Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Lung Cancer ; 190: 107506, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422883

ABSTRACT

BACKGROUND: First-line pembrolizumab plus chemotherapy has shown clinical benefit in patients with metastatic non-small cell lung cancer (NSCLC) regardless of tissue tumor mutational burden (tTMB) status. Blood tumor mutational burden (bTMB), assessed using plasma-derived circulating tumor DNA (ctDNA), may be a surrogate for tTMB. The KEYNOTE-782 study evaluated the correlation of bTMB with the efficacy of first-line pembrolizumab plus chemotherapy in NSCLC. METHODS: Previously untreated patients with stage IV nonsquamous NSCLC received pembrolizumab 200 mg plus pemetrexed 500 mg/m2 and investigator's choice of carboplatin area under the curve 5 mg/mL/min or cisplatin 75 mg/m2 for 4 cycles, then pembrolizumab plus pemetrexed for ≤31 additional cycles every 3 weeks. Study objectives were to evaluate the association of baseline bTMB with objective response rate (ORR) (RECIST v1.1 by investigator assessment; primary), progression-free survival (PFS; RECIST v1.1 by investigator assessment), overall survival (OS), and adverse events (AEs; all secondary). A next-generation sequencing assay (GRAIL LLC) with a ctDNA panel that included lung cancer-associated and immune gene targets was used to measure bTMB. RESULTS: 117 patients were enrolled; median time from first dose to data cutoff was 19.3 months (range, 1.0-35.5). ORR was 40.2 % (95 % CI 31.2-49.6 %), median PFS was 7.2 months (95 % CI 5.6-9.8) and median OS was 18.1 months (95 % CI 13.5-25.6). Treatment-related AEs occurred in 113 patients (96.6 %; grade 3-5, n = 56 [47.9 %]). Of patients with evaluable bTMB (n = 101), the area under the receiver operating characteristics curve for continuous bTMB to discriminate response was 0.47 (95 % CI 0.36-0.59). Baseline bTMB was not associated with PFS or OS (posterior probabilities of positive association: 16.8 % and 7.8 %, respectively). CONCLUSIONS: AEs were consistent with the established safety profile of first-line pembrolizumab plus chemotherapy in NSCLC. Baseline bTMB did not show evidence of an association with efficacy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Pemetrexed/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
PLoS One ; 18(4): e0283001, 2023.
Article in English | MEDLINE | ID: mdl-37058491

ABSTRACT

The analytical validation is reported for a targeted methylation-based cell-free DNA multi-cancer early detection test designed to detect cancer and predict the cancer signal origin (tissue of origin). A machine-learning classifier was used to analyze the methylation patterns of >105 genomic targets covering >1 million methylation sites. Analytical sensitivity (limit of detection [95% probability]) was characterized with respect to tumor content by expected variant allele frequency and was determined to be 0.07%-0.17% across five tumor cases and 0.51% for the lymphoid neoplasm case. Test specificity was 99.3% (95% confidence interval, 98.6-99.7%). In the reproducibility and repeatability study, results were consistent in 31/34 (91.2%) pairs with cancer and 17/17 (100%) pairs without cancer; between runs, results were concordant for 129/133 (97.0%) cancer and 37/37 (100%) non-cancer sample pairs. Across 3- to 100-ng input levels of cell-free DNA, cancer was detected in 157/182 (86.3%) cancer samples but not in any of the 62 non-cancer samples. In input titration tests, cancer signal origin was correctly predicted in all tumor samples detected as cancer. No cross-contamination events were observed. No potential interferent (hemoglobin, bilirubin, triglycerides, genomic DNA) affected performance. The results of this analytical validation study support continued clinical development of a targeted methylation cell-free DNA multi-cancer early detection test.


Subject(s)
Cell-Free Nucleic Acids , Neoplasms , Cell-Free Nucleic Acids/genetics , Sensitivity and Specificity , Early Detection of Cancer , Reproducibility of Results , DNA Methylation/genetics , Biomarkers, Tumor/genetics , Neoplasms/diagnosis , Neoplasms/genetics
3.
Cancer Cell ; 40(12): 1537-1549.e12, 2022 12 12.
Article in English | MEDLINE | ID: mdl-36400018

ABSTRACT

In the Circulating Cell-free Genome Atlas (NCT02889978) substudy 1, we evaluate several approaches for a circulating cell-free DNA (cfDNA)-based multi-cancer early detection (MCED) test by defining clinical limit of detection (LOD) based on circulating tumor allele fraction (cTAF), enabling performance comparisons. Among 10 machine-learning classifiers trained on the same samples and independently validated, when evaluated at 98% specificity, those using whole-genome (WG) methylation, single nucleotide variants with paired white blood cell background removal, and combined scores from classifiers evaluated in this study show the highest cancer signal detection sensitivities. Compared with clinical stage and tumor type, cTAF is a more significant predictor of classifier performance and may more closely reflect tumor biology. Clinical LODs mirror relative sensitivities for all approaches. The WG methylation feature best predicts cancer signal origin. WG methylation is the most promising technology for MCED and informs development of a targeted methylation MCED test.


Subject(s)
Cell-Free Nucleic Acids , Neoplasms , Humans , Cell-Free Nucleic Acids/genetics , Early Detection of Cancer , Neoplasms/diagnosis , Neoplasms/genetics , Biomarkers, Tumor/genetics , DNA Methylation
4.
JCO Precis Oncol ; 6: e2100372, 2022 08.
Article in English | MEDLINE | ID: mdl-35952319

ABSTRACT

PURPOSE: As immune checkpoint inhibitors (ICI) become increasingly used in frontline settings, identifying early indicators of response is needed. Recent studies suggest a role for circulating tumor DNA (ctDNA) in monitoring response to ICI, but uncertainty exists in the generalizability of these studies. Here, the role of ctDNA for monitoring response to ICI is assessed through a standardized approach by assessing clinical trial data from five independent studies. PATIENTS AND METHODS: Patient-level clinical and ctDNA data were pooled and harmonized from 200 patients across five independent clinical trials investigating the treatment of patients with non-small-cell lung cancer with programmed cell death-1 (PD-1)/programmed death ligand-1 (PD-L1)-directed monotherapy or in combination with chemotherapy. CtDNA levels were measured using different ctDNA assays across the studies. Maximum variant allele frequencies were calculated using all somatic tumor-derived variants in each unique patient sample to correlate ctDNA changes with overall survival (OS) and progression-free survival (PFS). RESULTS: We observed strong associations between reductions in ctDNA levels from on-treatment liquid biopsies with improved OS (OS; hazard ratio, 2.28; 95% CI, 1.62 to 3.20; P < .001) and PFS (PFS; hazard ratio 1.76; 95% CI, 1.31 to 2.36; P < .001). Changes in the maximum variant allele frequencies ctDNA values showed strong association across different outcomes. CONCLUSION: In this pooled analysis of five independent clinical trials, consistent and robust associations between reductions in ctDNA and outcomes were found across multiple end points assessed in patients with non-small-cell lung cancer treated with an ICI. Additional tumor types, stages, and drug classes should be included in future analyses to further validate this. CtDNA may serve as an important tool in clinical development and an early indicator of treatment benefit.


Subject(s)
Antineoplastic Agents, Immunological , Carcinoma, Non-Small-Cell Lung , Circulating Tumor DNA , Lung Neoplasms , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Circulating Tumor DNA/genetics , Clinical Trials as Topic , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Prognosis
5.
PLoS One ; 13(2): e0193476, 2018.
Article in English | MEDLINE | ID: mdl-29474437

ABSTRACT

INTRODUCTION: Non-invasive prenatal testing (NIPT) for aneuploidy using cell-free DNA in maternal plasma has been widely adopted. Recently, NIPT coverage has expanded to detect subchromosomal abnormalities including the 22q11.2 deletion. Validation of a SNP-based NIPT for detection of 22q11.2 deletions demonstrating a high sensitivity (97.8%) and specificity (99.75%) has been reported. We sought to further demonstrate the performance of a revised version of the test in a larger set of pregnancy plasma samples. METHODS: Blood samples from pregnant women (10 with 22q11.2-deletion‒affected fetuses and 390 negative controls) were successfully analyzed using a revised SNP-based NIPT for the 22q11.2 deletion. The sensitivity and specificity of the assay were measured. RESULTS: Sensitivity of the assay was 90% (9/10), and specificity of the assay was 99.74% (389/390), with a corresponding false positive-rate of 0.26%. DISCUSSION: The data presented in this study add to the growing body of evidence demonstrating the ability of the SNP-based NIPT to detect 22q11.2 deletions with high sensitivity and specificity.


Subject(s)
Chromosome Deletion , Genetic Testing/methods , Mothers , Plasma/metabolism , Polymorphism, Single Nucleotide , Prenatal Diagnosis/methods , Adult , Chromosomes, Human, Pair 22 , Female , Humans , Pregnancy
6.
Fetal Diagn Ther ; 40(3): 219-223, 2016.
Article in English | MEDLINE | ID: mdl-27028530

ABSTRACT

OBJECTIVE: To validate an updated version (Version 2) of a single-nucleotide polymorphism (SNP)-based noninvasive prenatal test (NIPT) and to determine the likelihood of success when testing for fetal aneuploidies following a redraw. METHODS: Version 2 was analytically validated using 587 plasma samples with known genotype (184 trisomy 21, 37 trisomy 18, 15 trisomy 13, 9 monosomy X, 4 triploidy and 338 euploid). Sensitivity, specificity and no-call rate were calculated, and a fetal-fraction adjustment was applied to enable projection of these values in a commercial distribution. Likelihood of success of a second blood draw was computed based on fetal fraction and maternal weight from the first draw. RESULTS: Validation of this methodology yielded high sensitivities (≥99.4%) and specificities (100%) for all conditions tested with an observed no-call rate of 2.3%. The no-call threshold for sample calling was reduced to 2.8% fetal fraction. The redraw success rate was driven by higher initial fetal fractions and lower maternal weights, with the fetal fraction being the more significant variable. CONCLUSIONS: The enhanced version of this SNP-based NIPT method showed a reduced no-call rate and a reduced fetal-fraction threshold for sample calling in comparison to the earlier version, while maintaining high sensitivity and specificity.


Subject(s)
Aneuploidy , Genetic Testing/methods , Maternal Serum Screening Tests/methods , Adult , Female , Gestational Age , Humans , Polymorphism, Single Nucleotide , Pregnancy
7.
Obstet Gynecol ; 124(2 Pt 1): 210-218, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25004354

ABSTRACT

OBJECTIVE: To estimate performance of a single-nucleotide polymorphism-based noninvasive prenatal screen for fetal aneuploidy in high-risk and low-risk populations on single venopuncture. METHODS: One thousand sixty-four maternal blood samples from 7 weeks of gestation and beyond were included; 1,051 were within specifications and 518 (49.3%) were low risk. Cell-free DNA was amplified, sequenced, and analyzed using the Next-generation Aneuploidy Test Using SNPs algorithm. Samples were called as trisomies 21, 18, 13, or monosomy X, or euploid, and male or female. RESULTS: Nine hundred sixty-six samples (91.9%) successfully generated a cell-free DNA result. Among these, sensitivity was 100% for trisomy 21 (58/58, confidence interval [CI] 93.8-100%), trisomy 13 (12/12, CI 73.5-100%), and fetal sex (358/358 female, CI 99.0-100%; 418/418 male, CI 99.1-100%), 96.0% for trisomy 18 (24/25, CI 79.7-99.9%), and 90% for monosomy X (9/10, CI 55.5-99.8%). Specificity for trisomies 21 and 13 was 100% (905/905, CI 99.6-100%; and 953/953, CI 99.6-100%, respectively) and for trisomy 18 and monosomy X was 99.9% (938/939, CI 99.4-100%; and 953/954, CI 99.4-100%, respectively). However, 16% (20/125) of aneuploid samples did not return a result; 50% (10/20) had a fetal fraction below the 1.5th percentile of euploid pregnancies. Aneuploidy rate was significantly higher in these samples (P<.001, odds ratio 9.2, CI 4.4-19.0). Sensitivity and specificity did not differ in low-risk and high-risk populations. CONCLUSIONS: This noninvasive prenatal screen performed with high sensitivity and specificity in high-risk and low-risk cohorts. Aneuploid samples were significantly more likely to not return a result; the number of aneuploidy samples was especially increased among samples with low fetal fraction. This underscores the importance of redraws or, in rare cases, invasive procedures based on low fetal fraction. LEVEL OF EVIDENCE: II.


Subject(s)
Aneuploidy , Chromosome Disorders/diagnosis , DNA/blood , Down Syndrome/diagnosis , Polymorphism, Single Nucleotide , Prenatal Diagnosis/methods , Trisomy/diagnosis , Turner Syndrome/diagnosis , Adolescent , Adult , Algorithms , Cell-Free System , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 18 , Female , Humans , Male , Middle Aged , Pregnancy , Risk Factors , Sensitivity and Specificity , Trisomy 13 Syndrome , Trisomy 18 Syndrome , Young Adult
8.
J Clin Invest ; 123(7): 2862-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23934129

ABSTRACT

During human pregnancy, a subset of placental cytotrophoblasts (CTBs) differentiates into cells that aggressively invade the uterus and its vasculature, anchoring the progeny and rerouting maternal blood to the placenta. In preeclampsia (PE), CTB invasion is limited, reducing placental perfusion and/or creating intermittent flow. This syndrome, affecting 4%-8% of pregnancies, entails maternal vascular alterations (e.g., high blood pressure, proteinuria, and edema) and, in some patients, fetal growth restriction. The only cure is removal of the faulty placenta, i.e., delivery. Previously, we showed that defective CTB differentiation contributes to the placental component of PE, but the causes were unknown. Here, we cultured CTBs isolated from PE and control placentas for 48 hours, enabling differentiation and invasion. In various severe forms of PE, transcriptomics revealed common aberrations in CTB gene expression immediately after isolation, including upregulation of SEMA3B, which resolved in culture. The addition of SEMA3B to normal CTBs inhibited invasion and recreated aspects of the PE phenotype. Additionally, SEMA3B downregulated VEGF signaling through the PI3K/AKT and GSK3 pathways, effects that were observed in PE CTBs. We propose that, in severe PE, the in vivo environment dysregulates CTB gene expression; the autocrine actions of the upregulated molecules (including SEMA3B) impair CTB differentiation, invasion and signaling; and patient-specific factors determine the signs.


Subject(s)
Gene Expression Regulation , Pre-Eclampsia/metabolism , Transcriptome , Trophoblasts/metabolism , Animals , COS Cells , Cell Differentiation , Cell Movement , Chick Embryo , Chlorocebus aethiops , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Female , Glycogen Synthase Kinase 3/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neovascularization, Pathologic/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Oligonucleotide Array Sequence Analysis , Placenta/pathology , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction , Trophoblasts/physiology , Vascular Endothelial Growth Factor A/physiology , beta Catenin/metabolism
9.
Am J Clin Exp Immunol ; 1(2): 70-89, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23304658

ABSTRACT

Profiling miRNA expression in cells that directly contribute to human disease pathogenesis is likely to aid the discovery of novel drug targets and biomarkers. However, tissue heterogeneity and the limited amount of human diseased tissue available for research purposes present fundamental difficulties that often constrain the scope and potential of such studies. We established a flow cytometry-based method for isolating pure populations of pathogenic T cells from bronchial biopsy samples of asthma patients, and optimized a high-throughput nano-scale qRT-PCR method capable of accurately measuring 96 miRNAs in as little as 100 cells. Comparison of circulating and airway T cells from healthy and asthmatic subjects revealed asthma-associated and tissue-specific miRNA expression patterns. These results establish the feasibility and utility of investigating miRNA expression in small populations of cells involved in asthma pathogenesis, and set a precedent for application of our nano-scale approach in other human diseases. The microarray data from this study (Figure 7) has been submitted to the NCBI Gene Expression Omnibus (GEO; http://ncbi.nlm.nih.gov/geo) under accession no. GSE31030.

10.
J Vis Exp ; (54)2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21847076

ABSTRACT

The broad involvement of miRNAs in critical processes underlying development, tissue homoeostasis and disease has led to a surging interest among the research and pharmaceutical communities. To study miRNAs, it is essential that the quantification of microRNA levels is accurate and robust. By comparing wild-type to small RNA deficient mouse embryonic stem cells (mESC), we revealed a lack of accuracy and robustness in previous published multiplex qRT-PCR techniques. Here, we describe an optimized method, including purifying away excessive primers from previous multiplex steps before singleplex real time detection, which dramatically increases the accuracy and robustness of the technique. Furthermore, we explain how performing the technique on a microfluidic chip at nanoliter volumes significantly reduces reagent costs and permits time effective high throughput miRNA expression profiling.


Subject(s)
Embryonic Stem Cells/chemistry , MicroRNAs/chemistry , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Embryonic Stem Cells/physiology , Mice , MicroRNAs/genetics , MicroRNAs/isolation & purification , Microfluidic Analytical Techniques , Nanotechnology/instrumentation , Nanotechnology/methods , Reverse Transcriptase Polymerase Chain Reaction/instrumentation
11.
Development ; 138(14): 2987-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21693515

ABSTRACT

Placental trophoblasts (TBs) invade and remodel uterine vessels with an arterial bias. This process, which involves vascular mimicry, re-routes maternal blood to the placenta, but fails in pre-eclampsia. We investigated Notch family members in both contexts, as they play important roles in arterial differentiation/function. Immunoanalyses of tissue sections showed step-wise modulation of Notch receptors/ligands during human TB invasion. Inhibition of Notch signaling reduced invasion of cultured human TBs and expression of the arterial marker EFNB2. In mouse placentas, Notch activity was highest in endovascular TBs. Conditional deletion of Notch2, the only receptor upregulated during mouse TB invasion, reduced arterial invasion, the size of maternal blood canals by 30-40% and placental perfusion by 23%. By E11.5, there was litter-wide lethality in proportion to the number of mutant offspring. In pre-eclampsia, expression of the Notch ligand JAG1 was absent in perivascular and endovascular TBs. We conclude that Notch signaling is crucial for TB vascular invasion.


Subject(s)
Placental Circulation/physiology , Pre-Eclampsia/physiopathology , Receptors, Notch/metabolism , Signal Transduction/physiology , Trophoblasts/physiology , Uterus/blood supply , Animals , Arteries/growth & development , Calcium-Binding Proteins/metabolism , DNA Primers/genetics , Ephrin-B2/metabolism , Female , Fluorescent Antibody Technique , Gene Deletion , Humans , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Pregnancy , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Trophoblasts/metabolism
12.
Methods Enzymol ; 445: 281-302, 2008.
Article in English | MEDLINE | ID: mdl-19022064

ABSTRACT

In eutherian mammals, the first functional organ is the placenta, a transient structure that is rapidly assembled in the extraembryonic compartment. By necessity the placenta develops in advance of the embryo, which it supports in utero by performing many of the same functions that the lungs, gastrointestinal tract, and urinary system carry out after birth. Specialized epithelial cells that arise from the placenta, termed cytotrophoblasts (CTBs), are responsible for redirecting maternal blood to the developing conceptus, which occurs as a result of the cells' aggressive invasion through the maternal endometrial stroma (interstitial invasion) and resident blood vessels (endovascular invasion). The latter process involves displacement of maternal endothelium and induction of apoptosis in the surrounding smooth muscle. Together, these events result in a reduction of blood vessel elasticity and increased blood flow. In the past, investigations of human CTB endovascular invasion have been limited to immunohistochemical examination of tissue sections. In this chapter, we will discuss the use of in vitro and in vivo techniques that have been recently adapted for the study of the complex events that occur during CTB endovascular invasion. As an introduction, we provide background on placental anatomy and the molecular basis of CTB behaviors. To follow, we present techniques used in the isolation and culture of primary CTBs and chorionic villous explants. Approaches for identifying trophoblast-modified blood vessels in placental tissue sections are also described. Next, we review methods used by other groups to study CTB/endothelial interactions in culture focusing on techniques that employ isolated cells and chorionic explants. Finally, we conclude with methods devised by our group and others to explore the complex heterotypic cell-cell interactions that occur as CTBs invade blood vessels in vivo in the nude mouse.


Subject(s)
Placenta/blood supply , Uterus/blood supply , Animals , Cell Movement , Cells, Cultured , Female , Humans , Mice , Neovascularization, Physiologic/physiology , Placenta/cytology , Placental Circulation/physiology , Pregnancy , Trophoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...