scholarly journals Impact of Charlson Comorbidity Index (CCI) and Refining the MIPI Index in Mantle Cell Lymphoma (MCL)

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2973-2973
Author(s):  
Andre Goy ◽  
Tommy Wu ◽  
Eric Hansen ◽  
Srikesh Arunajadai ◽  
Ewelina Protomastro ◽  
...  

Abstract Introduction: The outcome of MCL patients (pts) has improved over the last three decades, although this is debated outside clinical trials (Chandran, Leuk Lymphoma Aug 2012; Smith, Br J Cancer. April 2015). The Mantle Cell International Prognostic Index (MIPI) (Hoster, Blood Jan 2008) is based on 4 variables which predict survival: age (host factor), PS (tumor/host), LDH (tumor burden) and WBC (leukemic phase). The additional value of including co-morbidities into risk stratification has not been fully explored. Methods: Using the COTA database we retrospectively analyzed MCL cases treated at John Theurer Cancer Center and the affiliated practices of Regional Cancer Care Associates from 2004 to 2016. Clinical and treatment characteristics, including calculation of the CCI index (Charlson J Chronic Diseases 1987) were captured via the COTA platform by extracting data from the electronic health records. Results: 490 pts with MCL were evaluated and full longitudinal data from diagnosis is currently reported on 195 subjects. Pts characteristics included: male (66.15%), med age (65, range 34-94), stage IV (87%), LDH (elevated 26%; median 197, range 112-7950), MIPI (low 38%, interm 32%, high 30%), Ki-67 (≥30%: 51%, 86 NA), blastoid variant (16%, 35 NA), SOX-11 positive (87%, 143 NA), 17p abnormalities (p53 del or overexpression/mutation (24%, 88 NA) and b-2 microglobulin (b-2m) > 3 mg/L (52%). Frontline therapy consisted of R-Hyper-CVAD (with or without bortezomib on study) (36%), R-HyperCVAD or R-CHOP followed by high-dose therapy followed by autologous stem cell transplantation (ASCT) (9%), BR alone (8%), BR+ maintenance (8%), R-CHOP alone (4%), Rituximab (3%), R-BAC (3%), BR+ Ibrutinib vs placebo (2%), radiation (2%), R-Lenalidomide (1%), R-CHOP + maintenance (1%), other treatments (10%) while 10% of patients were treated expectantly Seventeen pts underwent ASCT consolidation (15 auto vs 2 allo (del17p/blastoid at presentation). Overall and progression free survival was computed using Kaplan Meier curves and significance tested using log-rank tests. The 5y OS for this entire cohort was 81. Overall, dose-intensive strategies (with or without ASCT) approach was associated with a 23 mo difference in PFS (median 74 mo, range 0-111 mo (intensive) vs median 51 mo, range 2-57 mo for the non-intensive group (p=0.37). The median OS was not reached for either group, with a 5y OS of 88% in intensive vs. 71% non-intensive regimens (p=0.14). Using MIPI as stratification, low/intermediate risk pts had similar outcome in intensive and non-intensive therapy groups (5y OS 88% vs 71%; p=0.13). Pts with high MIPI had a 5 year OS of 80% in the intensive therapy group vs 46% for non-intensive group (p=0.376). The CCI scores for the whole cohort were 0 in 16 pts (8%), 1-3 in 81 pts (41%), and ≥ 4 in 98 pts (50%). Baseline CCI score (pre-treatment) was highly predictive of outcome with a 5y OS of 90% in CCI 0-3 vs 62% in CCI 3+ (p=0.001) (Figure 1). CCI did not predict complete response rate (CR) to induction therapy (CCI 0-3 94% vs CCI 3+ 80%). The median MIPI scores were 5.7 for CCI 0-3 and 6.3 for CCI 3+. Age is a component of both indexes but more heavily weighted in the CCI. Adding CCI to MIPI defined a subset of pts among the high MIPI group who did better than expected with a 5y OS of 88% in combined high MIPI / CCI 0-3 vs 31% for high MIPI / CCI 4+/ (p=0.03). b-2m (cut-off 3mg/L) correlated with 5y OS 93% vs 80%; (p=0.04) as previously reported but did not add to MIPI or CCI risk stratification. Ki-67 (30% cut-off) was marginally associated with OS: 5-y 89% vs 77% (p=0.06). Conclusions: Our cohort is consistent with the improvement of MCL outcome comparing to historical controls and illustrates the importance of comorbidities captured at baseline. A combined CCI-MIPI approach might help identify pts who can still benefit from current therapy approaches in spite of age. Among the high MIPI score group, CCI further refines cohorts with significantly different outcomes. Figure 1 Figure 1. FIgure 2 FIgure 2. Disclosures Goy: Acerta: Consultancy, Membership on an entity's Board of Directors or advisory committees; Genentech: Other: Research funding for clinical trials through institution; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; COTA: Membership on an entity's Board of Directors or advisory committees; Janssen/Pharmacyclics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Research funding for clinical trials through institution, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Research funding for clinical trials through institution. Wu:COTA: Employment. Hansen:COTA: Employment. Arunajadai:COTA: Employment. Protomastro:COTA: Employment. Valentinetti:COTA: Employment. Murphy:COTA: Employment. Smith:COTA: Employment. Pe Benito:COTA: Employment. Hasan:COTA: Employment. Suryadevara:COTA: Employment. Feldman:Seattle Genetics: Consultancy, Speakers Bureau; Abbvie: Consultancy, Speakers Bureau; Pharmacyclics: Speakers Bureau; Celgene: Speakers Bureau. Skarbnik:Pharmacyclics: Consultancy; Genentech: Speakers Bureau; Seattle Genetics: Speakers Bureau; Gilead Sciences: Speakers Bureau; Abbvie: Consultancy. Leslie:Celgene: Speakers Bureau; Seattle Genetics: Speakers Bureau. Pecora:COTA: Employment, Equity Ownership. Goldberg:Bristol Myers Squibb, Novartis: Speakers Bureau; Neostem: Equity Ownership; Novartis: Consultancy; COTA Inc: Employment; Pfizer: Honoraria.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3543-3543 ◽  
Author(s):  
Alexis A. Thompson ◽  
Mark C. Walters ◽  
Janet L. Kwiatkowski ◽  
Suradej Hongeng ◽  
John B. Porter ◽  
...  

Background Transfusion-dependent β-thalassemia (TDT) is treated with regular, lifelong red blood cell (RBC) transfusions and despite iron-chelating therapy, carries a risk of serious organ damage from iron overload and other complications. Transplantation with autologous CD34+ cells encoding a βA-T87Q-globin gene (LentiGlobin for β-thalassemia) is being evaluated in patients with TDT. Interim results are presented here from the ongoing, international, single-arm, phase 3 Northstar-2 study (HGB-207; NCT02906202) of LentiGlobin gene therapy in pediatric, adolescent, and adult patients with TDT (defined by receiving ≥100 mL/kg/yr of RBCs or ≥8 RBC transfusions/yr) and non-β0/β0 genotypes. Methods Patients undergo hematopoietic stem cell (HSC) mobilization with G-CSF and plerixafor. Following apheresis, CD34+ cells are transduced with BB305 lentiviral vector and infused into patients after pharmacokinetic-adjusted, single-agent busulfan myeloablation. The primary efficacy endpoint is transfusion independence (TI; weighted average hemoglobin [Hb] ≥9 g/dL without RBC transfusions for ≥12 months). HSC engraftment, βA-T87Q-globin expression, Hb levels, detection of replication competent lentivirus (RCL), and adverse events (AE) are also assessed. Patients are followed for 2 years and offered participation in a long-term follow-up study. Summary statistics are presented as median (min - max). Results Twenty patients were treated in Northstar-2 as of 13 December 2018 and have been followed for a median of 8.1 (0.5 - 22.2) months. At enrollment, median age was 16 (8 - 34) years; 5 patients were <12 years of age. Median drug product cell dose was 8.0 (5.0 - 19.9) x106 cells/kg and vector copy number was 3.2 (1.9 - 5.6) copies/diploid genome. Time to neutrophil and platelet engraftment in the 18/20 and 15/20 evaluable patients was 22.5 (13 - 32) and 45 (20 - 84) days, respectively. Non-hematologic grade ≥3 AEs in ≥3 patients after LentiGlobin infusion included stomatitis (n=12), febrile neutropenia (n=6), pyrexia (n=4), epistaxis (n=3), and veno-occlusive liver disease (n=3). One serious AE of grade 3 thrombocytopenia was considered possibly related to LentiGlobin. No patient died, had graft failure, or had detection of RCL. No insertional oncogenesis has been observed. Gene therapy-derived HbAT87Q stabilized approximately 6 months after infusion. In adolescent and adult patients treated with LentiGlobin, median HbAT87Q at Months 6, 12 and 18 was 9.5 (n=11), 9.2 (n=8), and 9.5 (n=3) g/dL, respectively. The median total Hb without transfusions at Months 6, 12, and 18 were 11.9 (n=11), 12.4 (n=8), 12.3 (n=2) g/dL, respectively. At Month 6, 91% (10/11) of patients had total Hb of >11 g/dL without transfusions. Five adolescent and adult patients were evaluable for the primary endpoint of transfusion independence, 4 (80%) of whom achieved TI. The median weighted average Hb during TI was 12.4 (11.5 - 12.6) g/dL which compared favorably to pre-transfusion nadir Hb levels before enrollment (median 9.1 g/dL [7.5 - 10.0 g/dL]). At time of analysis, the median duration of TI was 13.6 (12.0 - 18.2) months. One patient who did not achieve TI stopped transfusions for 11.4 months but resumed transfusions due to recurrent anemia. This patient had a 71.4% reduction in RBC transfusion volume from Month 6 to Month 18 compared to baseline. Marrow cellularity and myeloid:erythroid (M:E) ratios were evaluated in 8 adolescent and adult patients with ≥12 months follow-up to assess the effect of LentiGlobin treatment on dyserythropoiesis. Seven of 8 patients had improved marrow M:E ratios at Month 12 (0.63 - 1.90) compared with baseline (0.14 - 0.48). In patients who stopped transfusions, soluble transferrin receptor levels were reduced by a median of 72% (58% - 78%) at Month 12 (n=6). Updated outcomes in adolescents and adults and outcomes in pediatric patients will be reported. Summary In this update of the Northstar-2 study of LentiGlobin gene therapy in patients with TDT and non-β0/β0 genotypes, transfusion independence was observed in 4/5 evaluable adolescent and adults and 10/11 treated patients had total Hb of >11 g/dL without transfusion support 6 months after LentiGlobin infusion. HbAT87Q stabilized approximately 6 months after treatment and patients who stopped RBC transfusions had improved erythropoiesis. A safety profile consistent with busulfan conditioning was observed after LentiGlobin gene therapy. Disclosures Thompson: bluebird bio, Inc.: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Baxalta: Research Funding. Walters:TruCode: Consultancy; AllCells, Inc: Consultancy; Editas Medicine: Consultancy. Kwiatkowski:bluebird bio, Inc.: Consultancy, Research Funding; Terumo: Research Funding; Celgene: Consultancy; Agios: Consultancy; Imara: Consultancy; Apopharma: Research Funding; Novartis: Research Funding. Porter:Protagonism: Honoraria; Celgene: Consultancy, Honoraria; Bluebird bio: Consultancy, Honoraria; Agios: Consultancy, Honoraria; La Jolla: Honoraria; Vifor: Honoraria; Silence therapeutics: Honoraria. Thrasher:Rocket Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Orchard Therapeutics: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Generation Bio: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; 4BIOCapital: Membership on an entity's Board of Directors or advisory committees. Thuret:BlueBird bio: Other: investigators for clinical trials, participation on scientific/medical advisory board; Celgene: Other: investigators for clinical trials, participation on scientific/medical advisory board; Novartis: Other: investigators for clinical trials, participation on scientific/medical advisory board; Apopharma: Consultancy. Elliot:bluebird bio, Inc.: Employment, Equity Ownership. Tao:bluebird bio, Inc.: Employment, Equity Ownership. Colvin:bluebird bio, Inc.: Employment, Equity Ownership. Locatelli:Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bellicum: Consultancy, Membership on an entity's Board of Directors or advisory committees; bluebird bio: Consultancy; Miltenyi: Honoraria.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2733-2733 ◽  
Author(s):  
Jorge E. Cortes ◽  
Akil Merchant ◽  
Catriona Jamieson ◽  
Daniel A Pollyea ◽  
Michael Heuser ◽  
...  

Abstract Background: In a previously reported Phase 2 randomized study of patients with acute myeloid leukemia (AML), addition of the investigational agent glasdegib (PF-04449913) to low-dose cytarabine (LDAC) improved overall survival (OS) when compared with LDAC alone. In a non-randomized study arm, glasdegib together with 7+3 chemotherapy was well tolerated and associated with clinical activity. We used a comprehensive biomarker analysis, evaluating gene expression, circulating cytokine levels, and gene mutations, to identify molecular drivers that predict overall response (OR) and OS. Methods: In this Phase 2 multicenter study (NCT01546038), patients with AML who were suitable for non-intensive therapy were randomized (2:1) to LDAC + glasdegib 100 mg QD or LDAC alone, and patients suitable for intensive therapy were assigned 7+3 plus glasdegib 100 mg QD. Whole blood, serum, and bone marrow aspirate samples were collected at baseline, and used to assess 19 genes for expression analysis, 38 analytes for circulating cytokine levels, and 109 genes for mutation analysis. Gene expression was analyzed using TaqMan Low Density Array Cards (TLDCs), cytokine levels were analyzed using quantitative, multiplexed immunoassays (Myriad RBM), and mutation analysis was performed using the Illumina® MiSeq instrument (San Diego, CA). All correlations were performed either for OS or for OR. For gene expression and cytokine analysis, a cut-off value above or below the median expression level for each treatment arm was used to separate samples into two subgroups (< or ≥ the median value) to explore the relationship of expression levels with OS data. Criteria for significance in the non-intensive cohort required one subgroup to have a p-value of <0.05 in the between-treatment arms comparison and the HR difference between the two subgroups to be ≥2 fold. Responses were defined as patients with a complete remission (CR), CR with incomplete blood count recovery (CRi), morphologic leukemia-free state, partial remission (PR), or PRi. For response correlations, genes or cytokines were considered to be differentially expressed if they had a p-value <0.05 and were differentially expressed by ≥2-fold. Results: Within the non-intensive arm (LDAC + glasdegib, n=68; LDAC alone, n=30), expression levels of several genes correlated with improved OS with glasdegib plus LDAC. Lower levels of expression of FOXM1 and MSI2, and higher expression levels of BCL2 and CCND2 correlated with improved OS with the combination. Additionally, lower levels of the cytokines 6CKINE (CCL21), ICAM-1, MIP-1α, and MMP-3 correlated with improved OS. An analysis of correlations of gene expression and cytokine levels with OR could not be completed due to the low number of responders in the LDAC only group (n=2). In the intensive treatment arm (glasdegib and 7+3, n=59), higher PTCH1 expression correlated with improved OS (p=0.0219, median OS 10.8 versus 39.5 months). In this cohort, lower levels of IL-8 (p=0.0225) and MIP-3β (p=0.0403) correlated with lower OS. Expression levels of no genes or cytokines significantly correlated with OR in this arm. We also examined correlations between gene mutation status and OS in both study arms. In the non-intensive arm (LDAC + glasdegib, n=58; LDAC alone, n=25), no genes mutated in at least 5 patients correlated with OS. In the intensive treatment arm (n=47), mutations in FLT3, TP53, CEP170, NPM1, and ANKRD26 correlated with OS (all p<0.05). Patients in this arm with FLT3 mutations responded better than patients with wild type FLT3 (p=0.0336, median OS of 13.1 months versus unreached for FLT3 mutant). Conclusions: In this biomarker analysis, we found that expression levels of a select number of genes and circulating cytokines implicated in AML correlated with OS in the non-intensive and the intensive arms. The improved response for patients with FLT3 mutations and high PTCH1 expression levels in the intensive arm deserves further investigation. These findings need to be verified in larger controlled studies, which are ongoing. Disclosures Cortes: Novartis: Consultancy, Research Funding; Daiichi Sankyo: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Astellas Pharma: Consultancy, Research Funding; Arog: Research Funding. Pollyea:Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy; Celyad: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Research Funding; Curis: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Heuser:Astellas: Research Funding; Daiichi Sankyo: Research Funding; Sunesis: Research Funding; Tetralogic: Research Funding; Bayer Pharma AG: Consultancy, Research Funding; StemLine Therapeutics: Consultancy; Janssen: Consultancy; Pfizer: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; BergenBio: Research Funding; Karyopharm: Research Funding. Chan:Pfizer: Employment, Equity Ownership. Wang:Pfizer: Employment, Equity Ownership. Ching:Pfizer Inc: Employment, Equity Ownership. Johnson:Pfizer Inc: Employment, Equity Ownership. O'Brien:Pfizer Inc: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2883-2883
Author(s):  
Matthew S. Davids ◽  
Andrew W. Roberts ◽  
William G. Wierda ◽  
Kathryn Humphrey ◽  
Debbie J Alter ◽  
...  

Abstract Introduction: Venetoclax is a selective, oral inhibitor of BCL2, a key regulator of the intrinsic apoptotic pathway. The dose-escalation phase 1 study of venetoclax in patients with relapsed/refractory non-Hodgkin lymphoma (NHL) enrolled 106 patients from June 2011, and the overall response rate across the entire NHL cohort was 44%. The highest response rate (75%) was seen in the 28 patients with mantle cell lymphoma (MCL) (Davids et al., J Clin Oncol. 2017). Here, we report longer-term outcomes for those patients, now with a median of 27 months (range: 0.2 - 59) follow up. Methods: Venetoclax was administered in dose cohorts ranging from a maximum dose of 300-1200 mg and continued until progressive disease (PD) or unacceptable toxicity; intra-patient dose escalation was allowed. Adverse events (AEs) were assessed by NCI-CTCAE v4.0 and responses were assessed using 2007 Cheson IWG response criteria, utilizing CT scans beginning at week 6. The data cut off for this analysis was June 4th, 2018. Results: For the 28 patients with MCL, the median age was 72 years (range: 35 - 85). They had received a median of 3 (range: 1 - 7) prior treatments; 5 patients received prior PI3K inhibitor (but no prior ibrutinib). The median time from the preceding treatment to start of venetoclax was 13 months (range: 2 - 148). The median dose of venetoclax was 400 mg/day; 25/28 received at least 400mg/day. Median time on study drug was 11 months (range: 0.2 - 59). Three patients have been on therapy for over 4 years. The overall response rate was 75%, with 6 (21%) patients achieving complete remission (CR) and 15 (54%) partial response (PR). The median duration of response was 16 months (95% CI: 4, 30) and median progression free survival was 11 months (95% CI: 5, 21) for all patients (Figure). The 2 year PFS estimate was 30% (95% CI: 14%, 47%) for all patients, 83% (95% CI: 27%, 97%) for patients who achieved CR and 14% (95% CI: 2%, 37%) for patients who achieved PR. One patient who achieved PR proceeded to allogeneic stem cell transplant and remained disease free at the last protocol defined follow-up (24 months after coming off study). Three patients developed progressive disease after receiving venetoclax for more than two years of therapy (time to progression: 31, 33, and 33 months). Two patients with CR continue on study without evidence of progression, currently at 47 and 59 months of venetoclax monotherapy. The most common (≥25% of patients with MCL) all grade treatment emergent AEs were nausea (57%), diarrhea (50%), fatigue (39%), constipation (29%) and upper respiratory infection (25%). The most common (≥10% of patients with MCL) grade 3/4 AEs were neutropenia (14%), anemia (14%), pneumonia (11%), and thrombocytopenia (11%). Biochemical tumor lysis syndrome (TLS), without accompanying clinical features, was reported in one patient considered high risk for TLS. Specific interventions were not required, and the patient continued on study drug. Conclusions: Venetoclax monotherapy leads to durable remission in a meaningful proportion of patients with pretreated MCL. Further studies in MCL are currently investigating potential biomarkers for durable response to venetoclax combination regimens, including a Phase 3 randomized study with ibrutinib (SYMPATICO, NCT03112174). Disclosures Davids: Roche/Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astra-Zeneca: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; BMS: Research Funding; Merck: Consultancy; AbbVie, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; TG Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Verastem: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy; MEI Pharma: Consultancy, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees; Surface Oncology: Research Funding. Roberts:Walter and Eliza Hall: Employment, Patents & Royalties: Employee of Walter and Eliza Hall Institute of Medical Research which receives milestone and royalty payments related to venetoclax; AbbVie: Research Funding; Genentech: Research Funding; Janssen: Research Funding. Wierda:Genentech: Research Funding; AbbVie, Inc: Research Funding. Humphrey:F. Hoffmann-La Roche Ltd: Employment, Equity Ownership. Alter:AbbVie, Inc: Employment, Equity Ownership. Masud:AbbVie, Inc: Employment, Equity Ownership. Buss:Abbvie, Inc: Employment, Equity Ownership. Verdugo:AbbVie, Inc: Employment, Equity Ownership. Seymour:Janssen: Honoraria, Research Funding; Celgene: Consultancy; Genentech Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; F. Hoffmann-La Roche Ltd: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Honoraria, Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 454-454 ◽  
Author(s):  
Amir T. Fathi ◽  
Harry P Erba ◽  
Jeffrey E Lancet ◽  
Eytan M Stein ◽  
Roland B. Walter ◽  
...  

Abstract Background Older patients with AML who are not candidates for intensive therapy are typically treated with hypomethylating agents (HMAs) or other low intensity therapy. HMAs have been shown to upregulate CD33 and to increase sensitivity to cytotoxic chemotherapy by decreasing apoptotic threshold in tumor cells. SGN-CD33A (or 33A) is a CD33-directed antibody conjugated to 2 molecules of a pyrrolobenzodiazepine (PBD) dimer. Upon binding, 33A is internalized and transported to the lysosomes where PBD dimer is released via proteolytic cleavage of the linker, crosslinking DNA, and leading to cell death. In preclinical studies combining 33A with an HMA (azacitidine and decitabine), synergy has been demonstrated in multidrug resistant AML models (Sutherland ASH 2014). Methods A combination cohort in a phase 1 study (NCT01902329) was designed to evaluate the safety, tolerability, pharmacokinetics (PK), and anti-leukemic activity of 33A in combination with an HMA. Eligible patients (ECOG 0-1) must have previously untreated CD33-positive AML, and have declined intensive therapy. A single dose level of 33A, 10 mcg/kg, was administered outpatient IV every 4 weeks on the last day of HMA (azacitidine or decitabine [5 day regimen], standard dosing). Patients with clinical benefit may continue treatment until relapse or unacceptable toxicity. Investigator assessment of response is per IWG criteria; CRi requires either platelet count of ≥100,000/µL or neutrophils of ≥1,000/µL (Cheson 2003). Results To date, 24 patients (63% male) with a median age of 77 years (range, 66-83) have been treated with the combination therapy. 42% of patients had adverse cytogenetics (MRC), 23 patients were treatment naïve and 1 patient had received prior low intensity therapy for MDS. At baseline, patients had a median of 60% BM blasts (range, 2%-90%) and a median of WBC of 2.2 (range, 1-132). At the time of this interim analysis, patients were on treatment for a median of 13.5+ weeks with 17 patients continuing treatment; no DLTs have been reported. Grade 3 or higher adverse events (AE) reported in >20% of patients were fatigue (54%), febrile neutropenia (46%), anemia (25%), neutropenia (25%), and thrombocytopenia (21%). Other treatment-emergent AEs regardless of relationship to study treatment reported in ˃20% of patients were nausea (29%), decreased appetite (25%), and constipation (21%). Thirty- and 60-day mortality rates are 0% and 4% respectively with no treatment-related deaths reported. Fifteen of the 23 efficacy evaluable patients (65%) achieved CR (5) or CRi (10). Remissions were generally obtained after 2 cycles of treatment and were observed in many patients with adverse risk including underlying myelodysplasia (6/8, 75%) and adverse cytogenetics (8/9, 89%). Median OS has not been reached with 20 patients alive at the time of this data cut. Conclusions The combination of 33A with HMA appears to be well-tolerated, active, and has no identified off-target toxicities. Activity with the combination compares favorably with historical experience with HMAs alone in this patient population. The CR+CRi rate of 65% in AML patients with poor risk factors with the observed low 60-day mortality (4%) are particularly encouraging. These promising data warrant further evaluation in future trials. Disclosures Fathi: Agios Pharmaceuticals: Other: Advisory Board participation; Merck: Other: Advisory Board participation; Seattle Genetics: Other: Advisory Board participation, Research Funding. Off Label Use: SGN-CD33A is an investigational agent being studied in patients with CD33-positive AML. SGN-CD33A is not approved for use.. Erba:GlycoMimetics; Janssen: Other: Data Safety & Monitoring Committees; Sunesis;Pfizer; Daiichi Sankyo; Ariad: Consultancy; Millennium/Takeda; Celator; Astellas: Research Funding; Seattle Genetics; Amgen: Consultancy, Research Funding; Novartis; Incyte; Celgene: Consultancy, Patents & Royalties. Lancet:Seattle Genetics: Consultancy; Pfizer: Research Funding; Boehringer-Ingelheim: Consultancy; Kalo-Bios: Consultancy; Amgen: Consultancy; Celgene: Consultancy, Research Funding. Stein:Seattle Genetics, Inc.: Membership on an entity's Board of Directors or advisory committees; Agios: Membership on an entity's Board of Directors or advisory committees. Walter:Pfizer, Inc.: Consultancy; Covagen AG: Consultancy; AstraZeneca, Inc.: Consultancy; CSL Behring: Research Funding; AbbVie, Inc.: Research Funding; Amgen: Research Funding; Amphivena Therapeutics, Inc.: Consultancy, Research Funding; Seattle Genetics, Inc.: Consultancy, Research Funding. DeAngelo:Incyte: Consultancy; Amgen: Consultancy; Pfizer: Consultancy; Ariad: Consultancy; Bristol Myers Squibb: Consultancy; Novartis: Consultancy; Celgene: Consultancy; Agios: Consultancy. Faderl:Celator: Research Funding; Ambit: Research Funding; BMS: Research Funding; Astellas: Research Funding; Karyopharm: Consultancy, Research Funding; Seattle Genetics, Inc.: Research Funding; JW Pharma: Consultancy; Celgene: Consultancy, Research Funding, Speakers Bureau; Pfizer: Research Funding; Onyx: Speakers Bureau. Jillella:Seattle Genetics, Inc.: Research Funding. Bixby:Seattle Genetics, Inc.: Research Funding. Kovacsovics:Seattle Genetics, Inc.: Research Funding. O'Meara:Seattle Genetics, Inc: Employment, Equity Ownership. Kennedy:Seattle Genetics, Inc.: Employment, Equity Ownership. Stein:Amgen: Speakers Bureau.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1950-1950 ◽  
Author(s):  
John A Lust ◽  
Charles Barranco ◽  
Saad Z Usmani ◽  
Frits van Rhee ◽  
Mehdi Hamadani ◽  
...  

Abstract Eukaryotic translation initiation factor 5A (eIF5A) has been implicated in the regulation of cell proliferation, apoptosis, and inflammation, and is the only known protein to be modified by hypusination. Hypusinated eIF5A, the predominant form of eIF5A in cancer cells, is involved in cell survival and activation of inflammatory pathways. In contrast, accumulation of the unhypusinated form of eIF5A is associated with apoptosis and mutants of eIF5A that cannot be hypusinated (e.g. eIF5AK50R) are pro-apoptotic. SNS01-T was designed to treat B-cell cancers and consists of two active components: a plasmid DNA expressing the pro-apoptotic eIF5AK50R under the control of a B cell-specific promoter, and an siRNA against an untranslated region of native eIF5A mRNA. When these two components are combined with linear polyethyleneimine (PEI), the nucleic acids are condensed into nanoparticles for protection from degradation in the blood and enhanced cellular delivery. The mode of action of SNS01-T is siRNA-mediated inhibition of hypusinated eIF5A and simultaneous over-expression of pro-apoptotic eIF5AK50R to induce cell death. In vitro cell studies and in vivo xenograft studies have demonstrated the efficacy of this approach. The safety and tolerability of intravenous administration of SNS01-T is being investigated in a first-in-human Phase1b/2a study in patients with relapsed or refractory multiple myeloma (MM), mantle cell lymphoma (MCL) or diffuse large B cell lymphoma (DLBCL). Eligible patients are being enrolled sequentially into four cohorts at increasing doses. Each patient receives an intravenous infusion of SNS01-T twice weekly for 6 consecutive weeks. Eligible patients must have been diagnosed with MM according to IMWG criteria, or with MCL or DLBCL with histologic confirmation. Patients also must have measurable disease, have relapsed or refractory disease after two or more prior treatment regimens, have a life expectancy of at least 3 months, and not be eligible to receive any other standard therapy known to extend life expectancy. The primary objective is to evaluate the safety and tolerability of multiple escalating doses of SNS01-T. Secondary objectives include analysis of pharmacokinetics, immunogenicity, pro-inflammatory cytokines, and therapeutic efficacy. The required 3 patients per cohort have completed the dosing schedule in cohorts 1 and 2 from a total of 10 patients enrolled (9 patients with MM and 1 with DLBCL). Of the ten patients enrolled, four completed the full treatment period, two did not complete dosing but were evaluable for safety, and four (three in cohort 1 and one in cohort 2) discontinued treatment after fewer than 8 doses and were not evaluable. There were no drug-related serious adverse events or dose limiting toxicities in either cohort 1 or 2. In cohort 1 (0.0125 mg/kg SNS01-T), two of three evaluable patients did not progress on treatment and were considered stable at week 3 and week 6, the end of the dosing regimen. The third patient progressed after receiving 10 of the 12 doses and was evaluable for safety. In cohort 2 (0.05 mg/kg), 3 patients (2 with MM and 1 with DLBCL) were evaluable for safety. Stabilization of serum monoclonal protein levels was observed in one MM patient of cohort 2. Two patients (1 with MM and 1 with DLBCL) progressed after receiving 8 of the 12 doses and were evaluable for safety. Results from ongoing pharmacokinetic studies, immunogenicity studies, and quantification of pro-inflammatory cytokines will be discussed. The planned dose levels for the third and fourth groups are 0.2 and 0.375 mg/kg, respectively. The results to date of this first-in-human clinical trial indicate that SNS01-T can be administered safely and the MTD has not yet been reached (Clinical Trials.gov Identifier: NCT01435720). Disclosures: Barranco: Senesco Technologies: Consultancy. Usmani:Celgene, Onyx, Millenium: Consultancy, Research Funding, Speakers Bureau. van Rhee:Jansen&Jansen: Research Funding. Thompson:Senesco Technologies: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Research Funding. Taylor:Senesco Technologies: stock options Other. Dondero:Senesco Technologies: Employment. Browne:Senesco Technologies Inc.: Employment, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees. Siegel:Celgene, Millenium, Onyx (same for all): Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3541-3541 ◽  
Author(s):  
Emily Leven ◽  
Loan Hsieh ◽  
Kavitha Ramaswamy ◽  
Catherine E. McGuinn ◽  
Diane Nugent ◽  
...  

Abstract Introduction The thrombopoietin (TPO) mimetic agents romiplostim and eltrombopag are used to stimulate platelet production and increase platelet counts in chronic immune thrombocytopenia (ITP) in adults. While two large, randomized trials investigating dosing, safety, and efficacy were ongoing in children, but not near completion, we conducted a retrospective IRB-approved analysis of 33 pediatric (≤21 years) chronic ITP patients from two centers treated with TPO agents off-study. Patients Patients initiated TPO therapy prior to 21 years of age (19 months to 19 years, median 14 years) after an average of 3.6 ITP therapies; 21 patients received romiplostim [11 at Children's Hospital of Orange County (CHOC), 10 at Weill Cornell Medical Center (WCMC)] and 12 eltrombopag [all at WCMC]. Median starting age was 11.5 years for patients on romiplostim and 16.5 years on eltrombopag. Primary response measures were: platelet counts ≥ 50 x 109 per liter or ≥20 x 109 per liter above baseline for 2 consecutive weeks and 50% of platelet counts ≥ 50 x 109 per liter. Duration of treatment and bone marrows with myelofibrosis (MF) consensus grade are shown in Figure 1. Results 27/33 (82%) patients responded to TPO agents; 18/21 to romiplostim and 9/12 to eltrombopag. Ten patients responded to ROMIPLOSTIM as a single agent; 8 romiplostim responders received concurrent medications including: mycophenolate mofetil (MMF), azathioprine (AZA), rituximab, and cyclosporine (CSA). One of two splenectomized patients was able to wean off TPO therapy with adequate counts. Two additional patients successfully discontinued TPO therapy and continued on MMF alone. Two patients (1 CSA, 1 AZA) successfully discontinued TPO therapy and then weaned off concurrent therapy. Eighteen of 21 patients had platelet counts ≥ 50 x 109 per liter or ≥20 x 109 per liter above baseline for 2 consecutive weeks and 18 had 50% of platelet counts ≥ 50 x 109 per liter (table). In this study, duration of successful romiplostim use outside of randomized clinical trials ranged from 6-44 months (11/18 ongoing). 3 patients did not respond and 2/3 went on to have therapeutic splenectomies. 1 patient with Evans syndrome had a transient 1 year response before relapsing. 5 responders to romiplostim had previously received eltrombopag with lesser or no effect. Nine of 12 patients responded to ELTROMBOPAG with platelet counts ≥ 50 x 109 per liter or ≥20 x 109 per liter above baseline for 2 consecutive weeks and 8/12 had 50% of platelet counts ≥ 50 x 109 per liter. One patient had been splenectomized; 7 responded to eltrombopag alone. 2 patients had concurrent therapy with prednisone and IV Anti-D and 1/2 was able to discontinue concurrent prednisone. One patient successfully discontinued eltrombopag with adequate counts for > 1 year. 5 patients attempted unsuccessfully to discontinue therapy. One responder, a previous romiplostim responder, switched to eltrombopag. No other patients were treated with TPO agents prior to starting eltrombopag. Duration of successful eltrombopag use outside of randomized clinical trials ranged from 23-53 months (7/12 are ongoing). Three patients did not respond to eltrombopag; 1/3 was HIV positive. One patient experienced a provoked DVT at site of ankle fracture while using eltrombopag. No other patients on either therapy experienced serious drug-related adverse events. Among 24 bone marrows (both agents; 21 reflecting off study use), only MF grades 0 -1 were seen. 10/24 marrows were performed after greater than 2 years of therapy. Conclusion Retrospective analysis of off-study use of eltrombopag and romiplostim in children shows that these TPO agents effectively increase platelet counts in more than 3/4 of children with chronic ITP, a result consistent with adult TPO use. Despite small sample size, the long-term duration of successful use (6-53 months) without tachyphylaxis supports efficacy. Re safety, the one DVT appeared to be precipitated by a fracture and there were no MF2/3 bone marrows in the 24 samples. In those patients who do not respond to one TPO agent, it may be beneficial to switch to the other form. Disclosures: Nugent: Bayer: Honoraria; CSL Behring: Honoraria; Novo Nordisk: Honoraria. Bussel:Shionogi: Membership on an entity’s Board of Directors or advisory committees, Research Funding; Eisai: Membership on an entity’s Board of Directors or advisory committees, Research Funding; Ligand: Membership on an entity’s Board of Directors or advisory committees, Research Funding; Immunomedics: Research Funding; IgG of America: Research Funding; Genzyme: Research Funding; Cangene: Research Funding; GlaxoSmithKline: Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Amgen: Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Sysmex: Research Funding; Symphogen: Membership on an entity’s Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2815-2815 ◽  
Author(s):  
Preetesh Jain ◽  
Shuangtao Zhao ◽  
Rashmi Kanagal-Shamanna ◽  
Lucy Navsaria ◽  
Holly Hill ◽  
...  

Introduction: Mantle cell lymphoma (MCL) patients (pts) who progress after ibrutinib and other lines of treatment "ultra-refractory MCL" have poor outcomes and exhibit BTK mutations infrequently (Jain P et al BJH 2018, Martin P et al Blood 2016). Venetoclax has shown promising efficacy in Phase I trial in NHL (Davids M et al JCO 2017) and is now under trials in MCL. Venetoclax response in pts with MCL after progression on ibrutinib was reported (Eyre T et al Haematologica 2018), however, genomic alterations associated with venetoclax resistance are not described. We present our experience in 24 pts with MCL treated with venetoclax and report their mutation profiles associated with progression on venetoclax. Methods: We collected data from 24 pts with MCL who were treated with venetoclax (off clinical trial) as a salvage measure after failing multiple lines of prior therapies. Pt characteristics were collected from the time of initiating venetoclax. Progression free survival (PFS) was calculated from the time of initiating venetoclax to the date of progression or to last follow up date/date of death while overall survival (OS) was calculated from the time of initiating venetoclax to the date of last follow up date/date of death. Post venetoclax survival was calculated from the date of discontinuing venetoclax to the date of last follow up/death. Whole-exome sequencing (WES) with SureSelect Human All Exon V6 was performed from evaluable biopsy samples from 7 pts (5 pts at/before starting venetoclax and 6 pts after progression of venetoclax), this included 5 pts who have pairs available for analysis (pre and post venetoclax). Results: Twenty four pts were treated with venetoclax (12 started as single agent and 8 started with combination with obinutuzumab and 3 with BTK inhibitors with/without obinutuzumab). Four pts had initial single agent venetoclax and later were rechallenged with combinations. Initial dose of venetoclax was dose escalation from 20 mg, then 50 mg then 100 mg PO daily up to 400 mg daily in 18/24 pts while in 3 pts it was 100 mg daily and in another 3 it was 400 mg daily. Median age at venetoclax start was 69 years (58-82). Median number of prior lines of therapy was 5 (range 1-11; including 17 pts who progressed on ibrutinib or other BTK inhibitors, 5 had exposure to ibrutinib and discontinued for intolerance, 4 had prior SCT and 2 had prior anti CD19 cellular therapy). At the baseline (pre/at-venetoclax start), 13 pts (54%) had blastoid/pleomorphic histology and 11 (46%) had classic variant morphology, the median Ki-67% was 60% (5-90%) and pts with Ki-67% ≥ 50 were 11 (55%), 4 pts did not have available Ki-67% values. Overall response rate (ORR) was 65% (13/20) - complete remission 25% (5/20) and partial remission 40% (8/20). Stable disease was observed in 10% (2/10), primary refractory were 25% (5/20). Four pts were not evaluable for response assessment. The median follow up after starting venetoclax was 17.5 months (1-27). The median PFS was 7.7 months (2 year 20%) and the median OS was 13.5 months (2 year 30%) Figure-1A-B. Pts in CR had a PFS of 15 months vs no CR 10 months (p=0.29). At the last follow up, 11 pts remained on venetoclax therapy (4 alive and 7 dead). Overall, 15 pts progressed and 14 pts were alive. The median post venetoclax survival was 6 months. Among 20 pts who discontinued venetoclax, 1 achieved CR and 3 PR on subsequent therapies. Among the 20 pts who discontinued venetoclax, 6 discontinued due to intolerance. In addition, we evaluated the somatic mutation profile in pts who progressed on venetoclax using WES. Figure-1C shows mutation spectrum. In our cohort, pts with MCL who progressed on venetoclax exhibited infrequent Bcl2 mutations (one pt at progression; 14 %; p.H3D) while the mutation frequency of other genes such as TP53 (71% vs. 40%), ATM (43% vs. 20%), KMT2D (57% vs. 20%), CELSR3 (57% vs. 20%), and KMT2C (43% vs. 20%) increased by > 2-fold at progression (compared to pretreatment samples, p=N.S due to small cohort size). The mutation of CARD11 (14%) and SMARCA4 (14%) was only observed at progression. Further details on copy number abnormalities will be presented. Conclusions: Venetoclax has promising results in refractory pts with MCL. Combination clinical trials with obinutuzumab, acalabrutinib are ongoing in MCL. We have characterized mutations and aneuploidy abnormalities in venetoclax resistant MCL pts and shown that unlike CLL, Bcl2 mutations are infrequent in venetoclax resistant MCL. Disclosures Nastoupil: Spectrum: Honoraria; TG Therapeutics: Honoraria, Research Funding; Novartis: Honoraria; Janssen: Honoraria, Research Funding; Gilead: Honoraria; Genentech, Inc.: Honoraria, Research Funding; Bayer: Honoraria; Celgene: Honoraria, Research Funding. Westin:Novartis: Other: Advisory Board, Research Funding; MorphoSys: Other: Advisory Board; Curis: Other: Advisory Board, Research Funding; Janssen: Other: Advisory Board, Research Funding; Kite: Other: Advisory Board, Research Funding; Juno: Other: Advisory Board; Celgene: Other: Advisory Board, Research Funding; Unum: Research Funding; Genentech: Other: Advisory Board, Research Funding; 47 Inc: Research Funding. Fowler:Abbvie: Membership on an entity's Board of Directors or advisory committees, Research Funding; TG Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding. Wang:Aviara: Research Funding; Dava Oncology: Honoraria; Juno Therapeutics: Research Funding; Celgene: Honoraria, Research Funding; BioInvent: Consultancy, Research Funding; Guidepoint Global: Consultancy; Kite Pharma: Consultancy, Research Funding; Acerta Pharma: Consultancy, Research Funding; MoreHealth: Consultancy, Equity Ownership; Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Pharmacyclics: Honoraria, Research Funding; AstraZeneca: Consultancy, Honoraria, Research Funding, Speakers Bureau; Loxo Oncology: Research Funding; VelosBio: Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4140-4140
Author(s):  
Alessia Castellino ◽  
Melissa C. Larson ◽  
Matthew J. Maurer ◽  
Susan L. Slager ◽  
Cristine Allmer ◽  
...  

Abstract Backgroud. The treatment landscape of mantle cell lymphoma (MCL) has significantly changed in last decades. Improvement in diagnosis and understanding of disease biology has been coupled with emergence of new therapeutic options, including targeted agents. While MCL outcome data comes primarily from clinical trials (CT), the impact of therapeutic advances on pattern of care (POC) and outcome in MCL in the general population is not well characterized. This study sought to characterize changes in pattern of care and outcomes of patients with MCL in a prospective observational series in the rituximab era. Methods. The study included consenting adult patients with newly diagnosed MCL that were prospectively enrolled into the University of Iowa/Mayo Clinic Lymphoma Specialized Program of Research Excellence (SPORE) Molecular Epidemiology Resource, from 09/01/2002 to 06/30/2015. Demographic, clinical and prognostic factors were abstracted, and all patients were actively followed for re-treatment, relapse and death. Since bendamustine-rituximab (BR) regimen starting to be used in 2010, we defined patients enrolled from 09/01/2002 to 12/31/2009 as era 1 and those enrolled 01/01/2010 to 06/30/2015 as era 2. Baseline characteristics and outcomes, evaluated in terms of event free survival (EFS), overall survival (OS) and cause of death, were analyzed and compared between the two eras identified. Results. 348 patients with newly diagnosed MCL were enrolled. Five patients had no available data for front-line treatment and were excluded. The analysis was thus conducted on 343 patients: 169 patients were diagnosed in era 1 with a median follow-up of 131.2 months vs. 174 in era 2 with a median follow-up of 58.9 months. Baseline clinical characteristics and MIPI score were similar across the two eras ( Table 1). Frontline induction treatment was significantly different in the two eras. BR use was 0 vs 49 (28.2% ), R-CHOP/CHOP like regimen in 89 (52.7%) vs 45 (25.9%), high-dose cytarabine (HiDAC)-based therapy in 1 (0.6%) vs 28 (16.1%), intensified regimens (HyperCVAD) in 16 (9.5%) vs 8 (4.5%), other regimens (including R-cladribine, R-fludarabine-mitoxantrone, rituximab monotherapy) in 35 (20.7%) vs 13 (7.5%) patients in era 1 vs era 2 respectively. Non-systemic treatment (observation, surgery or radiation only) was performed in 25 (14.8%) vs 31 (17.8%), while 9 (8%) vs 12 (7.7%) patients were enrolled in clinical trials, in era 1 vs era 2 respectively. Autologous stem cell transplantation (ASCT) as consolidation of first line treatment or use of Rituximab maintenance was not different between Era 1 and Era 2. Among the entire cohort of 343 MCL patients, 3y-EFS and 3y-OS were 51.9% (95% IC 46.7-57.6) and 73.5 (95% CI 68.8-78.4), respectively (Figure 1). 3y-EFS was 45.9% (95% CI 39.0-54.1%) vs 58.4 (95% CI, 51.2-66.6%) in Era 1 vs Era 2 (HR 0.69 (0.51-0.92), p=0.006), 3y-OS was 70.9% (95% CI, 63.4-78.1%) vs 76.1% (95% CI, 69.7-83.1%) in Era 1 vs Era 2 (HR 0.87 (0.61-1.24), p=0.26), respectively (Figure 1). In a univariate analysis, high risk simplified MIPI was prognostic of lower EFS and OS in both the era groups. Conclusion. The BR regimen entered front line therapy of MCL resulting in decline of R-CHOP use in the MER. While rates of ASCT remain similar over the two eras, high dose cytarabine usage in induction therapy has increased. While POC in MCL continue to evolve, the introduction of bendamustine and high-dose citarabine based regimens resulted in an improvement in EFS but not OS in this observational cohort-based analysis. Findings in this study are important for design and planning of future clinical trials incorporating novel agents in induction therapy of MCL. Figure 1. Figure 1. Disclosures Cohen: Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Infinity Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; BioInvent: Consultancy; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding; Bristol-Myers Squibb: Research Funding; Infinity Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Research Funding; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Research Funding; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; BioInvent: Consultancy; Seattle Genetics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Research Funding. Witzig:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Cerhan:Celgene: Research Funding; Jannsen: Other: Scientific Advisory Board; Nanostring: Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1765-1765
Author(s):  
Srdan Verstovsek ◽  
Jingbo Yu ◽  
Jonathan K. Kish ◽  
Dilan Chamikara Paranagama ◽  
Jill Kaufman ◽  
...  

Abstract Introduction Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by marrow fibrosis, extramedullary hematopoiesis, and leukoerythroblastosis. Clinical manifestations include severe anemia, splenomegaly, and symptoms. Median survival in patients with primary MF ranges from 2 to 11 years, depending on risk categorization. The National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines recommend the International Prognostic Scoring System (IPSS) for risk stratification at diagnosis; other systems including the Dynamic IPSS (DIPSS) and the DIPSS-Plus are also cited in the Guidelines. Treatment recommendations are risk-adapted in the NCCN Guidelines. The objective of this study is to describe how patients are risk stratified at diagnosis by community hematologists/oncologists and the impact that risk stratification has on the initiation of MF-directed treatments. Methods Medical chart reviews were conducted at community hematology/oncology practices in the OPEN network. Adult patients diagnosed with primary MF, post-PV MF, or post-ET MF between 1/2012 and 12/2016 and receiving care for at least 6 months were included. Data were collected with an electronic case report form (eCRF) with questions on clinical characteristics (symptoms, Hgb, WBC, blast %, and PLT) and risk assessment method used at diagnosis (IPSS, DIPSS, or DIPSS-Plus), treatments, and outcomes. A data-derived IPSS risk score was calculated for each patient. To assess the accuracy of the assigned risk, a data-derived risk score, corresponding to the system used by the provider, was also calculated. Patients were classified as treated at diagnosis if they received MF-directed therapy (hydroxyurea, interferon, ruxolitinib, or clinical trial) or allogeneic hematopoietic cell transplant (HCT) within 120 days of diagnosis. The methods and rates of risk stratification, accuracy of the provider-assigned risk versus data-derived risk, and treatment administered were reported. Results A total of 338 patients with MF from 28 community hematology/oncology practices were included. Mean (SD) age at diagnosis was 65.3 (11.8) years, 51.8% were male, and 68.3% had primary MF. JAK2, MPL, and CALR mutations were tested in 86.1%, 70.1%, and 60.9% of patients at diagnosis, of these, 71.1%, 23.2%, and 14.6% were positive, respectively; 18.4% (38/206) were triple negative. Median follow-up from diagnosis was 27.5 months (IQR, 18.5-42.6). Approximately 32% of patients did not have a risk classification in their medical records at diagnosis. A scoring system was used for risk assignment in 45.3% of patients; DIPSS (23.0%) and IPSS (21.3%) were most commonly used. Of all 338 patients, the corresponding data-derived risk classifications were: 5.6% low, 20.1% int-1, 18.3% int-2, and 55.9% high risk. Among those patients who were not assigned risk by their treating physicians (n=108), most had int-1 (28.7%), int-2 (17.6%), or high risk (43.5%) disease based on the data-derived IPSS risk classification. Of those who received a risk classification from their treating physician, 47.4% (n=109) received an inaccurate risk classification; among these patients, the risk was under-estimated for most (82.6%) (Table 1). Overall, 55.8% of patients (63.2% low-risk, 55.9% int-1, 52.5% int-2, 56.1% high-risk) received MF-directed pharmacological treatment or HCT within 4 months of diagnosis. Among all patients receiving MF-directed treatment, the mean time from diagnosis to treatment initiation was 5.3 months (SD=1.8), and the most common first pharmacological treatments were ruxolitinib (49.8%) and hydroxyurea (46.7%). Splenomegaly (81.3%), symptoms (72.6%), and anemia (65.6%) were top cited indications for treatment initiation. The treatment initiation rate was higher among those patients correctly risk classified compared to those incorrectly classified (64.2% versus 49.5%, p=0.032). Conclusions Nearly one-third of patients with MF did not receive a risk classification at diagnosis. When risk was assigned, almost half were incorrectly classified. Just over half of patients received treatment within four months of diagnosis. Patients who were correctly risk classified at diagnosis were more likely to start treatment promptly upon diagnosis versus those incorrectly risk classified, which may be attributable to the under-estimation of risk. Disclosures Verstovsek: Celgene: Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Italfarmaco: Membership on an entity's Board of Directors or advisory committees. Yu:Incyte Corporation: Employment, Equity Ownership. Kish:Cardinal Health: Employment. Paranagama:Incyte: Employment, Equity Ownership. Kaufman:Cardinal Health: Employment. Chung:Cardinal Health: Employment. Grunwald:Genentech: Research Funding; Incyte Corporation: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding; Alexion: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Medtronic: Equity Ownership; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Cardinal Health: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck: Consultancy, Membership on an entity's Board of Directors or advisory committees; Ariad: Consultancy, Membership on an entity's Board of Directors or advisory committees; Forma Therapeutics: Research Funding. Colucci:Incyte: Employment, Equity Ownership. Mesa:UT Health San Antonio - Mays Cancer Center: Employment; NS Pharma: Research Funding; Promedior: Research Funding; Gilead: Research Funding; Incyte Corporation: Research Funding; CTI Biopharma: Research Funding; Genentech: Research Funding; Celgene: Research Funding; Novartis: Consultancy; Pfizer: Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 660-660 ◽  
Author(s):  
Wiba Keke Wermann ◽  
Andreas Viardot ◽  
Sabine Kayser ◽  
Nael Alakel ◽  
Ahmet Elmaagacli ◽  
...  

Abstract Outcome of adult ALL has improved considerably during the past decades by intensive chemotherapy, which still remains a challenge in older pts. This may be partly due to comorbidities. So far there are no standards to differentiate pts who will be able to tolerate even age-adapted chemotherapy (fit vs unfit). In addition, little is known about the prevalence of comorbidities. Clinical trials with new compounds often represent a selection of pts w/o comorbidities. There is also no generally accepted tool for comorbidity scoring. The goal of this analysis is to provide reference data for pre-existing comorbidities in a large set of adult ALL pts, to compare two different tools and to evaluate the impact on early death (ED) in older pts. The German Multicenter Study Group for Adult ALL (GMALL) has collected data from trials for younger (18-55 y) and older (>55 y) pts and from a prospective registry. Trials had very limited exclusion criteria and in the registry there are no exclusion criteria. The Charlson Comorbidity Index (CCI) was assessed in the GMALL Elderly trial, whereas the Sorror Score (HCT-CI) was used in trials for younger pts and in the registry. 879 pts had a documented HCT-CI score from GMALL 08/2013 trial (N=282;group 1) and 3 groups from the registry: >55 y but eligible for intensive therapy (N=56, group 2), > 55 y in GMALL Elderly protocol (N=505, group 3) and >55 y in GMALL Frail protocol (N=36; group 4) (Table 1). In addition the CCI was documented in 333 pts treated in the GMALL Elderly Trial. HCT-CI-Score: The most frequent comorbidities were infections (17%), prior malignancies (16%), diabetes (16%), cardiac (14%) and moderate pulmonary disease (12%), obesity (11%) and mild liver disease (10%). Arrhythmias (<1%, 5%, 12% and 22% resp. in groups 1, 2, 3, 4), cardiac disease (2%, 7%, 19% and 42% resp.), prior malignancies (2%, 11%, 25% and 22% resp.) and diabetes (4%, 16%, 22% and 22% resp.) increased with age. Infections (15%, 18%, 22% and 22% resp.) or obesity (9%, 7%, 11% and 14% resp.) were not strongly correlated to age. Comorbidity rates were lower in pts >55 y (group 2) considered eligible for intensive therapy (57%) compared to those considered for the Elderly protocol (76%) (group 3). The proportion of low risk (LR) scores decreased with age (54%, 43%, 25% and 8% resp.;p=.01), whereas high risk (HR) increased (18%, 25%, 50% and 59% resp; p=.01). CCI: The most frequent comorbidities were prior malignancy (14%), diabetes (25%) with (3%) or w/o (22%) end organ damage, cardiac (11%) and vascular disease (8%). The incidence of prior malignancy within the last 5 y was 7%. Risk classification was: LR (0) 51%, intermediate risk (IMR) (1-2) 42% and HR (≥3) 7%. HCT-CI vs CCI in pts >55 y: With HCT-CI the incidence of heart diseases (21% arrhythmias, cardiac disease or valve damage) was higher compared to CCI (9%), which differentiated better into cardiac failure (7%) and myocardial infarction (4%). Peripheral vascular disease (8% with CCI) is not assessed by HCT-CI. Liver disease was less frequent with CCI (1.5%) vs HCT-CI (14%) due to different definitions, whereas moderate pulmonary disease (12%) or infections (18%) are not assessed by CCI. The incidences of prior malignancies and diabetes were comparable. Of note, the overall incidence of distinct comorbidities e.g. cardiac was lower than the sum of subentities because some pts had several comorbidities. ED in pts >55 y: ED rates in pts >55 y in group 3 and in GMALL Elderly trial were comparable (13% vs 12% resp). In group 3 ED rates in risk groups (HCT-CI) were 7% vs 13% vs 15% (p>.05). In the GMALL Elderly trial ED in risk groups (CCI) were 9%, 12% and 35% (p=.05; p=.003 LR/IMR vs HR). Overall the analysis reveals a high incidence of comorbidities in older (57-92%) and even in younger pts (46%), which partly would represent contraindications in clinical trials with novel compounds; thus real world data in pts with comorbidities are required after marketing authorisation. HCT-CI and CCI have a different focus and shortcomings. For ALL pts a more specific score with different organ modules would be helpful. Comorbidity is significantly correlated to ED risk. CCI allows to identify a small HR group (7%) with a mortality of 35%. HCT-CI (24% of pts) and even more CCI (51% of pts) allow to identify LR groups with <10% early mortality. It will be of interest to analyse the impact of individual comorbidities on ED rate. Overall structured comorbidity assessment should be part of all clinical trials in ALL. Disclosures Viardot: Gilead Kite: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Amgen: Consultancy; BMS: Consultancy, Honoraria; Roche: Consultancy, Honoraria. Fiedler:Teva: Other: support for meeting attendance; Gilead: Other: support for meeting attendance; Amgen: Other: support for meetíng attendance; Pfizer: Research Funding; Amgen: Research Funding; Amgen: Patents & Royalties; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees; ARIAD/Incyte: Membership on an entity's Board of Directors or advisory committees, support for meeting attendance; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; GSO: Other: support for meeting attendance; JAZZ Pharmaceuticals: Other: support for meeting attendance; Daiichi Sankyo: Other: support for meeting attendance. Stelljes:JAZZ: Honoraria; MSD: Consultancy; Amgen: Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Novartis: Honoraria. Serve:Bayer: Research Funding. Goekbuget:Kite / Gilead: Consultancy; Celgene: Consultancy; Novartis: Consultancy, Other: Travel support, Research Funding; Pfizer: Consultancy, Other: Travel support, Research Funding; Amgen: Consultancy, Other: Travel support, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document