Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Neurol ; 94(4): 736-744, 2023 10.
Article in English | MEDLINE | ID: mdl-37345334

ABSTRACT

OBJECTIVE: To determine early magnetic resonance imaging (MRI) features of new multiple sclerosis (MS) lesions that will develop into paramagnetic rim lesions (PRLs), which have been associated with progressive tissue injury in MS. METHODS: New contrast-enhancing lesions observed on routine clinical MRI were imaged at 7 T within 4 weeks of observation, and 3 and 6 months later. The 6-month MRI was used to classify PRL status (PRL or non-PRL). The relationship between early lesion characteristics and subsequent PRL status was assessed using generalized linear mixed effects models. Random forest classification was performed to classify early predictors of subsequent PRL status. RESULTS: From 93 contrast-enhancing lesions in 23 MS patients, 37 lesions developed into a PRL. In lesions that developed into PRLs compared with those that did not, the average lesion T1 on the initial 7 T MRI was 1994 ms compared with 1,670 ms (p-value <0.001), and the average volume was 168.7 mL compared with 44 mL (p-value <0.001) in lesions that did not. These volume differences were also found on 3 T scans (p-value <0.001), and for intensity-normalized T1 -w (p-value = 0.011) and fluid-attenuated inversion recovery (p-value = 0.005). The area under the receiver operating characteristic curve for the random forest classification with leave-one-out cross-validation was found to be 0.86 using initial 7 T features. INTERPRETATION: New MS lesions that evolve into PRLs can be identified early in lesion evolution. These findings suggest that biological mechanisms underlying PRL development begin early, which has important implications for clinical trials targeting PRLs development and subsequent therapeutics. ANN NEUROL 2023;94:736-744.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/pathology , Disease Progression , Magnetic Resonance Imaging/methods , Brain/pathology
2.
Cytotherapy ; 25(7): 718-727, 2023 07.
Article in English | MEDLINE | ID: mdl-37278683

ABSTRACT

BACKGROUND: Adoptive T cell therapy (ATCT) has been successful in treating hematological malignancies and is currently under investigation for solid-tumor therapy. In contrast to existing chimeric antigen receptor (CAR) T cell and/or antigen-specific T cell approaches, which require known targets, and responsive to the need for targeting a broad repertoire of antigens in solid tumors, we describe the first use of immunostimulatory photothermal nanoparticles to generate tumor-specific T cells. METHODS: Specifically, we subject whole tumor cells to Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) before culturing with dendritic cells (DCs), and subsequent stimulation of T cells. This strategy differs from previous approaches using tumor cell lysates because we use nanoparticles to mediate thermal and immunogenic cell death in tumor cells, rendering them enhanced antigen sources. RESULTS: In proof-of-concept studies using two glioblastoma (GBM) tumor cell lines, we first demonstrated that when PBNP-PTT was administered at a "thermal dose" targeted to induce the immunogenicity of U87 GBM cells, we effectively expanded U87-specific T cells. Further, we found that DCs cultured ex vivo with PBNP-PTT-treated U87 cells enabled 9- to 30-fold expansion of CD4+ and CD8+ T cells. Upon co-culture with target U87 cells, these T cells secreted interferon-É£ in a tumor-specific and dose-dependent manner (up to 647-fold over controls). Furthermore, T cells manufactured using PBNP-PTT ex vivo expansion elicited specific cytolytic activity against target U87 cells (donor-dependent 32-93% killing at an effector to target cell (E:T) ratio of 20:1) while sparing normal human astrocytes and peripheral blood mononuclear cells from the same donors. In contrast, T cells generated using U87 cell lysates expanded only 6- to 24-fold and killed 2- to 3-fold less U87 target cells at matched E:T ratios compared with T cell products expanded using the PBNP-PTT approach. These results were reproducible even when a different GBM cell line (SNB19) was used, wherein the PBNP-PTT-mediated approach resulted in a 7- to 39-fold expansion of T cells, which elicited 25-66% killing of the SNB19 cells at an E:T ratio of 20:1, depending on the donor. CONCLUSIONS: These findings provide proof-of-concept data supporting the use of PBNP-PTT to stimulate and expand tumor-specific T cells ex vivo for potential use as an adoptive T cell therapy approach for the treatment of patients with solid tumors.


Subject(s)
Glioblastoma , Nanoparticles , Humans , Leukocytes, Mononuclear , Immunotherapy, Adoptive/methods , CD8-Positive T-Lymphocytes , Glioblastoma/therapy , Cell Line, Tumor
3.
Epilepsia ; 64(5): 1236-1247, 2023 05.
Article in English | MEDLINE | ID: mdl-36815252

ABSTRACT

OBJECTIVE: Evaluating patients with drug-resistant epilepsy often requires inducing seizures by tapering antiseizure medications (ASMs) in the epilepsy monitoring unit (EMU). The relationship between ASM taper strategy, seizure timing, and severity remains unclear. In this study, we developed and validated a pharmacokinetic model of total ASM load and tested its association with seizure occurrence and severity in the EMU. METHODS: We studied 80 patients who underwent intracranial electroencephalographic recording for epilepsy surgery planning. We developed a first order pharmacokinetic model of the ASMs administered in the EMU to generate a continuous metric of overall ASM load. We then related modeled ASM load to seizure likelihood and severity. We determined the association between the rate of ASM load reduction, the length of hospital stay, and the probability of having a severe seizure. Finally, we used modeled ASM load to predict oncoming seizures. RESULTS: Seizures occurred in the bottom 50th percentile of sampled ASM loads across the cohort (p < .0001, Wilcoxon signed-rank test), and seizures requiring rescue therapy occurred at lower ASM loads than seizures that did not require rescue therapy (logistic regression mixed effects model, odds ratio = .27, p = .01). Greater ASM decrease early in the EMU was not associated with an increased likelihood of having a severe seizure, nor with a shorter length of stay. SIGNIFICANCE: A pharmacokinetic model can accurately estimate ASM levels for patients in the EMU. Lower modeled ASM levels are associated with increased seizure likelihood and seizure severity. We show that ASM load, rather than ASM taper speed, is associated with severe seizures. ASM modeling has the potential to help optimize taper strategy to minimize severe seizures while maximizing diagnostic yield.


Subject(s)
Drug Resistant Epilepsy , Seizures , Humans , Seizures/drug therapy , Drug Resistant Epilepsy/drug therapy , Electrocorticography , Length of Stay , Logistic Models
4.
Eur J Neurol ; 29(1): 237-246, 2022 01.
Article in English | MEDLINE | ID: mdl-34402140

ABSTRACT

BACKGROUND: Magnetic resonance imaging (MRI) provides insight into various pathological processes in multiple sclerosis (MS) and may provide insight into patterns of damage among patients. OBJECTIVE: We sought to determine if MRI features have clinical discriminative power among a cohort of MS patients. METHODS: Ninety-six relapsing remitting and seven progressive MS patients underwent myelin water fraction (MWF) imaging and conventional MRI for cortical thickness and thalamic volume. Patients were clustered based on lesion level MRI features using an agglomerative hierarchical clustering algorithm based on principal component analysis (PCA). RESULTS: One hundred and three patients with 1689 MS lesions were analyzed. PCA on MRI features demonstrated that lesion MWF and volume distributions (characterized by 25th, 50th, and 75th percentiles) accounted for 87% of the total variability based on four principal components. The best hierarchical cluster confirmed two distinct patient clusters. The clustering features in order of importance were lesion median MWF, MWF 25th, MWF 75th, volume 75th percentiles, median individual lesion volume, total lesion volume, cortical thickness, and thalamic volume (all p values <0.01368). The clusters were associated with patient Expanded Disability Status Scale (EDSS) (n = 103, p = 0.0338) at baseline and at 5 years (n = 72, p = 0.0337). CONCLUSIONS: These results demonstrate that individual MRI features can identify two patient clusters driven by lesion-based values, and our unique approach is an analysis blinded to clinical variables. The two distinct clusters exhibit MWF differences, most likely representing individual remyelination capabilities among different patient groups. These findings support the concept of patient-specific pathophysiological processes and may guide future therapeutic approaches.


Subject(s)
Multiple Sclerosis, Chronic Progressive , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Brain/pathology , Humans , Magnetic Resonance Imaging/methods , Multiple Sclerosis/complications , Multiple Sclerosis, Chronic Progressive/complications , Multiple Sclerosis, Relapsing-Remitting/complications , Myelin Sheath/pathology
5.
Pharmacoepidemiol Drug Saf ; 31(1): 46-54, 2022 01.
Article in English | MEDLINE | ID: mdl-34227170

ABSTRACT

BACKGROUND: Comparative-effectiveness studies using real-world data (RWD) can be susceptible to surveillance bias. In solid tumor oncology studies, analyses of endpoints such as progression-free survival (PFS) are based on progression events detected by imaging assessments. This study aimed to evaluate the potential bias introduced by differential imaging assessment frequency when using electronic health record (EHR)-derived data to investigate the comparative effectiveness of cancer therapies. METHODS: Using a nationwide de-identified EHR-derived database, we first analyzed imaging assessment frequency patterns in patients diagnosed with advanced non-small cell lung cancer (aNSCLC). We used those RWD inputs to develop a discrete event simulation model of two treatments where disease progression was the outcome and PFS was the endpoint. Using this model, we induced bias with differential imaging assessment timing and quantified its effect on observed versus true treatment effectiveness. We assessed percent bias in the estimated hazard ratio (HR). RESULTS: The frequency of assessments differed by cancer treatment types. In simulated comparative-effectiveness studies, PFS HRs estimated using real-world imaging assessment frequencies differed from the true HR by less than 10% in all scenarios (range: 0.4% to -9.6%). The greatest risk of biased effect estimates was found comparing treatments with widely different imaging frequencies, most exaggerated in disease settings where time to progression is very short. CONCLUSIONS: This study provided evidence that the frequency of imaging assessments to detect disease progression can differ by treatment type in real-world patients with cancer and may induce some bias in comparative-effectiveness studies in some situations.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Bias , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/epidemiology , Humans , Lung Neoplasms/diagnostic imaging , Progression-Free Survival
6.
Neuroimage ; 245: 118642, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34637901

ABSTRACT

Motor recovery following ischemic stroke is contingent on the ability of surviving brain networks to compensate for damaged tissue. In rodent models, sensory and motor cortical representations have been shown to remap onto intact tissue around the lesion site, but remapping to more distal sites (e.g. in the contralesional hemisphere) has also been observed. Resting state functional connectivity (FC) analysis has been employed to study compensatory network adaptations in humans, but mechanisms and time course of motor recovery are not well understood. Here, we examine longitudinal FC in 23 first-episode ischemic pontine stroke patients and utilize a graph matching approach to identify patterns of functional connectivity reorganization during recovery. We quantified functional reorganization between several intervals ranging from 1 week to 6 months following stroke, and demonstrated that the areas that undergo functional reorganization most frequently are in cerebellar/subcortical networks. Brain regions with more structural and functional connectome disruption due to the stroke also had more remapping over time. Finally, we show that functional reorganization is correlated with the extent of motor recovery in the early to late subacute phases, and furthermore, individuals with greater baseline motor impairment demonstrate more extensive early subacute functional reorganization (from one to two weeks post-stroke) and this reorganization correlates with better motor recovery at 6 months. Taken together, these results suggest that our graph matching approach can quantify recovery-relevant, whole-brain functional connectivity network reorganization after stroke.


Subject(s)
Connectome/methods , Magnetic Resonance Imaging/methods , Motor Cortex/diagnostic imaging , Motor Cortex/physiopathology , Recovery of Function , Stroke/diagnostic imaging , Stroke/physiopathology , Adult , Aged , Case-Control Studies , Female , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Male , Middle Aged
7.
Neuroimage ; 225: 117451, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33069865

ABSTRACT

We introduce the first-ever statistical framework for estimating the age of Multiple Sclerosis (MS) lesions from magnetic resonance imaging (MRI). Estimating lesion age is an important step when studying the longitudinal behavior of MS lesions and can be used in applications such as studying the temporal dynamics of chronic active MS lesions. Our lesion age estimation models use first order radiomic features over a lesion derived from conventional T1 (T1w) and T2 weighted (T2w) and fluid attenuated inversion recovery (FLAIR), T1w with gadolinium contrast (T1w+c), and Quantitative Susceptibility Mapping (QSM) MRI sequences as well as demographic information. For this analysis, we have a total of 32 patients with 53 new lesions observed at 244 time points. A one or two step random forest model for lesion age is fit on a training set using a lesion volume cutoff of 15 mm3 or 50 mm3. We explore the performance of nine different modeling scenarios that included various combinations of the MRI sequences and demographic information and a one or two step random forest models, as well as simpler models that only uses the mean radiomic feature from each MRI sequence. The best performing model on a validation set is a model that uses a two-step random forest model on the radiomic features from all of the MRI sequences with demographic information using a lesion volume cutoff of 50 mm3. This model has a mean absolute error of 7.23 months (95% CI: [6.98, 13.43]) and a median absolute error of 5.98 months (95% CI: [5.26, 13.25]) in the validation set. For this model, the predicted age and actual age have a statistically significant association (p-value <0.001) in the validation set.


Subject(s)
Brain/diagnostic imaging , Machine Learning , Multiple Sclerosis/diagnostic imaging , Adult , Contrast Media , Female , Gadolinium , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Reproducibility of Results , Time Factors
8.
Hum Genet ; 139(8): 1077-1090, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32266521

ABSTRACT

Our previous genome-wide association study (GWAS) for sagittal nonsyndromic craniosynostosis (sNCS) provided important insights into the genetics of midline CS. In this study, we performed a GWAS for a second midline NCS, metopic NCS (mNCS), using 215 non-Hispanic white case-parent triads. We identified six variants with genome-wide significance (P ≤ 5 × 10-8): rs781716 (P = 4.71 × 10-9; odds ratio [OR] = 2.44) intronic to SPRY3; rs6127972 (P = 4.41 × 10-8; OR = 2.17) intronic to BMP7; rs62590971 (P = 6.22 × 10-9; OR = 0.34), located ~ 155 kb upstream from TGIF2LX; and rs2522623, rs2573826, and rs2754857, all intronic to PCDH11X (P = 1.76 × 10-8, OR = 0.45; P = 3.31 × 10-8, OR = 0.45; P = 1.09 × 10-8, OR = 0.44, respectively). We performed a replication study of these variants using an independent non-Hispanic white sample of 194 unrelated mNCS cases and 333 unaffected controls; only the association for rs6127972 (P = 0.004, OR = 1.45; meta-analysis P = 1.27 × 10-8, OR = 1.74) was replicated. Our meta-analysis examining single nucleotide polymorphisms common to both our mNCS and sNCS studies showed the strongest association for rs6127972 (P = 1.16 × 10-6). Our imputation analysis identified a linkage disequilibrium block encompassing rs6127972, which contained an enhancer overlapping a CTCF transcription factor binding site (chr20:55,798,821-55,798,917) that was significantly hypomethylated in mesenchymal stem cells derived from fused metopic compared to open sutures from the same probands. This study provides additional insights into genetic factors in midline CS.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Craniosynostoses/genetics , Genetic Variation , Polymorphism, Single Nucleotide/genetics , Alleles , DNA Methylation , Genes, Reporter , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Introns/genetics , Linkage Disequilibrium , Promoter Regions, Genetic/genetics , Risk Factors
9.
Int J Hyperthermia ; 37(3): 34-49, 2020 12.
Article in English | MEDLINE | ID: mdl-33426992

ABSTRACT

Immune checkpoint blockade (ICB) comprising monoclonal antibodies (mAbs) against immune 'checkpoints', such as CTLA-4 and the PD1/PDL1 axis have dramatically improved clinical outcomes for patients with cancer. However, ICB by itself has failed to provide benefit in a wide range of solid tumors, where recurrence still occurs with high incidence. These poor response rates may be due to the therapeutic shortcomings of ICB; namely, a lack of cancer-specific cytotoxicity and ability to debulk tumors. To overcome these limitations, effective ICB therapy may require the combination with other complementary therapeutic platforms. Here, we propose that photothermal therapy (PTT) is an ideal therapeutic modality for combination with ICB because it can generate both tumor-specific cytotoxicity and immunogenicity. PTT elicits these specific effects because it is a localized thermal ablation technique that utilizes light-responsive nanoparticles activated by a wavelength-matched laser. While ICB immunotherapy alone improves cancer immunogenicity but does not generate robust antitumor cytotoxicity, nanoparticle-based PTT elicits targeted and controlled cytotoxicity but sub-optimal long-term immunogenicity. Thus, the two platforms offer complementary and potentially synergistic antitumor effects, which will be detailed in this review. We highlight three classes of nanoparticles used as agents of PTT (i.e., metallic inorganic nanoparticles, carbon-based nanoparticles and organic dyes), and illustrate the potential for nanoparticle-based PTT to potentiate the effects of ICB in preclinical models. Through this discussion, we aim to present PTT combined with ICB as a potent synergistic combination treatment for diverse cancer types currently refractory to ICB as well as PTT monotherapies.


Subject(s)
Antineoplastic Agents, Immunological , Neoplasms , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms/drug therapy , Photothermal Therapy
10.
Br J Nurs ; 29(Sup17): S14-S17, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32976024

ABSTRACT

Most patients with overt hepatic encephalopathy are managed in an acute hospital setting. The mainstays of treatment are non-absorbable disaccharides, To prevent a recurrence, and thus further hospital admission, the focus is on identifying and avoiding precipitants, optimising nutrition and prescribing medication including rifaximin-α*†.


Subject(s)
Hepatic Encephalopathy , Secondary Prevention , Hepatic Encephalopathy/nursing , Humans , Rifaximin/therapeutic use , Secondary Prevention/methods
11.
Hum Mol Genet ; 26(11): 2118-2132, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28369379

ABSTRACT

Twist transcription factors, members of the basic helix-loop-helix family, play crucial roles in mesoderm development in all animals. Humans have two paralogous genes, TWIST1 and TWIST2, and mutations in each gene have been identified in specific craniofacial disorders. Here, we describe a new clinical entity, Sweeney-Cox syndrome, associated with distinct de novo amino acid substitutions (p.Glu117Val and p.Glu117Gly) at a highly conserved glutamic acid residue located in the basic DNA binding domain of TWIST1, in two subjects with frontonasal dysplasia and additional malformations. Although about one hundred different TWIST1 mutations have been reported in patients with the dominant haploinsufficiency Saethre-Chotzen syndrome (typically associated with craniosynostosis), substitutions uniquely affecting the Glu117 codon were not observed previously. Recently, subjects with Barber-Say and Ablepharon-Macrostomia syndromes were found to harbor heterozygous missense substitutions in the paralogous glutamic acid residue in TWIST2 (p.Glu75Ala, p.Glu75Gln and p.Glu75Lys). To study systematically the effects of these substitutions in individual cells of the developing mesoderm, we engineered all five disease-associated alleles into the equivalent Glu29 residue encoded by hlh-8, the single Twist homolog present in Caenorhabditis elegans. This allelic series revealed that different substitutions exhibit graded severity, in terms of both gene expression and cellular phenotype, which we incorporate into a model explaining the various human disease phenotypes. The genetic analysis favors a predominantly dominant-negative mechanism for the action of amino acid substitutions at this highly conserved glutamic acid residue and illustrates the value of systematic mutagenesis of C. elegans for focused investigation of human disease processes.


Subject(s)
Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Abnormalities, Multiple , Acrocephalosyndactylia , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Child , Child, Preschool , Disease Models, Animal , Eye Abnormalities , Haploinsufficiency , Helix-Loop-Helix Motifs , Humans , Macrostomia , Male , Mutation , Nuclear Proteins/genetics , Phenotype , Protein Domains/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Twist-Related Protein 1/genetics
12.
Small ; 14(20): e1800678, 2018 05.
Article in English | MEDLINE | ID: mdl-29665282

ABSTRACT

A thermal "window" of immunogenic cell death (ICD) elicited by nanoparticle-based photothermal therapy (PTT) in an animal model of neuroblastoma is described. In studies using Prussian blue nanoparticles to administer photothermal therapy (PBNP-PTT) to established localized tumors in the neuroblastoma model, it is observed that PBNP-PTT conforms to the "more is better" paradigm, wherein higher doses of PBNP-PTT generates higher cell/local heating and thereby more cell death, and consequently improved animal survival. However, in vitro analysis of the biochemical correlates of ICD (ATP, high-motility group box 1, and calreticulin) elicited by PBNP-PTT demonstrates that PBNP-PTT triggers a thermal window of ICD. ICD markers are highly expressed within an optimal temperature (thermal dose) window of PBNP-PTT (63.3-66.4 °C) as compared with higher (83.0-83.5 °C) and lower PBNP-PTT (50.7-52.7 °C) temperatures, which both yield lower expression. Subsequent vaccination studies in the neuroblastoma model confirm the in vitro findings, wherein PBNP-PTT administered within the optimal temperature window results in long-term survival (33.3% at 100 d) compared with PBNP-PTT administered within the higher (0%) and lower (20%) temperature ranges, and controls (0%). The findings demonstrate a tunable immune response to heat generated by PBNP-PTT, which should be critically engaged in the administration of PTT for maximizing its therapeutic benefits.


Subject(s)
Hyperthermia, Induced , Neuroblastoma/pathology , Phototherapy , Animals , Cell Death , Cell Line, Tumor , Female , Ferrocyanides/chemistry , Humans , Mice , Nanoparticles/chemistry , Vaccination
13.
J Med Genet ; 54(4): 260-268, 2017 04.
Article in English | MEDLINE | ID: mdl-27884935

ABSTRACT

BACKGROUND: Craniosynostosis, the premature fusion of one or more cranial sutures, occurs in ∼1 in 2250 births, either in isolation or as part of a syndrome. Mutations in at least 57 genes have been associated with craniosynostosis, but only a minority of these are included in routine laboratory genetic testing. METHODS: We used exome or whole genome sequencing to seek a genetic cause in a cohort of 40 subjects with craniosynostosis, selected by clinical or molecular geneticists as being high-priority cases, and in whom prior clinically driven genetic testing had been negative. RESULTS: We identified likely associated mutations in 15 patients (37.5%), involving 14 different genes. All genes were mutated in single families, except for IL11RA (two families). We classified the other positive diagnoses as follows: commonly mutated craniosynostosis genes with atypical presentation (EFNB1, TWIST1); other core craniosynostosis genes (CDC45, MSX2, ZIC1); genes for which mutations are only rarely associated with craniosynostosis (FBN1, HUWE1, KRAS, STAT3); and known disease genes for which a causal relationship with craniosynostosis is currently unknown (AHDC1, NTRK2). In two further families, likely novel disease genes are currently undergoing functional validation. In 5 of the 15 positive cases, the (previously unanticipated) molecular diagnosis had immediate, actionable consequences for either genetic or medical management (mutations in EFNB1, FBN1, KRAS, NTRK2, STAT3). CONCLUSIONS: This substantial genetic heterogeneity, and the multiple actionable mutations identified, emphasises the benefits of exome/whole genome sequencing to identify causal mutations in craniosynostosis cases for which routine clinical testing has yielded negative results.


Subject(s)
Craniosynostoses/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Neoplasm Proteins/genetics , Craniosynostoses/diagnosis , Craniosynostoses/pathology , Exome/genetics , Genetic Testing , Humans , Mutation , Predictive Value of Tests
14.
Stat Appl Genet Mol Biol ; 15(3): 213-35, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26992202

ABSTRACT

When testing for differentially expressed genes between more than two groups, the groups are often defined by dose levels in dose-response experiments or ordinal phenotypes, such as disease stages. We discuss the potential of a new approach that uses the levels' ordering without making any structural assumptions, such as monotonicity, by testing for zero variance components in a mixed models framework. Since the mixed effects model approach borrows strength across doses/levels, the test proposed can also be applied when the number of dose levels/phenotypes is large and/or the number of subjects per group is small. We illustrate the new test in simulation studies and on several publicly available datasets and compare it to alternative testing procedures. All tests considered are implemented in R and are publicly available. The new approach offers a very fast and powerful way to test for differentially expressed genes between ordered groups without making restrictive assumptions with respect to the true relationship between factor levels and response.


Subject(s)
Gene Expression Profiling/methods , Models, Genetic , Computer Simulation , Phenotype
15.
Nanomedicine ; 13(2): 771-781, 2017 02.
Article in English | MEDLINE | ID: mdl-27826115

ABSTRACT

We describe "photothermal immunotherapy," which combines Prussian blue nanoparticle (PBNP)-based photothermal therapy (PTT) with anti-CTLA-4 checkpoint inhibition for treating neuroblastoma, a common, hard-to-treat pediatric cancer. PBNPs exhibit pH-dependent stability, which makes them suitable for intratumorally-administered PTT. PBNP-based PTT is able to lower tumor burden and prime an immune response, specifically an increased infiltration of lymphocytes and T cells to the tumor area, which is complemented by the antitumor effects of anti-CTLA-4 immunotherapy, providing a more durable treatment against neuroblastoma in an animal model. We observe 55.5% survival in photothermal immunotherapy-treated mice at 100days compared to 12.5%, 0%, 0%, and 0% survival in mice receiving: anti-CTLA-4 alone, PBNPs alone, PTT alone, and no treatment, respectively. Additionally, long-term surviving, photothermal immunotherapy-treated mice exhibit protection against neuroblastoma rechallenge, suggesting the development of immunity against these tumors. Our findings suggest the potential of photothermal immunotherapy in improving treatments for neuroblastoma.


Subject(s)
Immunotherapy/methods , Nanoparticles , Neuroblastoma/therapy , Phototherapy , Animals , CTLA-4 Antigen/immunology , Coloring Agents/chemistry , Mice , Nanomedicine , T-Lymphocytes
16.
Neuroimage ; 132: 198-212, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26923370

ABSTRACT

Magnetic resonance imaging (MRI) intensities are acquired in arbitrary units, making scans non-comparable across sites and between subjects. Intensity normalization is a first step for the improvement of comparability of the images across subjects. However, we show that unwanted inter-scan variability associated with imaging site, scanner effect, and other technical artifacts is still present after standard intensity normalization in large multi-site neuroimaging studies. We propose RAVEL (Removal of Artificial Voxel Effect by Linear regression), a tool to remove residual technical variability after intensity normalization. As proposed by SVA and RUV [Leek and Storey, 2007, 2008, Gagnon-Bartsch and Speed, 2012], two batch effect correction tools largely used in genomics, we decompose the voxel intensities of images registered to a template into a biological component and an unwanted variation component. The unwanted variation component is estimated from a control region obtained from the cerebrospinal fluid (CSF), where intensities are known to be unassociated with disease status and other clinical covariates. We perform a singular value decomposition (SVD) of the control voxels to estimate factors of unwanted variation. We then estimate the unwanted factors using linear regression for every voxel of the brain and take the residuals as the RAVEL-corrected intensities. We assess the performance of RAVEL using T1-weighted (T1-w) images from more than 900 subjects with Alzheimer's disease (AD) and mild cognitive impairment (MCI), as well as healthy controls from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. We compare RAVEL to two intensity-normalization-only methods: histogram matching and White Stripe. We show that RAVEL performs best at improving the replicability of the brain regions that are empirically found to be most associated with AD, and that these regions are significantly more present in structures impacted by AD (hippocampus, amygdala, parahippocampal gyrus, enthorinal area, and fornix stria terminals). In addition, we show that the RAVEL-corrected intensities have the best performance in distinguishing between MCI subjects and healthy subjects using the mean hippocampal intensity (AUC=67%), a marked improvement compared to results from intensity normalization alone (AUC=63% and 59% for histogram matching and White Stripe, respectively). RAVEL is promising for many other imaging modalities.


Subject(s)
Brain/anatomy & histology , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Algorithms , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Artifacts , Brain/pathology , Female , Humans , Image Processing, Computer-Assisted , Linear Models , Male , ROC Curve , Reproducibility of Results , Signal Processing, Computer-Assisted
17.
Neuroimage ; 133: 176-188, 2016 06.
Article in English | MEDLINE | ID: mdl-26732403

ABSTRACT

Quantitative T1 maps estimate T1 relaxation times and can be used to assess diffuse tissue abnormalities within normal-appearing tissue. T1 maps are popular for studying the progression and treatment of multiple sclerosis (MS). However, their inclusion in standard imaging protocols remains limited due to the additional scanning time and expert calibration required and susceptibility to bias and noise. Here, we propose a new method of estimating T1 maps using four conventional MR images, which are intensity-normalized using cerebellar gray matter as a reference tissue and related to T1 using a smooth regression model. Using cross-validation, we generate statistical T1 maps for 61 subjects with MS. The statistical maps are less noisy than the acquired maps and show similar reproducibility. Tests of group differences in normal-appearing white matter across MS subtypes give similar results using both methods.


Subject(s)
Algorithms , Brain/diagnostic imaging , Diffusion Tensor Imaging/methods , Image Interpretation, Computer-Assisted/methods , Models, Statistical , Multiple Sclerosis/diagnostic imaging , White Matter/diagnostic imaging , Adult , Brain/pathology , Computer Simulation , Data Interpretation, Statistical , Female , Humans , Image Enhancement/methods , Male , Middle Aged , Multiple Sclerosis/pathology , Regression Analysis , Reproducibility of Results , Sensitivity and Specificity , White Matter/pathology
18.
Am J Hum Genet ; 92(1): 81-7, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23246290

ABSTRACT

Hypotrichosis simplex (HS) comprises a group of hereditary isolated alopecias that are characterized by a diffuse and progressive loss of hair starting in childhood and shows a wide phenotypic variability. We mapped an autosomal-dominant form of HS to chromosome 1q31.3-1q41 in a Spanish family. By direct sequencing, we identified the heterozygous mutation c.1A>G (p.Met1?) in SNRPE that results in loss of the start codon of the transcript. We identified the same mutation in a simplex HS case from the UK and an additional mutation (c.133G>A [p.Gly45Ser]) in a simplex HS case originating from Tunisia. SNRPE encodes a core protein of U snRNPs, the key factors of the pre-mRNA processing spliceosome. The missense mutation c.133G>A leads to a glycine to serine substitution and is predicted to disrupt the structure of SNRPE. Western blot analyses of HEK293T cells expressing SNRPE c.1A>G revealed an N-terminally truncated protein, and therefore the mutation might result in use of an alternative in-frame downstream start codon. Subcellular localization of mutant SNRPE by immunofluorescence analyses as well as incorporation of mutant SNRPE proteins into U snRNPs was found to be normal, suggesting that the function of U snRNPs in splicing, rather than their biogenesis, is affected. In this report we link a core component of the spliceosome to hair loss, thus adding another specific factor in the complexity of hair growth. Furthermore, our findings extend the range of human phenotypes that are linked to the splicing machinery.


Subject(s)
Hypotrichosis/genetics , snRNP Core Proteins/genetics , Female , Genetic Linkage , Humans , Male , Mutation , Pedigree , Spliceosomes/genetics
19.
Mult Scler ; 22(12): 1578-1586, 2016 10.
Article in English | MEDLINE | ID: mdl-26769065

ABSTRACT

OBJECTIVE: To evaluate clinical fluid-attenuated inversion recovery (FLAIR)* 3T magnetic resonance imaging (MRI), which is sensitive to perivenular inflammatory demyelinating lesions, in diagnosing multiple sclerosis (MS). BACKGROUND: Central veins may be a distinguishing feature of MS lesions. FLAIR*, a combined contrast derived from clinical MRI scans, has not been studied as a clinical tool for diagnosing MS. METHODS: Two experienced MS neurologists evaluated 87 scan pairs (T2-FLAIR/FLAIR*), separately and side-by-side, from 68 MS cases, 8 healthy volunteers, and 11 individuals with other neurological diseases. Raters judged cases based on experience, published criteria, and a visual assessment of the "40% rule," whereby MS is favored if >40% of lesions demonstrate a central vein. Diagnostic accuracy was determined with area under the receiver operating characteristic curve (AUC), and inter-rater reliability was assessed with Cohen's kappa (κ). RESULTS: Diagnostic accuracy was high: rater 1, AUC 0.94 (95% confidence interval: 0.89, 0.97) for T2-FLAIR, 0.95 (0.92, 0.98) for FLAIR*; rater 2, 0.94 (0.90, 0.98) and 0.90 (0.85, 0.95). AUC improved when images were considered together: rater 1, 0.99 (0.98, 1.00); rater 2, 0.98 (0.96, 0.99). Inter-rater agreement was substantial for T2-FLAIR (κ = 0.68) and FLAIR* (κ = 0.74), despite low agreement on the 40% rule (κ = 0.47) ([Formula: see text] in all cases). CONCLUSIONS: Joint clinical evaluation of T2-FLAIR and FLAIR* images modestly improves diagnostic accuracy for MS and does not require counting lesions with central veins.


Subject(s)
Cerebral Veins/diagnostic imaging , Magnetic Resonance Imaging/methods , Multiple Sclerosis/diagnostic imaging , Adult , Female , Humans , Magnetic Resonance Imaging/standards , Male , Middle Aged
20.
Stroke ; 46(11): 3270-3, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26451031

ABSTRACT

BACKGROUND AND PURPOSE: The location of intracerebral hemorrhage (ICH) is currently described in a qualitative way; we provide a quantitative framework for estimating ICH engagement and its relevance to stroke outcomes. METHODS: We analyzed 111 patients with ICH from the Minimally Invasive Surgery Plus Recombinant-Tissue Plasminogen Activator for Intracerebral Evacuation (MISTIE) II clinical trial. We estimated ICH engagement at a population level using image registration of computed tomographic scans to a template and a previously labeled atlas. Predictive regions of National Institutes of Health Stroke Scale and Glasgow Coma Scale stroke severity scores, collected at enrollment, were estimated. RESULTS: The percent coverage of the ICH by these regions strongly outperformed the reader-labeled locations. The adjusted R(2) almost doubled from 0.129 (reader-labeled model) to 0.254 (quantitative location model) for National Institutes of Health Stroke Scale and more than tripled from 0.069 (reader-labeled model) to 0.214 (quantitative location model). A permutation test confirmed that the new predictive regions are more predictive than chance: P<0.001 for National Institutes of Health Stroke Scale and P<0.01 for Glasgow Coma Scale. CONCLUSIONS: Objective measures of ICH location and engagement using advanced computed tomographic imaging processing provide finer, objective, and more quantitative anatomic information than that provided by human readers. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00224770.


Subject(s)
Brain/diagnostic imaging , Cerebral Hemorrhage/diagnostic imaging , Stroke/diagnostic imaging , Aged , Cerebral Hemorrhage/complications , Cohort Studies , Female , Glasgow Coma Scale , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Stroke/etiology , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL