scholarly journals Associations between Microrna Expression, Disease Progression and Ex Vivo Drug Response in Multiple Myeloma

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3069-3069
Author(s):  
Romika Kumari ◽  
Ashwini Kumar ◽  
Alun Parsons ◽  
Minna Suvela ◽  
Juha Lievonen ◽  
...  

Targeted drug treatment strategies have significantly prolonged the overall survival rate among multiple myeloma (MM) patients. However, high relapse rates and multiple drug resistance still pose major challenges. Although, the underlying molecular features of the disease have been explored both at the genomic and transcriptomic levels, the functional role of microRNAs (miRNA) in MM disease progression and prognosis is yet to be investigated at a personalized level. In earlier studies, microRNAs have been implicated to regulate gene expression and were determined to play crucial roles in the biology of MM by acting as oncogenes or tumor suppressors. Nevertheless, considering the heterogeneity of MM, little is known about the roles of miRNAs in controlling MM disease progression and drug response at an individualized systems level. We collected bone marrow aspirates from MM patients at diagnosis (n=20) and relapse (n=25) after informed consent and following approved protocols in accordance with the Declaration of Helsinki. CD138+ plasma cells were enriched from the bone marrow samples and used for miRNA-sequencing and drug sensitivity and resistance testing (DSRT). The miRNA was prepared from the CD138+ cells and subjected to sequencing using Illumina compatible technologies. DSRT was performed and responses to 83 clinically approved drugs and investigational compounds were measured as drug sensitivity scores (DSS) as described previously (Majumder et al., Oncotarget 2017). The pairwise comparative analysis of miRNA expression and drug responses was performed using Spearman's rank-order correlations, to elucidate significant associations of miRNA expression with drug sensitivity and resistance. Additionally, using DEseq2 the differential miRNA expression was determined for the newly diagnosed and relapse samples to deconvolute the role of miRNAs in MM disease progression. The comparative analysis of the miRNA expression and drug sensitivity scores revealed statistically significant associations between miRNA expression and drug sensitivity measures with the Spearman coefficient (r) ranging from -0.71 to 0.64 (adjusted p-value ≤ 0.05) (Figure 1A). Negative associations were more prevalent, with 40 miRNAs negatively associated with ≥1 drug response from the total of 30 predicted drugs. miR-486, which is known to be an effective biomarker in diagnosis and prognosis of multiple cancer types (Jiang et al., Oncotarget 2018), was found to have significant negative correlation (r= -0.71 to -0.52, p-value ≤ 0.01) with the responses of 14 drugs. Similarly, negative correlation was observed for miR-144 with 12 drugs and miR-584 with 9 drugs. We observed that PI3K/mTOR inhibitors and HDAC inhibitors were common amongst all the significant negative correlations predicted. Specifically, the PI3K/mTOR inhibitors apitosilib, omipalisib and buparlisib were found to be negatively associated with the expression of 18, 14 and 7 miRNAs respectively. These observations can lead to the understanding of miRNA mediated regulation of molecular pathways involved in drug resistance. Differential miRNA expression analysis between newly diagnosed and relapse MM samples revealed the involvement of miRNAs in disease progression. The analysis resulted in total of 31 significant differentially expressed miRNAs with fold change ≥2 and adjusted p-value ≤ 0.1 (Figure 1B). Several miRNAs known to play crucial roles in cancer diagnosis and prognosis were found to be significantly upregulated in the relapse samples. In particular, 25 miRNAs were upregulated, including following miR-17/92 cluster members: miR-18b, miR-20a, miR-92b and miR-106a, which are known to have an oncogenic role in various cancer types (Mogilyansky & Rigoutsos, Cell Death and Differentiation 2013). Interestingly, 12/31 differentially regulated miRNAs were located on chromosome X. Although cytogenetic alteration data predicted that chromosome 1q gain is significantly prominent in the relapse samples (p-value = 0.009), only 3/31 differentially regulated miRNAs were located on chromosome 1. These results demonstrate the role of miRNAs in regulating drug response and disease progression in multiple myeloma. Monitoring miRNA expression profiles in MM patients can facilitate the assessment of treatment outcome and prognosis, and miRNAs could potentially be useful prognostic and treatment biomarkers for MM. Disclosures Silvennoinen: Amgen: Research Funding; Bristol-Myers Squibb (BMS): Research Funding; Takeda: Research Funding; Celgene: Research Funding. Heckman:Celgene: Research Funding; Novartis: Research Funding; Oncopeptides: Research Funding; Orion Pharma: Research Funding.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3092-3092
Author(s):  
Rafail Nikolaos Tasakis ◽  
Alessandro Lagana ◽  
Violetta Leshchenko ◽  
David Melnekoff ◽  
Itai Beno ◽  
...  

RNA editing is an epitranscriptomic modification of emerging relevance to disease development and manifestations. Here we identify a novel role of the RNA editing enzyme ADAR1 in multiple myeloma (MM) progression as inducer of cognate DNA mutations. We have previously demonstrated (Lagana et al, ASH 2017) that ADAR1, which resides on human chromosome 1q21, is an RNA editor whose over-expression, either by IFN induction or through gene amplification, is associated with poor outcomes in MM. We now demonstrate robust and reproducible ADAR-mediated RNA editing in MM that increases with disease progression. At the same time, since disease progression is also correlated with the acquisition of new mutations, we asked whether ADAR1 could play the dual role of RNA editor and DNA mutator in MM, especially in the context of relapse. In fact, previous work has revealed that ADAR can exert its functions by acting on DNA/RNA hybrids in vitro (Zheng et al, Nucleic Acids Research 2017), and that DNA mutations at edited sites occur more often than at unedited sites in human and D melanogaster (Popitsch et al, BioRxiv 2017). We performed a careful bioinformatic dissection of matched pre-and post-relapse samples from 21 patients in the MMRF CoMMpass Study. Samples were profiled both with whole-exome sequencing (WES) to identify DNA mutations, and with RNAseq to identify editing instances. WES raw data was processed according to GATK Best Practices to generate alignment files, which were then processed with Samtools to identify mutations. RNAseq data was mapped using the tool GSNAP and processed using REDItools to identify editing events. Downstream analysis revealed a correlation between locations of RNA editing at diagnosis and of DNA mutation at relapse, with regions mutated matching known MM mutational hotspots in genes participating in several pathways that are relevant in MM, such as IFNa, IFNg response, IL2-STAT5 and TNF-NFkB. Finally, we demonstrated that editing at those locations is reproducible in a number of tumor cell lines, and that targeted editing of those locations could also result in the generation of mutations, similar to those we observed from patient data. Overall, we have shown that the RNA editor ADAR1, can also mutate the DNA cognate to the targeted transcript, generating specific mutational signatures at predetermined locations. We further hypothesize that this dual role of RNA editor and DNA mutator might be shared by other deaminases, and we suggest that in some contexts, DNA mutation might be the result of collateral damage on the genome by an editing enzyme whose primary job is to re-code the cognate transcript toward specific functional outcomes. Disclosures Madduri: undation Medicine: Consultancy; Celgene: Consultancy; Abbvie: Consultancy; Takeda: Consultancy. Richter:Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Speakers Bureau; Bristol-Meyers Squibb: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees. Chari:Seattle Genetics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Research Funding; Oncoceutics: Research Funding; Novartis Pharmaceuticals: Research Funding; GlaxoSmithKline: Research Funding; Array Biopharma: Research Funding; Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium/Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Consultancy; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Cho:Agenus: Research Funding; Genentech: Honoraria, Research Funding; BMS: Consultancy; GSK: Consultancy; Takeda: Research Funding; Celgene: Honoraria, Research Funding; The Multiple Myeloma Research Foundation: Employment. Jagannath:Celgene: Consultancy; Novartis: Consultancy; Merck: Consultancy; Medicom: Speakers Bureau; Multiple Myeloma Research Foundation: Speakers Bureau; BMS: Consultancy. Parekh:Foundation Medicine Inc.: Consultancy; Karyopharm Inc.: Research Funding; Celgene Corporation: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3006-3006
Author(s):  
Muntasir Mamun Majumder ◽  
Raija Silvennoinen ◽  
Pekka Anttila ◽  
David Tamborero ◽  
Samuli Eldfors ◽  
...  

Abstract Introduction Response to treatment for multiple myeloma (MM) patients is variable and often unpredictable, which may be attributed to the heterogeneous genomic landscape of the disease. However, the effect of recurrent molecular alterations on drug response is unclear. To address this, we systematically profiled 50 samples from 43 patients to assess ex vivo sensitivity to 308 anti-cancer drugs including standard of care and investigational drugs, with results correlated to genomic alterations. Our results reveal novel insights about patient stratification, therapies for high-risk (HR) patients, signaling pathway aberrations and ex-vivo-in-vivo correlation. Methods Bone marrow (BM) aspirates (n=50) were collected from MM patients (newly diagnosed n=17; relapsed/refractory n=33) and healthy individuals (n=8). CD138+ plasma cells were enriched by Ficoll separation followed by immunomagnetic bead selection. Cells were screened against 308 oncology drugs tested in a 10,000-fold concentration range. Drug sensitivity scores were calculated based on the normalized area under the dose response curve (Yadav et al, Sci Reports, 2014). MM selective responses were determined by comparing data from MM patients with those of healthy BM cells. Clustering of drug sensitivity profiles was performed using unsupervised hierarchical ward-linkage clustering with Spearman and Manhattan distance measures of drug and sample profiles. Somatic alterations were identified by exome sequencing of DNA from CD138+ cells and skin biopsies from each patient, while cytogenetics were determined by fluorescence in situ hybridization. Results Comparison of the ex vivo chemosensitive profiles of plasma cells resulted in stratification of patients into four distinct subgroups that were highly sensitive (Group I), sensitive (Group II), resistant (Group III) or highly resistant (Group IV) to the panel of drugs tested. Many of the drug responses were specific for CD138+ cells with little effect on CD138- cells from the same patient or healthy BM controls. We generated a drug activity profile for the individual drugs correlating sensitivity to recurrent alterations including mutations to KRAS, DIS3, NRAS, TP53, FAM46C, and cytogenetic alterations del(17p), t(4;14), t(14;16), t(11;14), t(14;20), +1q and -13. Cells from HR patients with del(17p) exhibited the most resistant profiles (enriched in Groups III and IV), but were sensitive to some drugs including HDAC and BCL2 inhibitors. Samples from patients with t(4;14) were primarily in Group II and very sensitive to IMiDs, proteasome inhibitors and several targeted drugs. Along with known recurrently mutated genes in myeloma, somatic mutations were identified in genes involved in several critical signaling pathways including DNA damage response, IGF1R-PI3K-AKT, MAPK, glucocorticoid receptor signaling and NF-κB signaling pathways. The predicted impact of these mutations on the activity of the pathways often corresponded to the drug response. For example, all samples bearing NF1 (DSS=21±7.9) and 67% with NRAS (DSS=15±4.35) mutations showed higher sensitivity to MEK inhibitors compared to healthy controls (DSS=5±.21). However, sensitivity was less predictable for KRAS mutants with modest response only in 47% samples (DSS=7±2.14) . One sample bearing the activating V600E mutation to BRAF showed no sensitivity to vemurafenib, which otherwise has good activity towards V600E mutated melanoma and hairy-cell leukemia. Comparison of the chemosensitive subgroups with survival showed patients in Groups I and IV had high relapse rate and poor overall survival. The ex vivo drug sensitivity results were used to decide treatment for three HR patients with results showing good ex vivo -in vivo correlation. Summary Our initial results suggest that ex vivo drug testing and molecular profiling of MM patients aids stratification. Grouping of patients based on their ex vivo chemosensitive profile proved extremely informative to predict clinical phenotype and identify responders from non-responders. While some molecular markers could be used to predict drug response, others were less predictive. Nevertheless, ex vivo drug testing identified active drugs, particularly for HR and relapsed/refractory patients, and is a powerful method to determine treatment for this group of patients. Disclosures Silvennoinen: Genzyme: Honoraria; Sanofi: Honoraria; Janssen: Research Funding; Celgene: Research Funding; Research Committee of the Kuopio University Hospital Catchment Area for State Research Funding, project 5101424, Kuopio, Finland: Research Funding; Amgen: Consultancy, Honoraria. Porkka:Bristol-Myers Squibb: Honoraria; Celgene: Honoraria; Novartis: Honoraria; Pfizer: Honoraria. Heckman:Celgene: Honoraria, Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 749-749 ◽  
Author(s):  
Tarek H Mouhieddine ◽  
Jihye Park ◽  
Robert A. Redd ◽  
Christopher J. Gibson ◽  
Salomon Manier ◽  
...  

Abstract Introduction: Multiple Myeloma (MM) is a clonal plasma cell malignancy, accounting for 10% of all hematological malignancies. Genetic analyses of large populations revealed that blood-specific somatic mutations in hematopoietic stem cells (HSCs) are commonly acquired during aging, a new entity labeled: clonal hematopoiesis of indeterminate potential (CHIP). We sought to determine the role of CHIP on survival of MM patients, specifically those receiving immunomodulator (IMiD) maintenance (Lenalidomide or Thalidomide) post autologous stem cell transplant (ASCT). Methods: We collected the cryopreserved, growth factor mobilized peripheral blood of 629 MM patients who underwent ASCT between 2003 and 2011 at the Dana-Farber Cancer Institute (DFCI). Then, we performed targeted next-generation sequencing using a 224-gene panel at a mean depth of coverage of 978X and ultra-low pass whole-genome sequencing at 0.1X to account for tumor contamination. We downloaded (dbGAP # phs000748.v6.p4) the whole-exome sequencing (WES) data of a cohort of 1144 newly diagnosed, untreated MM patients from the Multiple Myeloma Research Foundation (MMRF) Clinical Outcomes in MM to Personal Assessment of Genetic Profile (CoMMpass, NCT0145429) study (MMRC) and the WES data of a cohort of 205 newly diagnosed, untreated MM patients from the Broad Institute dataset. We analyzed their peripheral blood (average coverage of 108X) and tumor (average coverage of 107X) data separately, looking for the same CHIP genes included in our target bait panel. Results: The DFCI cohort had a median age of 58 years [range, 24-83] at time of ASCT and median follow up post ASCT of 8 years [range, 0.1-14.5]. 204 patients (32%) in the DFCI cohort had CHIP at time of ASCT. The most commonly detected mutated genes were DNMT3A, TET2, TP53, ASXL1 and PPM1D. 24 patients (3.8%) developed a second hematological malignancy at a median of 4 years [range, 1-10] post ASCT, half of whom had CHIP. Around 48% of the DFCI cohort received IMiDs as part of induction therapy. Different induction regimens had no effect on CHIP prevalence at time of ASCT. Around 56% of the DFCI cohort received IMiD maintenance, 22% of which received maintenance for at least 3 years [range, 0.06-12.8]. Among those who did not receive IMiD maintenance, patients with CHIP had worse progression free survival (PFS) (p-value < 0.001) and overall survival (OS) (p-value = 0.005). In patients receiving IMiD maintenance, having CHIP had no effect on PFS or OS. On the other hand, the MMRF cohort had a median age of 63 years [range, 27-93] and median follow up of 3.03 years [range, 0-5.9] from time of diagnosis. Around 52% of that cohort underwent ASCT and around 76% of those received IMiD maintenance with a median follow up of 2.7 years [range, 0-5.5] from time of ASCT. Furthermore, 200 patients of the MMRF cohort have follow-up samples of both tumor and peripheral blood that had targeted sequencing done by a 562-gene panel that included our genes of interest. Similarly, when studying the genomic results of 139 out of 1144 MMRF patients, as well as the 205 patients from the Broad Institute dataset, we detected CHIP in 25.6% of them and the top 5 most commonly mutated genes were similar to those of our cohort. Conclusion: CHIP is a common entity among MM patients, reaching a prevalence of up to 32%, that predicts a worse PFS and OS in those who do not receive IMiD maintenance therapy post ASCT. As expected, IMiD maintenance improves outcome in MM patients, with and without CHIP. In patients with CHIP, the use of IMiDs abrogated the deleterious effect imposed by CHIP to a point that outcome is identical to that of patients without CHIP. Figure Figure. Disclosures Bustoros: Dava Oncology: Honoraria. Munshi:OncoPep: Other: Board of director. Anderson:Gilead: Membership on an entity's Board of Directors or advisory committees; OncoPep: Equity Ownership, Other: Scientific founder; Celgene: Consultancy; C4 Therapeutics: Equity Ownership, Other: Scientific founder; Bristol Myers Squibb: Consultancy; Millennium Takeda: Consultancy. Richardson:Oncopeptides: Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding. Soiffer:Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Ghobrial:Celgene: Consultancy; Janssen: Consultancy; BMS: Consultancy; Takeda: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1369-1369
Author(s):  
Yanyan Gu ◽  
Jonathan L. Kaufman ◽  
Lawrence H. Boise ◽  
Sagar Lonial

Abstract Abstract 1369 The 14-3-3 protein family includes seven members, β, γ, ε, η, σ, τ and ζ. With over 200 binding partners, 14-3-3 proteins act as integrators of diverse cell signaling pathways and participate in metabolism, cell cycle regulation, survival and apoptosis. 14-3-3ζ has been implicated in many cancers such as hepatocellular carcinoma, gastric cancer, breast cancer, lung carcinoma and lymphoma. However, the role of 14-3-3ζ in MM has not been extensively explored. Preliminary data from an affymatrix GEP profile of normal plasma cells (NPC), MGUS, Smoldering myeloma (SM) or multiple myeloma (MM) demonstrates statistically increased expression of 14-3-3 ζ in the transition between MGUS and SM. Among patients with newly diagnosed symptomatic MM, 14-3-3 ζ expression appears to be higher in the higher risk genetic subsets. These data suggest 14-3-3ζ plays a prominent role in the biology of MM especially among high risk myeloma patients. In order to identify the impact of 14-3-3 ζ signaling on MM proliferation and survival, we developed 14-3-3ζ silenced and over expressing stable cell lines to interrogate the biological role of 14-3-3ζ in MM. Using a library of human MM cell lines, we found that 14-3-3ζ is universally expressed in all MM cell lines examined. Knockdown of 14-3-3ζ significantly inhibits cell growth and proliferation in LP1 and U266 cells, which is partly related to G1 cell cycle arrest. Relevant signaling proteins such as Mcl-1, Bcl2, phospho-Akt and CDK6 decrease after silencing 14-3-3ζ. Furthermore, we performed gene expression profiling of LP1 scrambled and knockdown stable cell lines in order to identify key changes in gene regulation that may be mediated via 14-3-3ζ. The GEP data suggests that 14-3-3ζ is responsible for but not limited to several important signaling pathways, such as glycolysis/gluconeogenesis, p53 Signaling, NRF2-mediated oxidative stress response and death receptor signaling. In addition, we evaluated the effect of 14-3-3ζ expression on the drug sensitivity to commonly used chemotherapeutic compounds in MM treatment, such as bortezomib, etoposide, dexamethasone, melphalan, lenalidomide, doxorubicin and romidepsin. Knockdown 14-3-3ζ sensitizes cells to romidepsin- induced apoptosis, as demonstrated by Annexin V staining and western blot assay for caspase cleavage. However, bortezomib- induced apoptosis is significantly inhibited when 14-3-3ζ is silenced. Bortezomib (5nM)-induced apoptosis decreased from 37% in LP1 cells expressing shRNA with scrambled sequence to 14% in LP1 cells where 14-3-3 ζ is silenced. Moreover, 14-3-3ζ knockdown effectively inhibits bortezomib induced NOXA upregulation, suggesting a possible new molecular mechanism for the effects of 14-3-3ζ in bortezomib mediated apoptosis. Taken together, our work reveals the important biological function of 14-3-3ζ in MM growth, survival and proliferation; the data also provides valuable information for the development of new therapeutic strategies facilitating drug sensitivity and overcoming drug resistance. Disclosures: Kaufman: Millenium: Consultancy; Onyx Pharmaceuticals: Consultancy; Novartis: Consultancy; Keryx: Consultancy; Merck: Research Funding; Celgene: Research Funding. Lonial:Onyx: Consultancy; Bristol-Myers Squibb: Consultancy; Novartis: Consultancy; Celgene: Consultancy; Millennium Pharmaceuticals, Inc.: Consultancy; Merck: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 804-804 ◽  
Author(s):  
Mark Bustoros ◽  
Chia-jen Liu ◽  
Kaitlen Reyes ◽  
Kalvis Hornburg ◽  
Kathleen Guimond ◽  
...  

Abstract Background. This study aimed to determine the progression-free survival and response rate using early therapeutic intervention in patients with high-risk smoldering multiple myeloma (SMM) using the combination of ixazomib, lenalidomide, and dexamethasone. Methods. Patients enrolled on study met eligibility for high-risk SMM based on the newly defined criteria proposed by Rajkumar et al., Blood 2014. The treatment plan was designed to be administered on an outpatient basis where patients receive 9 cycles of induction therapy of ixazomib (4mg) at days 1, 8, and 15, in combination with lenalidomide (25mg) at days 1-21 and Dexamethasone at days 1, 8, 15, and 22. This induction phase is followed by ixazomib (4mg) and lenalidomide (15mg) maintenance for another 15 cycles. A treatment cycle is defined as 28 consecutive days, and therapy is administered for a total of 24 cycles total. Bone marrow samples from all patients were obtained before starting therapy for baseline assessment, whole exome sequencing (WES), and RNA sequencing of plasma and bone marrow microenvironment cells. Moreover, blood samples were obtained at screening and before each cycle to isolate cell-free DNA (cfDNA) and circulating tumor cells (CTCs). Stem cell collection is planned for all eligible patients. Results. In total, 26 of the planned 56 patients were enrolled in this study from February 2017 to April 2018. The median age of the patients enrolled was 63 years (range, 41 to 73) with 12 males (46.2%). Interphase fluorescence in situ hybridization (iFISH) was successful in 18 patients. High-risk cytogenetics (defined as the presence of t(4;14), 17p deletion, and 1q gain) were found in 11 patients (61.1%). The median number of cycles completed was 8 cycles (3-15). The most common toxicities were fatigue (69.6%), followed by rash (56.5%), and neutropenia (56.5%). The most common grade 3 adverse events were hypophosphatemia (13%), leukopenia (13%), and neutropenia (8.7%). One patient had grade 4 neutropenia during treatment. Additionally, grade 4 hyperglycemia occurred in another patient. As of this abstract date, the overall response rate (partial response or better) in participants who had at least 3 cycles of treatment was 89% (23/26), with 5 Complete Responses (CR, 19.2%), 9 very good partial responses (VGPR, 34.6%), 9 partial responses (34.6%), and 3 Minimal Responses (MR, 11.5%). None of the patients have shown progression to overt MM to date. Correlative studies including WES of plasma cells and single-cell RNA sequencing of the bone microenvironment cells are ongoing to identify the genomic and transcriptomic predictors for the differential response to therapy as well as for disease evolution. Furthermore, we are analyzing the cfDNA and CTCs of the patients at different time points to investigate their use in monitoring minimal residual disease and disease progression. Conclusion. The combination of ixazomib, lenalidomide, and dexamethasone is an effective and well-tolerated intervention in high-risk smoldering myeloma. The high response rate, convenient schedule with minimal toxicity observed to date are promising in this patient population at high risk of progression to symptomatic disease. Further studies and longer follow up for disease progression are warranted. Disclosures Bustoros: Dava Oncology: Honoraria. Munshi:OncoPep: Other: Board of director. Anderson:C4 Therapeutics: Equity Ownership; Celgene: Consultancy; Bristol Myers Squibb: Consultancy; Takeda Millennium: Consultancy; Gilead: Membership on an entity's Board of Directors or advisory committees; Oncopep: Equity Ownership. Richardson:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Oncopeptides: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding. Ghobrial:Celgene: Consultancy; Takeda: Consultancy; Janssen: Consultancy; BMS: Consultancy.


Cancers ◽  
2019 ◽  
Vol 11 (10) ◽  
pp. 1452 ◽  
Author(s):  
Yu Liu ◽  
Haocheng Yu ◽  
Seungyeul Yoo ◽  
Eunjee Lee ◽  
Alessandro Laganà ◽  
...  

Multiple myeloma (MM) is the second most prevalent hematological cancer. MM is a complex and heterogeneous disease, and thus, it is essential to leverage omics data from large MM cohorts to understand the molecular mechanisms underlying MM tumorigenesis, progression, and drug responses, which may aid in the development of better treatments. In this study, we analyzed gene expression, copy number variation, and clinical data from the Multiple Myeloma Research Consortium (MMRC) dataset and constructed a multiple myeloma molecular causal network (M3CN). The M3CN was used to unify eight prognostic gene signatures in the literature that shared very few genes between them, resulting in a prognostic subnetwork of the M3CN, consisting of 178 genes that were enriched for genes involved in cell cycle (fold enrichment = 8.4, p value = 6.1 × 10−26). The M3CN was further used to characterize immunomodulators and proteasome inhibitors for MM, demonstrating the pleiotropic effects of these drugs, with drug-response signature genes enriched across multiple M3CN subnetworks. Network analyses indicated potential links between these drug-response subnetworks and the prognostic subnetwork. To elucidate the structure of these important MM subnetworks, we identified putative key regulators predicted to modulate the state of these subnetworks. Finally, to assess the predictive power of our network-based models, we stratified MM patients in an independent cohort, the MMRF-CoMMpass study, based on the prognostic subnetwork, and compared the performance of this subnetwork against other signatures in the literature. We show that the M3CN-derived prognostic subnetwork achieved the best separation between different risk groups in terms of log-rank test p-values and hazard ratios. In summary, this work demonstrates the power of a probabilistic causal network approach to understanding molecular mechanisms underlying the different MM signatures.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1623-1623 ◽  
Author(s):  
Karen Dybkær ◽  
Hanne Due ◽  
Rasmus Froberg Brøndum ◽  
Ken H. Young ◽  
Martin Bøgsted

Background: Patients with Diffuse large B-cell lymphoma (DLBCL) in approximately 40% of cases suffer from primary refractory disease and treatment induced immuno-chemotherapy resistance demonstrating that standard provided treatment regimens are not sufficient to cure all patients. Early detection of resistance is of great importance and defining microRNA (miRNA) involvement in resistance could be useful to guide treatment selection and help monitor treatment administration while sparing patients for inefficient, but still toxic therapy. Concept and Aims: With information on drug-response specific miRNAs, we hypothesized that multi-miRNA panels can improve robustness of individual clinical markers and serve as a prognostic classifier predicting disease progression in DLBCL patients. Methods: Fifteen DLBCL cell lines were tested for sensitivity towards rituximab (R), cyclophosphamide (C), doxorubicin (H), and vincristine (O). Cell line specific seeding concentrations was used to ensure exponential growth and each cell line was subjected to 16 concentrations in serial 2-fold dilutions and number of metabolic active cells was evaluated after 48 hours of drug exposure using MTS assay. For each drug, we ranked the cell lines according to their sensitivity and categorized them as sensitive, intermediate responsive, or resistant. Differential miRNA expression analysis between sensitive and resistant cell lines identified 43 miRNAs to be associated with response to compounds of the R-CHOP regimen, by selecting probes with a log fold change larger than 2. Baseline miRNA expression data were obtained for each cell line in untreated condition, and differential miRNA expression analysis identified 43 miRNAs associated to response to R-CHOP. Using the Affymetrix HG-U133+2 platform, expression levels of the miRNA precursors were assessed in 701 diagnostic DLBCL biopsies, and miRNA-panel classifiers were build using multiple Cox regression or random survival forest. Results: Generated prognostic miRNA-panel classifiers were tested for predictive accuracies and were subsequently evaluated by Brier scores and time varying area under the ROC curves (tAUC). Progression-free survival (PFS) was chosen as the outcome, since it is a treatment evaluation parameter as closely as possible to the time of drug exposure and the tested miRNAs were all associated directly to drug specific response. Furthermore, overall survival (OS) was used for verification of findings. Comparison of analyses conducted for the respective cohorts (All DLBCL, ABC, and GCB patients) showed the lowest prediction errors for all models within the GCB subclass with a multivariate Cox miRNA-panel model including miR-146a, miR-155, miR-21, miR-34a, and miR-23a~miR-27a~miR-24-2 cluster performed the best and successfully stratified GCB-DLBCL patients into high- and low-risk of disease progression. In addition, combination of the miRNA-panel and international prognostic index (IPI) substantially increased prognostic performance in GCB classified patients, indicating a prognostic signal from the response-specific miRNAs independent of IPI. In conclusion: We found as proof of concept that adding gene expression data detecting drug-response specific miRNAs to the clinically established IPI improved the prognostic stratification of GCB-DLBCL patients treated with R-CHOP. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1850-1850 ◽  
Author(s):  
Klaus Podar ◽  
Alexander Zimmerhackl ◽  
Ursula Hainz ◽  
Mariateresa Fulciniti ◽  
Sonia Vallet ◽  
...  

Abstract Abstract 1850 Poster Board I-876 Multiple Myeloma (MM) is characterized by the clonal proliferation of malignant plasma cells in the bone marrow. Despite current therapeutic approach and prolongation of the median survival, new therapies are urgently needed. Integrins are cell surface receptors which mediate both cell-cell adhesion and cell-extracellular matrix (ECM) protein adhesion. beta1-integrins, including very-late antigen-4 (VLA-4;á4β1), are typically expressed on MM cells. In MM, VLA-4-mediated binding to ECMS and bone marrow stromal cells (BMSCs) confers protection against drug-induced apoptosis and triggers transcription and secretion of IL-6, the major MM growth and survival factor. In addition to up-regulation of cell surface-clustering, integrin activity can also be triggered by multiple agonists through ‘inside-out’ signaling, independent of changes in integrin expression levels. Importantly, VEGF-induced migration of MM cells on fibronectin is also associated with β1-integrin- and PI3-kinase- dependent PKC activation. Targeting VLA-4 is therefore of potential high therapeutic interest in MM. Indeed, an antibody against murine á4 induces inhibition of MM growth in a murine model. Natalizumab is a recombinant humanized IgG4 monoclonal antibody, which belongs to a new class of molecules known as selective adhesion molecule (SAM) inhibitors and binds to á4-integrin. Clinically, Natalizumab has demonstrated activity in patients with multiple sclerosis and Crohn's disease. Here we tested the potential therapeutic role of Natalizumab on MM cell survival, and migration in the BM microenvironment. VLA-4 is expressed by all MM cell lines investigated (NCIH929, RPMI8226, INA-6, MM.1S, and OPM2). Functionally, Natalizumab but not a control antibody, triggered dose-dependent inhibition of MM cell adhesion to fibronectin, BMSCs, and endothelial cells (ECs). Importantly, inhibition of adhesion to fibronectin, BMSCs, or ECs was observed in MM cells pretreated with Natalizumab. Moreover, inhibition of MM cell adhesion to fibronectin, BMSCs, or ECs was also observed when Natalizumab was added to already adherent MM cells. Taken together, Natalizumab decreases adhesion of non-adherent MM cells as well as binding of already adherent MM cells to non-cellular and cellular components of the microenvironment. Given the protective role of the microenvironment on MM cell survival, we next sought to evaluate the chemosensitizing activity of Natalizumab. Specifically, we investigated dose- and time- dependent effects of Natalizumab, alone and when combined with conventional and novel therapies, on MM cells. Our results show that Natalizumab alone did not inhibit growth or survival of MM cells when cultured without components of the microenvironment. However, Natalizumab enhanced sensitivity of tumor cells to both bortezomib and dexamethasone in MM-BMSC and, MM-EC co-cultures. These data indicate a potential role of Natalizumab in bortezomib- and dexamethasone-containing treatment regimens including MPV. Moreover, Natalizumab decreases IL-6 and VEGF secretion triggered in MM-BMSC co-cultures. Consequently, angiogenesis triggered by supernatants of Natalizumab- treated MM-BMSC co-cultures was inhibited. Moreover, Natalizumab blocked MM cell migration on fibronectin triggered by both VEGF and IGF-1. Finally, our previous results implicate an PKC signaling in MM cell migration on fibronectin, and our current results show that Natalizumab inhibits phosphorylation of á4 integrins and PKC induced by co-stimulation with VEGF/ fibronectin, IGF-1/ fibronectin, and patient serum. Taken together, our data indicate a potential therapeutic role of Natalizumab in MM. Ongoing studies evaluating the effect of Natalizumab in a SCID-hu murine model of MM will also be reported. Disclosures: Podar: Biogen Idec: Research Funding. Off Label Use: natalizumab, integrin inhibitor. Zimmerhackl:Biogen Idec: Research Funding. Olsen:Biogen Idec: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4735-4735
Author(s):  
Catherine D Williams ◽  
Irina Proskorovsky ◽  
Philip Lewis ◽  
K. Jack Ishak ◽  
Krista A Payne ◽  
...  

Abstract Abstract 4735 Introduction: Symptoms of multiple myeloma (MM) and the adverse events (AEs) associated with MM treatment can be debilitating on many levels. A better understanding of the extent to which patients are affected and how this in turn impacts global health-related quality of life (HRQOL) can improve management of patients. Methods: A survey in 11 centers in the United Kingdom and Germany gathered, among other items, data on HRQOL, measured by the European Organization for Research and Treatment of Cancer's (EORTC) generic cancer and MM modules (QLQ-C30 and QLQ-MY20 scales), from a cross-section of patients with multiple myeloma at various phases of the disease. The QLQ-C30 is comprised of a global QOL domain, 5 functional and 3 symptom domains, and 6 AE items; the QLQ-MY20 includes scales for disease symptoms, treatment side-effects, future perspective and body image. This analysis aimed to explore the association between individual QOL scales (from QLQ-C30 and QLQ-MY20) and global QOL. Values for each scale range from 0 to 100; higher values indicate better HRQOL for the global, functional, future perspective and body image scales, and worse HRQOL for the AE items, symptom domains, disease symptoms and side-effects scales. Scoring of the QLQ-C30 and MY-20 scales was described previously by Fayers et al. [i] and Cocks et al. [ii] respectively. The distribution and correlations (Spearman) between the various scales was explored. Moreover, a multiple linear regression analysis was carried out to assess the association between individual scales and global QOL (from QLQ-C30) with the aim to identify those that independently impact global QOL. Each scale was first considered alone as a predictor of global QOL; those with a statistically significant association at a p-value ≤ 0.10 were included in a multiple regression model. This was then trimmed to exclude scales that became non-significant (p-value > 0.10). Results: The survey included 154 patients: 63.0% were male and the mean age was 66.4 (SD: 10.0). Mean time since diagnosis was 3.7 years (SD: 3.7), 51.9% were currently on treatment, and 42.9% had at least one prior line of MM therapy. The mean global QOL score was 60.1 (SD: 25.5), with the middle two quartiles of patients scoring between 41.7 and 83.3. Cognitive and emotional functioning scores had means near or above 80, suggesting that these aspects of HRQOL were less affected than role (62.9 (IQR: 33.3–100)), social (63.9(IQR: 33.3–100)) and physical functioning (68.7(IQR:53.3-93.3)). While body image scores were generally high (77.9 (IQR:66.7-100)), future perspective appeared to be relatively more affected (59.9 (33.3-77.8)). Patients’ HRQOL is most affected by pain and fatigue (based on symptom and AE scales of the QLQ-C30), with means above 30, followed by insomnia and dyspnoea with means above 20, while diarrhea and nausea/vomiting scales had the lowest mean scores (below 10). The Disease Symptom (23.3 (IQR:0-38.9)) and Side Effect scale scores (19.5 (IQR:7.4-29.6)) from the QLQ-MY20 were consistent with the AE and symptom scales from the QLQ-C30. All of the domains except diarrhea and nausea/vomiting individually showed at least moderate correlations with global QOL (Spearman correlations above 0.25 in absolute value), but also exhibited strong correlations between themselves. The final multiple regression model retained physical and social functioning, fatigue, disease symptoms (QLQ-MY20) and future perspective scales (QLQ-MY20), all of which had relatively similar strength of association with global QOL. Conclusion: This study demonstrates that the impact of MM and treatment AEs can be seen on various dimensions of patients’ HRQOL, particularly reduced physical and social functioning, future perspective and various disease symptoms such as bone pain (as captured by the disease symptoms scale of the QLQ-MY20) and fatigue. Fayers P, Aaronson N, Bjordal K, Groenvold M, Curran D, Bottomley A: The EORTC QLQ-C30 Scoring Manual. 3 Edition EORTC Quality of Life Group, Brussels 2001. [ii]Cocks K, Cohen D, Wisloff F, et al. An international field study of the reliability and validity of a disease-specific questionnaire module (the QLQ-MY20) in assessing the quality of life of patients with multiple myeloma. Eur J Cancer 2007;43:1670-1678. Disclosures: Williams: Celgene: Honoraria; Jansen Cilag: Consultancy, Honoraria. Off Label Use: Some of the patients in the study received Thalidomide for the treatment of relapsed or refractory multiple myeloma. Proskorovsky:United BioSource Corporation: Consultancy, Research Funding. Lewis:Celgene International SARL: Employment. Ishak:United BioSource Corporation: Consultancy, Research Funding. Payne:United BioSource Corporation: Consultancy, Research Funding. Lordan:United BioSource Corporation: Consultancy, Research Funding. Davies:Celgene: Honoraria, Speakers Bureau; Ortho Biotech: Honoraria, Speakers Bureau. Peters:Celgene: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 136-136
Author(s):  
Ze Tian ◽  
Jian-Jun Zhao ◽  
Jianhong Lin ◽  
Dharminder Chauhan ◽  
Kenneth C. Anderson

Abstract Abstract 136 Investigational Agent MLN9708 Target Tumor Suppressor MicroRNA-33b in Multiple Myeloma Cells Ze Tian, Jianjun Zhao, Jianhong Lin, Dharminder Chauhan, Kenneth C. Anderson Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115 MicroRNAs (miRNAs) are 19–25 nucleotide-long noncoding RNA molecules that regulate gene expression both at the level of messenger RNA degradation and translation. Emerging evidence shows that miRNAs play a critical role in tumor pathogenesis by functioning as either oncogene or tumor suppressor genes. The role of miRNA and their regulation in response to proteasome inhibitors treatment in Multiple Myeloma (MM) is unclear. Here, we utilized MLN9708, a selective orally bio-available proteasome inhibitor to examine its effects on miRNA alterations in MM.1S MM cells. Upon exposure to aqueous solutions or plasma, MLN9708 rapidly hydrolyzes to its biologically active form MLN2238. Our previous study using both in vitro and in vivo models showed that MLN2238 inhibits tumor growth and triggers apoptosis via activation of caspases. Moreover, MLN2238 triggered apoptosis in bortezomib-resistant MM cells, and induced synergistic anti-MM activity when combined with HDAC inhibitor SAHA, dexamethasone, and lenalidomide. In the current study, we treated MM.1S cells with MLN2238 (12 nM) for 3 hours and harvested; total RNA was subjected to miRNA profiling using TaqMan® Array Human miRNA A-Card Set v3.0 and the data was analyzed using dChip analysis. Results showed that MLN2238 modulates miRNA expression with a total of 36 miRNA changing their expression profiling (δδCT>1.5 or δδCT <-1.5; 19 were upregulated and 17 showed a downregulation). Among all miRNA, miR-33b was highly (δδCT>7) upregulated in response to MLN2238 treatment. We therefore hypothesized that miR-33b may play a role in MM pathogenesis as well as during MLN2238-induced proteasome inhibition in MM cells. We first utilized quantitative polymerase chain reaction (q-PCR) to validate the changes in miRNA expression profiling. Results confirmed that MLN2238 treatment triggers significant increase in the miR-33b expression in MM.1S cells (2.1 and 2.2 folds at 3h and 6h, respectively; P<0.001). Examination of normal PBMCs and plasma cells showed higher expression of miR-33b than patient MM cells (P<0.001). We further investigated the functional role of miR-33b in MM cells at baseline and during MLN2238 treatment. Drug sensitivity, cell viability, apoptosis, colony formation, and migration assays were performed using cell TilTer-Glo, Annexin V-FITC/PI staining, MTT staining, and Transwell assays, respectively. Signaling pathways modulated post miR-33b overexpression were evaluated by q-PCR, immunoblot, and reporter assays. Our findings show that overexpression of miR-33b significantly decreased cell viability, cell migration, colony formation, as well as increased apoptosis and sensitivity of MM cells to MLN2238 treatment. Targetscan analysis predicted pim-1 as a putative downstream target of miR-33b. Overexpression of miR-33b downregulated pim-1 mRNA and protein expression. To further corroborate these data, we co-tranfected miR-33b and Pim-1-wt or Pim-1-mt in 293T and MM.1S cell lines. In concert with our earlier findings, miR-33b decreases pim-1-wt, but not pim-1-mt reporter activity in both cell lines. Reflecting the overexpression study results, MLN2238 treatment also decreases pim-1-wt, but not pim1-mt reporter activity. Moreover, a biochemical inhibitor of pim1/2 triggered apoptosis in MM cells. Finally, overexpression of miR-33b inhibits tumor growth (P<0.001) and prolongs survival (P<0.001) in both subcutaneous and disseminated human MM xenograft models. In summary, our study suggests that miR-33b is a tumor suppressor, which plays a role during MLN2238-induced apoptotic signaling in MM cells, and provide the basis for novel therapeutic strategies targeting miR-33b in MM. Disclosures: Anderson: Millennium: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees; Acetylon: Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document