Gene Expression Profiling Reveals 5-Azacytidine to Be a Novel, Potentially Effective Therapy for Poor-Prognosis Patients with Multiple Myeloma.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1833-1833
Author(s):  
Sascha Tuchman ◽  
Chaitanya Acharya ◽  
William Mostertz ◽  
William Barry ◽  
Cristina Gasparetto ◽  
...  

Abstract Abstract 1833 Poster Board I-859 Introduction Individualization of therapy for multiple myeloma (MM), based on gene expression profiling, has not yet been achieved. Methods We previously described a “metagene” genomic model that synergizes with standard clinical staging to robustly prognosticate in MM (ASCO 2009 meeting, oral abstract # 8521; submitted for publication). In the current work we queried the Connectivity Map (Lamb J et al., Science 313(5795):1929-35, 2006); http://www.broadinstitute.org/cmap) using the metagene model, to elicit novel therapies that are likely to mitigate the poor-prognosis clinical phenotype we can identify with that model. We then employed gene expression profiling, the metagene model, and probit binary regression analysis to classify 12 untreated MM cell lines as having a similar or dissimilar molecular phenotype to poor-prognosis patients. Lastly, we performed MTT tetrazolium viability assays to survey, in triplicate, the sensitivity of those cell lines to the drug identified as well as melphalan, which we used as a conventional therapeutic control. Results We used Connectivity Map to analyze the metagene model, which we had previously discovered and validated in silico using three patient cohorts with prospectively collected clinical follow-up data (n=624 total patients). In doing so, we elicited 5-azacytidine as a likely agent to reverse the poor-prognosis phenotype. Using the metagene model, we then characterized 8 untreated MM cell lines as having a “good prognosis” molecular phenotype (i.e., a gene expression profile dissimilar to poor-prognosis patients according to the metagene model), and 4 cell lines as having a “poor prognosis” molecular phenotype (i.e., similar to poor-prognosis patients based on the metagene model). “Poor-prognosis” cell lines were susceptible to viability reduction by 5-azacytidine in a dose-independent manner and, importantly, at a physiologically achievable dose in humans (5 μM), whereas ‘good-prognosis‘ cell lines were comparatively less susceptible (p=0.03 by repeated measures 2-way ANOVA). Conversely, melphalan, which we used as a clinically relevant control that was not elicited by our analysis of the metagene model as likely to be effective in poor-prognosis patients, reduced the viability of “good-prognosis” cell lines more than that of “poor-prognosis” cell lines (fig. 1; p=0.02). Conclusions Computational analysis of the extensively validated and clinically relevant metagene prognostic model indicates, and in vitro assays confirm, that 5-azacytidine is more effective in ameliorating a poor-prognosis phenotype than melphalan in a multiple myeloma pre-clinical model. This provides proof-of-concept that gene expression profiling may 1) reveal novel and effective therapeutic approaches for identifiable high-risk subgroups of patients, and 2) enable clinicians to decide prospectively which conventional agents, such as melphalan, are unlikely to be effective in certain patients' myeloma. Clinical trials are needed to study such individualized approaches to the treatment of this molecularly heterogeneous disease. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 606-606
Author(s):  
Jonathan J Keats ◽  
Marta Chesi ◽  
Esteban Braggio ◽  
Stephan Palmer ◽  
Angela Baker ◽  
...  

Abstract Abstract 606 Multiple myeloma is a complex malignancy with multiple underlying genetic events. Our group has spent considerable effort over the last 15 years to elucidate the genetic underpinnings of myeloma. Most recently, we used array-based comparative genomic hybridization (aCGH) as a discovery tool in 62 myeloma patients and 46 myeloma cell lines. In that preliminary screen using the Agilent 44B aCGH platform (∼70kb resolution) we identified a diverse array of abnormalities, which resulted in constitutive activation of the NF-kB pathways. That initial analysis concentrated on the 43 genes we identified as potential targets of the 13 homozygous deletion events detected in the patient samples. A pathway analysis of these genes revealed a single pathway involving TRAF3, TRAF2, BIRC2, BIRC3, and CYLD. This first analysis focused exclusively on abnormalities present in the patient samples as we worried some abnormalities identified exclusively in the cell lines might not be relevant to the pathogenesis of myeloma in patients. However, several abnormalities were equally or more frequent overall but occurred exclusively in cell lines including CDKN2C (14 samples), CDKN1B (4 samples), KDM6A/UTX (4 samples), RB1 (3 samples), TP53 (3 samples). Given the fact that KDM6A/UTX deletions were as frequent as many of the best-described tumor suppressors it seemed like a good candidate but in the absence of patient events or a known function at the time it was not prioritized for further study. Recently, as part of the Multiple Myeloma Research Consortium (MMRC) Genomics Initiative, we have completed the analysis of a cohort of 250 myeloma patient samples by aCGH using the Agilent 244A aCGH platform (∼15kb resolution) and gene expression profiling using the Affymetrix U133Plus2.0 genechip. In this cohort with a significantly improved aCGH platform we identified 17 genes that are recurrently inactivated by homozygous deletions including DIAPH2 (15 samples), CDKN2C (14 samples), TRAF3 (11 samples), CYLD (8 samples), BIRC2/3 (7 samples), KDM6A/UTX (6 samples), and RB1 (5 samples). Based on the significant improvement in resolution and data quality achieved with the Agilent 244A aCGH platform we rescreen all of the cell lines on this improved platform. This significantly changed the frequency of several homozygous deletions in this population with the most frequently targeted genes now being CDKN2C (20 samples), KDM6A/UTX (13 samples), DIAPH2 (7 samples), RB1 (4 samples), TP53 (4 samples), CDKN1B (4 samples), and TRAF3 (4 samples). Moreover, as part of the genomic characterization of a spontaneous myeloma mouse model that we have developed, Vk*-Myc, we have identified recurrent (∼50%) homozygous deletions of Kdm6a/Utx. Therefore, one of the genes most commonly targeted by a homozygous deletion in human and mouse myeloma is KDM6A/UTX. In late 2007 after we had identified the first patients with KDM6A/UTX deletions it was shown that UTX is a functional histone demethylase that removes methyl groups from histone H3 lysine 27 (H3K27me). Given the high incidence of deletions and the fact that MMSET, the overexpressed target gene of t(4;14) in myeloma, is predicted to methylate H3K27, H3K36, and/or H4K20 by evolutionary conservation we developed the hypothesis that myeloma is characterized by abnormalities that result in excessive H3K27me (typically a repressive chromatin mark). Given the extensive whole genome sequencing occurring in the MMRC genomics initiative we elected to focus our resequencing efforts on the cell lines exclusively. These studies identified an additional 4 samples with LOH and an inactivating mutation bringing the total percentage of inactivated cell lines to 33%. Clearly, in the expanded patient and cell line cohorts the inactivation of KDM6A/UTX is not independent of MMSET overexpression suggesting they may act independently or synergistically. We are currently attempting to identify the genes controlled by KDM6A/UTX inactivation to better understand the functional consequences of this highly recurrent event. However, in the mouse model unlike the patient or cell lines, the gene expression profiling has identified a gene expression signature that differentiates UTX inactivated and functional samples suggesting an oncogenic function of inactivation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2865-2865
Author(s):  
Ashima Shukla ◽  
Nagendra K Chaturvedi ◽  
Shantaram S Joshi ◽  
Philip Bierman ◽  
Adam Cornish ◽  
...  

Abstract Chronic Lymphocytic Leukemia (CLL) represents the most common adult leukemia in western countries, affecting approximately 10,000 individuals every year in United States of America. In order to develop effective therapeutic strategies there is a need to understand the precise molecular events associated with CLL development and progression. In an attempt to understand the process we performed transcriptome analysis of CLL cells from peripheral blood (PB) of 7 good prognosis and 8 poor prognosis patients. Additionally to validate the results further, we have utilized gene expression profiling data of CLL cells from 77 good or poor prognosis patients. Using transcriptome and gene expression profiling, we identified PR Domain Zinc Finger Protein1 (PRDM1) as one of the candidate tumor suppressor genes in CLL. PRDM1/BLIMP1 is a transcriptional repressor which is crucial for terminal differentiation of mature B cells into plasma cells. PRDM1 has been shown to promote differentiation by repressing genes essential for B cell receptor signaling and cellular proliferation. Our results demonstrated that PRDM1 expression was significantly (p < 0.05) decreased in CLL cells from poor prognosis compared to good prognosis CLL cases (Figure 1). In addition, to determine the clinical significance, the expression levels of PRDM1 were found to correlate with time to treatment in CLL patients (Figure 2). Lower expression of PRDM1 was associated (p=0.001) with shorter time to treatment in CLL (n = 40). Furthermore, using IPA analyses we identified PRDM1 interacting partners. Among those, there was increased expression of BCL2, PAX5, EHMT2, SPIB and decreased expression of BCL6, IRF4, BCR, CASP3, pFOS, TP53 and HDAC2 in CLL cells from poor prognosis patients in comparison with good prognosis. Also we have observed the differential expression of TLE1, a co-repressor molecule associated with PRDM1 and member of the Groucho family of proteins.Figure1Transcript levels of PRDM1 in Good Prognosis and Poor Prognosis.Figure1. Transcript levels of PRDM1 in Good Prognosis and Poor Prognosis.Figure2Clinical correlation of PRDM1 expression with time to treatment.Figure2. Clinical correlation of PRDM1 expression with time to treatment. Together these results are suggestive of a role for PRDM1 as a negative regulator of CLL aggressiveness and progression. These studies warranted additional investigation to elucidate the mechanism of PRDM1 mediated CLL progression and to identify a therapeutic target. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2781-2781
Author(s):  
Abigail M. Lee ◽  
Andrew Clear ◽  
Maria Calaminici ◽  
Finlay Macdougall ◽  
Lindsey Goff ◽  
...  

Abstract Increasing evidence supports the importance of the number and location of tumor infiltrating lymphocytes (TILs) in cancer. Although increased circulating T regulatory cells have been found in CLL, no reports to date have examined the nature of the immune microenvironment in the lymph node (LN) in CLL/SLL. We therefore constructed TMAs composed of 1mm cores from initial diagnostic lymph nodes in 35 patients with CLL/SLL. The diagnosis of CLL/SLL was confirmed in all cases. The goals of the study were 1) to examine the nature of TILs in this disease, 2) to compare any changes in protein expression with those we have identified by gene expression profiling in peripheral blood T cells, 3) to examine the immune microenvironment in those cases with and without a leukemic component, ie comparing SLL with CLL, and 4) to assess the ability to discriminate prognostic groups based upon immunohistochemistry of diagnostic lymph nodes. The first group of 17 patients were selected on the basis of poor outcome, with median survival of 38 months from diagnosis, with all patients dying of their disease. The median age of this group at presentation was 62 years, 12 were male and 5 female. The second group of 18 patients were selected on the basis of long survival and had median survival of more than 10 years. Their median age at presentation was 53 years, 11 were male and 7 female. Immunohistochemistry was performed on the TMAs using a panel of monoclonal antibodies including CD4, CD8, CD25, CD6, CD7, FOXP3, CD68, TIA-1 and Granzyme B. The immune infiltrates were scored based on immunophenotype, frequency and location. All cores were analyzed independently by two histopathologists and concordance was achieved in all cases. The majority of cases in both groups had >30 CD8 expressing cells per high power field (hpf) as well as a high frequency of FOXP3 expressing cells, whereas we observed low number of cells expressing TIA-1 and granzyme B. In keeping with our previous gene expression profiling data in peripheral blood T cells, CD6 was expressed in a lower frequency of cells in the poor prognosis group. The poor prognosis group also had fewer CD4 but increased CD25 expressing T cells, as well as increased CD68 expressing monocytes. Ongoing studies are assessing the use of TMA in LN and bone marrow biopsies in CLL/SLL in a larger cohort of patients as well as analyzing additional antibodies, particularly for those antigens previously identified by gene expression profiling. The results are in keeping with the hypothesis that there is a complex interaction between the disease and the immune microenvironment in the LN in CLL/SLL and that the immune microenvironment represents a rational therapeutic target in this disease. Antigen expression Good Prognosis Pts Poor Prognosis Pts CD4 >5 cells/hpf 14/17 cases (82%) >5 cells/hpf 8/12 cases (67%) <5 cells/hpf 3/17 cases (18%) <5 cells/hpf 4/12 cases (33%) CD25 >5 cells/hpf 11/16 cases (69%) >5 cells/hpf 11/13 cases (85%) <5 cells/hpf 5/16 cases (31%) <5 cells/hpf 2/13 cases(15%) CD68 >5 cells/hpf 9/17 cases (53%) >5 cells/hpf 11/13 cases (85%) <5 cells/hpf 8/17 cases (47%) <5 cells/hpf 2/13 cases(15%) CD6 >5 cells/hpf 14/17 cases (82%) >5 cells/hpf 7/12 cases (58%) <5 cells/hpf 3/17 cases (18%) <5 cells/hpf 5/12 cases (42%)


Biomolecules ◽  
2021 ◽  
Vol 11 (4) ◽  
pp. 588
Author(s):  
Adam Ustaszewski ◽  
Magdalena Kostrzewska-Poczekaj ◽  
Joanna Janiszewska ◽  
Malgorzata Jarmuz-Szymczak ◽  
Malgorzata Wierzbicka ◽  
...  

Selection of optimal control samples is crucial in expression profiling tumor samples. To address this issue, we performed microarray expression profiling of control samples routinely used in head and neck squamous cell carcinoma studies: human bronchial and tracheal epithelial cells, squamous cells obtained by laser uvulopalatoplasty and tumor surgical margins. We compared the results using multidimensional scaling and hierarchical clustering versus tumor samples and laryngeal squamous cell carcinoma cell lines. A general observation from our study is that the analyzed cohorts separated according to two dominant factors: “malignancy”, which separated controls from malignant samples and “cell culture-microenvironment” which reflected the differences between cultured and non-cultured samples. In conclusion, we advocate the use of cultured epithelial cells as controls for gene expression profiling of cancer cell lines. In contrast, comparisons of gene expression profiles of cancer cell lines versus surgical margin controls should be treated with caution, whereas fresh frozen surgical margins seem to be appropriate for gene expression profiling of tumor samples.


Blood ◽  
2010 ◽  
Vol 116 (14) ◽  
pp. 2543-2553 ◽  
Author(s):  
Annemiek Broyl ◽  
Dirk Hose ◽  
Henk Lokhorst ◽  
Yvonne de Knegt ◽  
Justine Peeters ◽  
...  

Abstract To identify molecularly defined subgroups in multiple myeloma, gene expression profiling was performed on purified CD138+ plasma cells of 320 newly diagnosed myeloma patients included in the Dutch-Belgian/German HOVON-65/GMMG-HD4 trial. Hierarchical clustering identified 10 subgroups; 6 corresponded to clusters described in the University of Arkansas for Medical Science (UAMS) classification, CD-1 (n = 13, 4.1%), CD-2 (n = 34, 1.6%), MF (n = 32, 1.0%), MS (n = 33, 1.3%), proliferation-associated genes (n = 15, 4.7%), and hyperdiploid (n = 77, 24.1%). Moreover, the UAMS low percentage of bone disease cluster was identified as a subcluster of the MF cluster (n = 15, 4.7%). One subgroup (n = 39, 12.2%) showed a myeloid signature. Three novel subgroups were defined, including a subgroup of 37 patients (11.6%) characterized by high expression of genes involved in the nuclear factor kappa light-chain-enhancer of activated B cells pathway, which include TNFAIP3 and CD40. Another subgroup of 22 patients (6.9%) was characterized by distinct overexpression of cancer testis antigens without overexpression of proliferation genes. The third novel cluster of 9 patients (2.8%) showed up-regulation of protein tyrosine phosphatases PRL-3 and PTPRZ1 as well as SOCS3. To conclude, in addition to 7 clusters described in the UAMS classification, we identified 3 novel subsets of multiple myeloma that may represent unique diagnostic entities.


Sign in / Sign up

Export Citation Format

Share Document