scholarly journals Localized- and advanced-stage follicular lymphomas differ in their gene expression profiles

Blood ◽  
2020 ◽  
Vol 135 (3) ◽  
pp. 181-190 ◽  
Author(s):  
Annette M. Staiger ◽  
Eva Hoster ◽  
Vindi Jurinovic ◽  
Stefan Winter ◽  
Ellen Leich ◽  
...  

Abstract The genetic background of follicular lymphomas (FLs) diagnosed in advanced clinical stages III/IV, and which are frequently characterized by t(14;18), has been substantially unraveled. Molecular features, as exemplified in the clinicogenetic risk model m7FLIPI, are important tools in risk stratification. In contrast, little information is available concerning localized-stage FL (clinical stages I/II), which accounts for ∼20% of newly diagnosed FL in which the detection rate of t(14;18) is only ∼50%. To investigate the genetic background of localized-stage FL, patient cohorts with advanced-stage FL or localized-stage FL, uniformly treated within phase 3 trials of the German Low-Grade Lymphoma Study Group, were comparatively analyzed. Targeted gene expression (GE) profiling of 184 genes using nCounter technology was performed in 110 localized-stage and 556 advanced-stage FL patients. By penalized Cox regression, a prognostic GE signature could not be identified in patients with advanced-stage FL, consistent with results from global tests and univariate regression. In contrast, it was possible to define robust GE signatures discriminating localized-stage and advanced-stage FL (area under the curve, 0.98) by penalized logistic regression. Of note, 3% of samples harboring an “advanced-stage signature” in the localized-stage cohort exhibited inferior failure-free survival (hazard ratio [HR], 7.1; P = .0003). Likewise, in the advanced-stage cohort, 7% of samples with a “localized-stage signature” had prolonged failure-free survival (HR, 2.3; P = .017) and overall survival (HR, 3.4; P = .072). These data support the concept of a biological difference between localized-stage and advanced-stage FL that might contribute to the superior outcome of localized FL.

2021 ◽  
Vol 12 ◽  
Author(s):  
Chao Sun ◽  
Xin Zheng ◽  
Yingxin Sun ◽  
Ju Yu ◽  
Minfeng Sheng ◽  
...  

N6-methyladenosine (m6A) RNA modification can alter gene expression and function by regulating RNA splicing, stability, translocation, and translation. It is involved in various types of cancer. However, its role in gliomas is not well known. This study aimed to determine the prognostic value of the m6A RNA methylation regulator in gliomas and investigate the underlying mechanisms of the aberrant expression of m6A-related genes.mRNA expression profiles and clinical information of 448 glioma samples were obtained from The Cancer Genome Atlas and cBioportal. The expression of m6A-related genes in normal controls and low-grade glioma and glioblastoma was obtained from Gene Expression Profiling Interactive Analysis. Further, m6A-related gene expression and its relationship with prognosis were obtained through The Chinese Glioma Genome Atlas (CGGA). Multivariate Cox regression analyses were performed, and a nomogram was built with potential risk factors based on a multivariate Cox analysis to predict survival probability. Online tools such as Gene Set Enrichment Analysis, STRING, Cytoscape, and Molecular Complex Detection were applied for bioinformatics analysis and to investigate the underlying mechanisms of the aberrant expression of m6A-related genes. The multivariate Cox regression analysis found that higher expression levels of YTHDC2 and insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3, also called IMP3) were independent negative and positive prognostic factors for overall survival (OS), respectively. Data from the CGGA database showed that IGF2BP3 expression increased when the tumor grade increased. Receiver operating characteristic (ROC) curve was used to evaluate the predictive specificity and sensitivity. The area under the ROC curve indicated that the OS prediction was 0.92 (1-year) and 0.917 (3-years), indicating that m6A-related genes could predict patient survival. In addition, IGF2BP3 was closely related to the shorter survival period of patients. Copy number variation and DNA methylation, but not somatic mutations, might contribute to the abnormal upregulation of IGF2BP3 in gliomas. Significantly altered genes were identified, and the protein–protein interaction network was constructed. Based on the data presented, our study identified several m6A-related genes, especially IGF2BP3, that could be potential prognostic biomarkers of gliomas. The study unveiled the potential regulatory mechanism of IGF2BP3 in gliomas.


2020 ◽  
Author(s):  
Liang Zhao ◽  
Jiayue Zhang ◽  
Zhiyuan Liu ◽  
Yu Wang ◽  
Shurui Xuan ◽  
...  

DNA methylation has been widely reported to associate with the progression of glioma. DNA methylation at the 5 position of cytosine (5-methylcytosine, 5mC), which is regulated by 5mC regulators ("writers", "erasers" and "readers"), is the most critical modification pattern. However, a systematic study on the role of these regulators in glioma is still lacking. In this study, we collected gene expression profiles and corresponding clinical information of gliomas from three independent public datasets. Gene expression of 21 5mC regulators was analyzed and linked to clinicopathological features. A novel molecular classification of glioma was developed using consensus non-negative matrix factorization (CNMF) algorithm, and the tight association with molecular characteristics as well as tumor immune microenvironment was clarified. Sixteen prognostic factors were identified using univariate Cox regression analysis, and a 5mC regulator-based gene signature was further constructed via the least absolute shrinkage and selection operator (LASSO) cox analysis. This risk model was proved as an efficient predictor of overall survival for diffuse glioma, glioblastoma (GBM), and low-grade glioma (LGG) patients in three glioma cohorts. The significant correlation between risk score and intratumoral infiltrated immune cells, as well as immunosuppressive pathways, was found, which explained the difference in clinical outcomes between high and low-risk groups. Finally, a nomogram incorporating the gene signature and other clinicopathological risk factors was established, which might direct clinical decision making. In summary, our work highlights the potential clinical application value of 5mC regulators in prognostic stratification of glioma and their potentialities for developing novel treatment strategies.


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii294-iii295
Author(s):  
Jovana Pavisic ◽  
Chankrit Sethi ◽  
Chris Jones ◽  
Stergios Zacharoulis ◽  
Andrea Califano

Abstract Diffuse intrinsic pontine glioma (DIPG) remains a fatal disease with no effective drugs to date. Mutation-based precision oncology approaches are limited by lack of targetable mutations and genetic heterogeneity. We leveraged systems biology methodologies to discover common targetable disease drivers—master regulator proteins (MRs)—in DIPG to expand treatment options. Using the metaVIPER algorithm, we interrogated an integrated low grade glioma and GBM gene regulatory network with 31 DIPG-gene expression signatures to identify tumor-specific MRs by differential expression of their transcriptional targets. Unsupervised clustering identified MR signatures of upregulated activity in RRM2/TOP2A in 13 patients, CD3D in 5 patients, and MMP7, TACSTD2, RAC2 and SLC15A1/SLC34A2 in individual patients, all of which can be targeted. Notably, intratumoral administration of etoposide by convection enhanced delivery was effective in murine proneural gliomas in which TOP2 was identified as a MR while RRM2—targetable by drugs such as cladribine—has been shown to be a positive regulator of glioma progression whose knock-down inhibits tumor growth. We also prioritized drugs by their ability to reverse MR-activity signatures using a large drug-perturbation database. Patients clustered by predicted drug sensitivities with distinct groups of tumors predicted to respond to proteasome inhibitors, Thiotepa or Volasertib all of which have early evidence in treating gliomas. We will refine this analysis in a multi-institutional study of >100 patient gene expression profiles to define MR signatures driving known biological/molecular disease subtypes, use DIPG cell lines recapitulating common MR architectures to optimize therapy prioritization, and validate our findings in vivo.


2020 ◽  
Author(s):  
Alena Moudra ◽  
Veronika Niederlova ◽  
Jiri Novotny ◽  
Lucie Schmiedova ◽  
Jan Kubovciak ◽  
...  

AbstractAntigen-inexperienced memory-like T (AIMT) cells are functionally unique T cells representing one of the two largest subsets of murine CD8+ T cells. However, differences between laboratory inbred strains, insufficient data from germ-free mice, a complete lack of data from feral mice, and unclear relationship between AIMT cells formation during aging represent major barriers for better understanding of their biology. We performed a thorough characterization of AIMT cells from mice of different genetic background, age, and hygienic status by flow cytometry and multi-omics approaches including analyses of gene expression, TCR repertoire, and microbial colonization. Our data showed that AIMT cells are steadily present in mice independently of their genetic background and hygienic status. Despite differences in their gene expression profiles, young and aged AIMT cells originate from identical clones. We identified that CD122 discriminates two major subsets of AIMT cells in a strain-independent manner. Whereas thymic CD122LOW AIMT cells (innate memory) prevail only in young animals with high thymic IL-4 production, peripheral CD122HIGH AIMT cells (virtual memory) dominate in aged mice. Co-housing with feral mice changed the bacterial colonization of laboratory strains, but had only minimal effects on the CD8+ T-cell compartment including AIMT cells.


2019 ◽  
Vol 80 (04) ◽  
pp. 240-249
Author(s):  
Jiajia Wang ◽  
Jie Ma

Glioblastoma multiforme (GBM), an aggressive brain tumor, is characterized histologically by the presence of a necrotic center surrounded by so-called pseudopalisading cells. Pseudopalisading necrosis has long been used as a prognostic feature. However, the underlying molecular mechanism regulating the progression of GBMs remains unclear. We hypothesized that the gene expression profiles of individual cancers, specifically necrosis-related genes, would provide objective information that would allow for the creation of a prognostic index. Gene expression profiles of necrotic and nonnecrotic areas were obtained from the Ivy Glioblastoma Atlas Project (IVY GAP) database to explore the differentially expressed genes.A robust signature of seven genes was identified as a predictor for glioblastoma and low-grade glioma (GBM/LGG) in patients from The Cancer Genome Atlas (TCGA) cohort. This set of genes was able to stratify GBM/LGG and GBM patients into high-risk and low-risk groups in the training set as well as the validation set. The TCGA, Repository for Molecular Brain Neoplasia Data (Rembrandt), and GSE16011 databases were then used to validate the expression level of these seven genes in GBMs and LGGs. Finally, the differentially expressed genes (DEGs) in the high-risk and low-risk groups were subjected to gene ontology enrichment, Kyoto Encyclopedia of Genes and Genomes pathway, and gene set enrichment analyses, and they revealed that these DEGs were associated with immune and inflammatory responses. In conclusion, our study identified a novel seven-gene signature that may guide the prognostic prediction and development of therapeutic applications.


2020 ◽  
Vol 11 ◽  
Author(s):  
Xin Qiu ◽  
Qin-Han Hou ◽  
Qiu-Yue Shi ◽  
Hai-Xing Jiang ◽  
Shan-Yu Qin

BackgroundIntratumoral oxidative stress (OS) has been associated with the progression of various tumors. However, OS has not been considered a candidate therapeutic target for pancreatic cancer (PC) owing to the lack of validated biomarkers.MethodsWe compared gene expression profiles of PC samples and the transcriptome data of normal pancreas tissues from The Cancer Genome Atlas (TCGA) and Genome Tissue Expression (GTEx) databases to identify differentially expressed OS genes in PC. PC patients’ gene profile from the Gene Expression Omnibus (GEO) database was used as a validation cohort.ResultsA total of 148 differentially expressed OS-related genes in PC were used to construct a protein-protein interaction network. Univariate Cox regression analysis, least absolute shrinkage, selection operator analysis revealed seven hub prognosis-associated OS genes that served to construct a prognostic risk model. Based on integrated bioinformatics analyses, our prognostic model, whose diagnostic accuracy was validated in both cohorts, reliably predicted the overall survival of patients with PC and cancer progression. Further analysis revealed significant associations between seven hub gene expression levels and patient outcomes, which were validated at the protein level using the Human Protein Atlas database. A nomogram based on the expression of these seven hub genes exhibited prognostic value in PC.ConclusionOur study provides novel insights into PC pathogenesis and provides new genetic markers for prognosis prediction and clinical treatment personalization for PC patients.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 545-545
Author(s):  
Han-Yu Chuang ◽  
Laura Rassenti ◽  
Trey Ideker ◽  
Thomas J. Kipps

Abstract The clinical course of patients with chronic lymphocytic leukemia (CLL) is heterogeneous. Several prognostic factors have been identified that can stratify patients into groups that differ in their relative tedancy for disease progression and/or survival. Microarray studies have highlighted differences in mRNA levels found between such CLL subgroups. We hypothesize that gene expression profiling might define a repertoire of transcriptional activity contributing to or resulting from the dynamic evolution of CLL cells. To evaluate for this, we profiled approximately 200 CLL patients (of >95% CD19+CD5+ peripheral blood mononuclear cells in each sample) on mRNA expression microarrays using Affymetrix HG-U133 plus 2 GeneChips. We first sought to develop an expression-based prognosis that assigns patients to “aggressive” (high-risk) or “indolent” (low-risk) groups based on their gene expression correlated to the treatment-free survival from the date of sample collection. Each of the ~22,000 genes was scored by the Cox metric, which measures the correlation between its gene expression level and treatment-free survival in a designated training set. Unsupervised 2-means clustering technique was then used to separate the training samples into two risk groups based on the similarity of their mRNA expression of the top Cox-scored genes. Patients in an independent test set were assigned to the “aggressive” or “indolent” groups based on their expression similarity to that of the training samples. The two risk groups defined by the gene signature displayed significantly different behaviors with respect to treatment-free survival (Fig. 1); however, neither of the two commonly-used prognostic factors, IgVH gene mutational status or leukemia-cell expression of ZAP-70 protein, could segregate these subgroups to the same degree of statistical significance. To achieve better prediction performance based upon biological-defendable models, we further adopted the network-based classification scheme we previously developed for predicting metastasis potential of breast cancers. The network-based approach identified prognostic markers not as individual genes but as subnetworks extracted from molecular interaction databases. Gene expression profiles from CLL patients were mapped to a large human molecular interaction network, consisting of 49,419 interactions (including protein-protein and protein-DNA interactions) among 9,795 genes/proteins, compiled from high-throughput screenings and curation of previous measurements reported in the literature. A search over this network was performed to identify prognostic subnetworks that could be used to predict treatment-free survival. Specifically, each subnetwork was scored by a vector of activities across all patients, where the activity for a given patient is a function of the expression levels of its member genes. A subnetwork’s prognostic power was computed as the uni-variate Cox score between the activity vector and the patient’s treatment-free survival. The resulting ~200 prognostic subnetworks identify new putative cancer markers and provide an array of “small-scale” models charting the molecular mechanisms correlated with CLL progression, e.g. subnetworks detailing interactions between proteins participating in Wnt signaling, Notch signaling, or cell death. Moreover, our network-based classification achieves higher accuracy in predicting duration of treatment-free survival in newly diagnosed patients than identified uniparameter prognostic markers or standard gene-expression array analyses. Thus, our network-based approach integrating protein interactions with CLL expression profiles leads to increased classification accuracy and, simultaneously, provides a view of the biological processes underlying cancer progression. Figure 1. Expression-based prognosis of CLL progression. Example of treatment-freesurvival analysis is shown in one pair of training and test set. p-values are derived from log-rank tests on the survival curves. Figure 1. Expression-based prognosis of CLL progression. Example of treatment-freesurvival analysis is shown in one pair of training and test set. p-values are derived from log-rank tests on the survival curves.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2686-2686 ◽  
Author(s):  
Luca Baldini ◽  
Alessandro Pulsoni ◽  
Giuseppe Rossi ◽  
Umberto Vitolo ◽  
Enrica Morra ◽  
...  

Abstract Abstract 2686 Poster Board II-662 Introduction: Fludarabine in combination with cyclophosphamide (FC) plus rituximab (R) is an effective treatment for newly diagnosed as well as relapsed low-grade non-Hodgin's lymphoma (NHL). The role of maintenance treatment with R has been demonstrated in relapsed/resistant follicular NHL improving overall and progression-free survival. We investigated efficacy and safety of the chemo-immunotherapy FCR followed by rituximab maintenance treatment in patients with advanced untreated indolent B-cell non follicular lymphomas (INFL). Patients and methods: from July 2005 to May 2007, 47 pts whit untreated advanced stage INFL (23 lymphocytic, 20 limphoplasmacytic and 4 nodal marginal zone NHL) were enrolled by 10 IIL centres, in an open label, single arm, multicenter phase II study. Treatment plan was: 6 courses of FC (Fludarabine, 25 mg/m2 i.v. plus Cyclophosphamide, 250 mg/m2, days 2–4) every 28 days plus 8 doses of R (375 mg/m2 , day 1 every FC cycle and day 14 of cycles 4 and 5) followed by R maintenance (375 mg/m2 every two months for 4 doses). Prophylactic antibiotic treatment with cotrimoxazole (two tablets three times a week) and antifungal profilaxis with itraconazole was planned from the beginning of chemotherapy to three months later or until normalization of CD4 count. The primary endpoint of this study is the percentage of failure free patients after two years from the treatment start. Results: all the patients were evaluable for safety analysis and 46/47 pts were evaluable in terms of intention to treat analysis. Median age was 59 years (31–68) and M/F ratio was 28/18; stages II/III/IV were 2/2/44; B symptoms and splenomegaly were observed in 11 and 14 pts respectively; FLIPI scores were: 0–1 in 16 pts (34.8%), score 2 in 19 pts (41.3%) and score ≥ 3 in 11 pts (23.9%). Forty-one patients (87.2%) completed the planned therapeutic program; the remaining 6 patients stopped the treatment for SAE (4 pts) or for other reasons (2 pts) after 9 courses (1 pt), 8 (1 pt), 6 (2 pts), 3 (1 pt) and 1 (1 pt). Overall response at the end of treatment was 80.4% with 60.9% CR and 19.5% PR. One patient relapsed during maintenance phase. All the patients are still alive. A total of 279 courses of FC were given to 47 patients. All the patients presented at least one toxic/adverse events (AE); 11 pts developed 12 serious AEs, but only 6 were related to therapy. Seventeen pts had to interrupt (4 pts), delay or reduce therapy. Three hundred twenty related AEs were registered: grade 1–2: 228 events; grade 3–4: 92 events. Among these last the most frequent was neutropenia: 30 pts presented 83 episodes whose grade 3–4 related to the therapy were 58. During maintenance phase, 4 episodes of neutropenia occurred (2 of grade 3–4). Sixteen pts presented 31 infective episodes; the most frequent were: 5 Herpes zoster infections, 5 pneumonia (1 mycotic) and 4 urinary tract infections. Conclusions: in a series of INFL at diagnosis, FCR regimen is effective with a very high CR rate. The toxicity was acceptable and the schedule can be considered safe although the frequence of neutropenia and infective events require a close surveillance. The next year of follow-up will allow us to establish the failure free survival after two years from the treatment start. Disclosures: Vitolo: Roche: Lecture fees. Morra:Roche: Lecture fees.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 5064-5064
Author(s):  
L. Ozbun ◽  
T. Bonome ◽  
M. E. Johnson ◽  
M. Radonovich ◽  
C. Pise-Masison ◽  
...  

5064 Background: The purpose of this study was to identify a predictive gene signature for chemoresponse in patients with advanced stage papillary serous ovarian cancer. Methods: Expression profiling was performed on 50 chemonaive, microdissected advanced stage papillary serous ovarian cancers using Affymetrix Human Genome U133 Plus 2.0 microarrays. Chemoresistance was defined as disease progression while the patients remained on primary chemotherapy. Nine normal human ovarian surface epithelial (HOSE) brushings were also assessed to quantify normal gene expression levels. Validation was performed by quantitative real time PCR using the HOSE isolates and microdissected ovarian tumor samples. Results: A supervised learning algorithm applied to genes differentially expressed between chemosensitive/resistance tumors (p < 0.001) using leave-one-out cross-validation (LOOCV), identified over 2000 genes associated with tumor chemosensitivity. The chemoresponsive gene list was further refined to 576 genes by including only genes used for all LOOCV iterations. An independent gene list was generated comparing expression profiles of chemoresistant tumors to HOSE. The two lists were compared to identify common genes, generating final classifier list of 75 genes that included genes involved in apoptosis, RNA processing, protein ubiquitination, transcription regulation, and other novel genes. We hypothesized genes identified in both data sets would be predictive and biologically relevant. Of these 75 genes, 20 were validated by real-time PCR. Validated genes were ranked by a univariate t-stat value to further resolve the predictor. 4 multivariate predictor algorithms demonstrated the 10 top ranked validated genes maximixed prediction accuracy (compound covariate, 91%; diagonal linear discriminant analysis, 91%; 3-nearest neighbor, 86%; nearest centroid, 95%). The predictive value of these genes will be evaluated on an independent sample set. Conclusions: Gene expression profiling can distinguish between chemosensitive and chemoresistant ovarian cancers. This signature can predict response to therapy and has identified novel biologically and clinically relevant targets. No significant financial relationships to disclose.


2020 ◽  
Author(s):  
Xing Chen ◽  
Junjie Zheng ◽  
Min ling Zhuo ◽  
Ailong Zhang ◽  
Zhenhui You

Abstract Background: Breast cancer (BRCA) represents the most common malignancy among women worldwide that with high mortality. Radiotherapy is a prevalent therapeutic for BRCA that with heterogeneous effectiveness among patients. Methods: we proposed to develop a gene expression-based signature for BRCA radiotherapy sensitivity prediction. Gene expression profiles of BRCA samples from the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) were obtained and used as training and independent testing dataset, respectively. Differential expression genes (DEGs) in BRCA tumor samples compared with their paracancerous samples in the training set were identified by using edgeR Bioconductor package followed by dimensionality reduction through autoencoder method and univariate Cox regression analysis to screen genes among DEGs that with significant prognosis significance in patients that were previously treated with radiation. LASSO Cox regression method was applied to screen optimal genes for constructing radiotherapy sensitivity prediction signature. Results: 603 DEGs were obtained in BRCA tumor samples, and seven out of which were retained after univariate cox regression analysis. LASSO Cox regression analysis finally remained six genes based on which the radiotherapy sensitivity prediction model was constructed. The signature was proved to be robust in both training and independent testing sets and an independent marker for BRCA radiotherapy sensitivity prediction. Conclusions: this study should be helpful for BRCA patients’ therapeutics selection and clinical decision.


Sign in / Sign up

Export Citation Format

Share Document