scholarly journals Protein Expression Signatures Predict Response, Relapse, and Survival after Venetoclax Containing Therapy in Adults with Acute Myeloid Leukemia

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 227-227
Author(s):  
Brandon Brown ◽  
Yihua Qiu ◽  
Fieke W Hoff ◽  
Steven M. Kornblau

Abstract Introduction When added to cytarabine (Ara-c) or hypomethylating agents (HMA), the BCL2 inhibitor, venetoclax (VTX), has been reported to improve response and overall survival (OS) rates. However, resistance and relapse still occur in the majority, and, although alterations in MCL1 and BCLXL are noted at relapse, identification of prognostic features remain unknown, notably not correlating with expression of the BCL2 target. Identification of prognostic markers could guide VTX use in patients and/or post-remission therapy. We searched for protein expression targets individually and collectively to predict VTX response and relapse in AML. Methods Reverse Phase Protein Array (RPPA) was performed on diagnostic leukemia samples of 818 adults with AML, of which 143 received VTX including 33 in combination with high dose Ara-C, 5 with standard dose Ara-C, 50 with HMA, and 13 with HMA and targeted therapy. Protein expression levels were evaluated using 390 validated antibodies were analyzed in the context of clinical data compiled by retrospective chart review. Pearson correlation was used to identify significant protein-protein correlations. Survival curves were generated by the Kaplan-Meier method and survival data was analyzed by multivariate cox regression model. Protein expression signatures were identified by hierarchical clustering and predictive models of classifiers were determined by classification and regression trees (CART) analysis. Results We queried the 390 proteins assayed in the 143 VTX treated patients to identify proteins individually prognostic (p<0.01) for OS (n=27) or remission duration (RD, n=44). Notably, neither MCL1, BCLXL nor BCL2 expression at diagnosis were prognostic of OS or RD. From these, unbiased hierarchical clustering revealed two cohorts (N=102 & 41 patients) for OS and RD. The clusters were similar for clinical features with no significant differences noted for, age, gender, performance status, cytogenetics, or the presence of molecular mutation markers FLT3.ITD, IDH1/2, NPM1 or TP53. The groups did not differ by therapy combination. Remission rates were insignificantly less in cluster 1 (61% vs 77%). Clear differences were observed for OS with estimated 3-yr overall survival 27% vs 66% (p=0.009, Figure 1) and relapse risk (RR) at 1-yr 45% vs 18% (p=0.001, Figure 2) in cluster 1 vs 2, respectively. In multivariate analysis, protein cluster membership was in independent prognostic factor for OS (along with TP53 and NPM1 mutations) but unfavorable cytogenetics was not. Prognostication did not vary based on cytogenetics or therapy received. For RD, protein cluster membership and unfavorable cytogenetics were the only independent predictors. Of the 44 proteins in the protein signature, CART modeling identified 3 - SPI1, NOTCH1.cle, and PTPN12 - that could predict clustering with a computed accuracy of 94.3%. Similarly, when these three proteins were used as training variables for random forest classification, the error rate was 3.7%. Several previously unrecognized potential therapeutic targets for preventing VTX resistance were also identified. Discussion Protein expression patterns, individually and in combination, were very highly predictive of outcome to VTX containing combination chemotherapy. A group with lower response rates, higher relapse rates, shorter RD and inferior OS was defined. A kit to prospectively determine cluster membership is in development. If validated this could be used to triage high-risk patients to alternate therapies, such as transplant, in CR1. Many new targets for combination therapy to prevent VTX resistance were identified and need to be tested in the laboratory for clinical relevance. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3813-3813 ◽  
Author(s):  
Steven M. Kornblau ◽  
Peter Ruvolo ◽  
Rui-Yu Wang ◽  
Vivian Ruvolo ◽  
Yihua Qiu ◽  
...  

Abstract Background: AML remains highly fatal, therefore understanding the mechanisms that cause chemoresistance is critical for developing more effective therapies. The leukemia bone marrow microenvironment (BME) and component MSC supports leukemia development and cell survival, implying a key role for MSC in resistance. We hypothesize that differences in the physiology of MSC in the leukemia BME (AML-MSC), relative to normal MSC (NL-MSC), exist and could be therapeutic targets. Methods: To compare the function of AML-MSC from NL-MSC a custom reverse phase protein array (RPPA) was made using cultured MSC from AML (N = 106) and healthy donor MSC (NL; N = 71) and probed with 151 antibodies against 114 total, 36 phospho sites (on 29 proteins) and 1 cleavage site. Both biased clustering and unbiased hierarchical clustering along with principal component analysis were used to analyze data. To examine the influence of age on protein expression, age matched AML and NL MSC were compared. Results: Comparison of protein expression in NL-MSC and AML-MSC identified 5 Sample Clusters (SC1..SC5) based on the differential expression of 83 of 151 proteins, which formed 5 Protein Clusters (PC1..PC5) (P < 0.000001, FDR = 0.0000017)(FIG 1). Distribution of NL-MSC was significantly skewed to SC1 (7 of 8) and SC3 (38 of 52) while AML-MSC dominated SC2 (37 of 45) and SC4 (45 of 59), (Χ2 = 45.3, df=4, P <0.0001). NL-MSC were characterized by low expression of proteins in PC1 and 2 and higher expression within PC 3,4 & 5 with SC1 having more extreme levels than SC3. Protein levels in AML dominant SC4 was opposite of SC1 and SC3 for all 5 PC and was designated as "AML". SC5 was a more extreme version of SC4, for PC2, 4 and 5 but resembled SC3 for PC1 and 3. In contrast, the AML dominant SC2 resembled NL-MSC dominant SC1 and SC3 for PC2, 4 and 5, resembling SC4 only in PC1 and 3. This cluster was designated as "NL-like AML". Proteins with universally higher expression in NL-MSC included: SMADs 1 and 4, STMN1, SIRT1, p-Foxo1/3 (S32), HSP90 and EIF2S1. AML MSC had higher levels of 18 proteins across all groups including CCND1, BCL-XL, STAT5, and PPP2R2A. Salvage cases were more often in SC2 (17 of 36) and SC4 (26 of 45) than in SC3 (3 of 15) (Χ2 = 6.44, df=2, P <0.04). The observed changes were similar within three age groups (<30, 30-49, 50-59) in 22 of 25 universally differentially expressed proteins, demonstrating age independence. MSC cluster membership correlated with cytogenetics: Unfavorable cytogenetics (41 % overall) comprised 30% of NL-Like SC2, 42% of "Normal" SC3 but 52% of "AML" SC4 cases (p= 0.04), and both favorable cytogenetics cases were in SC3. MSC subpopulation type was not associated with overall survival, remission duration, or AML mutation status (FLT3, NPM1, RAS). We confirmed higher differential expression of mRNA (by qRT-PCR) for some (BCL2L1, CCND1) but not all (p21) in 10 AML-MSC and 10 NL-MSC, suggesting that both transcriptional and translational mechanisms are involved. In a separate ASH submission Battula shows that AML-MSC cannot differentiate into adipocytes like NL-MSC. Ingenuity pathway analysis (IPA) of this dataset finds that PC3 members, which are highly expressed in NL-MSC SC1, 3 & 5, but low in AML SC2 & 4, are associated with adipogenesis. Notably PI3K/AKT and JAK/STAT signaling is higher in AML dominant SC4. Hierarchical clustering revealed that 9 proteins showed differential expression between diagnosis and salvage status ( P=0.05) with p-β catenin, p-RPS6, and Galectin 3 higher in salvage samples, while SMAD6, TCF4, LYN, integrin β3, p-EIF4BP1, and p-ELK1 were higher at diagnosis. IPA reveals these proteins are highly associated with osteoblast differentiation, molecular mechanism of cancer and stem cell pluripotency, suggesting potential mechanisms for how alterations in MSC protein expression could affect chemosensitivity. Summary: This study demonstrates that AML-MSC have two dominant protein expression signatures that are distinct from those of NL-MSC, with SC4 being associated with unfavorable cytogenetics and the salvage setting. Up-regulated pathways in AML-MSC are known to impact MSC cell survival and differentiation. Down regulated pathways may explain skewing towards osteogenic and away from adipogenic differentiation by AML-MSC. Experiments targeting MSC and assessing effects on AML blast survival are underway to determine if targeting MSC can reverse chemoresistance. Figure 1. Figure 1. Disclosures Konopleva: Novartis: Research Funding; AbbVie: Research Funding; Stemline: Research Funding; Calithera: Research Funding; Threshold: Research Funding. Andreeff:Oncoceutics, Inc.: Membership on an entity's Board of Directors or advisory committees.


BMC Cancer ◽  
2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Hanna Engqvist ◽  
Toshima Z. Parris ◽  
Anikó Kovács ◽  
Szilárd Nemes ◽  
Elisabeth Werner Rönnerman ◽  
...  

Abstract Background Ovarian cancer is the main cause of gynecological cancer-associated death. However, 5-year survival rates differ dramatically between the five main ovarian carcinoma histotypes. Therefore, we need to have a better understanding of the mechanisms that promote histotype-specific ovarian carcinogenesis and identify novel prognostic biomarkers. Methods Here, we evaluated the prognostic role of 29 genes for early-stage (I and II) ovarian carcinomas (n = 206) using immunohistochemistry (IHC). Results We provide evidence of aberrant protein expression patterns for Collagen type III alpha 1 chain (COL3A1), G protein-coupled receptor 158 (GPR158) and PITH domain containing 1 (PITHD1). Kaplan-Meier survival analysis revealed that COL3A1 expression was associated with shorter overall survival in the four major histotypes of epithelial ovarian carcinoma patients (P value = 0.026, HR = 2.99 (95% CI 1.089–8.19)). Furthermore, GPR158 and PITHD1 were shown to be histotype-specific prognostic biomarkers, with elevated GPR158 expression patterns in mucinous ovarian carcinoma patients with unfavorable overall survival (P value = 0.00043, HR = 6.13 (95% CI 1.98–18.98)), and an association with lower PITHD1 protein expression and unfavorable overall and disease-specific survival in clear-cell ovarian carcinoma patients (P value = 0.012, HR = 0.22 (95% CI 0.058–0.80); P value = 0.003, HR = 0.17 (95% CI 0.043–0.64)). Conclusions The novel biomarkers identified here may improve prognostication at the time of diagnosis and may assist in the development of future individualized therapeutic strategies for ovarian carcinoma patients.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2355-2355
Author(s):  
Steven M. Kornblau ◽  
David McCue ◽  
Kang L. Lu ◽  
Wenjing Chen ◽  
Kevin R. Coombes

Abstract Protein expression and activation determines the pathophysiology of leukemic cells in Myelodysplasia (MDS) and Acute Myelogenous Leukemia (AML) and is dependent on endogenous changes (e.g mutation, methylation) and exogenous signals from stromal interactions, cytokines (CTKN) and chemokines. We have previously performed proteomics on primary AML sample (using reverse phase protein arrays) and wanted to assess how cytokines affect protein expression and phosphorylation. Prior studies of CTKN expression in AML and MDS have generally measured individual CTKNs, but not provided an overall assessment of CTKN expression. We measured the level of 26 CTKN (IL-1RA, 1B, 2, 4 5, 6, 7 , 8 , 9, 10,12, 13, 15, 17, Eotaxin, FGFB, G-CSF, GM-CSF, IFNγ, IP10, MCP1, MIP1α, MIP1β, PDGF, TNFα and VEGF) using multiplex cytometry (Bioplex™, Biorad) in serum samples from 176 AML (138 untreated (New), 38 relapsed (REL)) and 114 MDS patients (97 New, 10 post biological therapy, 7 REL) and 19 normal (NL) subjects. Individual CTKN expression was not correlated with clinical features (e.g. age, gender, cytogenetics, FAB, HB, WBC, platelet etc). The levels of IL -1β, 4, 5, 6, 7,10,12, 13, 17, IFNγ, FGFB and MIP1α were significantly lower and IL-8 and 15 higher in AML/MDS compared to NL. The expression profiles of AML and MDS were statistically indistinguishable whether analyzed individually or by unsupervised hierarchical clustering, except for IL-8 and 13 (higher in AML) and VEGF (higher in MDS). When CTKN were evaluated individually in new AML cases higher levels of IL4, 5 and 10 correlated significantly with remission attainment, and higher levels of IL8, Il1Ra, IP-10, Mip1β, VEGF and ILR, correlated significantly with shorter survival. No CTKN predicted remission attainment or survival in MDS. Unsupervised hierarchical bootstrap clustering using Pearson correlation and average linkage of CTKN expression relative to other CTKN expression, where high levels of one CTKN correlated with high expression of the other, revealed 6 highly reproducible expression patterns: IL-1β 4, 7, 10, 12, 13, G-CSF, IFNγ, MIP1α and PDGF IL 1ra, 6, 8 Eotaxin, IP-10, MIP1β and VEGF, IL2, 9, 15 and GMCSF, IL5 IL-7, FGF-Basic, TNFα and MCP1. Similar unsupervised clustering of the samples based on CTKN expression using average linkage also revealed 5 disease clusters and a NL sample cluster (containing all 19 NL samples). Average expression levels of each CTKN in these 5 clusters show diminished expression of most CTKN that had high expression in the NL samples, with each group showing increase in expression in a distinct subset of CTKN relative to NL. Remission attainment was strongly associated with cytokine signature (P=0.005). Additional CTKN are being studied (SCF, TGFβ, IL3). Comparison of CTKN expression patterns with proteomic profiling of expression and phosphorylation status is pending. In summary, this is the largest sample set studied for multiple CTKN expression in AML and MDS and the first assessment of many of these CTKN in these diseases. Most CTKNs showed different expression in AML and MDS compared to NL. Interestingly, CTKN expression in AML and MDS were similar. Many CTKN are predictive of outcome individually. CTKN signatures distinguish groups of patients and are predictive of outcome. Correlation with proteomic profiling may suggest CTKN to target in combination with other targeted therapies to maximally affect activated pathways.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 656-656 ◽  
Author(s):  
Gareth J Morgan ◽  
Graham H Jackson ◽  
Faith E Davies ◽  
Mark T Drayson ◽  
Roger G Owen ◽  
...  

Abstract The role of maintenance therapy for the long term control of the plasma cell clone in patients induced into response with either intensive or conventional treatment is an important outstanding question. We addressed this in the MRC Myeloma IX study which incorporates intensive and non-intensive pathways selected according to PS and age. In the intensive pathway patients were randomised to either CTD or CVAD induction, followed by High Dose Melphalan (HDM) before being randomised to either thalidomide or no maintenance. In the non-intensive pathway patients were randomised to either MP or attenuated CTD prior to the maintenance randomisation. For patients randomised to thalidomide it was initiated at d100 following HDM or at the end of induction in the non-intensive arm with the aim of delivering 100mg daily until relapse. A dose reduction algorithm for side effects was used. Between the years of 2003–8, 820 patients were entered into the maintenance randomisation, median age 64 (intensive 59, non-intensive 73), median follow-up 32 months. Prognostic features were evenly distributed between the arms. FISH and cytogenetics were done using standard methods. Response was assessed by IWG criteria. For overall survival (OS) there was a non-significant trend in favour of the no maintenance arm, which enables us, by calculating confidence limits on the hazard ratio, to make the assertion that no maintenance could be up to 7% worse than thalidomide at 5 years (p=.005). Further analysis showed that there was no significant difference in OS in either the intensive or the non-intensive arm. The duration of time on thalidomide maintenance appeared to make no difference to OS. There was a non-significant improvement in progression free survival (PFS) across the maintenance randomisation as a whole and in the intensive pathway a significant benefit of maintenance was seen in the patients achieving less than a VGPR post initial induction therapy prior to HDM, (hazard ratio 1.9, p=.007). This PFS difference did not translate into a survival benefit because the survival after progression in the PR patients receiving maintenance thalidomide was poor (p=.002). In addition we looked at the time spent off thalidomide, the recovery time, (the time between stopping thalidomide and progression) as a possible predictor of survival after progression. Treated as a continuous variable in the Cox model this showed a trend for longer survival after relapse in those with longer recovery time (p=.056). In the non-intensive pathway a similar but less pronounced effect of thalidomide maintenance on PFS was seen. These results are consistent with a consolidation rather than a maintenance effect for thalidomide in this setting. The impact of maintenance in different cytogenetic subgroups was also determined [17p-, 13q-, 14q abnormalities including t(4;14), t(14;16), t(6;14), t(14;20) and t(11;14)]. For the 17p- group, the difference in OS between no thalidomide and thalidomide is large (HR = 4.55, p=.02) with the thalidomide patients faring worse, although this is based on only 30 patients. For the non 17p- group there is no difference in PFS (HR = 1.24, p=.37), in the 17p- group, however, the PFS is worse. In addition, of the 22, 17p- patients receiving CTD or CTDa as initial therapy, the 10 who received no thalidomide maintenance are all still alive, whereas 9/12 of those who went on to receive thalidomide maintenance have died. It seems that thalidomide given at induction and again in maintenance, may be particularly detrimental in 17p- patients. Although thalidomide maintenance may improve PFS, there is no demonstrable benefit on OS. It is important to identify 17p- in order to exclude these patients from receiving thalidomide maintenance.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e18550-e18550
Author(s):  
Matthew Risendal ◽  
Allison Hazlett ◽  
Roy T. Sabo ◽  
Priscilla Mpasi ◽  
Joan Bolling ◽  
...  

e18550 Background: Multiple myeloma (MM) is a plasma cell malignancy with variable prognosis depending on disease features such as β2m and cytogenetics. High dose therapy and stem cell transplantation (SCT) remains the current standard of care for MM, however the role of tandem SCT is controversial, particularly in the era of novel induction therapy. Methods: Our program has a practice of risk-stratified transplant allocation in MM patients referred for SCT, those with high-risk (HR) disease (β2m >5.5, adverse cytogenetics, >1st remission) are preferentially assigned tandem SCT, and those with standard risk (SR), a single SCT. Between 2008 and 2011, 129 MM patients underwent SCT, 43% SR patients received a single SCT (SRS), 22% HR received tandem SCT (HRT) & 36% HR a single SCT (HRS). Median age at SCT was 57 years. Maintenance therapy was administered in 51% SRS, 57% HRT & 67% HRS patients. Results: Complete response (CR) or very good partial response was achieved in 0.68, 0.72 and 0.80 for the HRT, HRS, and SRS groups. The HRT group (0.39) was more likely to achieve CR than HRS (0.20) (P=0.02). At a median follow up of 23.4 months, the overall survival for HRS was inferior to SRS (P=0.01) but there was no difference in the overall survival between HRT and SRS cohorts. Two-year survival rates were 0.81, 0.91 and 0.97 in the HRS, HRT and SRS cohorts (HRS vs. SRS P=0.02). This was attributable to a higher 1-year relapse rate in HRS (0.29) compared to HRT (0.07) and SRS (0.07) (P<0.01). Conclusions: Using a risk-stratified allocation system, we report that HR MM patients undergoing tandem SCT have outcomes comparable to SR patients. However, HR MM patients receiving a single SCT have inferior outcomes compared to those with SR. Notably, higher rate of CR and a lower relapse rate were observed in the HRT cohort when compared to HRS. This suggests that greater depth of remission achieved in HR patients undergoing tandem SCT may result in longer time to relapse and survival advantage compared to HR patients receiving a single SCT. In contrast, a single SCT may suffice for SR MM patients. These results demonstrate that risk-stratification based on disease prognostic features is an important treatment consideration when planning high dose therapy in MM patients.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 7521-7521 ◽  
Author(s):  
D. M. Weber ◽  
C. Chen ◽  
R. Niesvizky ◽  
M. Wang ◽  
A. Belch ◽  
...  

7521 Background: Lenalidomide is a novel, orally administered, immunomodulatory drug (IMiD) that has single-agent activity against MM and additive effects when combined with dexamethasone (Dex). Methods: In this phase 3, multicenter, double-blind trial, 354 patients (pts)with relapsed or refractory MM were treated with Dex 40 mg daily on days 1–4, 9–12, 17–20 every 28 days and were randomized to receive either lenalidomide (Len) 25 mg daily orally on days 1–21 every 28 days or placebo. Beginning with cycle 5, Dex was reduced to 40 mg daily on days 1–4 only, every 28 days. Patients were stratified with respect to B2M (≤ 2.5 vs. > 2.5 mg/mL), prior stem cell transplant (none vs. ≥ 1), and number of prior regimens (1 vs > 1). The treatment arms were well balanced for prognostic features. Results: The overall response rate was greater with Len-Dex than with Dex-placebo (59.4% vs. 21.1%; p < 0.001). Complete responses were achieved in 12.9% of pts treated with Len-Dex and 0.6% of pts treated with Dex-placebo. The median time to progression (TTP) for pts treated with Len-Dex was 11.1 months compared to 4.7 months for pts treated with Dex-placebo (p < 0.000001). Median overall survival was higher with Len-Dex (not reached) compared to Dex-placebo (24 months) (hazard ratio 1.76, p = .0125). Grade 3–4 neutropenia was more frequent with combination therapy than with Dex-placebo (24% vs. 3.5%), however ≥ grade 3 infections were similar in both groups. Thromboembolic events occurred in 15% of pts treated with Len-Dex and in 3.5% of pts treated with Dex-placebo. Atrial fibrillation occurred in 8 pts and CHF developed in 4 pts treated with Len-Dex. Conclusions: Considering the ease of oral administration, higher response rate, longer time to progression and overall survival benefit, the combination of lenalidomide-dexamethasone may very well represent the treatment of choice for early refractory or relapsing multiple myeloma. The relatively infrequent side effects should not detract from these improvements, but the use of prophylactic antithrombotic therapy should be considered for patients treated with the combination of lenalidomide and dexamethasone. [Table: see text]


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4262-4262 ◽  
Author(s):  
Heike Horn ◽  
Marita Ziepert ◽  
Thomas F.E. Barth ◽  
Heinz-Wolfram Bernd ◽  
Martin Wartenberg ◽  
...  

Abstract Purpose A number of retrospective analyses have looked into the prognostic implication of MYC, BCL2 and BCL6 rearrangements and protein expression for MYC, BCL2 and BCL6 in patients with diffuse large B-cell lymphoma (DLBCL). Many of these studies suffer from small patient cohorts and differing or unknown treatment strategies (with or without Rituximab) administered to patients under study. Furthermore, the median age of these patients was relatively high. We for the first time report on the prognostic consequences of MYC, BCL2 and BCL6 alterations in younger (18-60 years) high-risk patients (aaIPI 2 or 3) with DLBCL. Patients and Methods The MegaCHOEP study (Schmitz et al. Lancet Oncology 2012: 13, 1250) randomized patients to 8xCHOEP-14 or sequential high-dose therapy supported by repeated infusions of autologous stem cells. Both treatment arms included 6 infusions of R (375 mg/ sqm). The median patient age was 48 years, and 27% of patients scored an aaIPI of 3. Paraffin-embedded tumor samples from 112 de novo DLBCL were investigated using immunohistochemistry for MYC, BCL2, and BCL6, and fluorescence in-situ hybridisation (FISH) to detect breaks of MYC, BCL2 and BCL6. Results Rearrangements of MYC, BCL2 and BCL6 were detected in 13.6%, 20.7% and 30.9% of DLBCL, respectively. A double or triple hit constellation occurred in 10.8%. Presence of BCL2 breaks (RR=4.7, 95% CI: 1.8-12.2) and MYC breaks (RR=2.4, 95% CI: 0.8-7.5), but not of BCL6 breaks were associated with inferior overall survival in univariate and multivariate analyses adjusted for aaIPI and treatment arm. Protein overexpression of MYC ≥30% (RR=2.4, 95% CI: 0.9-6.5), but not BCL2 (≥60%) or BCL6 (≥30%) indicated inferior overall survival. BCL2 overexpression was associated with inferior EFS (RR=2.2, 95% CI: 0.9-5.5) and PFS (RR=2.8, 95% CI: 1.0-8.2). If the same cutpoint for BCL2 used for a similar analysis in elderly patients treated on the RICOVER-60 trial (Horn et al. Blood 2013) was applied, no differences in EFS or PFS were observed. Conclusion Rearrangements of MYC and BCL2 seem to be relevant prognostic factors also in young high-risk patients. The frequencies of MYC and BCL2 rearrangements are not substantially higher than reported for other, mostly elderly patient groups carrying IPI scores from 0 - 5; it seems unlikely, therefore, that the rearrangements described here (completely) explain the poor prognosis of young, high-risk patients. The prognostic role of BCL2, BCL6, and MYC as well as other biological risk factors must be validated in independent data sets. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii185-ii186
Author(s):  
Harish N Vasudevan ◽  
Calixto-Hope G Lucas ◽  
William Chen ◽  
Stephen Magill ◽  
Steve Braunstein ◽  
...  

Abstract BACKGROUND Malignant peripheral nerve sheath tumor (MPNST) is an aggressive neoplasm associated with neurofibromatosis type 1 (NF1). Despite multimodal therapy, clinical outcomes remain poor. To elucidate markers of MPNST treatment response, we retrospectively reviewed the medical records of MPNST patients at a single institution and performed histopathological and immunohistochemical (IHC) analysis for predictive and prognostic features. METHODS We identified 54 consecutive patients treated at University of California San Francisco between 1990 and 2018 that met diagnostic criteria for MPNST on pathologic review with sufficient tissue available for histology and immunohistochemistry (IHC) assays. IHC was performed for Ki-67, EGFR, p53, H3K27me3, neurofibromin, S100, p75NTR, SOX10, p16, and SOX2. Overall survival (OS), metastasis free survival (MFS), and locoregional failure free rate (LFFR), were estimated using the Kaplan-Meier method. Log-rank test, Cox Proportional Hazards regression, and hierarchical clustering were performed in R. RESULTS With a median follow up of 19.2 months, the 5-year OS, MFS, and LFFR were 58%, 68%, and 66%, respectively, with no significant differences between NF1 associated (n=32) and sporadic tumors (n=22). Radiation therapy significantly improved 5-year LFFR (80% versus 49%, p=0.05), but not OS or MFS. Tumor grade was associated with worse OS by Fédération Nationale des Centres de Lutte Contre Le Cancer (FNCLCC) grading (p=0.02). Furthermore, elevated Ki-67 index was associated with worse 5-year OS (39% versus 73% for Ki-67 index ³ 60 and Ki-67 index &lt; 60, p=0.01). Finally, hierarchical clustering of IHC data identified a predictive signature defined by elevated Ki-67 and EGFR expression associated with improved responses to radiation therapy (5-year OS 86% versus 10%, p=0.004). CONCLUSIONS Our data provide insights into the diagnosis and treatment of MPNST. Additional investigation is needed to understand the biologic mechanisms and generalizability of the signatures uncovered in our analysis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3230-3230
Author(s):  
Steven M. Kornblau ◽  
Yi Hua Qiu ◽  
Nianxiang Zhang ◽  
Kevin R Coombes ◽  
Susan O'Brien ◽  
...  

Abstract Abstract 3230 Background. Having previously shown that protein expression signatures (ProExpSig), based on the activation state of cell cycle, apoptosis and signal transduction (STP) regulating proteins, existed and were prognostic in AML, we extended this to evaluate protein expression patterns in ALL. Methodology. We have generated RPPA using protein derived from the leukemia-enriched fraction of 283 primary ALL samples from 216 cases with the goal of providing comprehensive proteomic based classification of ALL. Included are 253 samples from 194 newly diagnosed ALL patients (179 Marrow (BM), 95 blood (PB) samples, 41 with concurrent PB and BM specimens), and 14 relapse (REL) samples from these same cases. Phenotypes included 163 pre-B, 22 T-Cell and 9 Burkitt's cases. There were 42 PH1+ cases but only the 28 treated with TKIs were included for outcome analysis. All protein preps were made from fresh cells on the day of collection as we previously observed that protein expression changes dramatically in ALLs cells with cryopreservation. Present as controls were 16 CD34+ BM and 9 normal PB lymphocyte samples. Samples are printed as 5 serial 1:2 dilutions in duplicate using an Aushon 2470 Arrayer. Each array has a total of 6912 dots printed. Slides were probed with 128 antibodies (ABs) against apoptosis, cell cycle, signaling (STP), regulating proteins, integrins, phosphatases etc including 90 vs. total protein, 33 vs. phos-specific sites and 5 vs. caspase or PARP cleavage sites. Spot intensities were quantified using MicroVigene software. Data was analysed using R, with loading control and topographical background normalization being utilized. Results. Proteins were clustered using absolute Pearson correlation and Ward linkage. Based on the Gap statistic there appeared to be 15 constellations of protein with correlated expression. Many formed expected associations e.g., cleaved caspases, phosphorylated STATs, activated STPs. Based on the combination of expression of these 15 constellations, 7 distinct ProExpSig were defined containing between 18 and 37 cases. Phenotype was highly unbalanced with T cell and Burkitt's cases significantly overrepresented and B cell underrepresented in Signature 3 (O/E 5.7,4.2 and .3) and T cell ALL underrepresented in signatures 2, 4,5 and 6 (O/E .47, .76, .4 and .28). PH+ disease was strongly associated with signatures 5 (18 of 22) and 2 (11 of 375 cases). The PH+ dominated Sig5 was characterized by high expression of Myc, ARC, Survivin as well as high levels of activated STPs and Stats. The Ph+ cases in Sig 2 and 5 differed in that Sig5 had high expression of phospho PKCα, GAB2 and Stat 3 and 6, while Sig 2 had higher expression of proteins in the mTOR and TSC2 axis Almost all t(4:11) cases (9 of 11) were in Sig6, which was characterized by high levels of expression of most proteins excluding those that were highly expressed in the PH+ cases. Cases with Burkitt's were characterized by high expression of cyclins D1 and D3, pRB and by high levels of cleaved caspases. The WBC was markedly higher in Sig 3 and 6. The vast majority n=174) achieved remission with 9 primary refractory and 10 induction deaths occurring, so a signature predicting primary resistance was not observed. It was readily evident that there was a group of favorable signatures (1,2, 4 and 7) and another distinctly unfavorable group of signatures (3, 5, and 6). The favorable group had superior remission duration (median 172 vs. 96 weeks, p= 0.35), event free survival (median 132 vs. 72 weeks, p=0.03) and overall survival (median 180 vs. 93 weeks, P=0.04) compared to the unfavorable signatures. Conclusions. ALL is characterized by 7 distinct protein expression signatures which form a favorable and an unfavorable prognostic group. The adverse ProExpSig are associated with significantly shorter remission duration and survival. Differences within these ProExpSig can be used to direct the rational application of various targeted therapies to those patients most likely to benefit from them thereby improving outcome. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 93 (1) ◽  
pp. 51-54 ◽  
Author(s):  
D.S. Siegel ◽  
K.R. Desikan ◽  
J. Mehta ◽  
S. Singhal ◽  
A. Fassas ◽  
...  

Multiple myeloma (MM) typically afflicts elderly patients with a median age of 65 years. However, while recently shown to provide superior outcome to standard treatment, high-dose therapy (HDT) has usually been limited to patients up to 65 years. Among 550 patients with MM and a minimum follow-up of 18 months, 49 aged ≥65 years were identified (median age, 67; range, 65 to 76 years). Their outcome was compared with 49 younger pair mates (median, 52; range, 37 to 64 years) selected among the remaining 501 younger patients (<65 years) matched for five previously recognized critical prognostic factors (cytogenetics, β2-microglobulin, C-reactive protein, albumin, creatinine). Nearly one half had been treated for more than 1 year with standard therapy and about one third had refractory MM. All patients received high-dose melphalan-based therapy; 76% of the younger and 65% of the older group completed a second transplant (P = .3). Sufficient peripheral blood stem cells to support two HDT cycles (CD34 > 5 × 106/kg) were available in 83% of younger and 73% of older patients (P = .2). After HDT, hematopoietic recovery to critical levels of granulocytes (>500/μL) and of platelets (>50,000/μL) proceeded at comparable rates among younger and older subjects with both first and second HDT. The frequency of extramedullary toxicities was comparable. Treatment-related mortality with the first HDT cycle was 2% in younger and 8% among older subjects, whereas no mortality was encountered with the second transplant procedure. Comparing younger/older subjects, median durations of event-free and overall survival were 2.8/1.5 years (P = .2) and 4.8/3.3 years (P = .4). Multivariate analysis showed pretransplant cytogenetics and β2-microglobulin levels as critical prognostic features for both event-free and overall survival, whereas age was insignificant for both endpoints (P = .2/.8). Thus, age is not a biologically adverse parameter for patients with MM receiving high-dose melphalan-based therapy with peripheral blood stem cell support and, hence, should not constitute an exclusion criterion for participation in what appears to be superior therapy for symptomatic MM.


Sign in / Sign up

Export Citation Format

Share Document