Trends in All-Cause Mortality Among Older Patients with CML: A SEER Database Analysis

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3772-3772
Author(s):  
Andrew M. Brunner ◽  
Federico Campigotto ◽  
Benjamin J. Drapkin ◽  
Hossein Sadrzadeh ◽  
Donna S. Neuberg ◽  
...  

Abstract Abstract 3772 Introduction: Clinical outcomes for patients with chronic myeloid leukemia (CML) have dramatically improved over the course of the past ten years, following the advent of tyrosine kinase inhibitors (TKIs) that target BCR/ABL. Nonetheless, survival differences persist between age groups. Prior analyses suggested that this difference occurs in part due to treatment variation; in Sweden, patients older than age 79 had poorer relative survival, and more typically were treated with hydroxyurea rather than a TKI [Bjorkholm, J Clin Oncol 29:2514]. Other studies have noted gains made in relative survival up to the year 2004 [Brenner, Haematologica93:1544], but longer-term overall survival following the widespread use of TKI therapy is less well described among older patients in the U.S. We performed an epidemiologic study of patients registered in the Surveillance, Epidemiology, and End Results (SEER) database to estimate the 5 year overall survival (OS) of patients treated for CML in the era of TKI therapy to assess for differences in survival outcomes among patients within different age groups. Methods: Patients with a diagnosis of CML were identified using the SEER 19 registries database [www.seer.cancer.gov, 1973–2009, November 2011 submission]. We included patients with a diagnosis code of CML NOS (Code 9863) and BCR/ABL+ CML (Code 9875) diagnosed between January 2000 and December 2005. This interval brackets the FDA approval of imatinibin 2001, and its incorporation into NCCN guidelines in 2003. To reflect the evolution of CML treatment during this interval, we trended 5-year overall survival by the year of initial diagnosis. To evaluate the effect of age on survival, patients were divided into cohorts based on age at diagnosis: 15–44 years old, 45–64 years old, 65–74 years old, and 75–84 years old. Overall survival was estimated using the method of Kaplan and Meier. Cox proportional hazards regression was used to model OS to estimate the effects of year of diagnosis within each age group. All analyses were performed using SAS statistical software. Results: We identified 5,138 patients registered in the SEER database with a new diagnosis of CML between January 2000 and December 2005. The patients were 57.6% male; this was the first recorded primary malignancy for 88.4% of the cohort. The 5-year OS improved among patients in every age group between the years 2000 and 2005 (Table 1, Figure 1). Compared to patients diagnosed in the year 2000, patients between the ages of 15 and 44 years had the greatest improvement in 5 year OS (Figure 1; hazard ratio (HR) for dying 0.427, 95% CI: [0.278;0.655], P<0.0001). Patients between ages 75 and 84 also had significant survival gains; the OS estimate at 5 years increased from 19.2% in 2000 to 36.4% in 2005 (HR for dying 0.571, 95% CI: [0.443;0.736], P<0.0001). Discussion: Tyrosine kinase inhibitors targeting BCR/ABL have been FDA-approved for the treatment of CML since 2001 and are highly effective therapy for this disease. Since their advent, patient survival has improved among all age groups; intriguingly, this is also seen among older patients. Indeed, we found marked improvements in OS at 5 years among patients between the ages of 75 and 84, a group which historically has had very poor outcomes. Our data suggests that the advent of BCR/ABL tyrosine kinaseinhibitors has had a significant impact on the outcomes of older patients with CML, likely by providing them with tolerable and effective treatment options not previously available. Further study is needed to determine specific factors that contribute to this improvement in survival. In the future, older age groups are likely to experience ongoing benefit from novel and effective therapeutics with tolerable side effect profiles. Disclosures: Fathi: Teva Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Genzyme: Membership on an entity's Board of Directors or advisory committees.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 38-38 ◽  
Author(s):  
David Samuel diCapua Siegel ◽  
Susanna Jacobus ◽  
S. Vincent Rajkumar ◽  
Rafat Abonour ◽  
Natalie Scott Callander ◽  
...  

Abstract Abstract 38 INTRODUCTION: Lenalidomide and bortezomib have moved into the management of newly diagnosed multiple myeloma leading to dramatically improved outcomes. As a consequence, the role of upfront autologous peripheral blood stem cell transplant (ASCT) has become more controversial. The ECOG E4A03 clinical trial randomized newly diagnosed MM patients to lenalidomide with high-dose dexamethasone (LD) vs lenalidomide with low-dose dexamethasone (Ld) (Rajkumar et al Lancet Oncol 2010; 11: 29–37). Upon completing four cycles of therapy, pts had the option of ASCT or continuing on the assigned therapy. The purpose of this abstract is to determine the outcome of patients on this trial pursuing early ASCT according to various age-groups. MATERIALS and METHODS: This is a post hoc, retrospective analysis of overall survival within age subgroups stratified by early ASCT status. This is a landmark analysis including only pts surviving the first 4 cycles of therapy. RESULTS: In all three age-groups studied, 1, 2, and 3-year survival probability estimates with ASCT were excellent (Tables 1, 2, and 3). For patients under the age of 65 who survived the first 4 cycles of therapy, overall survival at 3-years was 94% with early ASCT, 78% in pts continuing protocol therapy. Although direct comparison with patients not going to early transplant is not possible because the assignment to early ASCT versus no early ASCT was not randomized, survival with ASCT at 3-years appeared higher. While we attempt to correct for age, the differences may be influenced by other factors such as performance status, comorbidities, response to therapy, etc. The presumption that treatment related mortality (TRM) should be more problematic for older pts undergoing ASCT is addressed by looking at the >65 and >70yo cohorts. In the >65 age group, one-year mortality is similar between the early ASCT population and the no early ASCT population. In the >70 age group, no adverse impact of early ASCT was seen in the first year on overall survival but the sample size is extremely small. In all age groups early ASCT seemed to mitigate some of the survival disadvantage associated with randomization to the LD arm. CONCLUSIONS: This analysis shows that the strategy of lenalidomide plus dexamethasone induction followed by early ASCT has a remarkably good outcome in terms of overall survival in all age groups studied and supports the continued role of early consolidative ASCT in newly diagnosed patients. The risk of early mortality was notably low in the first year in all age groups. The risk of early mortality seems to increase at 2 years for the LD pts not choosing early ASCT and at 3 years for the Ld pts not choosing early ASCT. Selection bias makes it difficult to compare results for pts that chose early ASCT directly to the patients who did not receive early ASCT in this trial. As such, these results emphasize the need for randomized trials investigating the timing of ASCT in myeloma in the era of novel therapy. Disclosures: Siegel: Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Millennium Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Merck: Membership on an entity's Board of Directors or advisory committees. Off Label Use: Lenalidomide for front line therapy. Abonour:Celgene: Speakers Bureau; Millennium Pharmaceuticals: Speakers Bureau. Callander:Millennium Pharmaceuticals: Research Funding. Fonseca:Amgen: Consultancy; Bristol-Myers Squibb: Consultancy; Celgene: Consultancy, Research Funding; Genzyme: Consultancy; Onyx: Research Funding; Otsuka: Consultancy; Medtronic: Consultancy. Vesole:Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 494-494
Author(s):  
Erica D Warlick ◽  
Brian McClune ◽  
Tanya L. Pedersen ◽  
Kwang W Awn ◽  
Joseph H. Antin ◽  
...  

Abstract Abstract 494 The introduction of tyrosine kinase inhibitors (TKI) and advent of RIC and non-myeloablative (NMA) conditioning hematopoietic cell transplants (HCT) have changed and the therapeutic strategy for patients with CML. We analyzed the post HCT outcomes of CML patients aged 40 and older undergoing RIC/NMA HCT in 2001–2007. Detailed information regarding pre HCT TKI use or rationale for timing of transplant was not available; however, the analysis time period captures the entry of TKIs into clinical practice. Outcomes were compared between age cohorts of 40–49, 50–59, and ≥ 60 years. Overall survival (OS), Day +100 acute graft versus host disease (aGVHD) grades II-IV, chronic (cGVHD), transplant-related mortality (TRM), relapse, and disease-free survival (DFS) were analyzed with multivariate analysis testing the impact of age, gender, disease status at HCT (CP1vs. CP2/accelerated phase (AP) vs. blast phase (BP), sex match, HLA match, GVHD prophylaxis, and conditioning intensity (RIC versus NMA as described by Bacigalupo et all 2009) on outcomes. A total of 306 CML patients underwent HCT at 125 centers: 117 (38%) aged 40–49; 119 (39%) aged 50–59; and 70 (23%) aged ≥ 60. At HCT most patients in the 40–49 age cohort were in CP1 (72%), while only 44% of patients aged 50–59 and 31% aged ≤ 60 were in CP1. Interval from diagnosis to HCT for CP1 patients was similar across age groups with a large percentage of each age cohort undergoing HCT ≥ 2 years (32%, 40%, and 45%, respectively). Sibling donors were the stem cell source for 56% of those aged 40–49; older cohorts had a higher percentage of unrelated donors (58 and 60%, respectively). Primarily peripheral blood grafts (78%, 80% and 90%) and RIC (78%, 76% and 70%) were used across age groups, respectively. GVHD prophylaxis was similar. Three year OS and cGVHD, Day +100 grade II-IV acute GVHD, and 1 year TRM were similar in all age cohorts. Three year relapse incidence increased and DFS decreased with age. Importantly in analysis of CP1 patients only, relapse and DFS were similar in each age cohort.Table 1:Univariate AnalysisOutcomeAge 40–49 Probability (95% CI)Age 50–59 Probability (95% CI)Age ≥ 60 Probability (95% CI)P-valueEntire CohortOSA54% (44–64)52% (42–61)41% (30–54)0.26aGVHDB II-IV26% (18–34)32% (24–40)32% (21–43)0.53cGVHDA58% (47–68)51% (41–61)43% (33–55)0.19TRMC18% (11–26)20% (13–27)13% (6–22)0.43RelapseA36% (27–46)43% (34–52)66% (53–77)0.001DFSA35% (26–45)32% (24–41)16% (7–27)0.01CP1 OnlyRelapseA34% (23–46)42% (28–56)51% (29–72)0.40DFSA43% (31–55)36% (23–51)39% (19–61)0.81A=3 year; B=Day +100; C=1 year Multivariate analysis confirmed the significant adverse impact of advanced CML (AP/CP2+ and BP) at HCT, NMA conditioning intensity, male gender, and older age on relapse and DFS. Overall survival was not impacted by age, but was significantly worse with advanced CML at HCT. (Table 2)Table 2:Multivariate AnalysisOutcomeRR95% CIP valueRelapse:Age:10.69–1.6420.000340–491.0641.441–3.60750–592.28≥60Disease Status:10.1319–0.9170.01CP11.3350.0036–1.403AP/CP2+2.823BPConditioning Intensity10.222–0.469<0.0001NMA0.323RICGender10.397–0.830.0032Male0.574FemaleOverall SurvivalAge:10.55–1.2260.4240–490.8210.683–1.61850–591.051≥60Disease Status:11.206–2.47<0.0001CP11.7263.199–10.0740.42AP/CP2+5.677BPGender:10.0189–0.4810.019Male0.671Female These data indicate that HCT is safe in older patients with CML with equivalent acute and chronic GVHD, TRM, and OS across age cohorts. Relapse increased in patients receiving NMA conditioning and in those aged 60 and above; most of whom had advanced disease. However, for HCT during CP1 relapse risks and DFS were similar, regardless of age. Allogeneic HCT using RIC conditioning for older patients with CP1 CML can control relapse with acceptable toxicity and survival. Comparison of outcomes with second line TKI versus HCT are as yet unreported but these favorable findings indicate appropriate consideration of HCT for older patients with CML. Disclosures: Gupta: Incyte: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 782-782 ◽  
Author(s):  
Pashna N. Munshi ◽  
Parameswaran Hari ◽  
David H. Vesole ◽  
Artur Jurczyszyn ◽  
Jan Zaucha ◽  
...  

Background: Autologous hematopoietic cell transplantation (AHCT) is an effective treatment to achieve deep and durable remission in multiple myeloma (MM). Typically, AHCT is offered to patients &lt;70 years of age, although the median age of diagnosis of MM is 70 years. We compared the outcomes of upfront AHCT across age groups for newly diagnosed MM in the era of novel therapies using the Center for International Blood and Marrow Transplant Research (CIBMTR) database. Methods: We analyzed the outcomes of 15,999 MM patients aged 20 years and older from the USA who received a single AHCT with melphalan conditioning within 12 months of diagnosis between 2013 and 2017 and reported to the CIBMTR. Multivariate analysis was performed for non-relapse mortality (NRM), relapse/progression (REL), progression-free survival (PFS) and overall survival (OS) using Cox proportional hazards model with age at transplant in decades as the main effect. Hazard ratio (HR) with 95% confidence intervals (CI) are reported. Because of the large sample size, a p-value of &lt;0.01 was considered significant a priori. Results: Table 1 shows patient, disease and treatment characteristics by age group at diagnosis. All age groups had similar distribution of gender, race, ethnicity, Karnofsky performance score (KPS), comorbidity index (HCT-CI), stage III by Durie-Salmon/International Staging System. There was a higher proportion of high-risk cytogenetics in patients ≥70 years (30%), compared to age group 40-49 years (24%) and 20-39 years (20%) in this population. Older patients were more likely to be White compared to younger patients: 85% ≥70 compared to 64% 20-39 years. While 82% of the overall population received melphalan 200 mg/m2, 58% of the ≥70% received Mel 140 mg/m2. There were more ASCT performed in the ≥70-year age group in 2017 (28%) compared to 2013 (15%). Univariate outcomes by age groups shown in Table 2 revealed that 100-day NRM was higher in ≥70 years at 1% compared to younger patients (p &lt;0.01) and 2-year OS was lower in ≥70 years at 86 (85-88)% compared to 60-69 years, 89 (88-89)% (p&lt;0.01). When adjusted for other variables (Table 3), compared to reference age group of 60-69 years, patients ≥70 had similar NRM (HR 1.3, 95% CI 1, 1.7, p 0.06), REL (HR 1.03, 95% CI 0.9, 1.1, p 0.6), PFS (HR 1.06, 95% CI 1, 1.2, p 0.2), and OS (HR 1.2, 95% CI 1, 1.4, p 0.02). The leading cause of death across all age groups was primary disease. Among the ≥70 years cohort, melphalan dose was a surrogate for worse outcomes including NRM at 100 days (Mel 140, 1 (1-2)% vs Mel 200 0 (0-1)%, p 0.003, PFS at 2 years Mel 140 64 (60-67)% vs Mel 200 69 (66-73)%, p 0.003, and OS at 2 years (Mel 140 85 (82-87)% vs Mel 200 89 (86-91)%, p 0.01) (Figure). Conclusions: This is the largest study of AHCT in older adults with MM. More MM patients ≥70 years are being transplanted in the US over time. While our data may highlight referral and access biases regarding which older patients may be referred for ASCT, our results confirm that patients ≥70 years can undergo transplant safely and achieve similar benefits as 60-69 years' old patients. Our results also suggest that melphalan 200 mg/m2 may be given safely in the ≥70 years population. While melphalan conditioning dose 140 mg/m2 in the ≥70 group is associated with worse outcomes, this is likely a surrogate for higher frailty and comorbidities in this cohort of patients. Our analysis confirms that AHCT has similar benefits in terms of disease control (REL and PFS) in both young and older MM patients. This benefit is seen even in a contemporaneous era where proteasome inhibitors and/or immunomodulator drugs are used in upfront treatment. Thus, AHCT remains a safe consolidation therapy across all age groups of MM patients. Disclosures Hari: Celgene: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Research Funding; Janssen: Consultancy, Honoraria; Kite: Consultancy, Honoraria; Amgen: Research Funding; Spectrum: Consultancy, Research Funding; Sanofi: Honoraria, Research Funding; Cell Vault: Equity Ownership; AbbVie: Consultancy, Honoraria. Kumar:Janssen: Consultancy, Research Funding; Takeda: Research Funding; Celgene: Consultancy, Research Funding. Shah:Poseida: Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Nkarta: Consultancy, Membership on an entity's Board of Directors or advisory committees; Kite: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene, Janssen, Bluebird Bio, Sutro Biopharma: Research Funding; University of California, San Francisco: Employment; Genentech, Seattle Genetics, Oncopeptides, Karoypharm, Surface Oncology, Precision biosciences GSK, Nektar, Amgen, Indapta Therapeutics, Sanofi: Membership on an entity's Board of Directors or advisory committees; Indapta Therapeutics: Equity Ownership; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Teneobio: Consultancy, Membership on an entity's Board of Directors or advisory committees. Qazilbash:Genzyme: Other: Speaker; Bioclinical: Consultancy; Amgen: Consultancy, Other: Advisory Board; Autolus: Consultancy. D'Souza:EDO-Mundapharma, Merck, Prothena, Sanofi, TeneoBio: Research Funding; Prothena: Consultancy; Pfizer, Imbrium, Akcea: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4554-4554
Author(s):  
Ning Dong ◽  
David S Siegel ◽  
Phyllis McKiernan ◽  
Noa Biran ◽  
Alan P Skarbnik ◽  
...  

Abstract Background: Autologous stem cell transplantation (ASCT) is increasingly offered to older patients with multiple myeloma (MM) based on clinical trials demonstrating improved outcome. Inherently, successful mobilization and collection of peripheral blood stem cells (PBSC) is necessary for ASCT. A direct comparison of the efficacy of mobilization between elderly and younger patients has not been reported. Retrospective studies demonstrated that older patients had lower CD34+ cell yield and a higher incidence of mobilization failure (Lee et al 2014, Muchtar et al 2016). In this retrospective study, we compared two age groups (>/=70 years and <70 years) in MM patients who received ASCT at the John Theurer Cancer Center. We compared the outcomes of PBSC mobilization and transplantation between the two groups. Methods: MM patients who received a single ASCT at our institution between 2005 and 2016 were included. More than 95% of patients received either cyclophosphamide-based chemotherapy with GCSF (CY-GCSF) or plerixafor with GCSF (Pleri-GCSF) and were included in the analysis. We identified 111 patients aged >/= 70 years and 315 patients <70 years. The total CD34+ cell yield, number of apheresis sessions and CD34+ yield per session were compared by age group and mobilization regimen using the student's t-test. Multivariate analysis was performed using the general linear model with age group, mobilization regimen, age and mobilization agent interaction, Durie-Salmon (DS) stage, disease status at mobilization, and mobilization within 12 months of diagnosis as covariates. Progression-free survival (PFS) and overall survival (OS) were compared using Cox proportional hazard model with the above-mentioned covariates. All analyses were done using SAS 9.4. Results: Patient characteristics and CD34+ cell yield by age group are summarized in Table 1. The majority of patients in both groups underwent mobilization within one year of diagnosis. Older patients were less likely to receive CY-GCSF and more likely to receive Pleri-GCSF compared to younger patients (p<0.001). Disease status at mobilization, time from diagnosis to mobilization and DS stage were not associated with choice of mobilization regimens (p>0.05 for all). The two groups had similar number of apheresis sessions regardless of mobilization regimen. When receiving Pleri-GCSF, the two age groups had similar CD34+ yield per apheresis (4.4x10e6 /kg and 3.9x10e6 /kg for age>/=70 and age<70, respectively, p=0.49). However when receiving CY-GCSF, the older patients had lower CD34+ yield per apheresis compared to the younger patients (5.4 x10e6 /kg and 7.5 x10e6 /kg, respectively, p=0.004). When comparing the CD34+ yield per session within the older cohort, the yields with CY-GCSF and Pleri-GCSF were similar (p=0.16). In the multivariate analysis including all patients, time from diagnosis to mobilization, disease stage and disease status at mobilization were not associated with CD34+ yield per session or total CD34+ yield. Neither age nor mobilization regimen was associated with PFS or OS, after adjusting for disease stage and disease status at ASCT (Table 2, Figure 1, Figure 2). Conclusions: CD34+ yield was comparable between younger patients and older patients receiving plerixafor + GCSF. In contrast, the CD34+ yield was lower in the older patients receiving cyclophosphamide + GCSF. The cause of the difference is not clear and warrants further study. The PFS and OS were comparable between the two groups after ASCT. The choice of mobilization regimen did not affect survival. We have previously advocated cyclophosphamide mobilization in most patients because of the higher CD34+ yield and lower cost compared to plerixafor + GCSF (Panchal et al., ASH 2017). However this study showed that the CD34+ yield was not significantly different for patients aged 70 years or older in response to the different mobilization regimens. It is reasonable to consider plerixafor + GCSF in the elderly when chemotherapy toxicity is of concern. Disclosures Siegel: Karyopharm: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Merck: Consultancy, Honoraria, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau; Amgen: Consultancy, Honoraria, Speakers Bureau; Novartis: Honoraria, Speakers Bureau; Takeda: Consultancy, Honoraria, Speakers Bureau; BMS: Consultancy, Honoraria, Speakers Bureau. Biran:Amgen: Consultancy, Speakers Bureau; Merck: Research Funding; Celgene: Consultancy, Honoraria, Speakers Bureau; BMS: Research Funding; Takeda: Consultancy, Speakers Bureau. Skarbnik:Gilead Sciences: Honoraria, Speakers Bureau; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Genentech: Honoraria, Speakers Bureau; Pharmacyclics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Jazz Pharmaceuticals: Honoraria, Speakers Bureau; Seattle Genetics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Honoraria, Speakers Bureau.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3129-3129
Author(s):  
Hans C. Lee ◽  
Sikander Ailawadhi ◽  
Cristina Gasparetto ◽  
Sundar Jagannath ◽  
Robert M. Rifkin ◽  
...  

Background: Multiple myeloma (MM) is common among the elderly, with 35% of patients (pts) diagnosed being aged ≥75 years (y). With increasing overall life expectancy, the incidence and prevalence of newly diagnosed and previously treated MM patients ≥80 y is expected to increase over time. Because elderly pts are often excluded from clinical trials, data focused on their treatment patterns and clinical outcomes are lacking. The Connect® MM Registry (NCT01081028) is a large, US, multicenter, prospective observational cohort study of pts with newly diagnosed MM (NDMM) designed to examine real-world diagnostic patterns, treatment patterns, clinical outcomes, and health-related quality of life patient-reported outcomes. This analysis reviews treatment patterns and outcomes in elderly pts from the Connect MM Registry. Methods: Pts enrolled in the Connect MM registry at 250 community, academic, and government sites were included in this analysis. Eligible pts were adults aged ≥18 y with symptomatic MM diagnosed ≤2 months before enrollment, as defined by International Myeloma Working Group criteria; no exclusion criteria were applied. For this analysis, pts were categorized into 4 age groups: <65, 65 to 74, 75 to 84, and ≥85 y. Pts were followed from time of enrollment to the earliest of disease progression (or death), loss to follow-up, or data cutoff date of February 7, 2019. Descriptive statistics were used for baseline characteristics and treatment regimens. Survival outcomes were analyzed using Cox regression. Time to progression (TTP) analysis excluded causes of death not related to MM. Results: Of 3011 pts enrolled (median age 67 y), 132 (4%) were aged ≥85 y, and 615 (20%) were aged 75-84 y at baseline. More pts aged ≥85 y had poor prognostic factors such as ISS stage III disease and reduced hemoglobin (<10 g/dL or >2 g/dL <LLN) compared with other age groups, although no notable differences between creatinine and calcium levels were observed across age groups (Table). A lower proportion of elderly pts (75-84 and ≥85 y) received triplet regimens as frontline therapy. More elderly pts received a single novel agent, whereas use of 2 novel agents was more common in younger pts (Table). The most common frontline regimens among elderly pts were bortezomib (V) + dexamethasone (D), followed by lenalidomide (R) + D, whereas those among younger pts included RVD, followed by VD and CyBorD (Table). No pt aged ≥85 y, and 4% of pts aged 75-84 y received high-dose chemotherapy and autologous stem cell transplant (vs 61% in the <65 y and 37% in the 65-74 y age group). The most common maintenance therapy was RD in pts ≥85 y (although the use was low) and R alone in other age groups (Table). In the ≥85 y group, 27%, 10%, and 4% of pts entered 2L, 3L, and 4L treatments respectively, vs 43%, 23%, and 13% in the <65 y group. Progression-free survival was significantly shorter in the ≥85 y age group vs the 75-84 y age group (P=0.003), 65-74 y age group (P<0.001), and <65 y age group (P<0.001; Fig.1). TTP was significantly shorter in the ≥85 y group vs the <65 y group (P=0.020); however, TTP was similar among the 65-74 y, 75-84 y, and ≥85 y cohorts (Fig. 2). Overall survival was significantly shorter in the ≥85 y group vs the 75-84 y, 65-74 y, and <65 y groups (all P<0.001; Fig. 3). The mortality rate was lowest (46%) during first-line treatment (1L) in pts aged ≥85 y (mainly attributed to MM progression) and increased in 2L and 3L (47% and 54%, respectively); a similar trend was observed in the younger age groups. The main cause of death was MM progression (29% in the ≥85 y vs 16% in the <65 y group). Other notable causes of death in the ≥85 y group included cardiac failure (5% vs 2% in <65 y group) and pneumonia (5% vs 1% in <65 y group). Conclusions: In this analysis, elderly pts received similar types of frontline and maintenance regimens as younger pts, although proportions varied with decreased use of triplet regimens with age. Considering similarities in TTP across the 65-74 y, 75-84 y, and ≥85 y cohorts, these real-world data support active treatment and aggressive supportive care of elderly symptomatic pts, including with novel agents. Additionally, further clinical studies specific to elderly patients with MM should be explored. Disclosures Lee: Amgen: Consultancy, Research Funding; GlaxoSmithKline plc: Research Funding; Sanofi: Consultancy; Daiichi Sankyo: Research Funding; Celgene: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Janssen: Consultancy, Research Funding. Ailawadhi:Janssen: Consultancy, Research Funding; Takeda: Consultancy; Pharmacyclics: Research Funding; Amgen: Consultancy, Research Funding; Celgene: Consultancy; Cellectar: Research Funding. Gasparetto:Celgene: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed ; Janssen: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed ; BMS: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed . Jagannath:AbbVie: Consultancy; Merck & Co.: Consultancy; Bristol-Myers Squibb: Consultancy; Karyopharm Therapeutics: Consultancy; Celgene Corporation: Consultancy; Janssen Pharmaceuticals: Consultancy. Rifkin:Celgene: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees. Durie:Amgen, Celgene, Johnson & Johnson, and Takeda: Consultancy. Narang:Celgene: Speakers Bureau. Terebelo:Celgene: Honoraria; Jannsen: Speakers Bureau; Newland Medical Asociates: Employment. Toomey:Celgene: Consultancy. Hardin:Celgene: Membership on an entity's Board of Directors or advisory committees. Wagner:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; American Cancer Society: Other: Section editor, Cancer journal. Omel:Celgene, Takeda, Janssen: Other: Patient Advisory Committees. Srinivasan:Celgene: Employment, Equity Ownership. Liu:TechData: Consultancy. Dhalla:Celgene: Employment. Agarwal:Celgene Corporation: Employment, Equity Ownership. Abonour:BMS: Consultancy; Celgene: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Janssen: Consultancy, Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2609-2609
Author(s):  
Muhned Alhumaid ◽  
Georgina S Daher-Reyes ◽  
Wilson Lam ◽  
Arjun Law ◽  
Tracy Murphy ◽  
...  

Introduction: Clinical outcomes of acute myeloid leukemia (AML) in adolescents and young adults (AYA) are rarely reported as an isolated subgroup. Treatments vary little across age groups, and treatment intensity depends upon comorbid conditions and performance status. Optimal treatment strategies focused on disease behavior, biological factors, and the distinct needs of this subset of AML patients remain elusive. The purpose of this retrospective analysis is to determine the characteristics and outcomes of AYA AML patients treated at a specialized adult leukemia cancer center in comparison to older adults with AML (40-60 years). Methods: A retrospective analysis was performed on all patients treated at Princess Margaret Cancer Center from 2008-2018. Patients with acute promyelocytic leukemia were excluded. Clinical characteristics, treatment strategies, and survival outcomes were recorded for all patients. Overall survival (OS) and disease-free survival (DFS) rates were calculated using the Kaplan-Meier product-limit method and the impact of covariates were assessed using the Log-rank test. Finally, we compared the outcomes of AYA patients treated at our centre between 2015-2018 with older patients. Results: A total of 175 patients aged 18-39 were identified. Patient characteristics are shown in (Table 1). Cytogenetic were available in 163 patients. Based on MRC criteria, 27 (16%) were favorable risk, intermediate in 95 (54%), adverse in 39 (22%), and missing/failed in 14(8%). NPM1 status was available in 110 patients of whom 38 (35%) were positive. FLT3-ITD was available in 67 patients with 24 (36%) positive. Both mutations were present in 13 (54%) patients. There were no significant differences in terms of risk stratification based on cytogenetic and molecular markers based on age (18-29 vs.30-39) (P= 0.98). Most patients 172 (98%) received induction, 157 (91%) with 3+7, and 15 (9%) with FLAG-IDA. Complete remission (CR) was achieved in 133 (77%) after first induction [120 (76%) after 3+7 and 11 (73%) after FLAG-IDA]. Induction related mortality was low (2%). Of the 39 who did not achieve CR, thirty-four patients received re-induction (13 FLAG-IDA, 16 NOVE-HiDAC, 5 others) with CR in 21 (62%). Overall, 154 (89.5%) achieved CR1. Sixty-four (42%) proceeded to hematopoietic stem cell transplantation (HSCT) in CR1. 59 (38%) patients relapsed in CR1 with 8 (12%) relapsing post HSCT. Fifty-five (5 post HSCT) patients received reinduction with 30 (51%) (2 after HSCT) achieving CR2. Fifteen patients received HSCT in CR2. OS and DFS at 2 years were 62% (95% CI 0.53-0.69) and 50% (95% CI 0.41-0.57), respectively. Stratified by cytogenetic risk, OS was 81% for favorable risk, 61% for intermediate, and 50% for adverse risk (P=0.0001), respectively. DFS in these groups was 85%, 57%, and 46 % (P=0.0025), respectively. We further compared outcomes in the 18-29y and 30-39y age groups. The OS was 61.9% compared to 62.5% (P=0.91) and DFS of 52.1% compared to 47% (P=0.65) respectively. On univariate analysis for OS and DFS, cytogenetic risk stratification was the only significant variable (P=0.0004 and P=0.0042). We then compared the outcomes 67 sequential patients aged I8-39 treated from 2014-2018, with those of 176 sequential patients aged 40-60 treated during the same period (table 2). OS at 2 years was not statistically higher in the younger group compared to the older group (66.7% vs. 61.2%, P=0.372). While relapse rate was lower in older patients (15.5% vs. 22.6%, P=0.093), NRM was higher in older patients (29.7% vs. 18.8%,P=0.094). Conclusion: AYA pts. occupy a unique niche amongst AML as a whole. While treatment responses have improved in general, there may be potential for further gains in these patients. Increased tolerance for more intense treatment strategies as well as the incorporation of novel agents into standard treatment protocols may provide a means to optimize care in AYA patients. Finally, research is needed to elucidate biological mechanisms and predictors of disease behavior instead of arbitrary, age-stratified treatment schema. Disclosures McNamara: Novartis Pharmaceutical Canada Inc.: Consultancy. Schimmer:Jazz Pharmaceuticals: Consultancy; Medivir Pharmaceuticals: Research Funding; Novartis Pharmaceuticals: Consultancy; Otsuka Pharmaceuticals: Consultancy. Schuh:Astellas: Honoraria, Membership on an entity's Board of Directors or advisory committees; AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Teva Canada Innovation: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Agios: Honoraria; Jazz: Honoraria, Membership on an entity's Board of Directors or advisory committees. Maze:Pfizer Inc: Consultancy; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees. Yee:Astellas: Membership on an entity's Board of Directors or advisory committees; Millennium: Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees; Astex: Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck: Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; MedImmune: Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Hoffman La Roche: Research Funding. Minden:Trillium Therapetuics: Other: licensing agreement. Gupta:Incyte: Honoraria, Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sierra Oncology: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2085-2085
Author(s):  
Chris L. Pashos ◽  
Christopher R Flowers ◽  
Mark Weiss ◽  
Nicole Lamanna ◽  
Charles M Farber ◽  
...  

Abstract Abstract 2085 Introduction. Although advanced patient age is commonly used as a factor in selecting therapy for patients with chronic lymphocytic leukemia (CLL), based on presumed associations with functional status, limited data exist regarding the relationships between age and physical, emotional, social, and functional well being. We examined the relationships between age and these domains of health-related quality of life (HRQOL) for CLL patients treated in US community practices. Methods. Baseline data were collected as part of Connect CLL®, a prospective observational registry initiated in March 2010 involving centers in the US. Data on patient demographics and clinical characteristics were provided by clinicians. HRQOL was self-reported by patients in the clinic at enrollment. Patients completed 3 psychometrically validated instruments: the Brief Fatigue Inventory (BFI), EQ-5D, and Functional Assessment of Cancer Therapy-Leukemia (FACT-Leu). Standard analyses were conducted of each instrument given clinical characteristics at that time. Reported mean BFI, EQ-5D and FACT-Leu scores were analyzed by age group (<65, 65–74, >74). Statistical significance of score differences among sub-cohorts was ascertained by ANOVA using SAS 9.1. Results. Baseline HRQOL data were reported by 604 patients, enrolled from 161 centers. Patients were predominantly male (62%) and white (90%) with mean age at 69.9 (standard deviation [SD] 11.2) yrs. HRQOL scores by age group are presented: There were no significant differences between the age groups in fatigue as measured by the BFI, or differences in overall HRQOL as measured by the EQ-5D Visual Analogue Scale (VAS) or the FACT-G. Anxiety/depression and self care are EQ-5D domains that also did not vary by age. Although mobility was most impaired in the oldest age group compared to the two younger groups, usual activities and pain/discomfort were worse in both the younger and older cohorts compared to those 65–74 years of age. FACT-Leu results indicated that the social/family domain scores did not vary by age, but that physical, emotional, and functional domains did vary statistically with the oldest typically doing better than the 65–74 year olds, but not necessarily better than those <65. Conclusions. Initial results from the Connect CLL® Registry indicate that HRQOL does not worsen monotonically with older age. In this cohort, both the youngest and oldest age groups had worse HRQOL in certain domains, presenting an inverted v-shaped relationship. Future analyses should be conducted on: (1) how HRQOL may be affected over time with changes in disease; and, (2) how HRQOL may be influenced by alternative therapies. Results reported here should serve as a useful baseline reference. Disclosures: Pashos: Celgene: Membership on an entity's Board of Directors or advisory committees. Flowers:Genentech/Roche (unpaid): Consultancy; Celgene: Consultancy; Millennium/Takeda: Research Funding; Wyeth: Research Funding; Novartis: Research Funding. Weiss:Celgene: Membership on an entity's Board of Directors or advisory committees. Lamanna:Celgene: Membership on an entity's Board of Directors or advisory committees. Farber:Celgene: Membership on an entity's Board of Directors or advisory committees. Kipps:Igenica: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Research Funding; Abbot Industries: Research Funding; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Genentech: Research Funding; GSK: Research Funding; Gilead Sciences: Consultancy, Research Funding; Amgen: Research Funding. Lerner:Celgene: Membership on an entity's Board of Directors or advisory committees. Kay:Celgene: Membership on an entity's Board of Directors or advisory committees. Sharman:Celgene: Membership on an entity's Board of Directors or advisory committees. Grinblatt:Celgene: Membership on an entity's Board of Directors or advisory committees. Flinn:Celgene: Membership on an entity's Board of Directors or advisory committees. Kozloff:Celgene: Membership on an entity's Board of Directors or advisory committees. Swern:Celgene Corporation: Employment, Equity Ownership. Kahn:Celgene Corporation: Employment, Equity Ownership. Street:Celgene: Employment, Equity Ownership. Sullivan:Celgene: Employment, Equity Ownership. Keating:Celgene: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3773-3773
Author(s):  
Adam Mendizabal ◽  
Paul H Levine

Abstract Abstract 3773 Background: Age at diagnosis of CML varies by race in the United States with median occurring around ages 54 and 63 among Black and White patients, respectively. The treatment paradigm shifted when Imatinib was approved in 2001 for treatment of CML. More recently, second generation tyrosine kinase inhibitors (TKI) have also been used for treatment of CML. Differences in outcomes by race have been previously reported prior to the TKI treatment period. We aimed to assess whether the earlier age at diagnosis resulted in differential trends in age-adjusted incidence rates and survival outcomes by race in the post-Imatinib treatment period. Methods: Data from the Surveillance, Epidemiology, and End Results (SEER) 18 Registries were extracted for diagnoses between 2002 and 2009 based on the assumption that cases diagnosed after 2002 would be treated with TKI's. CML was defined according to the International Classification of Diseases for Oncology 3rd edition code 9863 (CML-NOS) and 9875 (CML-Philadelphia Chromosome Positive). Cases diagnosed by autopsy or death certificate only were excluded. Incidence rates are expressed per 100,000 person-years and age-adjusted to the 2000 US Standard Population. Black/White incidence rate ratios (IRRBW) are shown with corresponding 95% confidence intervals (CI). Kaplan-Meier estimates of CML-specific survival (CPS) and overall survival (OS) were estimated at 5-years post-diagnosis with the event being time to CML-specific death or any death, respectively. Stratified Cox proportional hazards models were constructed to assess the impact of age and race on the risk of death expressed as a hazard ratio (HR). Results: Since 2002, 6,632 patients diagnosed with CML were reported to the SEER 18 registries including 5,829 White patients (87.9%) and 803 Black patients (12.1%) with 57% being male. The age-adjusted incidence rate for Blacks was 1.18 (95% CI, 1.10–1.27) per 100,000 and 1.12 (95% CI, 1.09–1.27) per 100,000 for Whites. The corresponding IRRBW was 1.06 (95% CI, 0.98– 1.14). When considering 20-year age-groups, Blacks had higher incidence rates in the 20–39 and 40–59 age groups; IRRBW of 1.26 (95% CI, 1.06–1.49; p=0.0073) and 1.23 (95% CI, 1.09–1.39; p=0.0007), respectively. No statistically significant differences in IRRBW were seen within the 0–19, 60–79 and 80+ age-groupings although Whites have higher non-significant incidence rates in the latter 2 age-groups. Differences in IRRBW prompted an assessment of survival to determine if the excess incidence observed in the younger age groups corresponded with a worse survival. CPS at 5-years was 85.5% (95% CI, 84.3–86.6). In univariate analysis, age was an important predictor of outcome (p<0.0001) with patients diagnosed after age 80 having the worse outcomes (OS: 58.3%), followed by patients diagnosed between 60 and 79 years (OS 84.7%), 0–19 years (OS: 87.1%), 40–59 years (OS: 90.2%), and 20–39 years (OS: 92.6%). When considering all age-groups, race was not a significant predictor of death (HR 0.91; 95% CI, 0.72–1.15). However, in a stratified analysis with 20-year age groups, Blacks had an increased risk of death as compared to Whites (Figure 1) in the 20–39 age group (HR: 2.94; 95% CI, 1.72–5.26; p<0.0001) and the 40–59 age group (HR: 1.67; 95% CI, 1.22–2.27; p=0.0069) while no differences were seen within the 0–19, 60–79 and 80+ age groups. Conclusions from OS models were similar to that of the CPS models. Conclusions: Through this analysis of population-based cancer registry data collected in the US between 2002 and 2009, we show that Blacks have a younger age at diagnosis with higher incidence rates observed in the 20–39 and 40–59 age-groups as compared to Whites. Both CPS and OS outcomes differed by race and age. Similar to the differences observed with the incidence rates, survival was worse in Blacks diagnosed within the 20–39 and 40–59 age-groups as compared to Whites. Although outcomes have globally improved in patients with CML since the advent of tyrosine kinase inhibitors, the persistence of incidence heterogeneity and poorer survival among Blacks warrants further attention. Access to care may be a possible reason for the differences observed but further studies are warranted to rule out biological differences which may be causing an earlier age at onset and poorer survival. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 137-137 ◽  
Author(s):  
Marc G Berger ◽  
Bruno Pereira ◽  
Charlotte Oris ◽  
Sandrine Saugues ◽  
Pascale Cony-Makhoul ◽  
...  

Abstract Context: The Tyrosine Kinase Inhibitors (TKIs) have revolutionized the treatment of chronic myeloid leukemia (CML) increasing dramatically the survival of CML patients and leading to a residual disease with a sustained and deep molecular response. In this subset of very good responder patients, the attempts of stopping treatment in different clinical trials were successfully achieved without relapse. The Swedish team in the EURO-SKI protocol already reported cases of musculoskeletal pain occurring after cessation of TKI (Richter et al., JCO, 2014). Since several clinical trials regarding TKI discontinuation have been also run in France, we decided to retrospectively collect data using the pharmacovigilance system of the different Trials collected prospectively. Method: 428 patients from STIM2 (n=204) and EURO-SKI (n=224) trials were systematically analyzed from the case report from each trial. For the EURO-SKI only French patients were included. Statistical analysis was performed using Stata 13 software (StataCorp LP, College Station, TX, US). Comparisons between the independent groups were realized using the Chi-squared or Fisher's exact tests for categorical variables, and using Student t-test or Mann-Whitney test for quantitative. Multivariate analyses were performed to take into account adjustment on covariates fixed according to univariate results and clinically relevance. Results: Among the 428 patients the main characteristics were as follow i,e; 208 (48.6%) men and 220 (51.4%) women, with a median age of 77.5 years (24-93). Sokal scores (n=449) were low in 187 (41.6%) patients, intermediate in 188 (41.9%) patients and high in 74 (16.5%) patients. A withdrawal TKI syndrome (WS) was reported for 102 (23.8%) patients (100 after imatinib and 2 after nilotinib). 2). The WS consists in bone and articular pains and arthritis and affects the upper limbs, shoulders and cervical rachis, with a grade 1 or 2 in most patients and grade 3 in 22% of patients . The prevalence of WS depends on the trials, 34.8% in EURO-SKI group and 13.8% in STIM2 group (p<0.001). The WS was treated by non-steroidal anti-inflammatory drugs, corticosteroids or by local infiltration. The median duration of WS was 7 months (range: 3-30 months, 24 exploitable cases). We did not observe any difference between WS group and the group without painful syndrome in terms of sex ratio (p=0.92), age (p=0.33), sokal score (p=0.15), BCR-ABL transcript (p=0.42) or duration of CML (p=0.24). However the median duration of TKI therapy appeared longer in this subgroup (median: 88.8 months vs 79.8 months (p=0.02). There was no biological inflammatory syndrome and the results of medical imaging were inconclusive. However, a medical history of osteoarticular pains or disease appeared as predisposing to withdrawal syndrome (22.9% in WS group vs 9.8% in control group; p=0.002). Finally the two factors, duration of treatment and medical history were confirmed using multivariate analysis (RR=1.73 and 1.76 respectively). Among 19 exploitable cases suffering CML relapse and requiring further TKI treatment, pain disappeared in 7 patients (37%) within a median period of 3.5 weeks. Conclusion: About 23% of patients who stopped TKIs experienced a TKI WS and all TKI seems to be concerned. The predisposing factors were a medical history of osteoarticular pain or disease, and the duration of treatment. So patients and physicians should be aware and recommendations should be proposed for patients who have treated longtime with a history of arthritis. Disclosures Legros: Novartis: Research Funding, Speakers Bureau; ARIAD: Speakers Bureau; BMS: Speakers Bureau. Nicolini:Ariad Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Rousselot:Novartis: Speakers Bureau; Pfizer: Consultancy, Speakers Bureau; ARIAD: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau. Rea:Novartis: Honoraria; BMS: Honoraria; Ariad: Honoraria; Pfizer: Honoraria. Mahon:Bristol-Myers Squibb: Consultancy, Honoraria; ARIAD: Consultancy; Novartis: Consultancy, Honoraria; Pfizer: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 48-48 ◽  
Author(s):  
Marie Balsat ◽  
Vincent Alcazer ◽  
Gabriel Etienne ◽  
Gaelle Fossard ◽  
Francoise Huguet ◽  
...  

Abstract Introduction Up to 10% of patients (pts) with chronic myeloid leukemia (CML) are already in accelerated phase (AP) at diagnosis and despite treatment advances in the field of tyrosine kinase inhibitors (TKIs), management of these pts is challenging. This study aims to examine the benefit of second generation BCR-ABL tyrosine kinase inhibitors (TKI2) as first-line treatment for newly diagnosed AP-CML. Methods Pts meeting criteria for AP-CML at diagnosis and treated with first-line TKI2 (i. e. Nilotinib or Dasatinib) were included in this retrospective multicenter observational national study. AP-CML were defined according to the ELN (Baccarani, Blood 2013) as hematological acceleration (HEM-AP, any of the following features: blasts in PB or marrow 15-29%, or blasts+promyelocytes in PB or marrow >30% with blasts <30%, basophils in PB ≥20%, or persistent thrombocytopenia <100×109/L (unrelated to therapy) and/or chromosomal abnormalities in addition to the Ph at diagnosis (ACA-AP). Pts initiated nilotinib at 6-800 mg BID or dasatinib at 100-140 mg QD with further dose adaptations according to toxicities or response. Overall survival (OS), progression-free survival (PFS) and failure-free survival [FFS= progression to blast crisis, death, loss of any previous response (CHR, CCyR, or MMR) discontinuation of TKI2 for toxicity], were analysed since TKI2 initiation in intention-to-treat. Results Sixty-six pts were analysed: 45 males (68%) and 21 females (32%) with a median age at diagnosis of 49 (15-78.5) years. The median follow-up of the cohort was 43.5 (1.7-117) months. We segregated the pts in HEM-AP (n=33) and ACA-AP (n=33) for further analyses. Nine pts with HEM-AP harboured ACA and were analysed in the HEM-AP group. Fusion transcripts were of the Major BCR in 57 pts, 6 pts had atypical BCR-ABL transcripts (2 e19a2, and 1 e1a2 in the HEM-AP group and 2 e19a2 and 1 Ma3 in the ACA-AP group), and 3 transcripts unknown. Not surprisingly, spleen enlargement was significantly greater in the HEM-AP group [10 (5-14.75) vs. 3 (0-10)cm, p=0.014]. PB basophils [median 10 (6-16) vs. 3 (2-5)%, p <0.001], PB blasts [median: 12.05 (7.5-15) vs. 1.5 (0-4)%, p<.001], as well as PB blasts+promyelocytes [median 14 (11-20) vs. 4 (1-7)%, p<.001]. Hemoglobin levels were significantly lower in the HEM-AP group [median 93 (6-113.5) vs 120 (100-134) g/L, p<0.001]. Neither WBC counts, platelets counts, nor BCR-ABL/ABL load differed significantly between the 2 groups. In the ACA-AP group, 10 (30%) pts harbored major route ACA and 23 (70%) pts harbored minor route ACA of whom 3 pts with i(17q) and 1 with 7q abnormalities. In the ACA-AP group, Sokal score was low in 42%, intermediate in 32% and high in 26% of pts (2 pts unknown). Dasatinib was initiated in 19/33 pts (57.5%) in the HEM-AP group and in 8/33 pts (24%) in the ACA-AP group. Treatment responses did not significantly differ between ACA-AP and HEM-AP group, regardless of the TKI2 administered, with 33/33 (100%) vs 31/33 (94%) pts achieving a CHR, 2/33 (6%) pts vs 0/33 (0%) pts achieving a MCyR, 5/33 (15%) pts vs 5/33 (15%) pts achieving CCyR, 9/33 (27%) pts vs 4/33 (12%) pts achieving a MMR respectively. However, 11/33 (33%) HEM-AP vs 22/33 (66%) ACA-AP pts achieved a deep molecular response (p=0.013, Fisher test). Median times to CHR and MMR were not significantly different between ACA-AP group and HEM-AP group with 1.05 vs 1.25 months (p=0.088) for CHR and 6 vs 7 months (p=0.156) for MMR, respectively. Overall, the estimated 7-yr FFS rate was 56.92% (95%CI: 40-81), 7-yr PFS was 83.42% (95% CI: 69.6-100%) and 7-yr OS was 87.14% (95%CI: 73.5-100%) (Figure 1.) with no significant differences between ACA-AP vs HEM-AP pts [7-yr FFS: 57.7 vs. 62%, p=0.739; 7-yr PFS: 84.7% vs. 84%, p=0.185; 7-yr OS: 88.9% vs 86.6%, p=0.132] respectively. There was also no difference in FFS, PFS and OS according to the type of TKI2. The only factors influencing negatively OS were the % of BM blasts (HR=1.17, 95%CI: 1.1-1.28, p<0.001) and the % of BM blasts+promyelocytes (HR=1.14, 95%CI: 1.06-1.22, p<0.001). We identified too few significant factors in univariate analysis to perform a multivariate analysis. Conclusion The initiation of a TKI2 in newly diagnosed AP-CML pts induces excellent response and survival rates, probably superior to that of Imatinib first-line, and counterbalances the negative impact of this advanced disease, particularly in HEM AP subgroup. Disclosures Etienne: Pfizer: Membership on an entity's Board of Directors or advisory committees, Other: Travel, Speakers Bureau; BMS: Membership on an entity's Board of Directors or advisory committees, Other: Travel, Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Incyte: Honoraria, Patents & Royalties, Speakers Bureau. Berger:Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Mahon:Incyte: Speakers Bureau; Pfizer: Speakers Bureau; Novartis: Speakers Bureau; BMS: Speakers Bureau. Rea:Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Incyte: Honoraria; Pfizer: Honoraria. Nicolini:BMS: Consultancy, Speakers Bureau; Incyte Biosciences: Consultancy, Speakers Bureau; Sun Pharma Ltd: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document