Risk Factors for Cardiac Toxicities Associated with Proteasome Inhibitor Chemotherapy during Treatment of Multiple Myeloma

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5363-5363
Author(s):  
John H. Chen ◽  
Daniel J. Lenihan ◽  
Sharon E. Phillips ◽  
Madan H. Jagasia ◽  
Stacey A. Goodman ◽  
...  

Abstract Introduction Proteasome inhibitors (PI) bortezomib (B) and carfilzomib (C) are cornerstone therapies for multiple myeloma (MM). An increased incidence of PI-induced cardiac adverse events (CAEs) has been reported in patients receiving C. However, risk factors for cardiac toxicity in this population remain unclear. Our objective is to evaluate the incidence of CAEs associated with C compared with B and identify risk factors for developing events. Patients and Method This was a retrospective analysis of 96 consecutive patients treated for MM at Vanderbilt University from 2011 to 2014 who received B (n=44) and/or C (n=52). Patients in the C group had been previously treated with B, whereas patients in the B group did not have exposure to C. No patients studied were included in both cohorts. We evaluated the clinical features and frequency of CAEs (grade II-IV heart failure, acute coronary syndrome, left ventricular dysfunction, atrial fibrillation/flutter, thromboembolism, systemic hypertension, pulmonary hypertension, orthostatic hypotension, or sudden cardiac death). To identify factors that predisposed patients to CAEs, we analyzed duration of PI therapy, 10-year atherosclerotic cardiovascular disease (ASCVD) risk (calculated risk of myocardial infarction or stroke), gender, use of antithrombotic (antiplatelet/anticoagulant) and antihypertensive medications, prior history of cardiac events, and disease cytogenetic profile. Patients with a prior history of cardiac events were followed by a cardio-oncologist during the course of treatment. Results Table 1 shows patient characteristics. Twenty-five patients experienced CAEs (B, 13% (n=12); C, 25% (n=13)). Cumulative incidence (CI) of CAEs was not significantly different in patients on C compared with B (log-rank test P = 0.41) (Figure 1). Heart failure was the most common type of CAE (Table 2). CAEs occurred after a median of 90 days (range, 4-456) with C and 63.5 days (range, 5-336) with B. By univariate analysis, more patients in the C group were prior smokers, underwent stem cell transplantation and had more prior lines of therapy. More patients in the B group used antithrombotic and ACE inhibitor agents. There were no other significant differences in the use of antihypertensive, antiarrhythmic, and lipid-lowering medications between cohorts. Multivariate analysis showed that male gender (HR 5.3, 95% CI 1.5-18.0, P = 0.007) was an independent risk factor for developing CAEs. Patients taking antithrombotic agents had a lower risk of CAE compared with those not on these therapies (HR 0.1, 95% CI 0.04-0.54, P = 0.004). While ASCVD risk was not predictive of CAEs, patients with a prior history of cardiac events who were followed by a cardio-oncologist experienced fewer CAEs (HR 0.2, 95% CI 0.05-0.72, P = 0.014). Longer duration of PI use resulted in decreasing risk of CAE (HR 0.8, 95% CI 0.7-0.9, P = 0.010). There were no interactions between these outcomes. Conclusions In this series, the incidence of CAEs associated with C did not differ significantly from that of B. We found that events occurred early in therapy. Male gender was an independent risk factor for CAEs. Use of antithrombotic therapy was associated with significantly reduced risk of CAEs. These data suggest that patients may benefit from antithrombotic therapy and follow-up by a cardio-oncologist while on PI therapy, particularly if there is a prior history of cardiac events. Table 1. Bortezomib % (n=44) Carfilzomib % (n=52) P-value ASCVD Risk 0.43 0-10% 46 50 10-20% 29 36 >20% 26 14 Male Gender 57 71 0.82 Median Age, y 61 (38-91) 60 (36-86) 0.20 Past Smoker 26 51 0.02 Type II Diabetes 11 17 0.41 Hyperlipidemia 27 27 0.97 Kidney Disease 9 12 0.70 Prior History of Cardiac Event 59 60 0.96 Median Duration on Bortezomib, d 229 203 0.67 Median Duration on Carfilzomib, d 87.5 ACE Inhibitor Use 32 13 0.03 Antithrombotic Use 48 23 0.01 ISS Stage 0.72 III 34 25 FISH Risk 0.13 Standard/Intermediate 93 85 High 7 15 Median Prior Lines of Therapy 0 (0-4) 2 (0-8) <0.001 Stem Cell Transplant 45 65 0.05 Table 2. Cardiac adverse events Bortezomib Carfilzomib P-value Total Cardiac Adverse Events* 19 17 0.08 Heart Failure 9 6 Acute Coronary Syndrome 1 2 Left Ventricular Dysfunction 0 1 Atrial Fibrillation/Flutter 2 2 Thromboembolism 2 2 Systemic Hypertension 3 3 Pulmonary Hypertension 0 1 Orthostatic Hypotension 2 0 Sudden Cardiac Death 0 0 *Some patients had multiple events Figure 1. Cumulative incidence of cardiac adverse events Figure 1. Cumulative incidence of cardiac adverse events Disclosures No relevant conflicts of interest to declare.

2008 ◽  
Vol 118 (5) ◽  
pp. 280-288
Author(s):  
Bożena Szyguła‑Jurkiewicz ◽  
Aleksander Owczarek ◽  
Agata Duszańska ◽  
Jacek Sikora ◽  
Andrzej Lekston ◽  
...  

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15075-e15075
Author(s):  
Brian Dreyfus ◽  
Samuel P Heilbroner ◽  
Reed Few ◽  
Christine Kratt ◽  
Andres Gomez

e15075 Background: Many oncology treatments have been associated with cardiovascular (CV) adverse events. Cases of CV events, including myocarditis have been reported for PD-1 and PD-L1 therapies. We created a machine learning model to predict potential CV events in PD-(L)1 patients using the CancerLinQ database. Methods: A XGBoosted decision tree model was trained to predict a patient’s risk of serious CV adverse events. The model was trained on 80% of all advanced non-small cell lung cancer (NSCLC), melanoma, and renal cell carcinoma (RCC) patients from our database including those who received PD-(L)1 therapy. Index date was defined as date of first PD-(L)1 administration or date of advanced diagnosis if no PD-(L)1 drug was given. The model contained approximately 400 potential risk factors for cardiac disease including elements of past medical history, social history, vitals, common labs, cancer history (e.g. stage, cancer type), medication history, and PD-(L)1 specific factors including PD-(L)1 expression status and PD-(L)1 therapy administered. The model was tested on two separate validation sets (patients not used in training): one using advanced NSCLC, melanoma, and RCC patients and another with PD-(L)1 patients only. Each factor’s importance to the model’s predictions was calculated using SHAP summary plots, a qualitative technique for interpreting machine learning models. Results: A total of 27,172 advanced cancer patients were included in our study. 4,966 received PD-(L)1 therapy. The model was trained on 21,758 patients and 5,414 patients were set aside for testing. The model predicted serious cardiac events within 100 days of index with an AUC-ROC of 0.75 in all patients and 0.79 in PD-(L)1 patients. The top predictors of cardiac risk in PD-(L)1 patients included a history of heart disease, weight loss, the % lymphocyte count, and median LDH. The % lymphocyte count and weight loss were noticeably more predictive in PD-(L)1 patients than in non-PD-(L)1 patients. However, in general SHAP summary plots of all and PD-(L)1 patients were nearly identical, suggesting that both cohorts’ cardiac risk is determined in a similar way. PD-(L)1 and autoimmune disease associated factors did not appear in the top 40 most predictive risk factors. Conclusions: Using traditional cardiac risk factors, our model was able to predict potential cardiac events in PD-(L)1 patients. Our model found that high lymphocyte count may be protective while weight loss and a history of cardiac disease (e.g. heart failure) could indicate a poor prognosis.


Circulation ◽  
2020 ◽  
Vol 142 (Suppl_3) ◽  
Author(s):  
Zaniar Ghazizadeh ◽  
Chad Gier ◽  
Avinainder Singh ◽  
Lina Vadlamani ◽  
Maxwell Eder ◽  
...  

Introduction: The prevalence and outcomes of patients hospitalized with COVID-19 with atrial fibrillation and atrial flutter (AF/FL) remains unclear. Methods: The Yale Cardiovascular COVID Registry is a cohort study of adult patients >=18 years hospitalized with COVID-19 in the Yale New Haven Health System. Retrospective medical record review was performed on consecutive patients from the registry admitted between March and June 2020. We calculated the rates of prior and in-hospital AF/FL and evaluated the unadjusted rates of in-hospital adverse events for both groups; we then calculated the adjusted odds of adverse events using logistic regression. Results: Among 396 patients, the mean age was 68.2, 52.3% were men, 56.4% were Caucasian, 28.4% Black and 16.9% Hispanic. 15.7% of patients had prior history of AF/FL. 19.9% of patients had in-hospital AF/FL, 7.83% of which did not have a prior history of AF/FL. Patients with in-hospital AF/FL had significantly more CV complications compared to those without including cardiac injury (78.5% vs 42.7%, p=0.000), type 2 myocardial infarction (53.3 vs 30.3%, p=0.002), and heart failure (32.9% vs 9.2%, p=0.000). In-hospital AF/FL was associated with significantly worse outcomes related to COVID-19 including ICU survival (OR 0.22 [0.08-0.59], p=0.002), heart failure (5.19 [2.56-10.5], p=0.000), myocardial injury (OR 2.87 [1.49-5.49], p=0.001), acute kidney injury (OR 2.02 [1.09-3.74], p=0.027), dialysis (OR 4.07 [1.38-12.03], p=0.011) and hospice/death (OR 2.47 [1.35-4.53], p=0.004). Conclusion: AF/FL are common in patients hospitalized with COVID-19 and these patients had significantly worse outcomes, including lower odds of ICU survival and higher odds of heart failure, acute kidney injury, dialysis and hospice/death.


Respiration ◽  
2021 ◽  
pp. 1-8
Author(s):  
Masato Kono ◽  
Yutaro Nakamura ◽  
Yasunori Enomoto ◽  
Hideki Yasui ◽  
Hironao Hozumi ◽  
...  

<b><i>Background:</i></b> Idiopathic pleuroparenchymal fibroelastosis (PPFE) is a rare form of idiopathic interstitial pneumonia that is characterized by predominantly upper lobe pleural and subpleural lung parenchymal fibrosis. Pneumothorax is one of the major respiratory complications in PPFE patients; however, its clinical features are poorly understood. <b><i>Objective:</i></b> We aimed to investigate the complication of pneumothorax in patients with idiopathic PPFE. <b><i>Methods:</i></b> A retrospective multicenter study involving 89 patients who had been diagnosed with idiopathic PPFE was conducted. We investigated the cumulative incidence, clinical features, and risk factors of pneumothorax after the diagnosis of idiopathic PPFE. <b><i>Results:</i></b> Pneumothorax developed in 53 patients (59.6%) with 120 events during the observation period (41.8 ± 35.0 months). The cumulative incidence of pneumothorax was 24.8, 44.9, and 53.9% at 1, 2, and 3 years, respectively. Most events of pneumothorax were asymptomatic (<i>n</i> = 85; 70.8%) and small in size (<i>n</i> = 92; 76.7%); 30 patients (56.6%) had recurrent pneumothorax. Chest drainage was required in 23 pneumothorax events (19.2%), and a persistent air leak was observed in 13 (56.5%). Patients with pneumothorax were predominantly male and frequently had pathological diagnoses of PPFE and prior history of pneumothorax and corticosteroid use; they also had significantly poorer survival than those without pneumothorax (log-rank test; <i>p</i> = 0.001). Multivariate analysis revealed that a higher residual volume/total lung capacity ratio was significantly associated with the development of pneumothorax after the diagnosis. <b><i>Conclusion:</i></b> Pneumothorax is often asymptomatic and recurrent in patients with idiopathic PPFE, leading to poor outcomes in some cases.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-35
Author(s):  
Jorge E. Cortes ◽  
Philipp D le Coutre ◽  
Carlo Gambacorti-Passerini ◽  
Henrik Hjorth-Hansen ◽  
Pia Raanani ◽  
...  

Introduction: Tyrosine kinase inhibitor therapy has been associated with cardiovascular (CV) events. BOS is approved for patients (pts) with newly diagnosed Philadelphia chromosome-positive (Ph+) chronic phase (CP) CML and Ph+ CML resistant/intolerant to prior therapy. We analyzed cardiac, vascular and hypertension treatment-emergent adverse events (TEAEs) in pts with newly diagnosed CP CML receiving BOS or IMA after 5 yrs follow-up in BFORE. Methods: In the open-label, phase 3 BFORE trial (NCT02130557), 536 pts with newly diagnosed CP CML were randomized 1:1 to receive first-line 400 mg once-daily BOS (n=268) or IMA (n=268; 3 untreated). The current analysis included all pts who received ≥1 dose of study drug. TEAEs were monitored from first dose of study drug until 28 d after last dose; monitoring of non-serious adverse events (AE) ended at the start of a new cancer treatment. Prespecified MedDRA terms (cardiac, n=133; vascular, n=502) comprised clusters of investigator-assessed TEAEs (Table). Multivariable proportional subdistribution hazard models predicting time to first TEAE included treatment group; baseline demographic information; history of cardiac events, vascular events, hypertension, diabetes, hyperlipidemia, and tobacco use; as well as hypertension, cardiac (for vascular model) and vascular (for cardiac model) TEAEs. CI excluding 1 was considered predictive of outcome. This analysis was based on 17-Apr-20 last pt last visit (12-Jun-20 database lock), 5 y (240 wks) after last enrolled pt. Results: At study completion, 59.7% of BOS pts and 58.1% of IMA pts were still on treatment. Median duration of treatment was 55 mo for pts receiving BOS or IMA; respective median (range) dose intensity was 393.6 (39-583) vs 400.0 (189-765) mg/d. In the BOS vs IMA arm, 57.8% vs 56.2% of pts had ≥1 CV risk factor at baseline; 20.9% vs 17.7% had ≥3 risk factors, respectively. The most common risk factors were a history of hypertension (BOS, 34.0%; IMA, 30.6%), BMI &gt;30 (BOS, 23.1%; IMA, 21.9%), age ≥65 y (BOS, 19.8%; IMA, 17.4%), and history of hyperlipidemia (BOS, 16.8%; IMA, 18.9%). Rates of cardiac, vascular and hypertension TEAEs; serious adverse events (AEs); AEs leading to treatment withdrawal and drug-related TEAEs (per investigator) were low in both arms (Table). Exposure-adjusted rates of cardiac, vascular and hypertension TEAEs were similar between arms (Table). Risk factors for cardiac TEAEs (HR; 95% CI) were age in years (1.04 [1.02-1.07]), race other than Asian or black compared to white (&lt;0.01 [&lt;0.01-&lt;0.01]), a history of tobacco use (6.94 [1.89-25.40]) and cardiac events (3.59 [1.68-7.65]), hypertension (3.07 [1.20-7.84]) and vascular TEAEs (5.27 [1.53-18.18]). Risk factors for vascular TEAEs (HR [95% CI]) were black race compared to white (&lt;0.01 [&lt;0.01-&lt;0.01]), a history of vascular events (4.65 [1.81-11.97]) and cardiac TEAEs (7.73 [2.31-25.84]). In multivariable analyses, treatment group was not predictive of time to initial cardiac (HR 0.87 [95% CI 0.45-1.70]) or vascular (HR 2.17 [95% CI 0.94-45.04) TEAEs. The most common cardiac, vascular, and hypertension TEAEs, respectively, were sinus bradycardia (2.2%), angina pectoris (3.0%) and hypertension (9.7%) with BOS vs electrocardiogram QT prolongation (3.8%), peripheral coldness (1.1%) and hypertension (10.9%) with IMA. One grade 5 cardiac (acute cardiac failure) and one vascular (myocardial ischemia) TEAE were reported with BOS, and one vascular (cerebrovascular accident) TEAE with IMA; none were considered related to study drug. Successful treatment rechallenge was achieved in the majority of pts with dose interruptions due to cardiac (BOS, 85.7%; IMA, 100%) and vascular (BOS, 100%; IMA, not applicable) TEAEs who were readministered study drug. No pts had dose interruptions due to hypertension TEAEs. Cardiac, vascular and hypertension TEAEs resolved in 57.7%, 75.0% and 35.7% of pts receiving BOS and 65.2%, 66.7% and 48.3% receiving IMA. Conclusions: In this final analysis of BFORE, the incidence of cardiac, vascular, and hypertension TEAEs in pts with newly diagnosed CP CML receiving BOS or IMA was low and was similar between treatment groups. These AEs infrequently led to treatment discontinuation. In addition to continued improved efficacy with BOS vs IMA after 5 yrs follow-up (Brümmendorf et al., ASH 2020), these safety results support the use of first-line BOS as a standard of care in pts with CP CML. Disclosures Cortes: Bristol-Myers Squibb: Research Funding; BiolineRx: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; BioPath Holdings: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Consultancy, Research Funding; Amphivena Therapeutics: Research Funding; Arog: Research Funding; Jazz Pharmaceuticals: Consultancy, Research Funding; Telios: Research Funding; Astellas: Research Funding; Sun Pharma: Research Funding; Merus: Research Funding; Daiichi Sankyo: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Immunogen: Research Funding. le Coutre:Incyte: Honoraria; Pfizer: Honoraria; Novartis: Honoraria. Gambacorti-Passerini:Bristol-Myers Squibb: Consultancy; Pfizer: Honoraria, Research Funding. Hjorth-Hansen:Pfizer: Honoraria, Research Funding; Austrian Orphan Pharma: Honoraria, Research Funding; Bristol-Myers Squibb: Research Funding. Garcia-Gutiérrez:Novartis: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Bristol-Myers Squibb: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Pfizer: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Incyte: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding. Kota:Ariad: Honoraria; Incyte: Honoraria; Xcenda: Honoraria; Novartis: Consultancy, Honoraria; Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical, Company Ltd, Cambridge, MA, USA: Honoraria; Pfizer: Consultancy, Honoraria. Purcell:Pfizer: Current Employment, Current equity holder in publicly-traded company. Viqueira:Pfizer: Current Employment, Current equity holder in publicly-traded company. Leip:Pfizer: Current Employment, Current equity holder in publicly-traded company. Brümmendorf:Takeda: Consultancy; Janssen: Consultancy; Merck: Consultancy; Pfizer: Consultancy, Honoraria, Other: Travel, Accommodation, Expenses, Research Funding; Novartis: Consultancy, Other: travel, accommodation, expenses, Research Funding.


BMJ Open ◽  
2018 ◽  
Vol 8 (8) ◽  
pp. e021681 ◽  
Author(s):  
Sander van Doorn ◽  
Annerien Tavenier ◽  
Frans H Rutten ◽  
Arno W Hoes ◽  
Karel G M Moons ◽  
...  

ObjectivesPatients with atrial fibrillation (AF) are at increased risk of many adverse events, notably stroke. To prevent all adverse outcomes, integrated AF care is advocated though the potential domain for such multidisciplinary management is still unclear. Therefore, insight in the systemic nature of AF and identifying patients at risk of adverse events after oral anticoagulation is needed. The aim of this study is to first describe the risk of hospitalisation and mortality in community-dwelling older patients with AF using anticoagulants, and second to assess the association between traditional cardiac risk factors and these outcomes.DesignA prospective cohort.SettingGeneral practice.Participants2068 patients with AF using oral anticoagulants.Outcome measuresWe calculated incidence rates (IRs) of ischaemic stroke, bleeding, hospitalisations and mortality, and compared risk factors using Cox regression between those with and without an adverse event, both for cardiac and non-cardiac causes.ResultsDuring a median follow-up of 2.7 (IQR 2.2–3.0) years, the IR per 100 person-years was 22.1 for hospitalisations and 6.7 for mortality. Non-cardiac events outnumbered cardiac events (IRs 15.7 vs 7.6 per 100 person-years for hospitalisation, p<0.001 and 5.0 vs 1.7, p<0.001 for mortality). As a comparison, the IRs for stroke and major bleeding were 1.7 and 0.8 per 100 person years, respectively. In multivariate models, high age, heart failure and vascular disease were independently associated with all-cause hospitalisation and— in addition to diabetes, previous stroke and renal disease—for all-cause mortality.ConclusionsIn anticoagulated community-dwelling patients with AF, stroke risk is effectively reduced and thus fairly low, whereas risks of hospitalisation and mortality remain high, importantly mainly for non-cardiac causes. Notably high age, heart failure and vascular disease are predictive for such outcomes and may be of value in identifying high-risk patients in the future.Trial registration numberNTR3741.


2005 ◽  
Vol 71 (5) ◽  
pp. 406-413 ◽  
Author(s):  
Benjamin K. Poulose ◽  
Marie R. Griffin ◽  
Yuwei Zhu ◽  
Walter Smalley ◽  
William O. Richards ◽  
...  

Identifying risk factors for adverse events after bariatric surgery (BaS) can help define high-risk groups to improve patient safety. We calculated cumulative incidence of adverse events and identified risk factors for these events using validated surgical patient safety indicators (PSIs) developed by the Agency for Healthcare Research and Quality. BaS patients ≥18 years old were identified using the 2002 Nationwide Inpatient Sample. Cumulative incidence at discharge was calculated for accidental puncture or laceration (APL), pulmonary embolus or deep venous thrombosis (PE/DVT), and postoperative respiratory failure (RF). Factors predictive of these PSIs were identified. From 7,853,982 discharges, a national cohort of 69,490 BaS patients was identified. During BaS hospitalization, the cumulative incidences per 1000 discharges of APL, PE/DVT, and RF were 12.6, 3.4, and 7.3, respectively. Risk factors for APL included male gender (odds ratio [OR] 1.6, 95% confidence interval 1.1–2.3, P < 0.05) and age of 40–49 years (OR 1.6 [1.1–2.3], P < 0.05) compared to ages 18–39 years. Patients aged 50–59 years (OR 3.5 [1.6–7.7], P < 0.05) had a higher chance of PE/DVT compared to those 18–39 years. Male gender (OR 1.8 [1.1–2.9], P < 0.05), ages 40–49 (OR 2.1 [1.1–4.2], P < 0.05) and 50–59 (OR 3.8 [2.1–6.9], P < 0.05), a history of chronic lung disease (OR 1.7 [1.1–2.7], P < 0.05), and Medicare coverage compared to private insurance (OR 2.2 [1.2–3.8], P < 0.05) were predictive of RF. This study established national measures for BaS adverse events. Further, risk factors associated with adverse events varied by gender, age, insurance status, and comorbidity. Evaluation of these higher risk BaS groups is needed to improve patient safety.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4490-4490
Author(s):  
Asifa Malik ◽  
Jorge E. Cortes ◽  
Hagop Kantarjian ◽  
Gloria Mattiuzzi ◽  
Gloria Iliescu ◽  
...  

Abstract Abstract 4490 Background: Imatinib is currently standard therapy for patients with success in Chronic Myeloid Leukemia (CML). Several instances of cardiac adverse events have been reported for patients while on therapy with imatinib. In many instances, these events occur in patients with pre-existing cardiac conditions. The aim of our study was to determine the occurrence of cardiac events in patients with CML treated with Imatinib, and the impact that cardiac risk factors and pre-existing cardiac conditions had on the risk of developing cardiac adverse effects. Methods: We reviewed the medical records of 51 patients with chronic phase CML who were treated with imatinib after failing prior therapies. For each patient we collected cardiac risk factors, pre-existing cardiac disease, pre-treatment EKG and echocardiogram (ECHO) readings, as well as post-treatment changes in EKG and ECHO findings. Results: Pre-existing cardiac conditions were found in 14 (27%) patients, including congestive heart failure in 2 (4%), myocardial infarction in 4 (8%), atrial fibrillation in 1 (2%), benign arrhythmias in 1(2%), aortic regurgitation in 1(2%), mitral valve prolapse in 1 (2%), mitral regurgitation in 1(2%), pericarditis in 1(2%), bradycardia in1(2%) and benign arrhythmia in 1(2%). Cardiac risk factors were present in 26 patients (51%), including smoking in 10 patients (20%), hypertension in 17 (33%), diabetes mellitus in 9 (18%), obesity in 2 (4%), hyperlipidemia in 5 (10%), stress (self-reported by patient or on anxiolytic therapy) in 5 (10%), alcohol in 11 (22%), atherosclerosis in 3 (6%), and positive family history for cardiac disease in 5 patients (9.8%). Cardiac events were noted in 19 patients (37%) of whom 11 (58%) had pre-existing cardiac conditions prior to initiating imatinib treatment and 14 patients (27%) had at least one cardiac risk factor. Congestive heart failure with clinical manifestations was seen in 9 patients (18%) with documentation of decreased ejection fraction on echocardiogram seen in 3 patients (6%) who had a decrease in LVEF from a median of 55% (range 50% to 72%) to a median of 45% (range 25% to 60%). Out of the patients who developed CHF while on treatment with imitanib, 3 patients (6%) had history of cardiac conditions (atrial fibrillation in 1 (2%), congestive heart failure in 2 (4%)). Myocardial infarction was documented in 3 patients (6%), one of which had prior myocardial infarction and pacemaker, another had history of mitral valve prolapse and hypertension; one patient had hypertension, diabetes mellitus and positive family history but no prior history of heart disease. Arrhythmia was seen in 3 patients (6%). Post-treatment EKG changes occurred in 14 patients (27%) including bradycardia, PAC's, PVC's, ST-T wave changes, tachycardia and other rhythm abnormalities. These changes were usually asymptomatic. Gated cardiac study done after a median of 63 months (range 29 to 83 months) after initiation of imatinib treatment showed EF below 60% in 9 patients (18%) with a median of 55% (range 36% to 59%). None of the patients died of cardiac conditions and none discontinued imatinib therapy because of cardiac events. Conclusion: Although cardiac events occur in some patients treated with imatinib, these are much more common among patients with pre-existing cardiac conditions and/or cardiac risk factors. These patients need to be monitored closely to minimize their risk and intervene early when new cardiac events arise. Disclosures: Cortes: novartis: Research Funding; BMS: Research Funding; Pfizer: Consultancy, Research Funding. Kantarjian:novartis: Research Funding; BMS: Research Funding. O'Brien:Novartis: Research Funding.


2021 ◽  
Vol 15 (1) ◽  
Author(s):  
Florence Jaguga

Abstract Background Stuttering is a rare side effect of clozapine. It has been shown to occur in the presence of one or more factors such as abnormal electrophysiological findings and seizures, extrapyramidal symptoms, brain pathology, and a family history of stuttering. Few case reports have documented the occurrence of clozapine-induced stuttering in the absence of these risk factors. Case presentation A 29-year-old African male on clozapine for treatment-resistant schizophrenia presented with stuttering at a dosage of 400 mg/day that resolved with dose reduction. Electroencephalogram findings were normal, and there was no clinical evidence of seizures. The patient had no prior history or family history of stuttering, had a normal neurological examination, and showed no signs of extrapyramidal symptoms. Conclusion Clinicians ought to be aware of stuttering as a side effect of clozapine, even in the absence of known risk factors. Further research should investigate the pathophysiology of clozapine-induced stuttering.


2021 ◽  
pp. 193864002199849
Author(s):  
Sumit Patel ◽  
Lauren Baker ◽  
Jose Perez ◽  
Ettore Vulcano ◽  
Jonathan Kaplan ◽  
...  

Background Nonunion is a postoperative complication after ankle arthrodesis (AA), which leads to increased morbidity and revision rates. Previous studies have identified risk factors for nonunion following AA, but no meta-analysis has been performed to stratify risk factors based on strength of evidence. Methods Abstracts and full-text articles were screened by 2 independent reviewers. Relevant data were extracted from the included studies. Random effects meta-analyses were summarized as forest plots of individual study and pooled random effect results. Results Database search yielded 13 studies involving 987 patients were included, and 37 potential risk factors for nonunion. Meta-analysis found 5 significant risk factors for nonunion post-AA. Strong evidence supports male gender (OR: 1.96; 95% CI: 1.13-3.41), smoking (OR: 2.89; 95% CI: 1.23-6.76), and history of operative site infection prior to arthrodesis (OR: 2.40; 95% CI: 1.13-5.09) as predictors for nonunion following AA. There was moderate evidence supporting history of open injury (OR: 5.95; 95% CI: 2.31-15.38) and limited evidence for preoperative avascular necrosis (OR: 13.16; 95% CI: 2.17-79.61) as possible risk factors for nonunion. Conclusion The results of our meta-analysis suggest that male gender, smoking, and history of operative site infection have strong evidence and that history of open injury and avascular necrosis also have evidence as risk factors for nonunion. Surgeons should be cognizant of these risks when performing AA and closely follow up with patients with the aforementioned risk factors to ensure postoperative success. Levels of Evidence: Level V: Systematic review of cohort and case-control studies


Sign in / Sign up

Export Citation Format

Share Document