The Utility Of Newer Risk Models In Predicting Outcomes Of Patients (pts) With Higher-Risk (HR) Myelodysplastic Syndromes (MDS) Treated With Azactidine (aza)

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2771-2771
Author(s):  
Amer M. Zeidan ◽  
Najla H Al Ali ◽  
Mohamed A. Kharfan-Dabaja ◽  
Eric Padron ◽  
Ling Zhang ◽  
...  

Abstract Background While aza is the only drug shown to prolong survival in MDS, only half of aza-treated pts achieve objective responses (10-20% complete remission), and 4-6 months might be needed before a response is seen. Therefore the ability to select pts with high and low likelihoods of benefit from aza therapy is a clinical and a research priority. No clinical or laboratory parameter consistently predicts response or survival with aza therapy. A French prognostic scoring system (FPSS) was proposed to predict survival among aza-treated pts with HR-MDS. We sought to compare the relative prognostic discriminatory power of the FPSS with that of the revised IPSS (IPSS-R) and the global MD Anderson prognostic scoring system (MDAPSS) in a large cohort of aza-treated pts with IPSS HR-MDS. Methods The MDS database at Moffitt Cancer Center (MCC) was used to identify patients with HR-MDS (International Prognostic Scoring System [IPSS] intermediate-2 [INT-2] and high-risk) who received aza therapy. Kaplan-Meier curves were used to depict survivals, and the log-rank test was used to compare median overall survival (OS). Akaike information criteria (AIC) were used to assess the relative goodness of fit of the models. Results We identified 259 patients HR-MDS (74.1% with INT-2 and 25.9% with high IPSS) treated with aza at the MCC. The median duration of follow up since diagnosis was 53 months (M) (95% confidence interval [CI], 49-59 M). The median number of aza cycles was 5 (range 1-72), with 75% of pts receiving 4 cycles of therapy or more. The median time from diagnosis to aza initiation was 1.5 M. The median OS for the entire cohort was 19 M (95%CI, 16-22 M), 64% of pts were males, 91% were white, 80% were older than 60 years, and 25% had therapy-related MDS. For the IPSS, the median OS was 23.5 M (CI, 18.4-28.6 M) for INT-2 and 15.9 M (CI, 13.6-18.2 M) for high-risk group (P=0.004). For the FPSS, the median OS was 29.5 M (CI, 14.1-44.9 M) for low risk (LR), 21.1 M (CI, 16.4-26.0 M) for intermediate risk (IR), and 14.1 M (CI, 8.7-19.5 M) for HR (P=0.001). For the MDAPSS the median OS was not reached (NR) for low, 53.5 M (CI, 37.5-69.4 M) for INT-1, 29.5 M (CI, 18.3-40.6 M) for INT-2, and 16.1 M (CI, 14.5-17.7 M) for high risk groups (P<0.0001). For the IPSS-R, the median OS 40.6 M (CI, 22.8-58.4 M) for low, 44.7 M (CI, 25.0-64.4 M) for INT, 23.5 M (CI, 20.3-26.8 M) for high, and 15.2 M (CI, 13.7-16.8 M) for very-high risk groups (P<0.0001). Scores generated using AIC to assess the relative goodness of fit (lower is better) were 1453 (IPSS-R), 1480 (MDAPSS), 1512 (FPSS), and 1522 (IPSS). The IPSS could not refine any of the other models. All three newer models refined IPSS INT-2 but not IPSS high-risk. The IPSS did not refine any of the newer models. The MDAPSS refined the FPSS LR and IR and the IPSS-R high and very-high groups. The FPSS refined IPSS-R very high risk group but not the not the MDAPSS. The R-IPSS did not refine the MDAPSS or the FPSS. Response rates were not statically significantly different within the prognostic groups in any of the scoring systems. Conclusions The IPSS-R, MDAPSS, and the FPSS all functioned well to separate aza-treated pts with IPSS HR-MDS into prognostic groups with different survivals, but the IPSS-R and MDAPSS appear superior to the FPSS. None of the prognostic systems predicted response to aza therapy. HR-MDS patients with poor projected survival with aza therapy might be considered for experimental approaches. Disclosures: Off Label Use: entinostat for MDS. Lancet:Celgene: Research Funding. List:Celgene: Research Funding. Komrokji:Celgene: Research Funding, Speakers Bureau.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4617-4617
Author(s):  
Jun Ho Jang ◽  
Kihyun Kim ◽  
Chul W. Jung ◽  
Keon Woo Park

Abstract Background Based upon the classification of FAB criteria, International Prognostic Scoring System(IPSS) has been a standard prognostic model to predict survival and progression in MDS. In 2000, the WHO has formulated a new classification of myelodysplastic syndrome(MDS). The aim of this study was to evaluate the prognostic value of WHO classification-based prognostic scoring system(WPSS) in MDS. Patients and methods One hundred forty-nine patients who were diagnosed as having de novo MDS at the Division of Hematology-Oncology, Samsung medical center, Seoul, Korea, between Dec. 1994 and Feb. 2007, were evaluated retrospectively for clinical and hamatologic features at diagnosis, transfusion dependence, overall survival(OS), and progression to leukemia(LFS). Risk group stratifications in MDS patients were done according to IPSS and WPSS. Results 18 patients(12.1%), 93 patients (62.4%), 29 patients(29%) and 9 patients(6%) had IPSS risk scores of low, intermediate-1(Int-1), intermediate-2(Int-2) and high, respectively. According to WPSS risk scores, 8 patients(5.4%), 30 patients(20.1%), 41 patients(27.5%), 57 patients(38.3%) and 13 patients(8.7%) were classified to very low, low, intermediate, high and very high risk group, respectively. In IPSS, median OSs of low, Int-1, Int-2 and high subgroup were 65.2, 32.9, 14.3 and 9.1 months respectively (p<0.001). According to WPSS, median OSs of very low, low, intermediate, high and very high risk subgroup were not reached, 55.4, 27.4, 19.0 and 6.2 months respectively (p<0.001). Between subgroups classified according to WPSS, significant differences in OS were noted in low vs. intermediate risk group (p=0.047), in intermediate vs. high risk group (p=0.046) and in high vs. very high risk group(p=0.003) but statistically not significant difference in OS was observed between very low and low risk group (p=0.08). The mean and median OS of the lowest risk group(low risk) in IPSS are 65.33 and 55.43 months, respectively. The mean and median OS of the lowest risk group(very low risk) in WPSS are 102.8 months and not reached, respectively. Conclusion These data show that WPSS with five risk groups might provide more refined prognostic stratifications of MDS than IPSS with four risk groups. Especially, new prognostic system appears to discriminate a subset of patients with very low risk, who could have long term survival.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3340-3340
Author(s):  
Piyanuch Kongtim ◽  
Uday R Popat ◽  
Marcos de Lima ◽  
Guillermo Garcia-Manero ◽  
Elias J. Jabbour ◽  
...  

Abstract MDS is a heterogeneous group of hematopoietic stem cell disorders. Various prognostic models have been established to categorize patients with MDS including the International Prognostic Scoring System (IPSS), the Revised-IPSS (r-IPSS) and MDACC Scoring System. In this analysis, we compared those three classification schemas for their outcome predictability after HSCT. We analyzed 291 MDS patients with a median age of 55 (interquartile range (IQR) 47-60.7 years) who underwent HSCT between January 2001 and December 2011. Histology by WHO classification included RA/RARS 48 (16.5%), RCMD 28 (9.6%), RAEB-1 59 (20.2%), RAEB-2 63 (21.7%), MDS unclassified 67 (23%), and CMML 26 (9%). Of 291, 117 patients (40.2%) had therapy related MDS (t-MDS). Conditioning regimen was myeloablative in 201 patients (69.1%) and reduced intensity in 90 patients (30.9%). Donors were matched related (MRD), matched unrelated (MUD), mismatched (MMD) in 131 (45%), 114 (39.2%) and 46 (15.8%) patients respectively. Risk categorization was performed by IPSS, r-IPSS and MDACC scoring systems at the time of diagnosis. IPSS, r-IPSS and MDACC scoring systems could be assessed in 239 (82.1%), 241 (82.8%) and 231 (79.4%) patients respectively. The median follow up time of 109 survivors was 45 months. The median time from diagnosis to HSCT was 7.3 months (IQR 4.6-12.4 months). Three-year overall survival (OS) was 38.1% (95%CI 32.3-43.9) with 3-year event free survival (EFS) of 34.2% (95%CI 28.4-40). Cumulative relapse incidence (RI) at 3-year was 28.8% (95%CI 23.3-34.5). Cumulative incidence of treatment related mortality (TRM) at 3 year post-transplant was 27.9% (95%CI 22.6-33.6). In univariate analysis, IPSS and r-IPSS were able to differentiate 2 risk groups for OS and EFS. High risk group per IPSS and very high risk group per r-IPSS had lower OS with hazard ratio (HR) of 2.4 to 3.1, lower EFS with HR of 2.2 to 2.7. While IPSS could not predict RI, very high risk group by r-IPSS had higher RI with HR of 3.6 compared with lower risk groups. Both IPSS and r-IPSS did not identify different risk groups for TRM. On the other hand, MDACC scoring system was able to identify 4 different risk groups for EFS and OS in univariate analysis. Three-year OS was 68%, 46.1%, 30.3% and 11.4% for patients with MDACC risk score of 0-4, 5-6, 7-8 and ≥9 respectively (p<0.001) (figure1). Three-year EFS with MDACC risk score of 0-4, 5-6, 7-8 and ≥9 was 61.7%, 40.8%, 28.1% and 7.4% respectively (p<0.001). For RI and TRM, only MDACC risk scores of ≥9 was associated with poor outcomes with 3-year RI of 38.9% and 3-year TRM of 41.7% compared with 13.3% and 15.5% in risk scores of 0-4 (p=0.01 and p=0.01 respectively). In multivariate analysis, MDACC score, matched unrelated and mismatched donors were associated with inferior OS (table1). As a summary, MDACC risk scoring system for MDS better differentiates prognostic groups than IPSS or r-IPSS. Considering the high frequency of t-MDS among transplanted MDS patients, we propose that MDACC scoring system should be used for prognostic classification for hematopoietic transplantation. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e20647-e20647
Author(s):  
Martina Torchio ◽  
Benvenuto Franceschetti ◽  
Carla Cavali ◽  
Sonia Zanirato ◽  
Angelo Olgiati ◽  
...  

e20647 Background: Venous thromboembolism (VTE), is a negative predictor of survival in pts with advanced cancer. International guidelines don’t recommend routine prophlaxis but suggest to consider pts, undergoing chemotherapy (CT), with high risk of VTE. Many clinical risk factors for cancer-associated VTE have been evaluated in a 5 parameter-based (body mass index, platelet and leucocyte counts, hemoglobin value and tumor site) scoring system, the Khorana score, utilized to indicate a prophylactic approach. We prospectively applied this score in cancer outpts beginning CT and an implementation based on 6 addictional factors analysis (sex, age, central venous catheter, CT-agents, antiangiogenetic drugs, erithropoiesis stimulating agent) to evaluate their impact in pts assignment into risk groups. Methods: We studied adult pts, followed at our Department from August 2011 to December 2012, with advanced cancers (breast, NSCLC, colorectal, pancreatic/gastric, urogenital, LNH, Hodgkin's disease, HD, and MM), receiving a first or second line standard CT. We stratified pts into three risk groups (score 0= low; score 1-2=intermediate; score 3-4-5=high) considering both the Khorana scoring system and its implementation. Results: We analyzed 169 pts (103F/66M, median age 62.3, range 35-80 yrs), pt population included: 38 breast, 32 colorectal, 31 LNH, HD and MM, 27 urogenital, 22 NSCLC and 19 pancreatic/gastric. With the Khorana score 49 pts were assigned to the low risk, 87 pts to the intermediate risk (57 with score=1, 28 with score=2), 16 pts (9.4%) to the high risk group (9 with score=3, 4 with score=4, 3 with score=5). When we considered 11 parameters 37 pts (21.8%) were assigned to the high risk group. Conclusions: A more comprehensive quantification of VTE risk, also considering new independent factors, is mandatory for a correct decision making of an antithrombotic-prophylactic approach.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2776-2776
Author(s):  
Andrea Kuendgen ◽  
Corinna Strupp ◽  
Kathrin Nachtkamp ◽  
Barbara Hildebrandt ◽  
Rainer Haas ◽  
...  

Abstract Abstract 2776 Poster Board II-752 Introduction: We wondered whether prognostic factors have similar relevance in different subpopulations of MDS patients. Methods: Our analysis was based on patients with primary, untreated MDS, including 181 RA, 169 RARS, 649 RCMD, 322 RSCMD, 79 5q-syndromes, 290 RAEB I, 324 RAEB II, 266 CMML I, 64 CMML II, and 209 RAEB-T. The impact of prognostic variables in univariate analysis was compared in subpopulations of patients defined by medullary blast count, namely <5%, ≥5% (table), ≥10%, and ≥20% (not shown), as well as 3 subpopulations defined by the cytogenetic risk groups according to IPSS (table). Multivariate analysis of prognostic factors was performed for cytogenetically defined subgroups and WHO-subtypes. Results: Strong prognostic factors in all blast-defined subgroups were hemoglobin, transfusion dependency, increased WBC, age, and LDH. However, all variables became less important in patients with ≥20% blasts (RAEB-T) and increased WBC was rare. Platelet count and cytogenetic risk groups were relevant in patients with <5%, ≥5%, and ≥10% marrow blasts, but not in RAEB-T. Marrow fibrosis was important in patients with <5% or ≥5% blasts, but not ≥10%. Gender and ANC <1000/μl were significant only in patients with a normal blast count. Furthermore, we looked for the effect of the karyotypes, relevant for IPSS scoring (-Y, del5q, del20q, others, del7q/-7, complex), and found a comparable influence on survival, irrespective whether patients had < or ≥5% marrow blasts. In subpopulations defined by cytogenetic risk groups, several prognostic factors were highly significant in univariate analysis, if patients had a good risk karyotype. These included hemoglobin, sex, age, LDH, increased WBC, transfusion need, and blast count (cut-offs 5%, 10%, and 20%). In the intermediate risk group only LDH, platelets, WBC, and blasts were significant prognostic factors, while in the high risk group only platelets and blast count remained significant. Multivariate analysis was performed for the cytogenetic risk groups and for subgroups defined by WHO subtypes. The analysis considered blast count (</≥5%), hemoglobin, platelets, ANC, cytogenetic risk group, transfusion need, sex, and age. In the subgroup including RA, RARS, and 5q-syndrome, LDH, transfusion, and age in descending order were independent prognostic parameters. In the RCMD+RSCMD group, karyotype, age, transfusion, and platelets were relevant factors. In the RAEB I+II subgroup, the order was hemoglobin, karyotype, age, and platelets, while in CMML I+II only hemoglobin had independent influence. In RAEB-T none of the factors examined was of independent significance. Looking at cytogenetic risk groups, in the favorable group, several variables independently influenced survival, namely transfusion, blasts, age, sex, and LDH (in this order). Interestingly, in the intermediate and high risk group, only blast count and platelets retained a significant impact. Conclusion: Univariate analysis showed prognostic factors (except ANC) included in IPSS and WPSS are relevant in most subgroups defined by marrow blast percentage. However, they all lose their impact if the blast count exceeds 20%. Regarding cytogenetic risk groups, several prognostic factors lose their influence already in the intermediate risk group. This underscores the prognostic importance of MDS cytogenetics. Multivariate analysis showed MDS subpopulations defined by WHO types also differ with regard to prognostic factors. In particular, CMML and RAEB-T stand out against the other MDS types. Disclosures: Kuendgen: Celgene: Honoraria. Hildebrandt:Celgene: Research Funding. Gattermann:Novartis: Honoraria, Participation in Advisory Boards on deferasirox clinical trials. Germing:Novartis, Celgene: Honoraria, Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2784-2784 ◽  
Author(s):  
Julie Schanz ◽  
Friederike Braulke ◽  
Katayoon Shirneshan ◽  
Kathrin Nachtkamp ◽  
Ulrich Germing ◽  
...  

Abstract Introduction Total (-7) or partial (7q-) monosomy 7 is frequent in malignant myeloid disorders, observed in around 12% of MDS/AML and up to 40% of therapy-associated MDS/AML. Monosomy 7 is associated with poor outcome, high susceptibility to infections and poor response to chemotherapy. A therapeutic benefit for 5-azacytidine was previously described (Fenaux et al., 2009). The present study was designed to analyze clinical features, prognosis and response to different therapeutic strategies in patients with monosomy 7 in a multicentric, retrospective German cohort study. Patients and methods Currently, 231 patients with MDS/AML following MDS and monosomy 7 were included. Inclusion criteria were defined as follows: Morphologic diagnosis of MDS/AML following MDS, age ≥18 years, bone marrow blast count ≤30% and presence of -7 or 7q-. The data was assembled from centers in Düsseldorf, (n=120; 52%), Cologne (n=38; 17%), Freiburg (n=31; 13%), Göttingen (n=14; 6%), Munich (n=13; 6%), Dresden (n=11; 5%) and Mannheim (n=4; 2%). The median age in the study cohort was 67 years, 65% of patients were males. 29/231 patients (13%) were diagnosed as AML following MDS. MDS/AML was therapy-associated in 24 patients (11%). Regarding IPSS, 38 (19%) were classified as low/intermediate 1 risk and 165 (81%) as intermediate-2/high-risk. According to IPSS-R, 2 (1%) were assigned to the very-low/low risk group, 31 (16%) to the intermediate group, 52 (27%) to the high-risk group and 107 (56%) to the very high risk group. The treatment was classified as follows: Best supportive care (BSC), low-dose Chemotherapy (LDC), high-dose chemotherapy (HDC), demethylating agents (DMA; either 5-azacytidine or decitabine), and others. Results A best supportive care regimen was chosen in nearly half of the patients (49%). The remaining patients received 1-4 sequential therapies (1: 29%; 2: 11%; 3: 10%; 4: 1%). As the first line therapy, 64 patients (54%) received DMA, 24 (20%) an allo-Tx, 9 (8%) HDC, 5 (4%) LDC, and 16 (14%) were treated with other therapies. The best prognosis was observed in patients eligible for allo-Tx: The median OS in transplanted patients was 924 days as compared to 361 days (p<0.01) in patients not eligible for transplantation. In the latter cohort, patients who received DMA at any course of their disease did not differ from those receiving other therapies: The median OS was 468 days in patients treated with DMA as compared to 325 on those with alternative therapies (p not significant) and the median time to AML-transformation was 580 versus 818 days (p not significant), respectively. However, by classifying patients according to IPSS- and IPSS-R, it became obvious that patients with an IPSS high-risk or an IPSS-R very high risk showed a clear benefit from DMA: In the first group, median OS was 444 days in DMA-treated and 201 days in non-DMA-treated patients (p=0.048), in the latter group, median OS 444 days in the DMA-treated and 203 days in the non-DMA treated cohort (p=0.017). Comparable results were observed regarding AML-free survival: Median time to AML was 580 (DMA) vs. 186 (no DMA) days in IPSS high risk patients (p=0.031) and 580 (DMA) vs. 273 (no DMA) days in the IPSS-R very high risk group (p not significant). Conclusions Patients with MDS, partial or total monosomy 7 and a high risk according to IPSS or a very high risk according to IPSS-R show a pronounced benefit when treated with DMA, regarding overall- as well as AML-free survival. Further results from the ongoing data analysis will be presented in detail. The study was supported by research funding from Celgene. Disclosures: Schanz: Celgene: Research Funding. Braulke:Celgene: Research Funding. Germing:Celgene: Honoraria, Research Funding. Schmitz:Novartis: Research Funding; Celegene: Consultancy, Research Funding, Speakers Bureau. Götze:Celgene: Honoraria. Platzbecker:Celgene: Honoraria, Research Funding. Haase:Celgene: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1419-1419 ◽  
Author(s):  
Susan L Heatley ◽  
Teresa Sadras ◽  
Eva Nievergall ◽  
Chung Hoow Kok ◽  
Phuong Dang ◽  
...  

Abstract Introduction: While remission rates for childhood acute lymphoblastic leukemia (ALL) now exceed 80%, relapsed ALL remains the leading cause of non-traumatic death in children. Recently, a high-risk group of B-progenitor ALL patients has been identified. Such cases exhibit a gene expression profile similar to that of BCR-ABL1 positive (Ph+) ALL but are BCR-ABL1 negative, and also experience poor treatment outcomes. This subset, termed Ph-like ALL, is characterised by a range of genetic alterations that activate cytokine receptor and kinase signalling, allowing potential targeting by available tyrosine kinase inhibitors (TKI). The frequency of Ph-like ALL in the Australian community and the prognosis in the setting of the first MRD (minimal residual disease) intervention trial by the Australian and New Zealand Children's Haematology/Oncology Group (ANZCHOG ALL8) is unknown. Method: We retrospectively screened 250 unselected samples that were available from children diagnosed with B-ALL, for Ph-like ALL. The children, aged between 1 and 18 years, were enrolled on the ANZCHOG ALL8 trial and recruited from 2002-2011. The criteria for stratification to the high-risk group, based upon Berlin-Frankfurt-Munster (BFM) protocols, were BCR-ABL1 or MLL t(4;11) translocation; poor prednisolone response at day 8; failure to achieve remission by day 33 or high MRD (>5 x10-4) at day 79. MRD was measured by RQ-PCR for patient-specific immunoglobulin and T-cell receptor rearrangements. All patients received a standard BFM four drug induction chemotherapy regimen including a prednisolone pre-phase and intrathecal methotrexate. High-risk patients received a further three novel intensive blocks of chemotherapy followed by transplant in most cases. Patients were screened for Ph-like ALL using a custom Taqman Low Density Array (TLDA) based upon previous reports. Fusions were then confirmed by RT-PCR for 30 known fusions, Sanger sequencing, mRNA sequencing and/or FISH. Results: Ten percent (25/250) of children in this cohort were identified as having Ph-like ALL, with most fusions converging on kinase activating pathways (Table 1). Three Ph-like ALL patients were considered high-risk, the remaining 22 (88%) were medium risk. Five children with Ph-like ALL, that did not have a fusion identified by RT-PCR, are currently under further investigation. Furthermore, 15 of the 20 (75%) of rearrangements involved CRLF2 with 10 (66%) of these children relapsing. Strikingly, 56% (14/25) of children in the ALL8 cohort who were identified as Ph-like subsequently relapsed compared to 16% (36/225) who were not, with significantly worse event free survival (p<0.0001) (Figure 1). Conclusion: Here we demonstrate a significantly higher frequency of relapse amongst Australian children with Ph-like ALL compared to non Ph-like disease despite a MRD-adjusted intensification regimen. In this cohort, these children should be classified as high-risk due to high treatment failure rates with standard/medium risk regimens. Importantly, rapid identification of these patients may guide future intervention with targeted therapies, such as TKI, matched to the causative genetic lesion in this high-risk group. Figure 1. Fusions identified in Ph-like ALL from ANZCHOG ALL8 cohort. Figure 1. Fusions identified in Ph-like ALL from ANZCHOG ALL8 cohort. Figure 2. Kaplan-Meier estimates of event free survival for patients with Ph-like ALL and non Ph-like ALL (all risk groups). Figure 2. Kaplan-Meier estimates of event free survival for patients with Ph-like ALL and non Ph-like ALL (all risk groups). Disclosures Hughes: ARIAD: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Mullighan:Incyte: Consultancy, Honoraria; Cancer Science Institute: Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Speakers Bureau; Loxo Oncology: Research Funding. White:Novartis: Honoraria, Research Funding; BMS: Honoraria, Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2970-2970 ◽  
Author(s):  
Martin van Vliet ◽  
Joske Ubels ◽  
Leonie de Best ◽  
Erik van Beers ◽  
Pieter Sonneveld

Abstract Introduction Multiple Myeloma (MM) is a heterogeneous disease with a strong need for robust markers for prognosis. Frequently occurring chromosomal abnormalities, such as t(4;14), gain(1q), and del(17p) etc. have some prognostic power, but lack robustness across different cohorts. Alternatively, gene expression profiling (GEP) studies have developed specific high risk signatures such as the SKY92 (EMC92, Kuiper et al. Leukemia 2012), which has shown to be a robust prognostic factor across five different clinical datasets. Moreover, studies comparing prognostic markers have indicated that the SKY92 signature outperforms all other markers for identifying high risk patients, both in single and multivariate analyses. Similarly, when assessing the prognostic value of combinations of various prognostic markers, the SKY92 combined with ISS was the top performer, and also enables detection of a low risk group (Kuiper et al. ASH 2014). Here, we present a further validation of the low and high risk groups identified by the SKY92 signature in combination with ISS on two additional cohorts of patients with diverse treatment backgrounds, containing newly diagnosed, previously treated, and relapsed/refractory MM patients. Materials and Methods The SKY92 signature was applied to two independent datasets. Firstly, the dataset from the Total Therapy 6 (TT6) trial, which is a phase 2 trial for symptomatic MM patients who have received 1 or more prior lines of treatment. The TT6 treatment regime consists of VTD-PACE induction, double transplant with Melphalan + VRD-PACE, followed by alternating VRD/VMD maintenance. Affymetrix HG-U133 Plus 2.0 chips were performed at baseline for n=55 patients, and OS was made available previously (Gene Expression Omnibus identifier: GSE57317). However, ISS was not available for this dataset. Secondly, a dataset of patients enrolled at two hospitals in the Czech Republic, and one in Slovakia (Kryukov et al. Leuk&Lymph 2013). Patients of all ages, and from first line up to seventh line of treatment were included (treatments incl Bort, Len, Dex). For n=73 patients Affymetrix Human Gene ST 1.0 array, OS (n=66), and ISS (n=58) was made available previously (ArrayExpress accession number: E-MTAB-1038). Both datasets were processed from .CEL files by MAS5 (TT6), and RMA (Czech), followed by mean variance normalization per probeset across the patients. The SKY92 was applied as previously described (Kuiper et al. Leukemia 2012), and identifies a High Risk and Standard Risk group. In conjunction with ISS, the SKY92 Standard Risk group is then further stratified into low and intermediate risk groups (Kuiper et al. ASH 2014). Kaplan-Meier plots were created, and the Cox proportional hazards model was used to calculate Hazard Ratios (HR), and associated 1-sided p-values that assess whether the SKY92 High Risk group has worse survival than SKY92 Standard Risk group (i.e. HR>1). Results Figure 1 shows the Kaplan Meier plots of the SKY92 High Risk and Standard Risk groups on the TT6 and Czech cohorts. On the TT6 dataset, the SKY92 signature identifies 11 out of 55 patients (20%) as High Risk. In both datasets, the SKY92 High Risk group has significantly worse overall survival, HR=10.3, p=7.4 * 10-6 (TT6), and HR=2.6, p=2.2 * 10-2 (Czech). In addition, the combination of SKY92 with ISS on the Czech dataset identifies a low risk group of 14 out of 61 patients (23%), with a five year overall survival estimate of 100% versus 28.7% in the SKY92 High Risk group (HR=inf). Robustness of the SKY92 signature is further demonstrated by the fact that it validates on both datasets, despite different microarray platforms being used. Conclusions The SKY92 high risk signature has been successfully validated on two independent datasets generated using different microarray platforms. In addition, on the Czech data, the low risk group (SKY92 Standard Risk combined with ISS 1) has been successfully validated. Together, this signifies the robust nature of the SKY92 signature for high and low risk prediction, across treatments, and with applicability in newly diagnosed, treated, and relapsed/refractory MM patients. Figure 1. Kaplan-Meier plots showing a significantly poorer overall survival in patients identified as SKY92 High Risk (red curves), relative to SKY92 Standard Risk, on both the TT6 (left), and Czech (middle) datasets, as well as a low risk group by SKY92 & ISS1 on the Czech dataset (green curve, right). Figure 1. Kaplan-Meier plots showing a significantly poorer overall survival in patients identified as SKY92 High Risk (red curves), relative to SKY92 Standard Risk, on both the TT6 (left), and Czech (middle) datasets, as well as a low risk group by SKY92 & ISS1 on the Czech dataset (green curve, right). Disclosures van Vliet: SkylineDx: Employment. Ubels:SkylineDx: Employment. de Best:SkylineDx: Employment. van Beers:SkylineDx: Employment. Sonneveld:Celgene: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Karyopharm: Research Funding; SkylineDx: Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4656-4656 ◽  
Author(s):  
Julie Schanz ◽  
Friederike Braulke ◽  
Ghulam J. Mufti ◽  
Elena Crisà ◽  
Austin Kulasekararaj ◽  
...  

Abstract Introduction: Total (-7) or partial (7q-) monosomy 7 is the second frequent abnormality in MDS, occurring in around 12% of MDS/AML and up to 40% of therapy-associated MDS/AML. The present study was designed to analyze clinical features, prognosis and response to different therapeutic strategies in patients with -7 or del(7q) in a multicentric, retrospective cohort study. Patients and Methods: 471 patients with MDS/AML following MDS and monosomy 7 were registered and retrospectively analyzed. The median observation time was 3.6 years. Inclusion criteria were defined as follows: Morphologic diagnosis of MDS/AML following MDS, bone marrow blast count <=30% and presence of -7 or 7q- proved by chromosome banding analysis (CBA) or fluorescence in situ-hybridization (FISH). The data was coalesced from 8 centers in London (n=140; 29.7%), Duesseldorf, (n=120; 25.5%%), Goettingen (n=118; 25.1%), Cologne (n=38; 8.1%), Freiburg (n=29; 6.2%), Munich (n=13; 2.8%), Dresden (n=10; 2.1%) and Mannheim (n=3; 0.6%). The median age in the study cohort was 66 years, 63% of patients were males. MDS/AML was therapy-associated in 53 (11%). According to IPSS-R, 9 (1.9%) were assigned to the low risk group, 39 (8.3%) to the intermediate group, 81 (17.2%) to the high-risk group and 133 (28.2%) to the very high risk group. The treatment was classified as follows: Best supportive care (BSC), low-dose Chemotherapy (LDC), high-dose chemotherapy (HDC), hypomethylating agents (HMA; either 5-azacytidine or decitabine), and others (e.g. valproic acid, steroids, lenalidomide or thalidomide). Survival analyses were performed regarding overall- (OS) as well as AML-free survival (AFS) using the Kaplan-Meier method. Results: 147 patients (31.2%) showed 7q-, 313 (66.5%) -7 and 11 (2.3) patients showed both abnormalities at the first cytogenetic examination. The abnormality was detected by CBA±FISH in 440 (93.4%) and by FISH only in 31 (6.6%). In the latter cases, the CBA was either unsuccessful or showed a normal karyotype. In 182 (38.6%) patients, -7/del7q was detected as a single abnormality, 77 (16.3%) showed two abnormalities and 184 (39.1%) showed a complex karyotype involving -7/7q-. As previously described (Schanz et al., 2012), untreated patients with an isolated 7q- as compared to an isolated -7 show a better prognosis regarding OS (median: 4.0 vs. 0.7 years; p<0.01) as well as AFS (median not reached vs. 2.3 years; p=0.062). Median hemoglobin level in the study cohort was 9.3 g/dl, ANC 0.98*103/μl, platelet count 73*103/μl and the median number of bone marrow blasts was 8%. Regarding the treatment, a best supportive care regimen was chosen in 195 (41%) patients. The remaining 276 (58.6) patients received 1-5 sequential therapies (one therapy: 31.6%; more than 1 therapy: 27.0%). 81 patients received an allogeneic bone marrow transplantation (ATX). Within the group of patients treated with HMA at any time of their disease (n=167), 147 (31.2%) received 5-Azacytidine, 8 (1.7%) Decitabine and 12 (2.5%) patients were treated with both drugs. As the first line therapy, 122 patients (25.9%) received HMA, 50 (10.6%) HDC, 28 (5.9%) ATX, 28 (5.9%) 11 (2.3%) LDC, and 28 (5.9%) were treated with other therapies. Patients eligible for ATX showed a significantly better prognosis as compared to any other therapy strategy: The median OS in was 2.1 years as compared to 1.1 years in non-transplanted patients (p<0.01). In patients not eligible for ATX, treatment with HMA at any course of their disease as compared to a BSC strategy was associated with a better OS (1.4 vs. 0.8 years, p=0.014). By comparing HMA to any other therapy, the OS did not differ significantly (1.4 years in HMA vs. 1.1 years in any other, p n.s.). In patients classified as very high risk according to IPSS-R, the median OS was significantly prolonged in patients receiving HMA as compared to BSC (1.1 vs. 0.6 years, p<0.01). This was also observed for the risk of AML-transformation in this subgroup of patients: The median time to AML was 1.8 years in HMA-treated patients versus 0.6 years in BSC (p=0.012). Conclusions: To our knowledge, the study describes the largest patient cohort with MDS/AML and monosomy 7 published to date. Further data regarding the clinical characteristics of this subgroup of patients and the treatment regimes applied will be presented in detail. The study was supported by research funding from Celgene Figure 1 Figure 1. Figure 2 Figure 2. Disclosures Schanz: Celgene: Honoraria, Research Funding, Travel grants: Celgene, Novartis, Lilly Other. Götze:Celgene Corp, Novartis Pharma: Honoraria. Nolte:Celgene, Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Travel grants Other.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 144-144 ◽  
Author(s):  
Ang Li ◽  
Qian V. Wu ◽  
Greg Warnick ◽  
Neil A Zakai ◽  
Edward N. Libby ◽  
...  

Abstract Introduction: Patients with newly diagnosed multiple myeloma (MM) have high risk of venous thromboembolism (VTE) when starting initial treatment that contains immunomodulatory drugs (IMID) such as lenalidomide or thalidomide. The National Comprehensive Cancer Network (NCCN) guideline recommends primary anticoagulant thromboprophylaxis for the high-risk patients. However, it is challenging to risk-stratify patients without a validated risk model. We have conducted a retrospective cohort study using the SEER-Medicare (Surveillance, Epidemiology, and End Results) database to derive a new VTE risk assessment model. Methods: We selected all patients 66 or older with newly diagnosed MM 2007 to 2013. Patients were included if they had a prescription of IMID within twelve months of diagnosis and complete enrollment for fee-for-service and prescription drug coverage. We ascertained baseline demographics and VTE risk factors from the current NCCN guideline using validated codes. The VTE outcome was defined as either one inpatient or two outpatient claims at least 30 days apart in combination with an anticoagulant prescription within 90 days. All patients were followed from the date of IMID initiation until first VTE occurrence or death and were censored for disenrollment from Medicare, discontinuation of IMID (after a grace period of 90 days), autologous transplantation, or the end of claims data (12/31/2014). Cause specific Cox regression models were used for time to VTE analysis. For variable selection, all risk factors with p-value <0.10 were considered candidates for inclusion in the final multivariable regression model. VTE history, recent surgery, and anticoagulant exposure were forced into the model, regardless of significance testing. Integer points were assigned according to the beta coefficients and subsequent risk groups were created. The model's discrimination was validated internally by the bias-corrected Harrell's c statistic and the 95% confidence interval was estimated from 200 bootstrap samples. Results: We identified 2397 MM patients on IMID that met the study criteria. The median time on IMID treatment was 116 days (IQR 28-279). The mean age of patients was 74, 49% were female, 80% were White, 13% were Black, 6.5% were Asian. Only 13% of patients had concurrent anticoagulant exposure (11% warfarin, 2% LMWH, 1% DOAC) with a median duration of 116 days (IQR 42-315 days). In the multivariable model built from candidate covariates, we identified history of VTE, recent surgery, cytotoxic (non-bortezomib) chemotherapy, higher dose dexamethasone, older age, and Black race, as important risk factors. Asian race and LMWH/DOAC use were associated with lower VTE risk (Table 1). We derived a risk assessment model that stratified patients into 2 prognostic risk groups (Table 1): 25% (n=581) in the very high-risk group (score 2 to 7), 75% (n=1816) in the standard-risk group (score -3 to 1). The incidence of VTE at 3 months and 6 months were 9.5% and 16.3% in the very high-risk group compared to 3.7% and 6.3% in the standard-risk group with a resulting hazard ratio of 2.73 (p<0.001) (Figure 1). The bias-corrected Harrell's c statistic for the product index was 0.63 (0.59-0.68). Conclusions: We have derived a VTE risk assessment model specifically for patients with MM starting IMID therapy. The HAS-RiSC score combines 7 clinical risk factors - History of VTE, Age 80+, Surgery within last 90 days, Race Black, race Asian, Steroid use, and Chemotherapy - into a simplified VTE risk assessment model that identifies a subgroup of patients at very high risk for VTE. External validation of this risk assessment model is currently in progress. Disclosures Garcia: Daiichi Sankyo: Research Funding; Incyte: Research Funding; Janssen: Consultancy, Research Funding; Pfizer: Consultancy; Retham Technologies LLC: Consultancy; Shingoi: Consultancy; Portola: Research Funding; Bristol Meyers Squibb: Consultancy; Boehringer Ingelheim: Consultancy. Lyman:Amgen: Other: Research support; Generex Biotechnology: Membership on an entity's Board of Directors or advisory committees; Halozyme; G1 Therapeutics; Coherus Biosciences: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2141-2141
Author(s):  
Piyanuch Kongtim ◽  
Simrit Parmar ◽  
Denái R. Milton ◽  
Jorge M. Ramos Perez ◽  
Gabriela Rondon ◽  
...  

Abstract Introduction Outcomes after allogeneic stem cell transplant (AHSCT) are influenced by both disease and patient related factors. We hypothesized that combining hematopoietic stem cell transplant comorbidity-age index (HCT-CI/Age) and the refined disease risk index (DRI-R) would better predict survival post-transplant and developed a hematopoietic cell Transplant-composite risk (HCT-CR) model, which we tested in a group of patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) treated at MD Anderson Cancer Center (MDACC). Methods The study included consecutively treated patients, 18 years of age or older, with AML and MDS who received first AHSCT at MDACC between 2005-2016. Donors were HLA-matched related (MRD), HLA-matched unrelated (MUD), 9/10 MUD (MMUD), haploidentical (HAPLO) and 9/10 MRD (MMRD). To develop this model, patients were assigned into 4 groups: 1. Patients with low/intermediate DRI-R and HCT-CI/Age </=3 (low-risk); 2. Patients with low/intermediate DRI-R and HCT-CI/Age >3 (intermediate-risk); 3. Patients with high/very high DRI-R and HCT-CI/Age </=3 (high-risk); and 4. Patients with high/very high DRI-R and HCT-CI/Age >3 (very high-risk). Primary endpoint was 5-year overall survival (OS); other outcomes assess were progression-free survival (PFS), non-relapse mortality (NRM) and relapse rate. The stability of the HCT-CR model was tested by bootstrap resampling. The discrimination power of the HCT-CR model on OS was compared with that of the DRI-R, HCT-CI/Age and cytogenetic risk model by the Harrell C-concordance index. Results The analysis included 942 patients (492 male and 450 female) with a median age of 53 years (range 18-65 years). Cytogenetic data at diagnosis was available in 928 (98.5%) patients and was favorable, intermediate and adverse cytogenetic risk in 63 (7%), 523 (56%) and 342 (37%), respectively. Fifty-five (6%), 399 (43%), 392 (42%) and 82 (9%) patients had low, intermediate, high and very high DRI-R, respectively. The HCT-CI/Age was available in 922 (98%) patients with the median score of 3 (range 0-18). Donor types included MRD (n=377), MUD (n=416), MMUD (n=68), HAPLO (n=73) and MMRD (N=8). Using the HCT-CR model, patients were stratified into 4 risk groups: low (N=272), intermediate (N=168), high (N=284) and very high-risk (N=184), with significantly different survival. The 5-year OS rates for patients in low, intermediate, high and very high-risk group were 57%, 48%, 34%, and 26%, respectively (P<0.001) (Figure 1). The probability of 5-year PFS rates were 55%, 46%, 30% and 23% for these 4 risk groups, respectively (P<0.001). Post-transplant outcomes of all 4 HCR-CR groups are summarized in Table 1. Compared with the low HCT-CR risk group, patients with intermediate, high and very high-risk group had a significantly increased risk of death with HR of 1.42 (95%CI 1.06-1.91; P=0.02), 2.11 (95%CI 1.65-2.70; P<0.001), and 3.02 (95%CI 2.32-3.92; P<0.001), respectively. The significant association between OS and the HCT-CR groups persisted after adjusting for potential confounders with adjusted HR of 1.37 (95%CI 1.02-1.85) for intermediate, 2.08 (95%CI 1.62-2.67) for high and 2.92 (95%CI 2.23-3.82) for very high-risk group when compared with low risk group. The stability of the hematopoietic cell transplant - composite risk model was confirmed in a bootstrap resampling procedure. Among 500 new datasets, on average, patients in intermediate, high and very high-risk group had significantly increased risk of death after transplant when compared with low risk group with HR of 1.39, 2.11 and 2.98, respectively. Results from the concordance test showed that the HCT-CR model provided better discriminative capacity for prediction of OS when compared with DRI-R, HCT-CI/Age and cytogenetic risk group models with C-index of 0.62 versus 0.60, 0.54 and 0.55, respectively. The goodness of fit test showed that the HCT-CR model fit the data significantly better than the other models (P<0.001). Conclusion Combining disease and patient-related factors provides better survival stratification for patients with AML/MDS receiving AHSCT. This novel HCT-CR model will be validated in patients with all diseases undergoing allogeneic hematopoietic stem cell transplantation and results will be presented at the meeting. Disclosures Oran: ASTEX: Research Funding; Celgene: Consultancy, Research Funding; AROG pharmaceuticals: Research Funding. Champlin:Otsuka: Research Funding; Sanofi: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document