scholarly journals A robust gene expression signature to predict proteasome inhibitor benefit in Multiple Myeloma

Author(s):  
Joske Ubels ◽  
Pieter Sonneveld ◽  
Martin H. van Vliet ◽  
Jeroen de Ridder

AbstractMany cancer drugs only benefit a subset of the patients that receive them, but are often associated with serious side effects. Predictive classification methods that can identify which patients will benefit from a specific treatment are therefore of great clinical utility. We here introduce a novel machine learning method to identify predictive gene expression signatures, based on the idea that patients who received different treatments but exhibit similar expression profiles can be used to model response to the alternative treatment. We use this method to predict proteasome inhibitor benefit in Multiple Myeloma (MM). In a dataset of 910 MM patients we identify a 14-gene expression signature that can successfully predict benefit to the proteasome inhibitor bortezomib, with a hazard ratio of 0.47 (p = 0.04) in class ‘benefit’, while in class ‘no benefit’ the hazard ratio is 0.91 (p = 0.68). Importantly, we observe a similar classification performance (HR class benefit = 0.46, p = 0.04) in an independent patient cohort which was moreover measured on a different platform, demonstrating the robustness of the signature. Moreover, we find that the genes in the discovered signature are essential, as no equivalent signature can be found when they are excluded from the analysis. Multiple genes in the signature are linked to working mechanisms of proteasome inhibitors or MM disease progression. In conclusion, our method allows for identification of gene expression signatures that can aid in treatment decisions for MM patients and provide insight into the biological mechanism behind treatment benefit.

2019 ◽  
Vol 17 (1) ◽  
Author(s):  
Yulong Zheng ◽  
Yongfeng Ding ◽  
Qifeng Wang ◽  
Yifeng Sun ◽  
Xiaodong Teng ◽  
...  

Abstract Background Brain metastases (BM) are the most common intracranial tumors. 2–14% of BM patients present with unknown primary site despite intensive evaluations. This study aims to evaluate the performance of a 90-gene expression signature in determining the primary sites for BM samples. Methods The sequence-based gene expression profiles of 708 primary brain tumors (PBT) collected from The Cancer Genome Atlas (TCGA) database were analyzed by the 90-gene expression signature, with a similarity score for each of 21 common tumor types. We then used Optimal Binning algorithm to generate a threshold for separating PBT from BM. Eighteen PBT samples were analyzed to substantiate the reliability of the threshold. In addition, the performance of the 90-gene expression signature for molecular classification of metastatic brain tumors was validated in a cohort of 48 BM samples with the known origin. For each BM sample, the tumor type with the highest similarity score was considered tissue of origin. When a sample was diagnosed as PBT, but the similarity score below the threshold, the second prediction was considered as the primary site. Results A threshold of the similarity score, 70, was identified to discriminate PBT from BM (PBT: > 70, BM: ≤ 70) with an accuracy of 99% (703/708, 95% CI 98–100%). The 90-gene expression signature was further validated with 18 PBT and 44 BM samples. The results of 18 PBT samples matched reference diagnosis with a concordance rate of 100%, and all similarity scores were above the threshold. Of 44 BM samples, the 90-gene expression signature accurately predicted primary sites in 89% (39/44, 95% CI 75–96%) of the cases. Conclusions Our findings demonstrated the potential that the 90-gene expression signature could serve as a powerful tool for accurately identifying the primary sites of metastatic brain tumors.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 719-719 ◽  
Author(s):  
Jacqueline E. Payton ◽  
Nicole R. Grieselhuber ◽  
Li-Wei Chang ◽  
Mark A. Murakami ◽  
Wenlin Yuan ◽  
...  

Abstract In order to better understand the pathogenesis of acute promyelocytic leukemia (APL, FAB M3), we sought to determine its gene expression signature by comparing the expression profiles of 14 APL samples to that of other AML subtypes (M0, M1, M2, M4, n=62) and to fractionated normal whole bone marrow cells (CD34 cells, promyelocytes, PMNs, n=5 each). We used ANOVA and SAM (Significance Analysis of Microarrays) to select genes that were highly expressed in APL cells and that displayed low to no expression in other AML subtypes. The APL signature was then further refined by filtering genes whose expression in APL was not significantly different from that of normal promyelocytes, yielding 1121 annotated genes that reliably distinguish APL from the other FAB subtypes using unsupervised hierarchical clustering, both in training and validation datasets. Fold change differences in expression between M3 and other AML FAB classes were striking, for example: GABRE 35.4, HGF 21.3, ANXA8 21.3, PTPRG 16.9, PTGDS 12.1, PPARG 11.1, STAB1 9.8. A large proportion of the APL versus other FAB dysregulome was recapitulated when we compared APL expression to that of the normal pattern of myeloid development. We identified 733 annotated genes with significantly different expression in APL versus normal myeloid cell fractions. These dysregulated genes were assigned to 4 classes: persistently expressed CD34 cell-specific genes, repressed promyelocyte-specific genes, prematurely expressed neutrophil-specific genes and genes with high expression in APL and low/no expression in normal myeloid cell fractions. Expression differences in several of the most dysregulated genes were validated by qRT-PCR. We then examined the expression of the APL signature genes in myeloid cell lines and tumors from a murine APL model. The bona fide M3 signature was not apparent in resting NB4 cells (which contain t(15;17), and which express PML-RARA), nor in PR-9 cells following Zn induction of PML-RARA expression, suggesting that neither cell line accurately models the gene expression signature of primary APL cells. Most of the nodal genes of the mCG-PML-RARA murine APL dysregulome (Yuan, et al, 2007) are similarly dysregulated in human M3 cells; however, the human and mouse dysregulomes do not completely coincide. Finally, we have begun investigating which APL signature genes are direct transcriptional targets of PML-RARA. The promoters of the APL signature genes were analyzed for the presence of known PML-RARA binding sites using multiple computational methods. The analyses demonstrated that several transcription factors (EBF3, TWIST1, SIX3, PPARG) have putative retinoic acid response elements (RAREs) in their upstream regulatory regions. Additionally, we examined the promoters of some of the most upregulated genes (HGF, PTGDS, STAB1) for known consensus sites of these transcription factors, and found that all have putative binding sites for at least one. These results suggest that PML-RARA may initiate a transcriptional cascade that relies not only on its own activity, but also on the actions of downstream transcription factors. In summary, our studies indicate that primary APL cells have a gene expression signature that is consistent and highly reproducible, but different from commonly used human APL cell lines and a mouse model of APL. The molecular mechanisms that govern this unique signature are currently under investigation.


2021 ◽  
Vol 17 (7) ◽  
pp. e1009228
Author(s):  
Kaiyi Zhu ◽  
Lingyi Cai ◽  
Chenqian Cui ◽  
Juan R. de los Toyos ◽  
Dimitris Anastassiou

During the last ten years, many research results have been referring to a particular type of cancer-associated fibroblasts associated with poor prognosis, invasiveness, metastasis and resistance to therapy in multiple cancer types, characterized by a gene expression signature with prominent presence of genes COL11A1, THBS2 and INHBA. Identifying the underlying biological mechanisms responsible for their creation may facilitate the discovery of targets for potential pan-cancer therapeutics. Using a novel computational approach for single-cell gene expression data analysis identifying the dominant cell populations in a sequence of samples from patients at various stages, we conclude that these fibroblasts are produced by a pan-cancer cellular transition originating from a particular type of adipose-derived stromal cells naturally present in the stromal vascular fraction of normal adipose tissue, having a characteristic gene expression signature. Focusing on a rich pancreatic cancer dataset, we provide a detailed description of the continuous modification of the gene expression profiles of cells as they transition from APOD-expressing adipose-derived stromal cells to COL11A1-expressing cancer-associated fibroblasts, identifying the key genes that participate in this transition. These results also provide an explanation to the well-known fact that the adipose microenvironment contributes to cancer progression.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5666-5666
Author(s):  
Angelique Bruyer ◽  
Alboukadel Kassambara ◽  
Paul Anziani ◽  
Donia El Bahlagui ◽  
Nicolas Robert ◽  
...  

Abstract Background: Inpatients with relaspsed/refractoryMultiple Myeloma (MM), outcomes are far from optimal, especially in patients refractory to current treatments Recent studies and clinical trials have highlighted the therapeutic potential of Palbociclib, a CDK4/6 inhibitor, in various cancers including MM. Deregulation of CDK4/6 is involved in the loss of cell cycle control in MM. Response to Palbociclib combined with bortezomib and dexamethasone was acquired in 20% of the relapsed/refractory MM patients, suggesting that biomarkers to identify patients that could benefit from this treatment are needed. Additional studies are required to understand the biological pathways associated with sensitivity or resistance of MM cells to Palbociclib. Methods: 14 human MM cell lines and 12 primary MM samples were tested for response to Palbociclib treatment. The concentration required to inhibit growth by 50% (IC50) was calculated. Gene expression signature associated with multiple myeloma response to Palbociclib, as well as, genes deregulated by the treatment have been analyzed using microarray and RNA-sequencing methods. Results: Palbociclib had an heterogeneous in vitro activity among the 14 human myeloma cell lines tested, which aggregated into three groups based on the distribution of the IC50 values: sensitive (n = 5, IC50: 0.2 - 0.3µM), intermediate (n = 3, IC50: 0.5 - 0.7µM) or more resistant group (n = 6, IC50: 0.9 - 2.4µM). The same holds true when testing the Palbociclib on primary multiple myeloma samples. The evaluation of the Palbociclib effect on cell cycle progression and the induction of the apoptosis, reveals that Palbociclib is essentially cytostatic, inducing prolonged G1 arrest in sensitive cell lines with a strong reduction of the percentage of cells in S phase. To better understand the molecular mechanisms associated with Palbociclib response, we identified a gene expression signature correlated with the response in both MM cell lines and primary myeloma cells from patients. Additionally, we have analyzed differentially expressed genes after Palbociclib treatment in human MM cell lines using RNA sequencing (n = 4). The physiological role of the downregulated genes after Palbociclib treatment is associated with cell cycle, mitosis and E2F mediated regulation of DNA replication. Significantly upregulated genes, after Palbociclib treatment, were enriched in genes encoding proteins involved in glutathione synthesis and recycling, and biological oxidations. Conclusion: Altogether, our data demonstrated a high heterogeneity in the response of MM cells to Palbociclib. We identified a gene expression signature associated with Palbociclib response in MM. These genes could help to identify MM patients that could benefit from Palbociclib treatment and provide novel targets for efficient combination therapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3719-3719
Author(s):  
Marta Sanchez-Martin ◽  
Alberto Ambesi-Impiombato ◽  
Luyao Xu ◽  
Yue Qin ◽  
Daniel Herranz ◽  
...  

Abstract Oncogenic NOTCH signaling is a major driver of T-cell transformation in T-cell acute lymphoblastic leukemia (T-ALL). However, clinical studies testing the efficacy of NOTCH1 inactivation with γ-secretase inhibitors (GSIs) have shown limited antileukemic activity for these drugs as single agents. Here we used an expression-based virtual screening approach and network perturbation analyses to identify and functionally characterize new highly active antileukemic drugs synergistic with NOTCH1 inhibition in T-ALL. Gene expression profiling studies have shown a prominent gene expression signature dominated by genes involved in growth and metabolism downstream of NOTCH1 in T-ALL. Notably, loss of the PTEN tumor suppressor gene confers resistance to GSI therapy and effectively rescues the gene expression signature induced by NOTCH1 inhibition in T-ALL. We hypothesized that drugs inducing transcriptional programs overlapping with those driven by NOTCH1 inhibition and antagonizing those resulting from PTEN loss could have synergistic antileukemic effects with GSIs in PTEN wild type and PTEN null leukemia cells. To address this question we generated gene expression signatures from Pten conditional-inducible knockout NOTCH1-driven leukemias in basal condition, upon NOTCH1 inhibition by GSI treatment and upon deletion of Pten. Connectivity Map (cMAP) analysis in this series identified 17 high scoring compounds as candidate antileukemic drugs (p<0.01). Reassuringly these included two inhibitors of the mTOR/PI3K/AKT pathway (rapamycin, wortmannin), but also histone deacetylase inhibitors (vorinostat, trichostatin A and valproic acid), phenothiazine antipsychotic drugs (trifluoperazine and thioridazine), antimalarial agents (astemizole, mefloquine) and compounds with less characterized activities such as withaferin A, parthenolide and pyrvinium pamoate. Transcriptional profiling followed by pairwise gene set enrichment analysis of these compounds identified groups of drugs with highly interconnected transcriptional programs suggestive of an overlapping mechanism of action (e.g. mTOR/PI3K inhibitors, HDAC inhibitors and phenothiazines), as well as compounds with more unique expression signatures suggestive of a more distinct mode of action (e.g. withaferin A, astemizole and mefloquine). Detailed characterization of the antileukemic effects of these 17 cMAP hits alone and in combination with the GSI DBZ in a broad panel of human NOTCH1-mutated T-ALL cell lines, identified withaferin A, rapamycin, wortmannin, parthenolide and vorinostat as the most active (lethal dose 50 <0.5 µM) and GSI-synergistic (combination index <0.4) drugs in this series. Among these, withaferin A, stood out as the most cytotoxic and GSI-synergistic compound against both PTEN positive and PTEN null T-ALL cell lines. Moreover, withaferin A treatment of primary mouse NOTCH1-induced T-ALLs and primary human T-ALL xenografts demonstrated strong and GSI-synergistic antileukemic activity in vivo. To address the mechanisms mediating the antileukemic effects of withaferin A we performed a detailed analysis of the gene expression signatures induced by this drug in T-ALL lymphoblasts. These studies revealed a strong enrichment of downregulated genes involved in translation regulation in T-ALL cells upon treatment with withaferin A (p<0.001). Mechanistically, transcriptional network perturbation analysis identified the eIF2A translation initiation complex as a potential effector of the antileukemic effects of withaferin A, and withaferin A treatment induced strong dose dependent phosphorylation of eIF2S1 in position S51, a modification responsible for blocking the activity of the eIF2A complex. Consistently, polysome profiling and nascent-protein assays revealed decreased translation in T-ALL cells treated with withaferin A. In this context, expression a phosphomimetic mutant form of eIF2S1 (S51D) impaired leukemia cell viability. Moreover, expression of a non-phosphorylatable form of eIF2S1 (eIF2S1 S51A) in T-ALL cells abrogated the antileukemic effects of withaferin A.These results support a direct role of eIF2S1 phosphorylation and the inhibition of eIF2A-dependent translation as a critical mediators of the antileukemic effects of withaferin A in T-ALL and a role for the combination of GSIs and inhibitors of protein translation for the treatment of high risk T-ALL. Disclosures Califano: Therasis Inc: Employment; Cancer Genetics Inc: Consultancy; Ipsen pharmaceuticals: Consultancy; Thermo Fischer Scientific: Consultancy.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 533-533
Author(s):  
S. Loi ◽  
B. Haibe-Kains ◽  
F. Lallemand ◽  
L. Pusztai ◽  
A. Bardelli ◽  
...  

533 Background: The phosphathidylinositol-3-kinase (PI3K) signaling pathway is frequently deregulated in tumor biology and is an attractive target for cancer therapy. Our aim was to characterize the molecular and clinical outcome effects of PIK3CA mutations in breast cancer (BC). Methods: We analyzed 173 BC samples for PIK3CA mutations. Corresponding gene expression profiles were used to understand its effects on the PI3K pathway. We validated a PIK3CA-GS in 2 independent BC cohorts (n = 183) with known PIK3CA mutation status and evaluated its correlation with clinical outcome in 1748 BC samples stratified by treatment and subtype. Results: 26% of BCs had a PIK3CA mutation. Tumors with PIK3CA mutation demonstrated a distinct gene expression signature (p = 0.03 after 1000 perm). In 2 datasets it could discriminate PIK3CA mutation carriers from wild-type (ROC 0.68, 0.71, p = 0.001for both). However, the PIK3CA-GS was correlated with deactivation of the PI3K pathway probably through a negative feedback loop. This observation was supported by: 1) the PIK3CA-GS was significantly correlated with gene expression changes induced by PI3K inhibitors (Connectivity Map, Gene set enrichment analyses) and 2) the PIK3CA-GS was anti-correlated with a GS of PTEN loss (R = -0.3; Saal et al, 2007). Higher levels of the PIK3CA signature were observed in HER-2+ and estrogen receptor positive (ER+), luminal BC subtypes. Whilst there was no association with mutation status alone and prognosis, increasing expression of the PIK3CA-GS (suggesting deactivation) was significantly associated with better clinical outcome in both untreated (p = 0.04) and particularly ER+, luminal-B, tamoxifen only-treated (p = 0.004) BC. Multivariate analysis (HR: 0.4; 95%CI: 0.3–0.7; p = 0.002) confirmed that the PI3KCA-GS provided independent prognostic information. Conclusions: Paradoxically, the PIK3CA-GS correlates with inhibition of the PI3K pathway in ER+ BC and identifies a subgroup of luminal B BCs with a favorable outcome. The PIK3CA-GS may be a better indicator of PI3K pathway dysfunction than mutation status, potentially indicating patients who may benefit from combined endocrine therapy and PI3K inhibition. No significant financial relationships to disclose.


2019 ◽  
Vol 25 ◽  
pp. 3247-3255 ◽  
Author(s):  
Fang-xiao Zhu ◽  
Yu-chan He ◽  
Jun-yan Zhang ◽  
Hang-fei Wang ◽  
Chen Zhong ◽  
...  

Leukemia ◽  
2012 ◽  
Vol 26 (11) ◽  
pp. 2406-2413 ◽  
Author(s):  
R Kuiper ◽  
A Broyl ◽  
Y de Knegt ◽  
M H van Vliet ◽  
E H van Beers ◽  
...  

Blood ◽  
2008 ◽  
Vol 111 (12) ◽  
pp. 5654-5662 ◽  
Author(s):  
Duane C. Hassane ◽  
Monica L. Guzman ◽  
Cheryl Corbett ◽  
Xiaojie Li ◽  
Ramzi Abboud ◽  
...  

Abstract Increasing evidence indicates that malignant stem cells are important for the pathogenesis of acute myelogenous leukemia (AML) and represent a reservoir of cells that drive the development of AML and relapse. Therefore, new treatment regimens are necessary to prevent relapse and improve therapeutic outcomes. Previous studies have shown that the sesquiterpene lactone, parthenolide (PTL), ablates bulk, progenitor, and stem AML cells while causing no appreciable toxicity to normal hematopoietic cells. Thus, PTL must evoke cellular responses capable of mediating AML selective cell death. Given recent advances in chemical genomics such as gene expression-based high-throughput screening (GE-HTS) and the Connectivity Map, we hypothesized that the gene expression signature resulting from treatment of primary AML with PTL could be used to search for similar signatures in publicly available gene expression profiles deposited into the Gene Expression Omnibus (GEO). We therefore devised a broad in silico screen of the GEO database using the PTL gene expression signature as a template and discovered 2 new agents, celastrol and 4-hydroxy-2-nonenal, that effectively eradicate AML at the bulk, progenitor, and stem cell level. These findings suggest the use of multicenter collections of high-throughput data to facilitate discovery of leukemia drugs and drug targets.


Sign in / Sign up

Export Citation Format

Share Document