scholarly journals Impact of Atrial Fibrillation on Cardiovascular and Economic Outcomes in Patients with Chronic Lymphocytic Leukemia

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4077-4077
Author(s):  
Anjana Mohan ◽  
Keri Yang ◽  
Sizhu Liu ◽  
Randall Dick ◽  
Boxiong Tang ◽  
...  

Abstract Introduction: Chronic lymphocytic leukemia (CLL) is the most frequently diagnosed hematologic cancer, with many patients diagnosed at a later stage in life and typically with comorbidities that often complicate CLL management. Atrial fibrillation (AF) is the most common arrhythmia in the United States and is associated with high hospitalization and mortality rates. While the relationship between CLL and AF has been reported, there is limited real-world evidence on the clinical and economic impact of AF in CLL patients. The objectives of this study were to examine the impact of AF on cardiovascular (stroke, bleeding events, heart failure) and economic (costs and healthcare resource utilization [HRU]) outcomes among patients with CLL. Methods: This retrospective observational study used the IBM MarketScan Research Databases in Treatment Pathways that includes at least a 10-million sample of the commercial and Medicare supplemental claims database. Patients who were at least 18 years, diagnosed with CLL (identified by ICD-9/ICD-10) and continuously enrolled from January 2009 to July 2020 were included. CLL patients were followed up for one year after their first CLL diagnosis date to examine the incidence of AF. Cardiovascular outcomes were evaluated as the incidence of stroke, bleeding events, and heart failure among the CLL patients with and without AF. Costs and HRU were assessed following up until 1 year after the first date of AF, stroke, bleeding events, and heart failure. HRU was evaluated for outpatient, emergency room (ER), inpatient, and pharmacy visits, and by length of stay (LOS) for hospitalizations. Costs were evaluated in overall population and by HRU type. Statistical differences in clinical outcomes between patients with and without AF were compared using chi-square tests. The Mann Whitney Wilcoxon test was used to compare the medians for the HRU and cost outcomes. The associations between AF and hospitalizations as well as costs were evaluated by multivariable logistic regression and generalized linear model, respectively. Statistical significance was determined at a P-value of less than 0.05. Results: Among the total of 23,756 newly diagnosed CLL patients (median age 68 years, 57.83% male) included in the study, 11% had AF within 1 year of the CLL diagnosis. CLL patients who had AF were significantly older (median age: 67 vs 82 years) and had more male patients (56.9% vs 65.1%) compared to CLL patients without AF. Significantly higher rates of stroke (12.67% vs. 4.97%), bleeding events (17.45% vs. 8.53%), and heart failure (31.14% vs. 4.70%) were observed in CLL patients with AF than those without (Figure 1). Compared to the CLL cohort without AF, adjusted regression analysis indicated that overall, CLL cohort with AF was twice as likely to be hospitalized (odds ratio [OR]=2.03) and incurred 44% higher total costs (cost ratio [CR]=1.44). Further examining the impact of AF-related cardiovascular outcomes, we observed that CLL patients with AF who developed stroke, bleeding events, and heart failure had statistically significant higher HRU (outpatient, ER, inpatient, and pharmacy visits), longer LOS, and costs (total, outpatient, pharmacy) than CLL patients without AF. After adjusting for demographic and clinical confounders, significantly higher hospitalization rates and costs were also reported in the CLL cohort who had AF with further stroke (OR=2.59; CR=1.36), bleeding events (OR=3.27; CR=2.03), and heart failure (OR=5.47; CR=2.07) compared to the CLL cohort without AF. Conclusions: The findings of this real-world study demonstrated significantly higher hospitalizations, cardiovascular events, and economic burden incurred by CLL patients who had AF than those without. The presence of stroke, bleeding events, heart failure among CLL patients with AF further increased the HRU and costs. This study highlighted the importance of better disease management, more intentional monitoring for AF and improved CLL therapeutics with a lower risk of AF or cardiovascular toxicity to prevent or minimize the incidence of AF in CLL patients. Figure 1 Figure 1. Disclosures Mohan: BeiGene, Ltd.: Current Employment. Yang: BeiGene, Ltd.: Current Employment. Liu: BeiGene, Ltd.: Current Employment. Dick: IBM Watson Health: Current Employment. Tang: BeiGene, Ltd.: Current Employment. Chanan-Khan: Ascentage: Research Funding; Alpha2 Pharmaceuticals, NonoDev, Starton: Current holder of stock options in a privately-held company; Cellectar: Current equity holder in publicly-traded company; Alpha2 Pharmaceuticals: Patents & Royalties: Tabi; Ascentage, Starton, Cellectar, NonoDev, Alpha2 Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; BeiGene, Jansen, Ascentage: Honoraria; BieGene, Jansen, Ascentage: Consultancy.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e19508-e19508
Author(s):  
Mohammad Ammad Ud Din ◽  
Samarthkumar Thakkar ◽  
Harsh P. Patel ◽  
Syed Ather Hussain ◽  
Aneeqa Zafar ◽  
...  

e19508 Background: With the increased use of novel agents like Bruton tyrosine kinase inhibitors (BTKi) for the treatment of chronic lymphocytic leukemia (CLL), the incidence of atrial fibrillation (AF) is on the rise in these patients. However, the excess burden added by AF to the morbidity and mortality of CLL patients is unclear. Methods: Using the appropriate ICD-9 and ICD-10 codes, the National Inpatient Sample (NIS) database was accessed to gather data of hospitalized CLL patients with AF from 2008 to 2019. Propensity-score matching (PSM) and logistic regression model were performed to control for baseline patient factors like age, sex, income, and the relevant co-morbidities to match 7265 CLL patient admissions with AF and 7265 CLL patient admissions without AF. The primary outcome was all-cause mortality (ACM), while secondary outcomes included stroke, acute heart failure (AHF), and total cost of hospital stay. Results: The mean age of the cohorts was 82 years. Females made up 44% of both groups. The AF group had similar prevalence of systemic hypertension (62.38% vs 62.10%; p= 0.73), diabetes mellitus (5.09% vs 5.43%; p= 0.35), congestive heart failure (5.57% vs 5.36%; p= 0.58), valvular heart disease (1.17% vs 1.44%; p= 0.14), and pulmonary hypertension (0.21% vs 0.14%; p= 0.31) compared to the group without AF. PSM revealed CLL patients with AF had a higher rate of ACM (6.06% vs 4.47%; p= <0.0001), AHF (7.50% vs 3.85%; p= <0.001), and stroke (3.09% vs 1.65%; p= <0.0001). Admission in the AF group also had a higher median total cost of hospital stay ($9097 vs $7646). A logistic regression model was done to adjust for confounders which revealed similar results for the AF group with increased adjusted odd’s ratio (aOR) of ACM (aOR:1.39, 95% confidence interval (CI): 1.19-1.61; p= <0.001), AHF (aOR: 2.16, 95% CI: 1.85-2.52; p= <0.001), and stroke (aOR:1.94, 95% CI: 1.54-2.44; P= <0.001) (Table). Conclusions: Our data suggest that hospitalized CLL patients with AF are at a significantly increased risk of all-cause mortality, AHF, and stroke. Several limitations like the inability to establish the temporal relationship between CLL and AF and the lack of data regarding medications of individual patients are important to keep in mind while interpreting the results.[Table: see text]


Author(s):  
Benjamin J. R. Buckley ◽  
Stephanie L. Harrison ◽  
Dhiraj Gupta ◽  
Elnara Fazio‐Eynullayeva ◽  
Paula Underhill ◽  
...  

Background Cardiomyopathy is a common cause of atrial fibrillation (AF) and may also present as a complication of AF. However, there is a scarcity of evidence of clinical outcomes for people with cardiomyopathy and concomittant AF. The aim of the present study was therefore to characterize the prevalence of AF in major subtypes of cardiomyopathy and investigate the impact on important clinical outcomes. Methods and Results A retrospective cohort study was conducted using electronic medical records from a global federated health research network, with data primarily from the United States. The TriNetX network was searched on January 17, 2021, including records from 2002 to 2020, which included at least 1 year of follow‐up data. Patients were included based on a diagnosis of hypertrophic, dilated, or restrictive cardiomyopathy and concomitant AF. Patients with cardiomyopathy and AF were propensity‐score matched for age, sex, race, and comorbidities with patients who had a cardiomyopathy only. The outcomes were 1‐year mortality, hospitalization, incident heart failure, and incident stroke. Of 634 885 patients with cardiomyopathy, there were 14 675 (2.3%) patients with hypertrophic, 90 117 (7.0%) with restrictive, and 37 685 (5.9%) with dilated cardiomyopathy with concomitant AF. AF was associated with significantly higher odds of all‐cause mortality (odds ratio [95% CI]) for patients with hypertrophic (1.26 [1.13–1.40]) and dilated (1.36 [1.27–1.46]), but not restrictive (0.98 [0.94–1.02]), cardiomyopathy. Odds of hospitalization, incident heart failure, and incident stroke were significantly higher in all cardiomyopathy subtypes with concomitant AF. Among patients with AF, catheter ablation was associated with significantly lower odds of all‐cause mortality at 12 months across all cardiomyopathy subtypes. Conclusions Findings of the present study suggest AF may be highly prevalent in patients with cardiomyopathy and associated with worsened prognosis. Subsequent research is needed to determine the usefulness of screening and multisdisciplinary treatment of AF in this population.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 69-69
Author(s):  
Carsten Utoft Niemann ◽  
Julie Dubois ◽  
Christian Brieghel ◽  
Sabina Kersting ◽  
Lisbeth Enggaard ◽  
...  

Abstract Background: For patients with Relapsed/Refractory Chronic Lymphocytic Leukemia (R/R CLL), targeted time-limited treatment options are warranted. Thus far, therapeutic options are based on a (one-size-fits-all) fixed treatment duration or treatment until progression independent of individualized responsiveness. Post-treatment minimal residual disease (MRD) predicts outcome for patients receiving venetoclax-based therapy. Aim: To evaluate Progression-Free Survival (PFS) for patients with R/R CLL 12 months after MRD guided treatment cessation of venetoclax + ibrutinib (V+I) treatment (arm B) with the option to reinitiate V+I based on MRD reappearance. Methods: Patients (BTK inhibitor naïve) received ibrutinib lead-in (420 mg daily) for two (28-day) cycles. Venetoclax ramp-up was initiated during cycle 3, reaching 400 mg daily at cycle 4 with continued V+I during cycle 4-15. Patients reaching at least partial remission (PR) and undetectable (u)MRD (&lt;10 -4) in blood (PB) and bone marrow (BM, by central 8-color flow cytometry) at cycle 15 day 15 were randomized 1:2 between ibrutinib maintenance (arm A) or treatment cessation (arm B). Patients later becoming MRD positive (&gt;10 -2) in arm B reinitiated V+I. MRD positive patients at cycle 15 remained on ibrutinib until progression. Results: 225 patients were enrolled: median age 68 years (range 36-87), median CIRS score 2 (range 0-12), Binet stage B/C 84%, IGHV unmutated: 64%, TP53 aberrations (deletions and/or mutations): 24%. Planned treatment until randomization was completed by 194 (86%) patients, 8 patients went off protocol due to toxicity, 5 (2%) patients died during the first two cycles of ibrutinib lead-in (unknown cause, myocardial infarction, intracranial hemorrhage, Richter's transformation and tick-borne viral encephalitis), 15 patients went off protocol due to refusal to continue or other reasons and 3 patients progressed. At cycle 15, 81 (36%) patients achieved uMRD in both PB (112, 50%) and BM (84, 37%) and overall responses were 86%, of which 145 pts (64%) CR(i). Patients with uMRD were randomized to ibrutinib maintenance arm A (n 24 11%) or observation arm B (n 48 21%). Patients who did not achieve uMRD (n 125, 56%) continued ibrutinib maintenance without randomization while 5 patients went off protocol. The primary endpoint of the trial was met; PFS was achieved for 47 (98%) of 48 patients randomized to observation arm B at 12 months after randomization (27 months after starting therapy). One patient died during observation due to myelodysplastic syndrome and 1 patient, without sign of progression, reinitiated per protocol due to MRD positivity. A similar proportion of uMRD patients randomized to ibrutinib maintenance (arm A) or observation (arm B) remained uMRD after 12 months (month 27 after treatment start); 75% and 71%, respectively; see figure. No difference in blood uMRD at cycle 15 was seen neither between TP53 aberrated/wildtype (uMRD: 46%, 52%) nor between IGHV unmutated/mutated patients (uMRD: 50%, 50%). The most frequent adverse event (AE, only grade ≥2) was infections with 68 (30%) grade 2 and 62 (28%) grade ≥3 during the first 15 cycles, while 14 (58%) and 6 (12%) infections appeared after randomization in the treatment arm A and observation arm B, respectively. Atrial fibrillation was reported for 29 (13%) patients during the first 15 cycles, for 4 (3%) during ibrutinib maintenance (arm A + non-randomized) while no patients were reported with atrial fibrillation in arm B. Hypertension was reported for 23 (10%) patients during the first 15 cycles and for 10 (7%) patients during ibrutinib maintenance while no events were reported in arm B. One fatal bleeding event was reported in addition to 32 (14%) grade 2-3 bleeding events during the first 15 cycles; 12 (9%) grade 2-3 bleeding events were reported during ibrutinib maintenance, while no events were reported in arm B. Laboratory tumor lysis grades 2-3 was reported for 11 (5%) of patients. Conclusion: MRD guided time-limited treatment with ibrutinib and venetoclax in the setting of R/R CLL is feasible and demonstrates a favorable benefit-risk profile. No new safety signals were detected. No patients progressed after treatment cessation while patients becoming MRD positive successfully reinitiated therapy, thus proving MRD-guided cessation and reinitiation of targeted therapy feasible in CLL. Figure 1 Figure 1. Disclosures Niemann: CSL Behring, Genmab, Takeda, Octapharma: Consultancy; Abbvie, AstraZeneca, Janssen: Consultancy, Research Funding; Novo Nordisk Foundation: Research Funding. Dubois: Abbvie: Research Funding; Genentech: Research Funding; Roche: Research Funding. Brieghel: AstraZeneca: Consultancy. Kersting: Cellgene: Other: travel grant. Enggaard: Abbvie: Consultancy; Janssen: Consultancy. Mellink: Financial support related to microarray analysis of Murano samples: Research Funding; Genome Diagnostics Laboratory, AUMC: Current Employment; Cytogenetic Field: Consultancy. Poulsen: Janssen: Consultancy; Abbvie: Consultancy. Frederiksen: Abbvie: Research Funding; Janssen Pharmaceuticals: Research Funding; Gilead: Research Funding; Alexion: Research Funding; Novartis: Research Funding. Janssens: Sanofi: Consultancy; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie, Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Trael Grant, Speakers Bureau; Beigene, AstraZeneca: Consultancy, Speakers Bureau. Mattsson: Gilead: Research Funding. Bellido: Janssen: Other: educational funding. Tran: Novartis, Janssen, Abbvie, Takeda, CSL Bering: Consultancy; Astra Zeneca: Consultancy. Levin: Roche, Janssen, Abbvie: Other: Travel Expenses, Ad-Board. Kater: Genmab, LAVA: Other: Ad Board, Steering Committee; BMS, Roche/Genentech: Other: Ad Board, , Research Funding; Janssen, AstraZeneca: Other: Ad Board, steering committee, Research Funding; Abbvie: Honoraria, Other: Ad Board, Research Funding. OffLabel Disclosure: The combination of ibrutinib and venetoclax is not registered for treatment of CLL, both drugs are separately registered for treatment of CLL


Haematologica ◽  
2018 ◽  
Vol 103 (9) ◽  
pp. 1511-1517 ◽  
Author(s):  
Anthony R. Mato ◽  
Meghan Thompson ◽  
John N. Allan ◽  
Danielle M. Brander ◽  
John M. Pagel ◽  
...  

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 8024-8024 ◽  
Author(s):  
John C. Byrd ◽  
Jennifer Ann Woyach ◽  
Richard R. Furman ◽  
Peter Martin ◽  
Susan Mary O'Brien ◽  
...  

8024 Background: The next-generation Bruton tyrosine kinase inhibitor acalabrutinib was approved in patients (pts) with treatment-naïve (TN) and relapsed/refractory chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) based on two complementary phase 3 studies, ELEVATE-TN and ASCEND. This report of ACE-CL-001 (NCT02029443), the first phase 2 study of acalabrutinib, provides the longest safety and efficacy follow-up to date in symptomatic TN CLL pts. Methods: Adults with TN CLL/SLL were eligible if they met iwCLL 2008 criteria for treatment, were inappropriate for/declined standard chemotherapy and had ECOG performance status 0–2. Pts received acalabrutinib 100 mg BID or 200 mg QD, later switching to 100 mg BID, until progressive disease (PD) or unacceptable toxicity. Primary endpoint was safety. Events of clinical interest (ECI) were based on combined AE terms for infections, bleeding events, hypertension, and second primary malignancies (SPM) excluding non-melanoma skin, and on a single AE term for atrial fibrillation. Additional endpoints included investigator-assessed overall response rate (ORR), duration of response (DOR), time to response (TTR), and event-free survival (EFS). Results: Ninety-nine pts (n = 62 100 mg BID; n = 37 200 mg QD), were treated [median age: 64 years, 47% Rai stage 3–4 disease, 10% del(17p), 62% unmutated IGHV]. At median follow-up of 53 months (range, 1–59), 85 (86%) pts remain on treatment; most discontinuations were due to AEs (n = 6) or PD (n = 3 [n = 1 Richter transformation]). Most common AEs (any grade) were diarrhea (52%), headache (45%), upper respiratory tract infection (44%), arthralgia (42%), and contusion (42%). All-grade and grade ≥3 ECIs included infection (84%, 15%), bleeding events (66%, 3%), and hypertension (22%, 11%). Atrial fibrillation (all grades) occurred in 5% of pts (incidence: 1% in years 1, 2, 4; 3% in year 3). SPMs excluding non-melanoma skin (all grades) occurred in 11%. Serious AEs were reported in 38% of pts; those in > 2 pts were pneumonia (n = 4) and sepsis (n = 3). ORR was 97% (7% complete response; 90% partial response). Median TTR was 3.7 months (range, 2–22). Response rates were similar across high-risk groups. Median DOR and median EFS were not reached; 48-month DOR rate was 97% (95% CI, 90%–99%), and 48-month EFS rate was 90% (95% CI, 82%–94%). Conclusions: Long-term data from ACE-CL-001 further support the favorable results with acalabrutinib in phase 3 studies and demonstrate durable responses with no new long-term safety issues. Clinical trial information: NCT02029443 .


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3209-3209
Author(s):  
Yaser Alkhatib ◽  
Shahzaib Nabi ◽  
Edward Peres

Abstract INTRODUCTION Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in the United States with a higher incidence in Caucasians compared to African Americans (AA). Few studies have looked into the impact of race on the outcome of patients with CLL, with some reporting a more aggressive nature of disease in AA compared to Caucasians and others reporting no difference in outcome between the two groups. In addition, variation in genetic mutations in different races is not well studied in patients with CLL. Hence, we performed a study to evaluate the racial differences in genetic mutations and outcome in patients with CLL. METHODS We reviewed the charts of patients with CLL treated in our institution between 2004 and 2014. We collected data on age, gender, race, genetic mutations, rai stage, time to first treatment (TTFT), and time to second treatment (TTST). Both conventional cytogenetics and florescence in-situ hybridization (FISH) analysis were done for all patients. Only Caucasian and African American patients were included in our analysis. We used Fisher's exact test to assess the statistical significance of any difference. P-values <0.05 were considered significant. RESULTS A total of 177 patients with CLL were analyzed, of which 112 patients were Caucasians and 65 patients were AA. For patients with favorable cytogenetics, deletion 13q14 was seen more often in Caucasian patients compared to AA (41.9% versus 18.4%, p= .001), while trisomy 12 was observed more often in AA patients compared to Caucasians (35% versus 17%, p= .009). There was no difference in patients harboring normal cytogenetics between the two groups. For unfavorable cytogenetics, there was no difference between Caucasian and AA patients in deletion 11q23 (8.9% versus 10.9%, p= .68), or deletion 17p/p53 abnormality (11.6% and 9%, p =.62). There was no difference in Rai stage at the time of diagnosis between Caucasians and AA (Rai stage ≤2: 86.6% versus 76.9%, and Rai >2: 13.4% versus 23.1%, p= .1). No difference was observed between the two groups in regards to TTFT (mean TTFT for Caucasians was 32.2 months versus 32.3 months in AA) or TTST (mean TTST for Caucasians was 50.1 months versus 50.6 months in AA). DISCUSSION Racial differences in CLL including genetic mutations and outcome are poorly studied. Falchi et al has reported that AA patients tended to have a higher risk feature profile at the time of presentation, with an overall worse overall survival. In a SEER database analysis reported by Shenoy et al, authors found that AA had an inferior overall survival compared to non-African Americans. In our analysis, we aimed to assess for any difference in the incidence of prognostic cytogenetics between Caucasians and AA. We also evaluated the difference in outcome in terms of TTFT and TTST. We found that despite the presence of some differences in the favorable cytogenetic profile at time of diagnosis between both races, it did not translate into a difference in rai stage at time of presentation or the overall outcome as measured by TTFT and TTST. Our results confirm the most recent analysis by Nabhan et al, which utilized the Mayo Clinic CLL database, and found no difference in the outcome to either AA or Caucasian CLL patients when treated similarly. Conclusion Based on our analysis, a possible difference in some prognostic cytogenetics might be present between different races which did not affect the overall outcome. Hence, racial difference should not be taken into account when treating patients with newly diagnosed CLL, since there is no evidence that outcomes are different when treated with conventional therapy. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document