A multiple biomarker risk score for guiding clinical decisions using a decision curve approach

2011 ◽  
Vol 19 (4) ◽  
pp. 874-884 ◽  
Author(s):  
Maria F Hughes ◽  
Olli Saarela ◽  
Stefan Blankenberg ◽  
Tanja Zeller ◽  
Aki S Havulinna ◽  
...  

Aims: We assessed whether a cardiovascular risk model based on classic risk factors (e.g. cholesterol, blood pressure) could refine disease prediction if it included novel biomarkers (C-reactive protein, N-terminal pro-B-type natriuretic peptide, troponin I) using a decision curve approach which can incorporate clinical consequences. Methods and results: We evaluated whether a model including biomarkers and classic risk factors could improve prediction of 10 year risk of cardiovascular disease (CVD; chronic heart disease and ischaemic stroke) against a classic risk factor model using a decision curve approach in two prospective MORGAM cohorts. This included 7739 men and women with 457 CVD cases from the FINRISK97 cohort; and 2524 men with 259 CVD cases from PRIME Belfast. The biomarker model improved disease prediction in FINRISK across the high-risk group (20⊟40%) but not in the intermediate risk group, at the 23% risk threshold net benefit was 0.0033 (95% CI 0.0013−0.0052). However, in PRIME Belfast the net benefit of decisions guided by the decision curve was improved across intermediate risk thresholds (10⊟20%). At pt = 10% in PRIME, the net benefit was 0.0059 (95% CI 0.0007⊟0.0112) with a net increase in 6 true positive cases per 1000 people screened and net decrease of 53 false positive cases per 1000 potentially leading to 5% fewer treatments in patients not destined for an event. Conclusion: The biomarker model improves 10-year CVD prediction at intermediate and high-risk thresholds and in particular, could be clinically useful at advising middle-aged European males of their CVD risk.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Peter Piko ◽  
Zsigmond Kosa ◽  
Janos Sandor ◽  
Roza Adany

AbstractCardiovascular diseases (CVDs) are the number one cause of death globally, and the early identification of high risk is crucial to prevent the disease and to reduce healthcare costs. Short life expectancy and increased mortality among the Roma are generally accepted (although not indeed proven by mortality analyses) which can be partially explained by the high prevalence of cardiovascular risk factors (CVRF) among them. This study aims to elaborate on the prevalence of the most important CVD risk factors, assess the estimation of a 10-year risk of development of fatal and nonfatal CVDs based on the most used risk assessment scoring models, and to compare the Hungarian general (HG) and Roma (HR) populations. In 2018 a complex health survey was accomplished on the HG (n = 380) and HR (n = 347) populations. The prevalence of CVRS was defined and 10-year cardiovascular risk was estimated for both study populations using the following systems: Framingham Risk Score for hard coronary heart disease (FRSCHD) and for cardiovascular disease (FRSCVD), Systematic COronary Risk Evaluation (SCORE), ACC/AHA Pooled Cohort Equations (PCE) and Revised Pooled Cohort Equations (RPCE). After the risk scores had been calculated, the populations were divided into risk categories and all subjects were classified. For all CVD risk estimation scores, the average of the estimated risk was higher among Roma compared to the HG independently of the gender. The proportion of high-risk group in the Hungarian Roma males population was on average 1.5–3 times higher than in the general one. Among Roma females, the average risk value was higher than in the HG one. The proportion of high-risk group in the Hungarian Roma females population was on average 2–3 times higher compared to the distribution of females in the general population. Our results show that both genders in the Hungarian Roma population have a significantly higher risk for a 10-year development of cardiovascular diseases and dying from them compared to the HG one. Therefore, cardiovascular interventions should be focusing not only on reducing smoking among Roma but on improving health literacy and service provision regarding prevention, early recognition, and treatment of lipid disorders and diabetes among them.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4613-4613
Author(s):  
Naheed Alam ◽  
Eshetu G Atenafu ◽  
Wei Xu ◽  
Jieun Uhm ◽  
Vikas Gupta ◽  
...  

Introduction Steroid refractory acute GVHD (SR-aGVHD) occurs frequently and affects transplant outcomes adversely associated with high morbidities and mortalities. The present study attempted to develop a risk model predicting the risk of SR-aGVHD using both candidate single nucleotide polymorphism (SNP) and clinical risk factors. Methods A total of 268 patients were included who had diagnosis of acute GVHD and treated with systemic steroids. SR-aGVHD was defined with followings: 1) progression of GVHD after 3 days of systemic steroids; 2) No change after 7 days treatment; 3) incomplete response after 14 days of treatment initation. Patients were randomly divided into training (n=180) and validation sets (n=88) adjusted for the presence of SR-aGVHD, disease risk, grade 3/4 aGVHD, presence of gastrointestinal and liver involvement. A total of 259 SNPs in 53 genes were genotyped as previously described (Kim, Transplantation 2012). Clinical risk factors were also included to generate risk model for SR-aGVHD. Results Overall, 132 (47.3%) patients developed SR-aGVHD which was equally distributed in training and validation sets. In the training set, 85 patients (47.2%) developed SR-aGVHD. In univariate analysis, gut involvement (p<0.0001) and grade 3/4 aGVHD (p<0.0001) were identified as risk factors as well as donor genotypes of IL6 (rs1800797; p=6.15x10-4) and IFNG (rs2069727; p=4.37x10-4). Multivariate analysis confirmed that these two SNPs along with gut GVHD were independent risk factors for SR-aGVHD, but not grade 3/4 acute GVHD. A combined risk model was generating using 2 SNPs of IL6 (rs1800797), IFNG (rs2069727) and clinical risk factor of gut GVHD. A score of one was assigned to each of above risk factors and patients were divided based on these scores. Overall, the risk of SR-aGVHD increased as scores increased. Then we divided the patients into low risk (score 0, n=74) versus high risk groups (score 1, 2 and 3, n=106). Higher incidence of SR-aGVHD was noted in high risk group (61.3%; 65/106) vs low risk group (27%; 20/74; p<0.0001, OR 4.28 [95% CI 2.25-8.16]). The combined risk model was successfully replicated to stratify the groups risk of SR-aGVHD in the validation set (p=0.0045, OR 3.74 [95% CI 1.47-9.52]): incidence of SR-aGVHD was 57% in high risk group (31/54) vs 26% low risk group (9/34) in the validation set. When the combined risk model was used, using SNPs along with clinical risk factor, the risk model showed AUC of 0.738 in training set with sensitivity of 76 % and specificity of 56%. In the validation set, it showed AUC of 0.773 with sensitivity of 77 % and specificity of 52%. Conclusion The present study suggested that this risk model could identify high risk patient for SR-aGVHD with following information including donor genotype of IL6 (rs1800797) and IFNG (rs2069727) with gut involvement of GVHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1022-1022 ◽  
Author(s):  
Gary H. Lyman ◽  
Eva Culakova ◽  
Marek S. Poniewierski ◽  
Jeffrey Crawford ◽  
David C. Dale ◽  
...  

Abstract Abstract 1022 Background: Neutropenic complications represent important dose-limiting toxicities of cancer chemotherapy. We recently developed and validated a risk model for neutropenic complications in patients with solid tumors or lymphoma receiving cancer chemotherapy (Lyman et al. Cancer 2011). While practice guidelines recommend primary colony-stimulating factor (CSF) prophylaxis for patients at >20% risk of febrile neutropenia (FN), many patients receive chemotherapy regimens associated with an intermediate risk (10–20%) of FN. For these patients, the decision to give or withhold primary CSF prophylaxis is based on clinical judgment. We report here the ability of the risk model to identify patients with individual characteristics placing them at high risk for neutropenic complications among patients receiving intermediate risk chemotherapy regimens. Methods: A prospective cohort study was conducted of consenting patients initiating a new chemotherapy regimen at 115 randomly selected US oncology practices between 2002–2006. The risk of cycle 1 severe or febrile neutropenia was estimated [95% CI] utilizing logistic regression analysis adjusting for key clinical factors including: planned chemotherapy, prior chemotherapy, age, abnormal hepatic or renal function, low pretreatment white blood count, and immunosuppressive medications. The cumulative incidence of severe neutropenia and FN across 4 cycles was estimated by the product limit method of Kaplan and Meier. Results: Among 3,760 patients with cancers of breast, lung, ovary, colon, or lymphoma, 2,270 received an intermediate risk chemotherapy regimen based on NCCN guidelines. Overall, in the subpopulation receiving intermediate risk regimens, severe or febrile neutropenia occurred in cycle 1 in 21.4% while FN over 4 cycles was observed in 11%, and primary CSF prophylaxis was utilized in 16.4%. The performance of the risk model was good in this subgroup with a c-statistic of 0.82 [0.80–0.84]. Among the half of patients classified as high risk based on the model despite receiving an intermediate risk chemotherapy regimen, cycle 1 severe or febrile neutropenia occurred in 38% [35%–41%] compared to 5% [4%–6%] of patients classified as low risk based on the model receiving such regimens. Model sensitivity and specificity were 89% and 61%, respectively. The cumulative risk of FN over 4 cycles of chemotherapy was 20% in predicted high risk group versus 5% in the low risk group (Figure). The majority of severe or febrile neutropenia events (67%) and FN events (55%) were observed in cycle 1. One-half of high risk patients who did not receive primary CSF prophylaxis in cycle 1 received CSF during subsequent cycles following a neutropenic event. Conclusions: Our model for predicting neutropenic complications can identify patients at high individual risk for severe neutropenia in cycle 1 or FN in the first 4 cycles of chemotherapy when receiving intermediate risk chemotherapy. This analysis emphasizes the potential value of determining an individual patient's risk of chemotherapy complications based on a validated risk model. Disclosures: Lyman: Amgen: Research Funding. Crawford:Amgen: Consultancy, Honoraria, Research Funding. Dale:Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Kuderer:Amgen: Research Funding.


2016 ◽  
Vol 34 (26) ◽  
pp. 3150-3156 ◽  
Author(s):  
Norbert Schmitz ◽  
Samira Zeynalova ◽  
Maike Nickelsen ◽  
Roopesh Kansara ◽  
Diego Villa ◽  
...  

Purpose To develop and validate a risk score for relapse in the CNS in patients with diffuse large B-cell lymphoma (DLBCL). Patients and Methods A total of 2,164 patients (18 to 80 years old) with aggressive B-cell lymphomas (80% DLBCL) treated with rituximab and CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone)-like chemotherapy, who were enrolled in studies from the German High-Grade Non-Hodgkin Lymphoma Study Group and the MabThera International Trial, were analyzed for occurrence of relapse/progression in the CNS. The resulting risk model was validated in an independent data set of 1,597 patients with DLBCL identified in the British Columbia Cancer Agency Lymphoid Cancer database. Results The risk model consists of the International Prognostic Index (IPI) factors in addition to involvement of kidneys and/or adrenal glands (CNS-IPI). In a three-risk group model, the low-risk group (46% of all patients analyzed), the intermediate-risk group (41%), and the high-risk group (12%) showed 2-year rates of CNS disease of 0.6% (CI, 0% to 1.2%), 3.4% (CI, 2.2% to 4.4%), and 10.2% (CI, 6.3% to 14.1%), respectively. Patients from the validation British Columbia Cancer Agency data set showed similar rates of CNS disease for low-risk (0.8%; CI, 0.0% to 1.6%), intermediate-risk (3.9%; CI, 2.3% to 5.5%), and high-risk (12.0%; CI, 7.9% to 16.1%) groups. Conclusion The CNS-IPI is a robust, highly reproducible tool that can be used to estimate the risk of CNS relapse/progression in patients with DLBCL treated with R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) chemotherapy. Close to 90% of patients with DLBCL belong to the low- and intermediate-risk groups and have a CNS relapse risk < 5%; they may be spared any diagnostic and therapeutic intervention. In contrast, those in the high-risk group have a > 10% risk of CNS relapse and should be considered for CNS-directed investigations and prophylactic interventions.


2021 ◽  
Author(s):  
Hui-Juan Zuo ◽  
Xian-Tao Song ◽  
Jin-Wen Wang ◽  
Hong-Xia Yang ◽  
Jie Lin

Abstract Background: Ischemic cardiovascular disease (ISCVD) is a massive public health problem. ISCVD risk prediction models based on traditional risk factors as predictors is limited. Carotid atherosclerosis plays a fundamental value in the occurrence of ISCVD. The aim of this study was to evaluate the value of risk stratification plus carotid plaque improving the prediction of ISCVD. Methods: Between June 2016 and June 2017, 3998 subjects with hypertension were prospectively recruited and completed traditional risk factors survey and carotid ultrasound measurements in Anzhen Hospital, Beijing, China. Results: A total of 2010 (50.3%) subjects were detected carotid plaque. Among patients free from ISCVD (n=3479), there were 884 patients (25.4%) at high risk for ISCVD, and 868 (25.0%), 1727 (49.6%) was classified as intermediate risk or low risk according to Chinese cardiovascular risk score chart. The detected rate of carotid plaque was 64.7%, 53.7%, and 38.5% among patients at high risk to low risk, respectively. Carotid plaques and risk stratification alone or in combination were significantly associated with ischemic stroke, and negatively correlated with coronary heart disease (all P>0.05). Adding carotid plaque to risk stratification, the ischemic stroke prevalence increased from 5.3% to 9.1% in the low-risk group (P=0.001), 5.4% to 12.3% in the intermediate-risk group (P<0.001) and 8.2% to 14.4% than in the high-risk group (P=0.004). Intermediate risk plus carotid plaque (443/3998) were reclassified to a new high-risk group, high risk only (749/3998) and low risk plus carotid plaque (353/3998) were reclassified to a new intermediate risk group; and intermediate risk only (553/3998) were reclassified to a new low risk group. According to the reclassification, there were 1635 subjects (40.9%) at high risk, and 1102 (27.6%), 1261 (31.5%) was classified as intermediate risk or low risk. Conclusions: Carotid plaque has an important position as it plus risk stratification may improve the risk assessment of ischemic stroke and have resulted in reclassification.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4650-4650
Author(s):  
Rami S Komrokji ◽  
Eric Padron ◽  
Najla H Al Ali ◽  
Jeffrey E Lancet ◽  
Jeff Hall ◽  
...  

Abstract Introduction The World Health organization (WHO) MDS classification proposes presence of 2-4% peripheral myeloblasts (PB) as criteria for Refractory anemia with excess blasts I (RAEB-I) and 5-19% PB for RAEB-II classification, while 1% PB persistently is recognized as MDS unclassified. The most widely used clinical prognostic models such as IPSS, R-IPSS, and MD Anderson risk model (MDAS) have not incorporated PB as a prognostic variable. We evaluated the prognostic value of PB in a large MDS cohort and provide a proposal to incorporate presence of PB in R-IPSS. Methods The MDS database at Moffitt Cancer Center (MCC) was used to identify MDS patients (pts). Pts were classified into two groups based on presence of PB (1% or more) at time of diagnosis as PB-MDS group and those without PB called BM-MDS. Kaplan-Meier curves were used to depict survivals, and the log-rank test was used to compare median overall survival (OS). We collaborated with Genoptix Medical Laboratory to assess the correlation between presence of PB and gene mutations identified by next generation sequencing (21 myeloid gene panel) in a cohort of de-identified MDS pts. Results There were 1905 pts included from MCC MDS database among whom 260 pts (14%) had evidence of PB near time of diagnosis. PB-MDS patients were younger, more likely to have trilineage cytopenia, complex karyotype, transfusion dependent and more in therapy related MDS. Among PB-MDS pts 175 (67%) received hypomethylating agent (HMA) compared to 977 (59%) in the BM-MDS group, p 0.017 According to IPSS risk stratification PB-MDS pts were more likely to be classified as higher risk (HR-MDS) 157 (63%) compared to 445 (28%) in the BM-MDS group (p<0.005). Pts with PB-MDS were also HR-MDS by MDAS (p <0.005). The rate of AML transformation was 46% (n=120) compared to 25% (n=418) in PB-MDS and BM-MDS group respectively, p <0.005. Median overall survival (OS) was 46 month (95% CI 42 to 49.6) in the absence of PB compared to 17.5 mo (95% CI 14.9-20) with PB (p < 0.005). Impact on OS was greater in IPSS lower risk MDS patients where median OS was 34 mo in PB-MDS compared to 60 mo in BM-MDS patients (p < 0.005), while in IPSS HR- MDS the median OS was 16.5 in PB-MDS compared to 18 month BM-MDS (p 0.018) We then examined prognostic discrimination of PB among each R-IPSS category. Median OS for very low risk R-IPSS was 104 mo in absence of PB compared to 37 mo in BM-MDS (p 0.032). Among low risk patients the median OS was 69 mo in absence of PB compared to 40 mo (p 0.07). In the intermediate risk group, median OS was 40 mo in absence of PB compared to 23 mo with PB (p 0.001), whereas median OS in the high risk group was 24 mo without PB compared to 20 mo with PB (p 0.11). Finally, in the very high risk R-IPSS the median OS was 15 mo compared to 13 mo respectively (p 0.44). The presence of PB upgraded pts with very low or low risk R-IPSS to intermediate risk. The outcome of intermediate risk group with PB was similar to the high risk group. In Cox regression analysis the presence of PB was an independent prognostic covariate for OS after adjusting for R-IPSS and age, HR 1.5 (95% CI 1.3-1.8). Among HR-MDS pts treated with HMA (n=470) presence of PB was independent prognostic variable for OS. (HR 1.3, p 0.027) We next created a R-IPSS+PB risk model where one additional point was awarded for presence of PB and pts categorized into risk groups based on the same lump score suggested by R-IPSS for each risk category. We applied this score for 245 pts with PB where R-IPSS score was known (Table-1). Sixty three pts (26%) were upstaged to high or very high risk group and most pts were upstaged to next risk group. Among 51 pts in Genoptix Medical Laboratory database with known PB, the rate of at least single gene mutation identification in pts with PB-MDS was 100%, (4 out of 4 with PB) compared to 81% in those without PB (38 out of 47 without PB, 12 of those were single SF3B1 gene mutation in ring sideroblasts MDS subtype). The gene mutations in PB-MDS included U2AF1 gene mutation in 2 pts, SRSF2, TET-2, ASXL-1, and RUNX-1. Two pts had two gene mutations. Conclusions Presence of PB in MDS is an adverse independent prognostic variable that refines prognostic discrimination in Low to Intermediate risk R-IPSS groups. Accounting for presence of PB particularly the intermediate risk group, prioritizes disease altering therapeutic strategies. TableRisk groupR-IPSSR-IPSS+PBNOverall survival (mo)NOverall survival (mo)Very low91low2440854intermediate51231238High63204330Very high991318115 Disclosures Hall: Genoptix Medical Laboratory: Employment. Kwok:Genoptix, Inc., a Novartis company: Employment, Equity Ownership.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e17016-e17016
Author(s):  
You Wu ◽  
Li Nan ◽  
Jusheng An ◽  
Manni Huang ◽  
Shaokang Ma ◽  
...  

e17016 Background: The aim of this study was to evaluate the efficacy and safety of paclitaxel/carboplatin as an adjuvant systemic chemotherapy for stage IB-IIA cervical adenocarcinoma patients after radical hysterectomy and pelvic lymphadenectomy with or without radiotherapy(RT). Methods: The cases of all patients (n = 152) with FIGO IB-IIA cervical adenocarcinoma who were treated by radical hysterectomy and pelvic lymphadenectomy with or without RT at Chinese academy of medical sciences cancer institute and hospital between January 2008 and January 2017 were reviewed. Of these, 35 patients displayed high-risk prognostic factors (high-risk group), 65 displayed intermediate-risk prognostic factors meeting sedlis criteria(intermediate-risk group),and 54 cases presented poorly differentiated status or LVSI, without other risk factors. In the high-risk group,18 patients received adjuvant radiotherapy and concurrent cisplatin chemoradiotherapy(RT-CCRT ) and 17 received adjuvant radiotherapy plus systemic chemotherapy using Paclitaxel Plus Carboplatin (RT-CT ). In the intermediate-risk group, 47 patients were treated with RT-CCRT and 16 were treated with RT-CT ,among the patients with poorly differentiated status or LVSI ,20 were treated with adjuvant chemotherapy using Paclitaxel Plus Carboplatin (CT ) and 34 received no further treatment (NFT). Results: In the high-risk group, adjuvant RT-CT was significantly superior to RT-CCRT with regard to median PFS and 2-year PFS rate(93.07months vs 52,44months, 94.1% vs 72.2%), but the recurrence rate and survival rate showed no significantly difference(11.8% vs 33.3%,100% vs 83.3%) . In the intermediate-risk group, Patients receiving RT-CT showed a better 2-year PFS rate and recurrence rate compared to those with RT-CCRT (100% vs 90.7%, 6.3% vs 12.6%), but the differences were not statistically significant. Among the patients with poorly differentiated status or LVSI, addition of adjuvant systemic chemotherapy resulted in significantly improved 2-year PFS rate compared with the NFT group (95% vs. 75.2%,p = 0.032). Conclusions: Adjuvant systemic chemotherapy using Paclitaxel Plus Carboplatin improved the prognosis of FIGO stage IB-IIA cervical adenocarcinoma patients in the high-risk group and patients who presented 2 or more intermediate-risk factors. Early cervical adenocarcinoma patients with low differentiation or LVSI can also benefit from postoperative adjuvant TC chemotherapy.


Author(s):  
L. N. Suslov ◽  
O. G. Sukonko ◽  
L. V. Mirilenko

The objective of this study is to devise a prognostic classification of AKI in the postoperative period based on preoperative factors for patients after partial nephrectomy for renal masses in the solitary kidney (SK). This method will allow identifying patients in a high risk group and reducing this unfavorable postoperative outcome by a careful treatment planning.We present a series of 136 patients with SK tumor who underwent open partial nephrectomy in situ performed at the N. N. Alexandrov National Cancer Centre of Belarus in 2000–2016. During the early postoperative period, AKI occurred in 28 (20.6 %) patients. Three risk factors associated with a risk of developing AKI were included in the multivariate analysis: categorized risk factors such as tumor size and serum potassium and dichotomous – multifocality. On the basis of the multivariant model presented, 3 risk factors were assigned a weighted score. Depending on the score, the cohort of patients was divided into 3 groups. Patients with a score from 0 to 2 were classified as a low-risk group, 3 points – an intermediate-risk group and ≥4 points – a high-risk group with the development of AKI in the postoperative period.The devised prognostic classification allows one during the preoperative period with a predictive accuracy of 82.3 % to determine a risk of development of AKI after partial nephrectomy of SK. In the low-risk group, the probability of developing AKI after surgery is 5.6 %, in the intermediate-risk group – 2.9 %, in the high-risk group – 68.2 % (р < 0.001).


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4618-4618
Author(s):  
Guangshuai Teng ◽  
Yuhui Zhang ◽  
Chao Gao ◽  
Naibo Hu ◽  
Chenxiao Du ◽  
...  

Abstract Thrombosis is a major cause of morbidity and mortality in polycythemia vera (PV). Post PV myelofibrosis (MF) is the advanced phase in the natural progression of PV. To explore the risk factors for thrombosis in post-PV MF, clinical characteristics, laboratory characteristics, the incidence of thrombosis and survival were retrospectively analyzed in a cohort of 163 Chinese patients with post-PV MF. The Kaplan-Meier method and multivariate Cox analysis were used to identify the risk factors and a risk model for thrombosis was established. Among the 163 patients, the median follow-up duration was 6 (1-18) years. During follow-up, 84 (51.5%) patients developed thrombosis, 11 (6.7%) patients progressed to acute leukemia, and 35 (21.5%) patients died (20% of whom died due to thrombosis). The 5-year, 10-year, and 15-year thrombosis-free survival (TFS) rates were 59.8%, 28.2%, and 9%, respectively. The TFS time of the post-PV MF patients was significantly lower than that of the age- and sex-matched PV patients (P&lt;0.001). The incidence of venous thrombosis was significantly higher after the diagnosis of post-PV MF than before or at the time of the diagnosis, and the proportions of patients with JAK 2 V617F allele burden ≥75% or absolute monocyte count ≥1.5×10 9/L was significantly higher in the venous thrombosis group than in the group without venous thrombosis (P&lt;0.05). Multivariate analysis showed that palpable splenomegaly (P=0.008, HR=3.284, 95% CI [1.373,7.855]), age ≥60 years (P=0.048, HR=1.604, 95% CI [1.004,2.56]), and a history of thrombosis (P&lt;0.001, HR=2.767, 95% CI [1.735, 4.412]) were risk factors for thrombosis in post-PV MF patients, then a risk model for thrombosis was established according to these data. The median TFS durations in patients in the extremely high-risk group, high-risk group, intermediate-risk group, and low-risk group were 2 years, 4 years, 9 years, and 13 years, respectively. In summary, post-PV MF patients have a higher incidence of thrombosis. Reducing the volume of the spleen and the allele burden of JAK2 V617F is critical to prevent thrombosis in post-PV MF patients. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e20005-e20005
Author(s):  
Camille Tessier ◽  
Thomas Allard ◽  
Jean-Samuel Boudreault ◽  
Rayan Kaedbey ◽  
Flechere Fortin ◽  
...  

e20005 Background: Smoldering multiple myeloma (SMM) is an asymptomatic plasma cell disorder associated with a high risk of transformation to symptomatic multiple myeloma (MM). SMM risk of progression to MM is highly heterogeneous and several models have been suggested to predict this risk, but none have yet been adopted internationally. Lakshman et al. recently proposed a risk stratification model based on three markers: bone marrow plasma cell (BMPC) percentage > 20%, free light chain ratio (FLCr) > 20 and serum M protein > 20 g/L. In this “20/20/20” model, patients with 0, 1 or ≥ 2 risk factors are respectively considered at low, intermediate and high-risk of progression. The goal of our study was to test this risk model in our population and to determine if similar results could be obtained in another cohort of SMM patients. Methods: We conducted a retrospective, single center study with 89 patients diagnosed with SMM between January 2008 and December 2019. Patients were identified by query of the electronic medical records and the 2014 International Myeloma Working Group (IMWG) diagnostic criteria for SMM were used to determine eligibility. The main endpoint was progression to symptomatic multiple myeloma or amyloidosis. Results: All three markers proposed by Lakshman et al. were associated with an increased risk of progression: BMPC percentage ≥ 20% (hazard ratio [HR]: 4.28 [95% C.I., 1.90 – 9.61]; p < 0.001), serum M protein ≥ 20g/L (HR: 4.20 [95% C.I., 1.90 – 15.53]; p = 0.032) and FLCr ≥ 20 (HR: 3.25 [95% C.I., 1.09 – 9.71]; p = 0.035). Immunoparesis (HR: 2.61 [95% C.I., 1.07 – 6.41]; p = 0.036) was also an independent risk factor in our population. The estimated median time to progression (TTP) was not reached for the low and intermediate risk groups and was 29.1 months (95% C.I., 3.9 – 54.4) in the high-risk group (p = 0.006). The estimated mean TTP for the low-risk group, the intermediate-risk group and the high-risk group were respectively 78.4 months (95% C.I., 68.3 – 88.5), 48.3 months (95% C.I., 31.9 – 64.8) and 35.2 months (95% C.I., 19.1 – 51.2). Sex, IgA isotype, positive Bence-Jones, abnormal β2-microglobulin and MGUS prior to SMM did not result in an increased risk of progression. The estimated proportion of progression-free patients at 1, 2 and 5 years were 96.8%, 93.4% and 77.5% for the low-risk group, 80.0%, 80.0% and 62.2% for the intermediate risk group and 70.0%, 58.3% and 29.2% for the high-risk group. Conclusions: When Mayo Clinic’s new 20/20/20 risk model was applied to our population, it adequately predicted the risk of progression to symptomatic disease at 2 years. As it relies on readily available biological parameters, this model is easy to use and can be applied in most clinical settings. We believe this model could be used to further study therapeutic approaches in higher risk SMM.


Sign in / Sign up

Export Citation Format

Share Document