targeting/methods , Animals , Base Sequence , Binding Sites/genetics , CHO Cells , Cricetinae , Cricetulus , DNA/genetics , DNA/metabolism , Genes, Reporter , Genetic Techniques , Luciferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed/methods , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/genetics , Peptide Nucleic Acids/metabolism , Recombination, Genetic
12.
Front Biosci ; 12: 4288-97, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485375

ABSTRACT

Triple-helix DNA structures can form endogenously at mirror repeat polypurine/polypyrimidine sequences or by introduction of triplex-forming oligonucleotides (TFOs). Recent evidence suggests that triple helices are sources of genetic instability, and are subject to increased rates of mutagenesis and recruitment of repair factors. Indeed, observations using TFOs suggest that triple helices provoke a variety of biological processes which can be harnessed to modulate gene expression and induce heritable changes in targeted genes. This review surveys the biological applications of TFOs, with particular attention to their recombinogenic and mutagenic potential, and summarizes available evidence for the mechanism of triplex and triplex-associated repair.


Subject(s)
DNA Repair , DNA/physiology , Recombination, Genetic , DNA/chemistry , Nucleic Acid Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...