Single Cell Network Profiling (SCNP) of Early-Stage Chronic Lymphocytic Leukemia (CLL): Association with p53, IGHV Mutational Status and Time to First Treatment (TTFT)

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1783-1783
Author(s):  
Jason Ptacek ◽  
Erik Evensen ◽  
Rachael E. Hawtin ◽  
Greg Friedland ◽  
Jodi R. Ware ◽  
...  

Abstract Abstract 1783 Background CLL follows a highly variable clinical course. Evidence suggests that BCR signaling is a driving event in disease onset and progression. SCNP is a multiparametric flow cytometry-based assay that measures changes in intracellular signaling proteins in response to modulators, providing a functional measure of pathway activity. In prior studies using samples collected from patients with Binet Stage A CLL but at different time points in the natural history of their disease, an association between elevated BCR signaling and shorter TTFT was observed (Cesano et al. ASH 2011 Abstract 2834). In order to assess the correlation of CLL biology (measured by SCNP) and clinical course in a clinically more homogeneous population, samples collected as part of a clinical trial from elderly patients with previously untreated CLL prior to therapy initiation were assessed. Objectives 1) To confirm the association between αIgM-induced (→) p-ERK signaling and TTFT in an independent CLL patient cohort, and 2) to identify additional associations between signaling and prognostic, molecularly defined clinical subgroups (i.e. IGHV and p53 mutational status). Methods: 29 evaluable cryopreserved CLL peripheral blood patient samples were collected between November 2008 and January 2010 as part of a Phase II trial. These studies were not part of the trial. Of the 29 samples, 20 were IGHV unmutated and 12 had unfavorable cytogenetics (del11q22.3 and/or del17p13) with 2 carrying p53 mutations. SCNP analysis was performed to quantitatively measure 18 intracellular signaling proteins within CD19+CD5+ CLL cells using a panel of 14 disease-relevant modulators. Cox Proportional Hazards regression was performed and Kaplan–Meier curves were used to assess signaling associations with TTFT in the 24 Binet A/B samples, as the prior TTFT association with BCR signaling was identified in Binet A samples. Wilcoxon signed-rank test and logistic regression were used to identify signaling associations with p53 and IGHV mutational status. Results: Consistent with prior studies, αIgM→p-ERK signaling was associated with TTFT (p=0.05, likelihood ratio (LR) test). Notably, the combination of SDF1α and αIgM modulation induced greater p-ERK signaling than observed with either agent alone and displayed a stronger association with TTFT (p=0.02) (Figure 1A). For this cohort, only the IGHV mutational status (p=0.01) (Figure 1B) and not cytogenetic risk categories, CD38, or ZAP-70 showed a significant association with TTFT. In addition, combining IGHV with αIgM→p-ERK did not improve predictive power. Significant associations to IGHV unmutated status (Figure 2A) included multiple nodes modulated by αIgM (p-ERK, p-PLCγ2, p-SYK). The strength of this relationship was greater using concurrent stimulation with αIgM+SDF1α. R848 (TLR7/8 agonist) and thapsigargin (Ca2+ influx) signaling were also increased in the unmutated samples. Finally, since the induction of p21 is in part regulated by p53 we tested the hypothesis that the lack of p21 induction by the DNA damaging agent, bendamustine, will be associated with p53 mutations. Samples with high spontaneous apoptosis in the absence of drug were removed prior to unblinding. Of the 13 evaluable samples, there was a significant association to p53 mutational status and p21 induction by bendamustine (p=0.0125, LR test, Figure 2B). Conclusions: These data confirm the association of BCR and BCR+SDF1α signaling with disease progression in CLL, and the potential for SCNP to identify patients more likely to require early treatment. These data support the potential utility of SCNP to: (1) identify in one assay those patients with a more aggressive form of CLL, including both unmutated IGHV and p53 pathway alterations, and (2) identify patients with signaling profiles who may be more likely to respond to targeted therapies. Disclosures: Ptacek: Nodality, Inc.: Employment, Equity Ownership. Evensen:Nodality, Inc.: Employment, Equity Ownership. Hawtin:Nodality, Inc.: Employment, Equity Ownership. Friedland:Nodality, Inc.: Employment, Equity Ownership. Ware:Nodality, Inc.: Employment, Equity Ownership. Cordeiro:Nodality, Inc.: Employment, Equity Ownership. Cesano:Nodality, Inc: Employment, Equity Ownership.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2876-2876
Author(s):  
Jason Ptacek ◽  
Erik Evensen ◽  
Greg Friedland ◽  
James Cordeiro ◽  
Jodi R. Ware ◽  
...  

Abstract Abstract 2876 Background: B-CLL follows a variable clinical course, with a subset of patients progressing quickly. The reasons for this outcome disparity are not fully understood; however, evidence suggests that B-cell receptor (BCR) signaling is a driving event in disease onset and progression. B-CLL cells also receive survival signals through additional receptors. SCNP is a multiparametric flow cytometry-based assay that measures, quantitatively at the single cell level, changes in intracellular signaling proteins in response to extracellular modulators. This provides a functional measure of pathway activity and intraclonal signaling differences within the larger B-CLL cell population. In prior studies, we observed higher αIgM-induced (→) p-ERK signaling in B-CLL samples from patients who had a shorter time to first treatment (TTFT) (Cesano et al. ASH 2011 Abstract 2834). Herein we examined the feasibility for SCNP to further define patient risk stratification. Objectives: The current study was designed to 1) map signaling profiles in early-stage B-CLL and to 2) identify signaling associations with clinical subgroups defining B-CLL prognosis (IGHV mutational status, cytogenetic risk, CD38 / ZAP-70 expression). Methods: Between 2006 and 2007, peripheral blood mononuclear cells (PBMCs) were collected and cryopreserved from a cohort of 39 untreated B-CLL patients, Rai stage 0 - II, at different time points during their clinical course. Of the 39 samples evaluated; 15 and 20 expressed CD38 (≥30% of cells) and ZAP-70 (≥20% of cells), respectively; 19 were IGHV unmutated (98% cutoff); and cytogenetic risk groups were evenly represented. SCNP analysis quantitatively measured 22 intracellular signaling proteins within CD19+CD5+ B-CLL cells, using a panel of 14 disease-relevant modulators. The Wilcoxon rank-sum test was used to identify signaling associations with CD38 and ZAP-70 expression, cytogenetic risk categories, del17p (includes p53 gene deletion) and IGHV mutational status. Results: Significant associations between patient risk categories and signaling are summarized in Table 1. IGHV unmutated and ZAP-70+ samples showed (1) elevated α-IgM or α-IgD-induced BCR signaling, either alone or in combination, (2) decreased CpGβ → p-ERK induction and (3) increased thapsigargin-induced (Ca2+ signaling) signaling. Of note, CD38+ samples did not show the same associations, but shared with unmutated IGHV samples a higher responsiveness to IFNα and weaker induction of p21 in response to bendamustine. The unfavorable cytogenetic group samples showed increased αIgM→p-ERK and had higher basal p-S6 that further increased with IgD crosslinking. Lack of p21 induction was also associated with unfavorable cytogenetics, which includes deletion of p53 (del17p), a regulator of p21 expression. Conclusions: This is the second, independent SCNP analysis of B-CLL signaling showing decreased bendamustine→p21 and increased αIgM→p-ERK signaling in samples with unfavorable cytogenetics. Further associations with IGHV unmutated status included increased BCR signaling in multiple nodes, altered TLR9 responsiveness, and decreased drug-induced p21. These data support the potential utility of SCNP in: (1) identifying in one assay those patients with a more aggressive form of B-CLL, including both unmutated IGHV and p53 pathway alteration, and (2) identification of patients with signaling profiles that may be more likely to respond to targeted therapies. Disclosures: Ptacek: Nodality, Inc.: Employment, Equity Ownership. Evensen:Nodality, Inc.: Employment, Equity Ownership. Friedland:Nodality, Inc.: Employment, Equity Ownership. Cordeiro:Nodality, Inc.: Employment, Equity Ownership. Ware:Nodality, Inc.: Employment, Equity Ownership. Cesano:Nodality, Inc: Employment, Equity Ownership. Hawtin:Nodality, Inc.: Employment, Equity Ownership.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2834-2834
Author(s):  
Alessandra Cesano ◽  
Erik Evensen ◽  
Jason Ptacek ◽  
James Cordeiro ◽  
Rachael E. Hawtin ◽  
...  

Abstract Abstract 2834 Background: B-cell chronic lymphocytic leukemia (B-CLL) follows a highly variable clinical course, with some patients having indolent disease and not requiring treatment, whereas others experience rapid disease progression, treatment resistance, and death within 2 to 3 years. Widely accepted staging systems, biological parameters, and prognostic indices help stratify patients into risk groups, yet there remains substantial intragroup heterogeneity. Single cell network profiling (SCNP) is a multiparametric flow cytometry-based assay that simultaneously measures, quantitatively at the single cell level, both extracellular surface marker levels and changes in intracellular signaling proteins in response to extracellular modulators (Kornblau et al. Clin Cancer Res 2010). Previously, we reported the use of this assay to functionally characterize BCR signaling in 21 cryopreserved samples from Binet Stage A B-CLL patients. In that study, a panel of signaling nodes (a node is defined as the combination of extracellular modulator and corresponding intracellular readout and is abbreviated node→readout) was examined and increased anti-IgM-induced phospho (p)-Erk signaling (anti-IgM→p-Erk) was associated with shorter time from diagnosis to first treatment (TTFT) (Scupoli et al. EHA 2010). Objectives: The present study was undertaken to independently verify the association between BCR responsiveness and shorter TTFT, to assess repeatability of SCNP measurements in B-CLL cells, and to explore additional signaling nodes relevant to B-CLL biology. Methods: SCNP was performed as previously described on cryopreserved peripheral blood mononuclear cells (PBMC) collected between 2004 and 2009 from 32 patients with untreated Binet Stage A B-CLL at various points during their clinical follow up; at the time of the analysis, 9 (28%) progressed to active disease, requiring treatment. Median follow-up was 47 months (range 4 to 179 months). SCNP analysis of 21 signaling nodes investigating B cell receptor, survival and NFkB signaling in B-CLL cells was performed in replicate, on separate plates run on the same day. Repeatability was assessed by regressing paired induced signaling measurements from experimental replicates. Associations between SCNP measurements and TTFT were determined using Cox Proportional Hazards regression. Results: Excellent repeatability was observed for SCNP signaling measurements in B-CLL cells (e.g., anti-IgM→p-Erk: R2=0.97, slope=1.01). Consistent with the previous study, an association between BCR responsiveness, measured by anti-IgM→p-Erk, and shorter TTFT was also observed in this study, although the pre-specified significance criteria were not met (likelihood ratio (LR) Chi square (χ2) test p=0.07). A post hoc analysis excluding two samples with low viability (low % aqua negative cells) was also performed (Figure 1a) showing a statistically significant association between anti-IgM→p-Erk signaling and TTFT (p=0.03, Figure 1b). Among the new nodes investigated in this study, an additional significant association between increased anti-IgD→p-NFkB and shorter TTFT was identified (p=0.02). Conclusions: This study confirms, in an independent data set, the association between increased anti-IgM→p-Erk signaling and shorter TTFT in B-CLL that was observed previously in a separate study. In addition, a newly identified association between anti-IgD→p-NFkB and TTFT was observed, thus supporting the role of BCR signaling in the pathogenesis of the disease and the utility of SCNP assay in elucidating these signaling deregulations with the potential for the development of prognostic and predictive tests. Disclosures: Cesano: Nodality: Employment, Equity Ownership. Evensen:Nodality: Employment, Equity Ownership. Ptacek:Nodality Inc.: Employment, Equity Ownership. Cordeiro:Nodality Inc.: Employment, Equity Ownership. Hawtin:Nodality Inc.: Employment, Equity Ownership. Ware:Nodality Inc.: Employment, Equity Ownership.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1588-1588 ◽  
Author(s):  
M. D. Minden ◽  
Steven M. Kornblau ◽  
David B. Rosen ◽  
Aileen Cleary Cohen ◽  
Urte Gayko ◽  
...  

Abstract Abstract 1588 Poster Board I-614 Background Mutations in the receptor tyrosine kinase (RTK) Fms-like tyrosine kinase 3 (FLT3) gene are among the most common somatic mutations in AML with FLT3 internal tandem duplications (ITDs) occurring in 20-35% of adult and 5-15% of pediatric AML. While the presence of FLT3 ITD mutation does not appear to influence outcome to induction chemotherapy, this mutation has been shown to confer a poor prognosis with significantly shorter disease free and relapse free survival. For patients with intermediate risk cytogenetically normal AML, molecular testing for FLT3 ITD has recently been incorporated into the National Comprehensive Cancer Network (NCCN) guidelines for clinical practice. However, while molecular testing can identify a subset of patients at high risk for relapse, there remains clinical heterogeneity likely due to differences in activation of signal transduction networks. Objectives This study tested the ability to use single cell network profiling (SCNP), in which cells are perturbed with extracellular modulators and their response ascertained by multiparametric flow cytometry, to identify a more clinically predictive functional readout of activation state, intracellular signaling capabilities and pathway dysregulation in the context of FLT3 mutational status. Methods Modulated SCNP was performed sequentially on two independent sets of patient samples (n=32 peripheral blood and n=85 bone marrow samples respectively). 304 and 201 “node-metric” i.e. modulated read outs of dynamic elements on individual proteins in signaling pathways were measured in the two sets respectively. These were derived from pathways known to be relevant to Flt3 WT and Flt3-ITD signaling (e.g. Ras-Raf-Erk-S6, PI3K-Akt-S6, STATs), as well as in-vitro chemotherapeutic induction of apoptosis (cleaved PARP, cleaved caspases), phosphatases, drug transporters (e.g. MDR-1, ABCG2) and expression of growth factor RTKs (e.g. Flt3R, c-Kit). Results In the first study, univariate analysis revealed 76 nodes out of 304 tested that distinguished FLT3 ITD from FLT3 WT patient samples (i.e. AUC of ROC >0.7; p<0.05). Analysis of false discovery rate showed this frequency to be significantly greater than the number of nodes that can be expected by chance (p=0.0009). Although several nodes were found to be correlated, many were independent of each other and represented multiple signaling pathways. Importantly, multivariate analysis showed that combinations of independently predictive nodes improved stratification over the single nodes (AUC of ROC up to 0.99) with respect to distinguishing WT and ITD FLT3 samples. Independent analysis of a second set of samples, revealed several nodes in common between the 2 studies which distinguish FLT3 ITD from WT, including etoposide/c-PARP (apoptosis), IL-27/p-STAT3, 5 (JAK/STAT pathways) and Flt3L/p-S6 (Ras/Erk/mTOR/S6 or PI3K/mTor/S6 pathways). In both sample sets, Flt3 receptor expression did not differ significantly between FLT3 ITD and FLT3 WT samples. Conclusions Pathway analysis by SCNP revealed significant differences in signaling in FLT3 ITD relative to WT AML samples across multiple pathways. We propose that a functional signature of FLT3 signaling is distinct from the existing molecular typing and may improve the ability to predict prognostic outcomes in individual AML patients. The impact of other important prognostic, molecular markers within the FLT3 context (e.g. NPM1) are currently under investigation. Disclosures Kornblau: Nodality, Inc.: Consultancy. Rosen:Nodality, Inc.: Employment, Equity Ownership. Cleary Cohen:Nodality Inc.: Employment, Equity Ownership. Gayko:Nodality, Inc.: Employment, Equity Ownership. Putta:Nodality, Inc.: Employment, Equity Ownership. Woronicz:Nodality, Inc.: Employment, Equity Ownership. Fantl:Nodality, Inc.: Employment, Equity Ownership. Cesano:Nodality, Inc.: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1528-1528
Author(s):  
Sebastian Stasik ◽  
Jan Moritz Middeke ◽  
Michael Kramer ◽  
Christoph Rollig ◽  
Alwin Krämer ◽  
...  

Abstract Purpose: The enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase and key epigenetic regulator involved in transcriptional repression and embryonic development. Loss of EZH2 activity by inactivating mutations is associated with poor prognosis in myeloid malignancies such as MDS. More recently, EZH2 inactivation was shown to induce chemoresistance in acute myeloid leukemia (AML) (Göllner et al., 2017). Data on the frequency and prognostic role of EZH2-mutations in AML are rare and mostly confined to smaller cohorts. To investigate the prevalence and prognostic impact of this alteration in more detail, we analyzed a large cohort of AML patients (n = 1604) for EZH2 mutations. Patients and Methods: All patients analyzed had newly diagnosed AML, were registered in clinical protocols of the Study Alliance Leukemia (SAL) (AML96, AML2003 or AML60+, SORAML) and had available material at diagnosis. Screening for EZH2 mutations and associated alterations was done using Next-Generation Sequencing (NGS) (TruSight Myeloid Sequencing Panel, Illumina) on an Illumina MiSeq-system using bone marrow or peripheral blood. Detection was conducted with a defined cut-off of 5% variant allele frequency (VAF). All samples below the predefined threshold were classified as EZH2 wild type (wt). Patient clinical characteristics and co-mutations were analyzed according to the mutational status. Furthermore, multivariate analysis was used to identify the impact of EZH2 mutations on outcome. Results: EZH2-mutations were found in 63 of 1604 (4%) patients, with a median VAF of 44% (range 6-97%; median coverage 3077x). Mutations were detected within several exons (2-6; 8-12; 14-20) with highest frequencies in exons 17 and 18 (29%). The majority of detected mutations (71% missense and 29% nonsense/frameshift) were single nucleotide variants (SNVs) (87%), followed by small indel mutations. Descriptive statistics of clinical parameters and associated co-mutations revealed significant differences between EZH2-mut and -wt patients. At diagnosis, patients with EZH2 mutations were significantly older (median age 59 yrs) than EZH2-wt patients (median 56 yrs; p=0.044). In addition, significantly fewer EZH2-mut patients (71%) were diagnosed with de novo AML compared to EZH2-wt patients (84%; p=0.036). Accordingly, EZH2-mut patients had a higher rate of secondary acute myeloid leukemia (sAML) (21%), evolving from prior MDS or after prior chemotherapy (tAML) (8%; p=0.036). Also, bone marrow (and blood) blast counts differed between the two groups (EZH2-mut patients had significantly lower BM and PB blast counts; p=0.013). In contrast, no differences were observed for WBC counts, karyotype, ECOG performance status and ELN-2017 risk category compared to EZH2-wt patients. Based on cytogenetics according to the 2017 ELN criteria, 35% of EZH2-mut patients were categorized with favorable risk, 28% had intermediate and 37% adverse risk. No association was seen with -7/7q-. In the group of EZH2-mut AML patients, significantly higher rates of co-mutations were detected in RUNX1 (25%), ASXL1 (22%) and NRAS (25%) compared to EZH2-wt patients (with 10%; 8% and 15%, respectively). Vice versa, concomitant mutations in NPM1 were (non-significantly) more common in EZH2-wt patients (33%) vs EZH2-mut patients (21%). For other frequently mutated genes in AML there was no major difference between EZH2-mut and -wt patients, e.g. FLT3ITD (13%), FLT3TKD (10%) and CEBPA (24%), as well as genes encoding epigenetic modifiers, namely, DNMT3A (21%), IDH1/2 (11/14%), and TET2 (21%). The correlation of EZH2 mutational status with clinical outcomes showed no effect of EZH2 mutations on the rate of complete remission (CR), relapse free survival (RFS) and overall survival (OS) (with a median OS of 18.4 and 17.1 months for EZH2-mut and -wt patients, respectively) in the univariate analyses. Likewise, the multivariate analysis with clinical variable such as age, cytogenetics and WBC using Cox proportional hazard regression, revealed that EZH2 mutations were not an independent risk factor for OS or RFS. Conclusion EZH mutations are recurrent alterations in patients with AML. The association with certain clinical factors and typical mutations such as RUNX1 and ASXL1 points to the fact that these mutations are associated with secondary AML. Our data do not indicate that EZH2 mutations represent an independent prognostic factor. Disclosures Middeke: Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees. Rollig:Bayer: Research Funding; Janssen: Research Funding. Scholl:Jazz Pharma: Membership on an entity's Board of Directors or advisory committees; Abbivie: Other: Travel support; Alexion: Other: Travel support; MDS: Other: Travel support; Novartis: Other: Travel support; Deutsche Krebshilfe: Research Funding; Carreras Foundation: Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees. Hochhaus:Pfizer: Research Funding; Incyte: Research Funding; Novartis: Research Funding; Bristol-Myers Squibb: Research Funding; Takeda: Research Funding. Brümmendorf:Janssen: Consultancy; Takeda: Consultancy; Novartis: Consultancy, Research Funding; Merck: Consultancy; Pfizer: Consultancy, Research Funding. Burchert:AOP Orphan: Honoraria, Research Funding; Bayer: Research Funding; Pfizer: Honoraria; Bristol Myers Squibb: Honoraria, Research Funding; Novartis: Research Funding. Krause:Novartis: Research Funding. Hänel:Amgen: Honoraria; Roche: Honoraria; Takeda: Honoraria; Novartis: Honoraria. Platzbecker:Celgene: Research Funding. Mayer:Eisai: Research Funding; Novartis: Research Funding; Roche: Research Funding; Johnson & Johnson: Research Funding; Affimed: Research Funding. Serve:Bayer: Research Funding. Ehninger:Cellex Gesellschaft fuer Zellgewinnung mbH: Employment, Equity Ownership; Bayer: Research Funding; GEMoaB Monoclonals GmbH: Employment, Equity Ownership. Thiede:AgenDix: Other: Ownership; Novartis: Honoraria, Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1263-1263
Author(s):  
Erik Evensen ◽  
Adam Palazzo ◽  
Ying-Wen Huang ◽  
Alessandra Cesano ◽  
Laura Z. Rassenti ◽  
...  

Abstract Abstract 1263 Poster Board I-285 Background In conjunction with antigen-driven responses, ligand-independent signaling (termed tonic signaling) through both the pre-B cell receptor and B-cell receptor has an important role in B cell development, maturation and survival. In addition to the recognized role of CD79 alpha and CD79 beta BCR signaling, tyrosine phosphatases can impact tonic BCR signaling (Wienands et al. PNAS, 93 p.7865 (1996), Monroe Nat. Rev. Immunol. 6 p.283 (2006)). We previously subjected chronic lymphocytic leukemia (CLL) cells with modulators of BCR signaling and monitored their responses using flow cytometry-based Single Cell Network Profiling (SCNP). Of the many signaling modulators studied, hydrogen peroxide treatment (a general inhibitor of tyrosine phosphatase activity) augmented BCR signaling in a subset of CLL patient samples evaluated. In the remaining samples there was an apparent lack of response to hydrogen peroxide. These data suggested that differential phosphatase activity proximal to BCR signaling was driving the biology of these two patient groups. Objectives Studies were designed to evaluate whether there were any associations between tonic and/or ligand-dependent BCR signaling and in vitro sensitivity to fludarabine, as well as whether such response profiles showed a relationship to the hydrogen peroxide-dependent signaling we observed previously. Methods 23 CLL samples and 7 healthy PBMCs were treated with anti-m alone, hydrogen peroxide alone or the combination for 10 minutes. Separate aliquots of the same sample were exposed to F-ara-A for 48 hours. SCNP was carried out on gated B cells with quantitation of single cell measures of intracellular phosphorylated kinases and adaptor proteins downstream of the BCR. Additionally, the relative activation status of several protein markers of the apoptotic cascade (cytoplasmic cytochrome C, cleaved caspase 3, and cleaved PARP) was measured. Results As previously observed, CLL samples could be segregated into one of two groups exhibiting either responsive or refractory signaling after exposure to hydrogen peroxide alone. Moreover, responsive signaling in CLL cells was correlated in that all the measured components of the canonical B cell receptor network (p-Lyn, p-Syk, p-BLNK, p-PLC-gamma-2, p-Erk and p-Akt) showed the same phosphorylation response: either augmented in unison, or not activated at all. In vitro F-ara-A treatment (48 hours in the presence of 1mM F-ara-A) of parallel samples from these same CLL patients identified distinct populations of apoptosis responsive and refractory cells. Surprisingly, the capacity of patient samples to show augmented BCR signaling in response to hydrogen peroxide was associated prominently with the ability of cells in these patients to exhibit apoptotic proficiency to F-ara-A in vitro. This implies a link between mechanisms governing apoptosis in these CLL cells, survival pathways, and cell states that govern the role of phosphatase activity and BCR signaling potential. Conclusions This study reveals a link between tonic BCR signaling and regulation of apoptosis pathways. This suggests that the subgroup of CLL patients with active phosphatase activity (which suppresses BCR responses) have cell populations that are responsive to F-ara-A, a standard drug in CLL therapy. Conversely, the presence of CLL cells in a patient sample that remain unresponsive to hydrogen peroxide repression of phosphatase activity appear to identify patient samples which cannot undergo apoptosis in response to in vitro F-Ara-A exposure. The clinical implications of this work will be the focus of future translational studies. Disclosures Evensen: Nodality Inc.: Employment, Equity Ownership. Palazzo:Nodality Inc.: Employment, Equity Ownership. Huang:Nodality Inc.: Employment, Equity Ownership. Cesano:Nodality Inc.: Employment, Equity Ownership. Fantl:Nodality, Inc.: Employment, Equity Ownership.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2693-2693
Author(s):  
Alessandra Cesano ◽  
David B. Rosen ◽  
Santosh Putta ◽  
Urte Gayko ◽  
Larry Cripe ◽  
...  

Abstract Abstract 2693 Background: Single Cell Network Profiling (SCNP) is used to measure simultaneously the effects of multiple modulators (including drugs) on signaling cascades at the single cell level. Using this technology, ECOG in collaboration with Nodality is developing several novel biomarker assays with the aim to find blast functional signaling profiles predictive of response to induction therapy and risk of relapse in AML patients. To date, such assays utilized patient bone marrow (BM) as the sample source of blasts. However, in about 65% of patients with AML, circulating peripheral blasts are detected and peripheral blood (PB) sampling is easier and less invasive for patients than BM sampling. Objectives: The objective of this study was to compare by SCNP the functional effects of a panel of compounds simultaneously on different signaling pathways (such as the phosphoinositide 3-kinase (P13K )and the Janus Kinases (Jak) signal transducers and activators of transcription (Stat) pathway) relevant both to the biology of the disease and the development of new therapeutics, in paired, diagnostic, cryopreserved PB mononuclear cells (PBMC) and BMMC samples from 44 AML patients. A paired sample was defined as a BMMC and PBMC specimen collected from the same patient on the same day. Methods: Modulated SCNP using a multiparametric flow cytometry platform was used to evaluate the activation state of intracellular signaling molecules in leukemic blasts under basal conditions and after treatment with specific modulators (Table 1). The SCNP phosphoflow assay was performed on 88 BMMC/PBMC pairs from ECOG trial, E3999. The relationship between readouts of modulated intracellular proteins (“nodes”) between BMMC and PBMC was assessed using linear regression, Bland-Altman method or Lin's concordance correlation coefficient. Results: Table 1 shows the goodness of fit (R2) values from the linear regression analysis for both the basal levels and the modulated levels of intracellular signaling proteins. Most of the signaling nodes show strong correlations (R2 >0.64) with several of the exceptions belonging to nodes with weak response to modulation (e.g. SCF -> p-Akt) or antibodies with dim fluorphores (i.e. Alexa 647). The lack of response is however, consistent between the tissue types for the weak response nodes. Using a rank based metric that is less sensitive to the absolute intensity levels seem to perform better for the antibodies with dim fluorophores. Results from other methods; Bland Altman and Lin's Concordance also show good concordance between the tissue types. Conclusions: The data presented here demonstrate: 1) Specimen source (BM or PB) does not affect proteomic signaling in patients with AML and circulating blasts. 2) PB myeloblasts can be used as a sample source for Nodality SCNP assays to identify functionally distinct leukemic blats cell populations with distinct sensitivities to therapy. 3) The ability to use PB as a sample source will greatly improve the utility of these assays. In particular, our results will facilitate the monitoring of cellular signaling effects following the administration of targeted therapies, e.g., kinase inhibitors, at time-points when BM aspirates are not clinically justifiable. Disclosures: Cesano: Nodality Inc.: Employment, Equity Ownership. Rosen:Nodality Inc.: Employment, Equity Ownership. Putta:Nodality Inc.: Employment, Equity Ownership. Gayko:Nodality Inc.: Employment, Equity Ownership.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 57-57 ◽  
Author(s):  
Jan A. Burger ◽  
Susan O'Brien ◽  
Nathan Fowler ◽  
Ranjana Advani ◽  
Jeff Porte Sharman ◽  
...  

Abstract Abstract 57 Introduction: Bruton's tyrosine kinase (Btk) is a downstream mediator of B-cell receptor (BCR) signaling and is not expressed in T-cells or NK-cells. As such, Btk represents an ideal therapeutic target for B-cell malignancies dependent upon BCR signaling. Chronic lymphocytic leukemia (CLL)/small lymphocytic leukemia (SLL) has been reported to have constitutively active BCR signaling. PCI-32765 is a potent, selective, irreversible and orally bioavailable small molecule inhibitor of Btk that has pre-clinical activity in B-cell malignancies (Proc Natl Acad Sci 2010;107(29):13075-80). PCI-32765 was therefore moved forward to a Phase 1 study in B-cell malignancies including patients (pts) with CLL/SLL. A subsequent CLL/SLL-specific Phase 1b study was initiated to further explore safety, pharmacokinetics (PK), pharmacodynamics (PD), and efficacy of PCI-32765. This report includes a composite summary of the CLL/SLL experience in both of these studies. Pts and Methods: Pts with CLL/SLL who had relapsed or refractory disease after >1 prior treatment regimens were eligible for treatment in each of the studies whereas the second Phase 1b study also included a cohort of elderly pts (aged ≥ 65 years) with CLL/SLL who required treatment and were “treatment-naive”. Responses were assessed by the investigator using the International Working Group CLL criteria (Hallek et al, Blood 2008 for pts with CLL) and the International Workshop to Standardize Response Criteria for Non-Hodgkin's Lymphomas (Cheson et al, J Clin Oncol 2007 for pts with SLL). Results: To date, 30 CLL/SLL patients (including 4 treatment-naive) have been enrolled across the 2 studies. Eighty-four percent of subjects are men with an overall median age of 68 (range 44–82) years. Of the subjects with prior therapy for CLL/SLL the median number of prior therapies is 3 (range 1–4). Treatment has been well-tolerated; Grade ≥ 3 toxicities have been infrequent (10/30 pts; 33%). Two study-drug related serious adverse events have been reported: 1 case of viral adenitis (Grade 3) and 1 case of viral infection (Grade 2). Two adverse events have led to discontinuation of study drug: a small bowel obstruction (Grade 3) and exacerbation of chronic obstructive disease (Grade 3); both events were reported as unrelated to study drug. No study-drug related deaths have reported. There has been no change in either NK cell or T cell counts. Target inhibition as measured by a probe of Btk drug occupancy showed inhibition of Btk at PCI-32765 exposure levels of ≥ 245 ng•h/mL. Of the 14 patients currently evaluable for response using the pre-defined criteria, the overall response rate is 64% (1 complete remission [CR], 8 partial remissions [PR], and 4 SD). Both studies are ongoing and open to enrollment. An update on response rate, response duration, safety, and PD information will be presented on enrolled patients based on a November 2010 database cut-off. Conclusion: PCI-32765 is a novel oral and selective “first-in-human” inhibitor of Btk that induces objective partial and complete responses in a substantial proportion of pts with CLL/SLL and has a favorable safety profile. These data support further studies of both monotherapy and also combination treatment with PCI-32765 in CLL/SLL. Disclosures: O'Brien: Pharmacyclics, Inc: Honoraria, PI grant. Fowler:Pharmacyclics: Consultancy, Research Funding. Advani:Pharmacyclics, Inc: Honoraria, PI grant. Sharman:Pharmacyclics, Inc: Honoraria, PI grant. Furman:Pharmacyclics, Inc: PI grant. Izumi:Pharmacyclics, Inc: Employment. Buggy:Pharmacyclics, Inc: Employment, Equity Ownership. Loury:Pharmacyclics: Employment, Equity Ownership. Hamdy:Pharmacyclics, Inc: Employment, Equity Ownership.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2512-2512
Author(s):  
Alessandra Cesano ◽  
Santosh Putta ◽  
Kavita Mathi ◽  
David B. Rosen ◽  
Urte Gayko ◽  
...  

Abstract Abstract 2512 Background: FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations (FLT3 ITD+) result in constitutive activation of this receptor and have been shown to increase the risk of relapse in patients (pts) with AML; however, substantial heterogeneity in clinical outcomes still exists within both the FLT3 ITD+ and FLT3 ITD- AML subgroups, suggesting alternative mechanisms of disease relapse not accounted for by FLT3 mutational status. Single Cell Network Profiling (SCNP) is a multiparametric flow cytometry-based assay that simultaneously measures, in a quantitative fashion and at the single cell level, both extracellular surface marker levels and changes in intracellular signaling proteins in response to extracellular modulators (Kornblau et al. Clin Cancer Res 2010). Previously, we reported the use of this assay to functionally characterize FLT3 receptor signaling in healthy bone marrow and AML samples (Rosen et al. PLoS One 2010). By applying it to a separate cohort of samples collected from elderly non-M3 AML pts at diagnosis, a subclassification of AML samples beyond their “static” molecular FLT3 ITD status was generated (Rosen et al. ASH 2010 Abstr 2739). Specifically, FLT3 ITD- AML samples displayed a wide range of induced signaling, with a fraction having signaling profiles comparable to FLT3 ITD+ AML samples. Conversely, FLT3 ITD+ AML samples displayed more homogeneous induced signaling, with the exception of those with low mutational load, which had profiles more analogous to FLT3 ITD- AML samples. Due to the small numbers of pts in that exploratory study (n=44 [38 FLT3 ITD- and 6 FLT3 ITD+ pts]), an independent study was undertaken to confirm the observations, as well as to evaluate their clinical relevance (i.e., association with disease free survival (DFS) following anthracycline/cytarabine-based induction therapy). Methods: SCNP was performed as previously described on cryopreserved bone marrow or peripheral blood samples collected prior to anthracycline/cytarabine-based induction therapy from 104 elderly (>60y) non-M3 AML pts enrolled on ECOG trial 3999 or 3993 for whom ITD mutational status (including % mutational load), response and DFS data were available. Samples included 85 FLT3 ITD- and 19 FLT3 ITD+ AML, 30 and 8 of which, respectively, were collected from patients who achieved complete remission (CR). Objectives: The primary study objective was to confirm that levels of FLT3 ligand (FLT3L)-induced signaling (as measured by changes in intracellular phospho-S6 level) are more homogeneous in FLT3 ITD+ than in FLT3 ITD- myeloblasts. Four FLT3 ITD+ groups were pre-defined based on % mutation load (>0, 30%, 40%, or 50%). In addition, FLT3 ITD mutational status and signaling data from the SCNP assay (FLT3L and stem cell factor-induced phospho-S6 signaling and cytarabine/daunorubicin-induced apoptosis [cleaved PARP]) were combined to mathematically model their association with DFS among pts who achieved CR. DFS was defined as time from date of confirmed CR to date of relapse or death. Results: As shown in Figure 1a, our previous observations that variance in FLT3L-induced signaling is higher in FLT3 ITD- AML samples than in FLT3 ITD+ ones and that variance is decreased with increasing mutational load were verified in this study (Levene Test for FLT3 ITD- vs FLT3 ITD+ 50 p value=0.023). Further, when the association of DFS with FLT3 ITD mutational status and signaling data from the SCNP assay was measured using a Cox Proportional-Hazards model, the SCNP data were shown to provide independent information from FLT3 ITD mutational status (p =0.0115 for FLT3L-induced phospho-S6 signaling, Figure 1b). Conclusions: These data add to the growing body of evidence that, even within currently accepted risk stratification groups, AML is a heterogeneous disease. Functional characterization of FLT3 receptor signaling deregulation using SCNP provides prognostic information independent from FLT3 ITD mutational status and allows for more accurate pt stratification by functionally defining DFS risk sub-groups. Characterization of FLT3 signaling deregulation by SCNP could ultimately aid in the improved clinical management of AML pts and help identify candidates for FLT3 receptor inhibitor studies. Disclosures: Cesano: Nodality: Employment, Equity Ownership. Putta:Nodality Inc.: Employment, Equity Ownership. Mathi:Nodality: Employment. Rosen:Nodality Inc.: Employment, Equity Ownership. Gayko:Nodality Inc.: Employment, Equity Ownership. Hawtin:Nodality: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1721-1721
Author(s):  
Sabine Jeromin ◽  
Wolfgang Kern ◽  
Richard Schabath ◽  
Tamara Alpermann ◽  
Niroshan Nadarajah ◽  
...  

Abstract Background: Relapse or refractory disease is a challenging clinical problem in the majority of chronic lymphocytic leukemia (CLL) patients. Treatment influences the clonal composition by selection and eventually induction of additional genetic abnormalities. Aim: To characterize the clonal evolution in relapsed CLL patients by deep-sequencing analysis of mutations in ATM, BIRC3, NOTCH1, POT1, SF3B1, SAMHD1 and TP53. Patients and Methods: Sequential samples of 20 relapsed CLL patients at three time-points were evaluated: A: at diagnosis (n=16) or in untreated state (n=4), B: at first relapse (n=20) and C: at second relapse (n=2). Patients were treated with diverse treatment schemes and had temporarily achieved either complete or partial remission during the course of the disease. The median time from diagnosis to first-line treatment was 13 months (1 - 169 months). All geneswere sequenced by a deep sequencing approach (Illumina, San Diego, CA). IGHV mutational status was determined by direct Sanger sequencing at time-point A. Chromosome banding analysis (CBA) and FISH data on del(17p), del(11q), trisomy 12 (+12), and del(13q) were available in 33/42 and 36/42 samples, respectively. Results: Initially, samples at first relapse were sequenced. Mutations in SF3B1 (6/20, 30%), TP53 (5/20, 25%), ATM (5/20, 25%), NOTCH1 (4/20, 20%), and SAMHD1 (3/20, 15%) were detected at high frequencies. No mutations were detected in BIRC3 and POT1. In total, 75% of cases presented with at least one mutation (Figure 1): 8 (40%) cases had one, 6 (30%) cases had two and one patient had three genes concomitantly mutated (mut). Patients were also analyzed for IGHV mutational status at diagnosis and presented with unmutated status at a frequency of 85% (17/20). Subsequently, samples from cases with mutations were analyzed at time-point A. In 12/15 (80%) cases the mutations at relapse were already detectable at time-point A with a similar load indicating presence of the main clone before and after chemotherapy. However, in 7/15 (47%) patients new gene mutations were acquired either additionally to existing mutations (n=4) or in previously wild-type cases (n=3). In 5/7 (71%) cases mutations were located in TP53. TP53 mut were the only mutations that were not detected in samples before treatment (sensitivity of 3%). Thus, TP53 mutations might have been initiated by chemotherapy or exist in a minor subclone subsequently selected by chemotherapy. Furthermore, only 4 cases had low-level mutations (3-6% mutation load) at diagnosis in either SAMHD1 or SF3B1 that eventually increased in their burden during disease course. Of note, in two patients a multibranching clonal evolution could be identified (#2, #9). For patient #2 three time-points were analyzed. At diagnosis 2 ATM mutations were detected with mutation loads of about 20%, each. In the course of the disease these mutations were lost, whereas SF3B1 mut showed a stable mutation load in all three time-points of about 40%. In contrast, mutation load of SAMHD1 increased over time from 4% to 87%. CBA was performed at diagnosis and detected independent clones with del(11q) and del(13q). Accordingly, del(11q) detected by FISH at diagnosis was lost and the percentage of cells with del(13q) increased from diagnosis to time-point C. Therefore, patient #2 shows different genetic subclones in parallel that were eradicated or selected by chemotherapy. In patient #9 two SF3B1 mutations were initially detected with the same mutation load of 10%. After treatment one mutation was lost, whereas the load of the second mutation increased indicating at least two different subclones with only one of them being sensitive to chemotherapy. This might be due to different additional aberrations. Indeed, CBA identified two clones: one with +12 alone and one in combination with del(13q). FISH revealed unchanged percentage of +12 at time-point B, whereas del(13q) positive cells were diminished. Conclusions: In 75% of relapsed CLL cases mutations in SF3B1, TP53, ATM, NOTCH1, and SAMHD1 are present at high frequencies. 80% of these mutations are already detectable before treatment initiation representing the main clone. Remarkably, TP53 mutations were the only mutations that were not detected before but only after chemotherapy. Figure 1. Distribution of gene mutations in 15 CLL cases with mutations at diagnosis or before treatment (D) and at relapse (R). Red = mutated, grey = wild-type, white = not analyzed. Figure 1. Distribution of gene mutations in 15 CLL cases with mutations at diagnosis or before treatment (D) and at relapse (R). Red = mutated, grey = wild-type, white = not analyzed. Disclosures Jeromin: MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schabath:MLL Munich Leukemia Laboratory: Employment. Alpermann:MLL Munich Leukemia Laboratory: Employment. Nadarajah:MLL Munich Leukemia Laboratory: Employment. Meggendorfer:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2052-2052
Author(s):  
Steven D Levin ◽  
Hieu Nguyen ◽  
Joseph Kuijper ◽  
Rebecca P Wu ◽  
Ryan Swanson ◽  
...  

Abstract Background: T cell receptor (TCR)-engineered T cell therapies hold great promise as personalized, adoptive anticancer treatments, but clinical experience to date has demonstrated generally only modest efficacy, attributed in large part to unfavorable factors in the tumor microenvironment. This includes the presence of receptors, such as PD-L1, that can inhibit T cell responses, and/or insufficient engineered T cell longevity. Common strategies to address such limitations have involved attempts to provide additional costimulatory signals to T cells. However, addition of costimulatory signals alone may not be sufficient to overcome PD-L1-mediated inhibition. We have utilized our variant immunoglobulin domain (vIgD) platform, based on the directed evolution of immunoglobulin superfamily (IgSF) members, to develop PD-1 domains with higher affinity for PD-L1 and then substituted costimulatory intracellular signaling domains for the native PD-1 inhibitory intracellular region with the hypothesis that PD-L1 engagement of these Transmembrane Immune-modulatory Proteins (TIPs) would result in costimulation rather than inhibitory signaling. Such "Switch" TIPs that consist of a checkpoint-inhibitory extracellular PD-1 vIgD and intracellular costimulatory domains may therefore improve the activity of TCR-engineered T cells by providing costimulation while preventing inhibitory signaling through native PD-1. Methods: Variant PD-1 extracellular domains (PD-1 vIgDs) were generated using random mutagenesis and FACS-based selection of yeast displayed proteins selecting for increased binding to PD-L1. These variants were then fused to intracellular signaling domains from CD28, ICOS and CD137 either singly or together in combinations. These constructs were expressed via lentivirus in primary human T cells along with a TCR recognizing an HPV16 E6 peptide (E6 TCR). Surface expression was confirmed by flow cytometry, and activity was assessed against HPV+ tumor cell lines via proliferation, cytokine production (IFNg, TNFa and IL-2) and cytotoxicity. Results: TIPs including PD-1 vIgDs selected for increased PD-L1 binding and various combinations of intracellular signaling domains potently enhanced the activity of E6 TCR-engineered T cells, including killing of HPV+ target cells (Figure 1a) as well as target-driven proliferation and cytokine production (Figure 1b). Activity was consistently superior to TIPs consisting of only a PD-1 extracellular vIgD, or TIPs which included a wild-type extracellular PD-1 domain. Importantly, TIP activity was abrogated in the presence of an anti-PD-L1 antibody, demonstrating PD-L1 dependence of the costimulatory activity. Conclusions: "Switch" TIPs consisting of high-affinity PD-1 vIgD extracellular domains and costimulatory intracellular signaling domains potently augment the antitumor activity of TCR-engineered T cells as judged by proliferation, cytokine production and cytotoxicity. Ongoing studies continue to explore this and analogous strategies with other IgSF-based vIgDs and/or costimulatory domains and will hopefully significantly enhance the clinical efficacy of engineered T cells in both solid and hematological malignancies. Disclosures Levin: Alpine Immune Sciences: Employment, Equity Ownership. Nguyen:Alpine Immune Sciences: Employment, Equity Ownership. Kuijper:Alpine Immune Sciences: Employment, Equity Ownership. Wu:Alpine Immune Sciences: Employment, Equity Ownership. Swanson:Alpine Immune Sciences: Employment, Equity Ownership. Peng:Alpine Immune Sciences: Employment, Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document