Skip to main content
Erschienen in: BMC Cancer 1/2024

Open Access 01.12.2024 | Research

Development of a risk assessment model for cardiac injury in patients newly diagnosed with acute myeloid leukemia based on a multicenter, real-world analysis in China

verfasst von: Linlu Ma, Qian Wang, Xinqi Li, Yufeng Shang, Nan Zhang, Jinxian Wu, Yuxing Liang, Guopeng Chen, Yuxin Tan, Xiaoyan Liu, Guolin Yuan, Fuling Zhou

Erschienen in: BMC Cancer | Ausgabe 1/2024

Abstract

Background

Studies have revealed that acute myeloid leukemia (AML) patients are prone to combined cardiac injury. We aimed to identify hematological risk factors associated with cardiac injury in newly diagnosed AML patients before chemotherapy and develop a personalized predictive model.

Methods

The population baseline, blood test, electrocardiogram, echocardiograph, and genetic and cytogenetic data were collected from newly diagnosed AML patients. The data were subdivided into training and validation cohorts. The independent risk factors were explored by univariate and multivariate logistic regression analysis respectively, and data dimension reduction and variable selection were performed using the least absolute shrinkage and selection operator (LASSO) regression models. The nomogram was generated and the reliability and generalizability were verified by receiver operating characteristic (ROC) curves, the area under the curve (AUC) and calibration curves in an external validation cohort.

Results

Finally, 499 AML patients were included. After univariate logistic regression, LASSO regression and multivariate logistic regression analysis, abnormal NT-proBNP, NPM1 mutation, WBC, and RBC were independent risk factors for cardiac injury in AML patients (all P < 0.05). The nomogram was constructed based on the above four variables with high accuracy. The area under the curve was 0.742, 0.750, and 0.706 in the training, internal validation, and external validation cohort, respectively. The calibration curve indicated that the model has good testing capability. The Kaplan-Meier curve showed that the higher the risk of combined cardiac injury in AML patients, the lower their probability of survival.

Conclusions

This prediction nomogram identifies hematological risk factors associated with cardiac injury in newly diagnosed AML patients and can help hematologists identify the risk and provide precise treatment options.
Hinweise
LM, QW and XL contributed equally to this work and should be considered as co−first authors.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Cancer and cardiovascular disease (CVD) are the two most common diseases in the world with a higher mortality rate [1]. Leukemia is a life-threatening hematological cancer, with 474,519 new cases and 311,594 deaths worldwide in 2020 [2]. Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with a 5-year survival rate of less than 40% [3]. From 1990 to 2017, the global number of disability-adjusted life years caused by AML increased by 56.14% [4]. Especially for the AML cases over 60 years old, accounting for the vast majority of AML patients, the 5-year overall survival (OS) is only 10–20% [5].
Greater overlap between cancer and CVD is observed due to an aging population and the sharing of common risk factors and biological pathways [6]. The common risk factors of both disease entities [7] include tobacco smoking, obesity, unhealthy diet, hypertension, diabetes, etc. The potential mechanisms contributing to this overlap include chronic inflammation, oxidative stress, a prothrombotic state, metabolic derangements, genetic predisposition, and clonal hematopoiesis of indeterminate potential (CHIP) [8]. Additionally, autopsies conducted on individuals who succumbed to acute leukemia (AL) revealed that a considerable proportion, up to 37%, exhibited leukemic infiltrates within the cardiac structures, including the walls of cardiac chambers, pericardia and subepicardial adipose tissue [911]. Furthermore, the occurrence of leukemic cardiac infiltration was notably higher in AML compared to acute lymphoblastic leukemia (ALL) [12]. It is worth noting that while certain leukemia patients were admitted to the hospital due to cardiac manifestations as their initial [1315] or accompanying symptoms [1618], the reported incidence of cardiac injury in the conducted studies may only represent a fraction of the actual prevalence, primarily due to the absence of early cardiac monitoring [19].
Hence, it is postulated that leukemia per se induces some cardiac injury through the release of excessive cytokines or infiltration of leukemic cells, even before the administration of anti-tumor medications. Our team conducted a cardiac assessment of newly diagnosed AL patients in the Chinese population and found that the myocardial enzyme levels in AL patients were significantly higher than those in healthy controls [20]. Consequently, we intend to investigate additional hematological risk factors linked to cardiac injury in newly diagnosed AML patients, with the ultimate goal of constructing a prediction model that would assist hematologists in identifying varying levels of risk and offering precise treatment options for individual patients.

Materials and methods

Participants of inclusion

Data for this study were divided into three data sets from two medical centers. We analyzed the information of newly diagnosed AML patients between January 2017 and December 2022 at Wuhan University Zhongnan Hospital and Xiangyang City Centre Hospital. AML patients from Wuhan University Zhongnan Hospital were randomly divided into a training set and a testing set according to a ratio of 7:3, and those from Xiangyang City Centre Hospital were used as an external validation set. This study complied with the ethical guidelines of the Declaration of Helsinki and was approved by the ethics review committee of Wuhan University Zhongnan Hospital.
All included patients were diagnosed with AML according to the 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia [21] and did not receive chemotherapy. Patients younger than 18 years old, with prior or concurrent malignancy, with a known history of cardiomyopathy, coronary artery disease (CAD) or heart failure (HF) were excluded from this study. The cases without cardiac biomarkers, including values of high-sensitivity troponin I (hs-TNI) or creatinine kinase-myocardial band (CK-MB) or electrical abnormal markers, were excluded.

Definition of cardiac injury and outcomes

Previous studies have defined cardiac injury based on cardiac biomarkers obtained through blood sampling and the abnormalities on ECG [2224]. Combined with literature and clinical experience, cardiac injury was defined when the following abnormalities were present at the same time: (1) cardiac biomarkers (abnormal CK-MB or hs-TNI, above the 99th-percentile upper reference limit); (2) electrical abnormal markers (cardiac arrhythmias, such as: atrial flutter/fibrillation, supraventricular tachycardia, ventricular tachycardia and ventricular fibrillation, bundle branch block, T wave flattening/inversion, ST-segment elevation/depression and QT interval prolongation were first detected or recently developed on ECG). In addition, we divided these newly diagnosed AML patients into two groups according to whether they had concomitant cardiac injury, assessed factors associated with cardiac injury, and identified patients at high risk of cardiac injury to determine appropriate prevention strategies.

Risk factors exploration and nomogram establishment

Gender, age, cardiovascular risk factors (history of smoking, hypertension, and diabetes), laboratory tests: complete blood count, myocardial enzymes, N-terminal pro-B-type natriuretic peptide (NT-proBNP), hs-TNI, bone marrow (BM) manifestation, molecular genetics, cytogenetics, ECG and echocardiogram data were all obtained from electronic medical records with standardized data collection forms. The data were reviewed by a team of trained physicians who also followed up the patients’ survival status and survival time.
Univariate logistic regression was used to identify independent variables, and nonzero coefficients were identified by the least absolute shrinkage and selection operator (LASSO) regression analysis. Then, multivariate logistic regression was performed to acquire variables. A nomogram prediction model was developed, and its efficacy was assessed through internal and external validation in the testing set and external validation set respectively. The predictive accuracy of the nomograms was assessed by using receiver operating characteristic curve (ROC) analysis and calculating the area under the curve (AUC). Calibration curves were used to compare the agreement of the predicted and actual probabilities of the nomogram. We calculated all risk scores from the nomogram and classified them as low and high risk, respectively. Finally, we also assessed the prognosis by risk stratification.

Statistical analysis

All statistical analyzes were performed using IBM SPSS version 20.0 and R version 4.2.2. Continuous variables were reported as median with interquartile ranges (IQR), while categorical variables were reported as numbers with proportions. Patient characteristics were compared using the chi-square test or Fisher’s exact test for categorical variables, and the Mann-Whitney U test for continuous variables using IBM SPSS statistics software. The survival rate of AML patients with or without cardiac injury was analyzed by Kaplan-Meier (KM) curve. Univariate logistic regression, imputation of a small amount of missing data, the LASSO regression and multivariate logistic regression were performed by R to screen meaningful variables to develop a predictive model. Nomograms, calibration curves and ROC were performed or plotted using R version 4.2.2 ultimately. All reported p-values were two-sided with a significance level of 0.05.

Results

Basic characteristics of patients

Finally, 499 patients with newly diagnosed AML from two medical centers, including 272 males and 227 females, were included in this study. Data from 399 patients from Wuhan University Zhongnan Hospital were used for the training and testing sets, and data from 100 patients from Xiangyang City Centre Hospital were used for the external validation set. The analysis flow chart of this study was shown in Fig. 1.

Comparison of basic characteristics between AML patients with or without cardiac injury

The cohort of newly diagnosed AML patients was divided into two groups based on the occurrence of cardiac injury for characteristic assessment. The clinical, molecular and biochemical characteristics of the above two groups of patients were summarized in detail below (Table 1). There were no significant differences in gender, age, cardiovascular risk factors (the smoking history, hypertension history and diabetes history) between the two groups. The AML patients with cardiac injury had higher WBC counts (P < 0.001) and marrow blasts (P = 0.007), and a higher percentage of gene mutations, such as NPM1 (P < 0.001), FLT3 (P < 0.001), Ras (P = 0.021), WT1 (P = 0.003) and JAK2 (P = 0.002), when compared to the patients without cardiac injury. Moreover, the patients with cardiac injury were more often diagnosed with M5 (P = 0.004), but less diagnosed with M1 compared with other subgroups (P = 0.026). However, no statistical differences were observed between the two groups in terms of cytogenetics. In addition, AML patients with cardiac injury had a higher rate of abnormal NT-proBNP (P < 0.001) and lower EF values. Finally, to determine the prognostic effect of combined cardiac injury on AML patients, we performed a KM analysis of the two groups, and the KM curves showed that patients with newly diagnosed AML combined with cardiac injury had a poorer prognosis (P = 0.025), see Fig. 2.
Table 1
Basic characteristics of newly diagnosed AML patients with or without cardiac injury
Characteristics
Total (n = 399)
No. (%) / Median (IQR)
Comparison
With cardiac injury (n = 161)
No. (%) / Median (IQR)
Without cardiac injury (n = 238)
No. (%) / Median (IQR)
P
Gender
   
0.863
 Male
216(54.1)
88(54.7)
128(53.8)
 
 Female
183(45.9)
73(45.3)
110(46.2)
 
Age (years)
   
0.309
 Age < 60
218(54.6)
83(51.6)
135(56.7)
 
 Age ≥ 60
181(45.4)
78(48.4)
103(43.3)
 
Smoking history
67(16.8)
27(16.8)
40(16.8)
0.992
History of hypertension
67(16.8)
28(17.4)
39(16.4)
0.792
History of diabetes
29(7.3)
16(9.9)
13(5.5)
0.091
WBC (109/L)
10.14(2.2-37.13)
24.6(5.87–77.2)
5.27(1.72-15.0425)
< 0.001
RBC (1012/L)
2.19(1.7–2.62)
2.32(1.83–2.87)
2.12(1.65–2.56)
0.002
Hb (g/L)
70(56.4–85.8)
70(59.35–87.4)
69.55(54.6-84.85)
0.152
PLT (109/L)
38(21–73)
33(19-77.5)
39(21–73)
0.602
BM blast ratio (%)
59.5(35.5–80.5)
64(37-83.25)
57(33.375-76)
0.007
FAB subtypes
    
 M1
32(8)
7(4.3)
25(10.5)
0.026
 M2
176(44.1)
66(41.0)
110(46.2)
0.302
 M3
52(13)
19(11.8)
33(13.9)
0.548
 M4
22(5.5)
8(5.0)
14(5.9)
0.695
 M5
72(18)
40(24.8)
32(13.4)
0.004
 M6
5(1.3)
3(1.9)
2(0.8)
0.658
 Unkown
40(10)
18(11.2)
22(9.2)
0.527
Gene mutations
    
 NPM1
63(15.8)
42(26.1)
21(8.8)
< 0.001
 CEBPA
42(10.5)
17(10.6)
25(10.5)
0.986
 FLT3
67(16.8)
43(26.7)
24(10.1)
< 0.001
 C-kit
15(3.8)
9(5.6)
6(2.5)
0.114
 TP53
24(6.0)
12(7.5)
12(5.0)
0.320
 RUNX1
17(4.3)
7(4.3)
10(4.2)
0.943
 Ras
20(5.0)
13(8.1)
7(2.9)
0.021
 TET2
26(6.5)
14(8.7)
12(5.0)
0.147
 IDH
37(9.3)
18(11.2)
19(8.0)
0.280
 WT1
235(58.9)
109(67.7)
126(52.9)
0.003
 JAK2
11(2.8)
10(6.2)
1(0.4)
0.002#
 AML1-ETO
29(7.3)
14(8.7)
15(6.3)
0.366
 CBFB-MYH11
17(4.3)
6(3.7)
11(4.6)
0.664#
 BCR-ABL
14(3.5)
9(5.6)
5(2.1)
0.063#
 MLL
23(5.8)
8(5.0)
15(6.3)
0.575
Cytogenetics
    
 Abnormal karyotype
209(52.4)
92(57.1)
117(49.2)
0.117
 Complex karyotype
71(17.8)
26(16.1)
45(18.9)
0.480
 Monosomy karyotype
44(11.0)
16(9.9)
28(11.8)
0.568
 t8-21
23(5.8)
13(8.1)
10(4.2)
0.103
 inv16
15(3.8)
6(3.7)
9(3.8)
0.977#
 t9-22
5(1.3)
3(1.9)
2(0.8)
0.658#
 plus8
38(9.5)
16(9.9)
22(9.2)
0.817
 del5q
29(7.3)
14(8.7)
15(6.3)
0.366
 del7
25(6.3)
9(5.6)
16(6.7)
0.647
 del17p
14(3.5)
6(3.7)
8(3.4)
0.846#
Abnormal NT-proBNP
101(25.3)
58(36.0)
43(18.1)
< 0.001
Echocardiogram indexes
    
 LA (mm)
34(30–38)
34(31-37.5)
33(29.75-38)
0.357
 LV (mm)
45(42–48)
45(43–48)
46(42–48)
0.871
 IVS (mm)
10(9–11)
10(9–11)
10(9–11)
0.019
 EDV (ml)
97(78–113)
95(80-108.5)
97(78–118)
0.254
 ESV (ml)
31(25–40)
31(24–41)
30(25–39)
0.910
 SV (ml)
66(55–78)
64(53–75)
67(56–80)
0.172
 EF (%)
65(60–70)
64(60-70.5)
66(60–70)
0.062
#: Fisher’s exact test
NPM1: nucleophosmin 1; CEBPA: CCAAT/enhancer-binding protein alpha; FLT3: Fms-like tyrosine kinase 3; TP53, Tumor Protein P53; RUNX1: runt-related transcription factor 1; TET2: tet methylcytosine dioxygenase 2; IDH: Isocitrate dehydrogenase; WT1: Wilm tumor gene1; JAK2: Janus kinase 2; AML1-ETO: acute myeloid leukemia 1 Eight-twenty-one; CBFB-MYH11: core-binding factor, beta subunit-myosin heavy chain 11; BCR-ABL: breakpoint cluster region-Abelson; MLL: Mixed lineage leukemia; NT-pro BNP: N-terminal pro-B-type natriuretic peptide; WBC: White blood cells; RBC: Red blood cells; Hb: Hemoglobin; PLT: Platelets; LA: Left atrium; LV: Left ventricle; IVS: Interventricular septum; EDV: End-diastolic volume; ESV: End-systolic volume; SV: stroke volume; EF: Ejection fraction; BM: bone marrow

Baseline characteristics of the training set and testing set

The data of 399 AML patients from Wuhan University Zhongnan Hospital were grouped into a training set and a testing set by the random digital grouping method, as shown in Table 2. Of the 279 patients in the training set, 113 (40.5%) had cardiac injury, and of the 120 patients in the testing set, 48 (40%) had cardiac injury. There were no statistical differences in baseline characteristics of the patients between the training and testing set (P > 0.05). In addition, of the 100 cases in the externally validated cohort, 48 were combined with cardiac injury. The comparison between AML patients with and without cardiac injury in the training set, testing set, and external validation set was detailed in Table 3.
Table 2
Comparisons of baseline characteristics between training set and testing set
Characteristics
Training set (n = 279)
No. (%) / Median (IQR)
Testing set (n = 120)
No. (%) / Median (IQR)
P
Cardiac injury
113(40.5)
48(40.0)
0.925
Gender
  
0.993
 Male
151(54.1)
65(54.2)
 
 Female
128(45.9)
55(45.8)
 
Age (years)
  
0.593
 Age < 60
150(53.8)
68(56.7)
 
 Age ≥ 60
129(46.2)
52(43.3)
 
Smoking history
47(16.8)
20(16.7)
0.965
History of hypertension
47(16.8)
20(16.7)
0.965
History of diabetes
21(7.5)
8(6.7)
0.761
WBC (109/L)
10.34(2.23–40.62)
8.835(2.005–25.98)
0.395
RBC (1012/L)
2.2(1.7–2.66)
2.185(1.7–2.58)
0.827
Hb (g/L)
70(56.2–86.2)
69.85(56.725–84.45)
0.946
PLT (109/L)
37(20–71)
40(21-81.5)
0.307
BM blast ratio (%)
59.5(35.5–81.5)
59.25(35.75–78.05)
0.740
Gene mutations
   
 NPM1
43(15.4)
20(16.7)
0.753
 CEBPA
31(11.1)
11(9.2)
0.562
 FLT3
47(16.8)
20(16.7)
0.965
 C-kit
12(4.3)
3(2.5)
0.562
 TP53
17(6.1)
7(5.8)
0.920
 RUNX1
11(3.9)
6(5.0)
0.632
 Ras
16(5.7)
4(3.3)
0.313
 TET2
16(5.7)
10(8.3)
0.335
 IDH
25(9.0)
12(10.0)
0.743
 WT1
165(59.1)
70(58.3)
0.881
 JAK2
6(2.2)
5(4.2)
0.427
 AML1-ETO
19(6.8)
10(8.3)
0.591
 CBFB-MYH11
13(4.7)
4(3.3)
0.548
 BCR-ABL
9(3.2)
5(4.2)
0.864
 MLL
14(5.0)
9(7.5)
0.329
Cytogenetics
   
 Abnormal karyotype
149(53.4)
60(50.0)
0.532
 Complex karyotype
44(15.8)
27(22.5)
0.107
 Monosomy karyotype
28(10.0)
16(13.3)
0.335
 t8-21
15(5.4)
8(6.7)
0.612
 inv16
12(4.3)
3(2.5)
0.562
 t9-22
4(1.4)
1(0.8)
0.997
 plus8
25(9.0)
13(10.8)
0.559
 del5q
22(7.9)
7(5.8)
0.469
 del7
17(6.1)
8(6.7)
0.828
 del17p
9(3.2)
5(4.2)
0.864
Abnormal NT-proBNP
70(25.1)
31(25.8)
0.875
Echocardiogram indexes
   
 LA (mm)
34(30–38)
34(30–38)
0.823
 LV (mm)
45(42–48)
46(42.25-48)
0.678
 IVS (mm)
10(9–11)
10(9–11)
0.312
 EDV (ml)
97(78–111)
97(82.25–113)
0.930
 ESV (ml)
31(25–40)
30(24.25-41)
0.704
 SV (ml)
64(54–77)
68(56-78.75)
0.256
 EF (%)
65(60–70)
65.5(60–71)
0.839
NPM1: nucleophosmin 1; CEBPA: CCAAT/enhancer-binding protein alpha; FLT3: Fms-like tyrosine kinase 3; TP53, Tumor Protein P53; RUNX1: runt-related transcription factor 1; TET2: tet methylcytosine dioxygenase 2; IDH: Isocitrate dehydrogenase; WT1: Wilm tumor gene1; JAK2: Janus kinase 2; AML1-ETO: acute myeloid leukemia 1 Eight-twenty-one; CBFB-MYH11: core-binding factor, beta subunit-myosin heavy chain 11; BCR-ABL: breakpoint cluster region-Abelson; MLL: Mixed lineage leukemia; NT-pro BNP: N-terminal pro-B-type natriuretic peptide; WBC: White blood cells; RBC: Red blood cells; Hb: Hemoglobin; PLT: Platelets; LA: Left atrium; LV: Left ventricle; IVS: Interventricular septum; EDV: End-diastolic volume; ESV: End-systolic volume; SV: stroke volume; EF: Ejection fraction; BM: bone marrow
Table 3
Distribution and comparisons of AML patients with or without cardiac injury in the training set, testing set and external validation set
 
Training set
Testing set
External validation set
Characteristics
Cardiac injury
(n = 113)
No cardiac injury (n = 166)
P
Cardiac injury
(n = 48)
No cardiac injury
(n = 72)
P
Cardiac injury
(n = 48)
No cardiac injury
(n = 52)
P
Gender
  
0.652
  
0.708
  
0.393
 Male
63(55.8)
88(53.0)
 
25(52.1)
40(55.6)
 
29(60.4)
27(51.9)
 
 Female
50(44.2)
78(47.0)
 
23(47.9)
32(44.4)
 
19(39.6)
25(48.1)
 
Age (years)
  
0.159
  
0.764
  
0.030
 Age < 60
55(48.7)
95(57.2)
 
28(58.3)
40(55.6)
 
21(43.8)
34(65.4)
 
 Age ≥ 60
58(51.3)
71(42.8)
 
20(41.7)
32(44.4)
 
27(56.2)
18(34.6)
 
Smoking history
20(17.7)
27(16.3)
0.753
7(14.6)
13(18.1)
0.617
14(29.2)
10(19.2)
0.245
History of hypertension
21(18.6)
26(15.7)
0.522
7(14.6)
13(18.1)
0.617
9(18.8)
6(11.5)
0.313
History of diabetes
13(11.5)
8(4.8)
0.038
3(6.2)
5(6.9)
1.000
6(12.5)
3(5.8)
0.409
WBC (109/L)
27.56(6.53-76.505)
5.52(1.7675–16.065)
< 0.001
21.205(5.0675–86.14)
4.6950(1.6375–14.2175)
< 0.001
28.115(3.77-64.7125)
4.315(1.9-23.7675)
0.011
RBC (1012/L)
2.32(1.83–2.88)
2.14(1.6325-2.56)
0.018
2.295(1.84-2.8575)
2.07(1.655-2.5)
0.033
2.385(1.8075–2.945)
2.23(1.7975-2.56)
0.210
Hb (g/L)
70(58.65-87)
69.85(54.75-85.425)
0.331
70.85(59.775–90.375)
68.8(54.375–82.95)
0.263
78.4(61-88.7)
72.2(58.775–88.5)
0.558
PLT (109/L)
31(17.5–70)
38(22-72.25)
0.357
39(22-89.5)
43.5(20.25-73)
0.682
29.5(22.25-96)
37.5(23.25–71.5)
0.959
BM blast ratio (%)
65.2(37.95–83.75)
56.15(31.875–77.35)
0.015
59.75(34.825-83)
57.75(36.625–72.875)
0.247
65.75(48.47-85.6675)
62.25(24.7775-79)
0.013
Gene mutations
         
 NPM1
29(25.7)
14(8.4)
< 0.001
13(27.1)
7(9.7)
0.012
14(29.2)
9(17.3)
0.159
 CEBPA
13(11.5)
18(10.8)
0.863
4(8.3)
7(9.7)
1.000
10(20.8)
7(13.5)
0.327
 FLT3
30(26.5)
17(10.2)
< 0.001
13(27.1)
7(9.7)
0.012
16(33.3)
9(17.3)
0.064
 C-kit
7(6.2)
5(3.0)
0.324
2(4.2)
1(1.4)
0.720
4(8.3)
5(9.6)
1.000
 TP53
8(7.1)
9(5.4)
0.570
4(8.3)
3(4.2)
0.578
5(10.4)
2(3.8)
0.371
 RUNX1
4(3.5)
7(4.2)
1.000
3(6.2)
3(4.2)
0.932
4(8.3)
3(5.8)
0.913
 Ras
10(8.8)
6(3.6)
0.065
3(6.2)
1(1.4)
0.350
5(10.4)
5(9.6)
1.000
 TET2
7(6.2)
9(5.4)
0.785
7(14.6)
3(4.2)
0.092
7(14.6)
4(7.7)
0.271
 IDH
12(10.6)
13(7.8)
0.423
6(12.5)
6(8.3)
0.664
5(10.4)
4(7.7)
0.900
 WT1
77(68.1)
88(53.0)
0.012
32(66.7)
38(52.8)
0.131
22(45.8)
22(42.3)
0.723
 JAK2
5(4.4)
1(0.6)
0.082
5(10.4)
0(0)
0.020
3(6.2)
1(1.9)
0.554
 AML1-ETO
9(8.0)
10(6.0)
0.528
5(10.4)
5(6.9)
0.736
5(10.4)
0(0)
0.054
 CBFB-MYH11
5(4.4)
8(4.8)
0.878
1(2.1)
3(4.2)
0.917
2(4.2)
4(7.7)
0.749
 BCR-ABL
5(4.4)
4(2.4)
0.555
4(8.3)
1(1.4)
0.162
1(2.1)
2(3.8)
1.000
 MLL
5(4.4)
9(5.4)
0.708
3(6.2)
6(8.3)
0.944
2(4.2)
2(3.8)
1.000
Cytogenetics
         
Abnormal karyotype
66(58.4)
83(50.0)
0.167
26(54.2)
34(47.2)
0.456
31(64.6)
29(55.8)
0.369
Complex karyotype
16(14.2)
28(16.9)
0.542
10(20.8)
17(23.6)
0.721
9(18.8)
7(13.5)
0.471
Monosomy karyotype
11(9.7)
17(10.2)
0.890
5(10.4)
11(15.3)
0.443
7(14.6)
3(5.8)
0.257
 t8-21
8(7.1)
7(4.2)
0.298
5(10.4)
3(4.2)
0.331
2(4.2)
0(0)
0.228
 inv16
5(4.4)
7(4.2)
1.000
1(2.1)
2(2.8)
1.000
3(6.2)
4(7.7)
1.000
 t9-22
2(1.8)
2(1.2)
1.000
1(2.1)
0(0)
0.400
1(2.1)
2(3.8)
1.000
 plus8
10(8.8)
15(9.0)
0.957
6(12.5)
7(9.7)
0.631
4(8.3)
2(3.8)
0.601
 del5q
11(9.7)
11(6.6)
0.344
3(6.2)
4(5.6)
1.000
5(10.4)
2(3.8)
0.371
 del7
7(6.2)
10(6.0)
0.953
2(4.2)
6(8.3)
0.601
4(8.3)
3(5.8)
0.913
 del17p
3(2.7)
6(3.6)
0.920
3(6.2)
2(2.8)
0.641
2(4.2)
3(5.8)
1.000
Abnormal NT-proBNP
41(36.3)
29(17.5)
< 0.001
17(35.4)
14(19.4)
0.050
21(43.8)
5(9.6)
< 0.001
Echocardiogram indexes
         
LA (mm)
34(31–38)
33(29–38)
0.459
34.5(31.25-37)
34(30–38)
0.588
35.5(31–38)
33(29–39)
0.445
LV (mm)
45(43–48)
45(42–48)
0.700
46(41.5–48)
46(42.25-48)
0.778
43.5(38.25-49)
45(41–48)
0.663
IVS (mm)
10(9–11)
10(9–11)
0.070
10(9.25-12)
10(9–11)
0.125
10(9–12)
10(9–11)
0.371
EDV (ml)
95(83-107.5)
97(78–118)
0.471
93.5(78-110.5)
99(83–118)
0.314
101.5(87-118.25)
100(88-108.75)
0.942
ESV (ml)
32(25–41)
30(25–39)
0.404
30.5(22–44)
30(25–39)
0.271
35(28-43.25)
31(25.25-39)
0.168
SV (ml)
64(53–73)
67(55–80)
0.151
68.5(56-77.75)
67.5(56.75–82.25)
0.799
68(56.25-78)
68(55.5–79)
0.855
EF (%)
63(60–69)
66.5(60–70)
0.045
64.5(60–71)
66(59.25–71.75)
0.674
60(57–66)
65(61.25–70.75)
< 0.001
NPM1: nucleophosmin 1; CEBPA: CCAAT/enhancer-binding protein alpha; FLT3: Fms-like tyrosine kinase 3; TP53, Tumor Protein P53; RUNX1: runt-related transcription factor 1; TET2: tet methylcytosine dioxygenase 2; IDH: Isocitrate dehydrogenase; WT1: Wilm tumor gene1; JAK2: Janus kinase 2; AML1-ETO: acute myeloid leukemia 1 Eight-twenty-one; CBFB-MYH11: core-binding factor, beta subunit-myosin heavy chain 11; BCR-ABL: breakpoint cluster region-Abelson; MLL: Mixed lineage leukemia; NT-pro BNP: N-terminal pro-B-type natriuretic peptide; WBC: White blood cells; RBC: Red blood cells; Hb: Hemoglobin; PLT: Platelets; LA: Left atrium; LV: Left ventricle; IVS: Interventricular septum; EDV: End-diastolic volume; ESV: End-systolic volume; SV: stroke volume; EF: Ejection fraction; BM: bone marrow

Risk factors and predictive models of cardiac injury

In the training set, variables with P < 0.1 were screened by univariate logistic regression analysis. These 12 variables were analyzed by LASSO regression, and 10 non-zero coefficients in lambda.1se were selected as candidate variables. The lambda.1se corresponding to λ value of 0.0447 was chosen and ten predictors were selected the model (Fig. 3A-B). The coefficients for each parameter were as follows: 0.4029 for abnormal NT-pro BNP, 0.1706 for history of diabetes, 0.6376 for NPM1, 0.0294 for FLT3, 0.0383 for Ras, 0.1220 for WT1, 0.5206 for JAK2, 0.0055 for WBC, 0.0942 for RBC, and − 0.0007 for EF. Then, by multivariate analysis, abnormal NT-pro BNP, NPM1, WBC and RBC were still independent risk factors for cardiac injury, and the odds ratio (OR) of the four variables were shown in Table 4.
Table 4
Multivariate logistic regression analyses of cardiac injury in AML patients from training set
Variables
Coefficients
SE
OR
95%CI
P
History of diabetes
     
 No
Ref.
 
Ref.
  
 Yes
0.875133
0.529612
2.4
0.85–6.77
0.098
Genetic mutations
     
 NPM1
     
  No
Ref.
 
Ref.
  
  Yes
1.10079
0.39808
3.01
1.38–6.56
0.006
 FLT3
     
  No
Ref.
 
Ref.
  
  Yes
0.240088
0.406053
1.27
0.57–2.82
0.554
 Ras
     
  No
Ref.
 
Ref.
  
  Yes
0.678078
0.581091
1.97
0.63–6.15
0.243
 WT1
     
  No
Ref.
 
Ref.
  
  Yes
0.487583
0.306741
1.63
0.89–2.97
0.112
 JAK2
     
 No
Ref.
 
Ref.
  
 Yes
2.178846
1.139206
8.84
0.95–82.41
0.056
NT-proBNP
     
 Normal
Ref.
 
Ref.
  
 Abnormal
0.911099
0.340318
2.49
1.28–4.85
0.007
WBC (109/L)
0.008208
0.002956
1.0082
1.0024–1.0141
0.005
RBC (1012/L)
0.376219
0.190353
1.46
1-2.12
0.048
EF (%)
-0.02026
0.018682
0.98
0.94–1.02
0.278
NPM1: nucleophosmin 1; FLT3: Fms-like tyrosine kinase 3; WT1: Wilm tumor gene1; JAK2: Janus kinase 2; NT-pro BNP: N-terminal pro-B-type natriuretic peptide; WBC: White blood cells; RBC: Red blood cells; EF: Ejection fraction

Nomogram establishment and validation

Based on the above analysis results, abnormal NT-pro BNP, NPM1, WBC and RBC were included in the prediction model as independent risk factors. We generated a cardiac injury risk model and presented it in the form of a nomogram to visually illustrate the probability of cardiac injury in newly diagnosed AML patients (Fig. 3C).
By using ROC analysis, the model showed high diagnostic accuracy in the training set, testing set, and external validation set (Fig. 4A-C). Our model had an AUC of 0.742 (0.683–0.802) on the training set. Meanwhile, the AUCs in the testing set and external validation set were 0.750 (0.658–0.842) and 0.706 (0.601–0.811), respectively, showing good prediction accuracy. The numbers on the subfigures represent the Youden index and its corresponding specificity and sensitivity. The performance of the model was validated by calibration curves, and no significant deviation from the reference line was observed on the training, testing, and external validation sets (after repetitive Bootstrap self-sampling 1,000 times show that the mean absolute errors of the simulated curves and the actual curves were 0.018, 0.029, and 0.031, respectively), suggesting good consistency between predictions in the training set and actual observations (Fig. 4D-F).

Risk score in Nomogram

We calculated nomogram scores for all newly diagnosed AML patients using R. Here, we showed all scores for each variable in a nomogram (Table 5). Patients were divided into low risk (points ≤ 17.85) and high risk (points > 17.85) according to the optimal threshold of the training set ROC curve. We found that the degree of cardiac injury in newly diagnosed AML patients increased with increasing risk (P < 0.001). KM curves were applied to better visualize the relationship between risk stratification and survival prognosis, which indicated that AML patients with higher risk of cardiac injury had a worse prognosis (P = 0.001, Fig. 4G-H).
Table 5
Points given for each variable in the nomogram
Variables
Nomogram points
NT-proBNP
 
 Abnormal
18
 Normal
0
NPM1-mutation
 
 Yes
24
 No
0
WBC (109/L)
 
0
 0
 50
9
 100
18
 150
27
 200
36
 250
45
 300
55
 350
64
 400
73
 450
82
 500
91
 550
100
RBC (1012/L)
 
 0.5
0
 1
4
 1.5
7
 2
11
 2.5
14
 3
18
 3.5
21
 4
25
 4.5
28
 5
32

Discussion

In this study, we developed and validated a personalized predictive nomogram and risk score for the risk of cardiac injury before chemotherapy in patients with newly diagnosed AML, using cost-effective and readily available variables to help hematologists identify high-risk patients. The prediction model includes four variables: WBC, RBC, NT-proBNP, and NPM1 mutations. Internal and external validation showed that our nomogram and risk scores have good predictive performance.
In recent years, the field of cardio-oncology has made significant progress in elucidating the intricate relationship between tumors and the heart. While much attention has been devoted to studying cardiac injury resulting from anti-tumor therapy, injury caused by the tumor itself is also coming into the limelight [25]. A large Danish HF Cohort study reported that when diagnosed with cancer, their all-cause mortality rate was higher than that of without HF [26]. Xiao W et al. found that newly diagnosed AL patients did experience some cardiac-related lesions prior to chemotherapy, and this injury persisted after adjusting for confounding factors. In addition, cardiac enzyme abnormalities were more severe in the hyperleukocytic leukemia subgroup (WBC count ≥ 100 × 109/L), and the proportion of blasts was positively correlated with cardiac injury [20]. In agreement with our result, a retrospective study from Harvard Medical School also found that AL was associated with cardiac alterations before chemotherapy [27], which together with other similar studies [28, 29], suggests that leukemia itself may cause direct damage to the heart prior to anti-tumor therapy.
From an etiological perspective, several factors may explain the development of cardiac injury in AL patients before chemotherapy. First, these two entities have overlapping risk factors, such as hypertension, diabetes, genetic risk factors, etc. [30]. Second, both share common systemic pathogenic pathways and mechanisms, such as abnormal production of pro-inflammatory cytokines, metabolic reprogramming in the tumor environment, clonal hematopoiesis of uncertain potential, crosstalk between stromal cells and the extracellular environment, etc. [8]. Finally, the direct infiltration of leukemic cells into the heart has been identified as another potential mechanism [11, 12, 3133]. Additionally, the investigation of cytokine networks in AML has gained increasing attention in recent years. Some pro-inflammatory mediators such as IL-1β, TNF-α and IL-6 tend to increase AML aggressiveness and can promote the survival and proliferation of AML cells [34]. And these can also cause heart damage [35, 36].
As previously stated, the bidirectional relationship between AML and cardiac injury status makes the diagnosis of cardiac injury potentially rather delayed, in part because symptoms of the former are attributable to the latter and vice versa [37]. Certain studies have suggested that the assessment of global longitudinal strain peak during systole using speckle tracking echocardiography may sensitively monitor early cardiotoxic alterations [38, 39]. However, this approach is limited to hospitals equipped with the necessary resources. Furthermore, cardiac biomarkers, including troponin, BNP, and cardiac enzymes [40] or other potential biomarkers such as glycogen phosphorylase BB [41], total antioxidant status, circulating microRNAs [42] and endothelial dysfunction [43] should be comprehensively assessed. The complexity and specialization involved make it easy for non-cardiac doctors to miss tests when focusing on the patient’s pre-treatment cardiac status and therefore slightly less clinically implementable.
We developed and validated a nomogram of cardiac injury risk before chemotherapy in patients with newly diagnosed AML. The model demonstrated good predictive performance. Nomograms combine several modeling algorithms to calculate continuous probabilities for specific outcomes. Among the many predictive tools currently available, nomograms have higher accuracy and better discriminative features [44]. We used LASSO regression and the multivariate logistic regression in the screening process, considering the collinearity and interaction of the screened variables. Besides, we performed a complete evaluation of the model for discrimination, clinical use and calibration, as well as an external validation of the model. Moreover, for further convenience of clinical use, we also established a risk score, and risk stratification was performed.
In clinical practice, AML patients are applied chemotherapy regimens very quickly after being definitively diagnosed, which often directly exacerbates the occurrence of adverse cardiac effects [19]. Generally, we attribute these cardiac injuries to the treatment, ignoring the heart’s injury state. Therefore, it is essential to promptly recognize the risk of cardiac injury in patients with incipient AML. In this study, we provide a prediction model that can help clinicians identify patients with incipient AML at high risk of cardiac injury and help them choose more appropriate treatment options promptly. Our nomogram model and risk score are routine clinical variables that are readily available to hematology clinicians, and as such, they can be easily applied in practice.
Our model includes four indicators: NT-proBNP, NPM1 mutations, WBC, and RBC. The level of NT-proBNP was closely related to the state of the heart [45]. AML with NPM1 mutations, although considered to have a good prognosis, results in low patient survival and high relapse rates due to its frequent coexistence with FLT3-ITD [46]. In addition, it often combines to exhibit higher leukocyte levels, percentage of BM blasts, cell invasiveness, and an incidence of extramedullary involvement [47]. AML patients with leukocytosis were prone to vascular stagnation and accumulation. In addition, the increase in metabolic byproducts in the state of blast cells will also increase the risk of cardiac injury [48]. The erythropoietic activity of AML patients was more active, especially those with BM involvement [49]. Furthermore, under certain conditions of excess oxidative stress, erythrocytes may switch the redox balance, promoting oxidative stress and being detrimental to resident and circulating adjacent cell types [50]. The latest research found that RBCs may be carriers of cytokines, which can induce significant damage to endothelial function, thereby causing vascular dysfunction [51]. This provides a new idea for cardiac injury in AML state.
Although we have developed a predictive model for the risk of cardiac injury before chemotherapy in patients with newly diagnosed AML patients, and tried to predict the risk with a minimum number of markers, this real-world study still has certain limitations. First, although hs-TNI is a more sensitive indicator of the degree of myocardial damage, it was combined with the diagnostic value of CK-MB in order to investigate this scientific question as it is not routinely examined in AML patients. Second, we used multiple imputations to replace missing values. This has the potential to lead to bias. Third, compared with other published disease prediction models, nomogram makes it difficult to deal with some nonlinear effects in the algorithm [5254]. In the future, we can consider collaborating with other professionals to optimizing the model algorithm and verifying the model effect with multiple calibrations to screen the optimal model. Fourth, our sample size is small and focuses on Asian people, which affects the extrapolation of the model to some extent. Although there is much room for improvement in this study, we believe that as cardiac oncology develops, this interdisciplinary model of collaboration will eventually bridge the gap between the two fields. It still needs multi-center, large-sample clinical studies or other related work to test it before it can be widely accepted or applied.

Conclusion

We developed and validated a personalized predictive nomogram for cardiac injury risk before chemotherapy in patients with newly diagnosed AML. The nomogram has good predictability and generalizability. It can provide individualized risk assessment for patients and help clinicians manage related decisions. In addition, substantial clinical and other related work is required before this nomogram can be widely accepted and used.

Acknowledgements

The authors thank the patients from Zhongnan Hospital of Wuhan University and the Xiangyang Central Hospital who participated in the study.

Declarations

The Studies involving human subjects were reviewed and approved by the Ethics Committee at Zhongnan Hospital of Wuhan University. The patients provided their written informed consent to participate in this study. All methods were carried out in accordance with declaration of Helsinki.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sturgeon KM, Deng L, Bluethmann SM, Zhou S, Trifiletti DM, Jiang C, et al. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur Heart J. 2019;40(48):3889–97.PubMedPubMedCentralCrossRef Sturgeon KM, Deng L, Bluethmann SM, Zhou S, Trifiletti DM, Jiang C, et al. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur Heart J. 2019;40(48):3889–97.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
3.
Zurück zum Zitat Potenza L, Borelli E, Bigi S, Giusti D, Longo G, Odejide O, et al. Early Palliative Care in Acute myeloid leukemia. Cancers (Basel). 2022;14(3):478.PubMedCrossRef Potenza L, Borelli E, Bigi S, Giusti D, Longo G, Odejide O, et al. Early Palliative Care in Acute myeloid leukemia. Cancers (Basel). 2022;14(3):478.PubMedCrossRef
4.
Zurück zum Zitat Ou Z, Yu D, Liang Y, He W, Li Y, Zhang M, et al. Analysis of the Global Burden of Disease study highlights the trends in death and disability-adjusted life years of leukemia from 1990 to 2017. Cancer Commun (Lond). 2020;40(11):598–610.PubMedCrossRef Ou Z, Yu D, Liang Y, He W, Li Y, Zhang M, et al. Analysis of the Global Burden of Disease study highlights the trends in death and disability-adjusted life years of leukemia from 1990 to 2017. Cancer Commun (Lond). 2020;40(11):598–610.PubMedCrossRef
5.
Zurück zum Zitat Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef
6.
Zurück zum Zitat Velusamy R, Nolan M, Murphy A, Thavendiranathan P, Marwick TH. Screening for coronary artery disease in Cancer survivors: JACC: CardioOncology State-of-the-art review. JACC: CardioOncology. 2023;5(1):22–38.PubMedPubMedCentral Velusamy R, Nolan M, Murphy A, Thavendiranathan P, Marwick TH. Screening for coronary artery disease in Cancer survivors: JACC: CardioOncology State-of-the-art review. JACC: CardioOncology. 2023;5(1):22–38.PubMedPubMedCentral
7.
8.
Zurück zum Zitat de Boer RA, Hulot JS, Tocchetti CG, Aboumsallem JP, Ameri P, Anker SD, et al. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur J Heart Fail. 2020;22(12):2272–89.PubMedCrossRef de Boer RA, Hulot JS, Tocchetti CG, Aboumsallem JP, Ameri P, Anker SD, et al. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur J Heart Fail. 2020;22(12):2272–89.PubMedCrossRef
9.
Zurück zum Zitat Javier BV, Yount WJ, Crosby DJ, Hall TC. Cardiac metastasis in lymphoma and leukemia. Dis Chest. 1967;52(4):481–4.PubMedCrossRef Javier BV, Yount WJ, Crosby DJ, Hall TC. Cardiac metastasis in lymphoma and leukemia. Dis Chest. 1967;52(4):481–4.PubMedCrossRef
10.
Zurück zum Zitat Kirshbaum JD, Preuss FS. Leukemia: a clinical and pathologic study of one hundred and twenty-three fatal cases in a series of 14,400 necropsies. Arch Intern Med. 1943;71(6):777–92.CrossRef Kirshbaum JD, Preuss FS. Leukemia: a clinical and pathologic study of one hundred and twenty-three fatal cases in a series of 14,400 necropsies. Arch Intern Med. 1943;71(6):777–92.CrossRef
11.
Zurück zum Zitat Roberts WC, Bodey GP, Wertlake PT. The heart in acute leukemia. A study of 420 autopsy cases. Am J Cardiol. 1968;21(3):388–412.PubMedCrossRef Roberts WC, Bodey GP, Wertlake PT. The heart in acute leukemia. A study of 420 autopsy cases. Am J Cardiol. 1968;21(3):388–412.PubMedCrossRef
12.
Zurück zum Zitat Sumners JE, Johnson WW, Ainger LE. Childhood leukemic heart disease. A study of 116 hearts of children dying of leukemia. Circulation. 1969;40(4):575–81.PubMedCrossRef Sumners JE, Johnson WW, Ainger LE. Childhood leukemic heart disease. A study of 116 hearts of children dying of leukemia. Circulation. 1969;40(4):575–81.PubMedCrossRef
13.
Zurück zum Zitat Luo Z, Cheng J, Wang Y. Cardiac infiltration as the First Manifestation of Acute Lymphoblastic Leukemia: a systematic review. Front Oncol. 2022;12:805981.PubMedPubMedCentralCrossRef Luo Z, Cheng J, Wang Y. Cardiac infiltration as the First Manifestation of Acute Lymphoblastic Leukemia: a systematic review. Front Oncol. 2022;12:805981.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Cohen Y, Amir G, Da’as N, Gillis S, Rund D, Polliack A. Acute myocardial infarction as the presenting symptom of acute myeloblastic leukemia with extreme hyperleukocytosis. Am J Hematol. 2002;71(1):47–9.PubMedCrossRef Cohen Y, Amir G, Da’as N, Gillis S, Rund D, Polliack A. Acute myocardial infarction as the presenting symptom of acute myeloblastic leukemia with extreme hyperleukocytosis. Am J Hematol. 2002;71(1):47–9.PubMedCrossRef
15.
Zurück zum Zitat da Silva GL, Valle S, Pinto FJ, Almeida AG. Leukaemic myocardial infiltration presenting as acute heart failure. Eur Heart J Cardiovasc Imaging. 2015;16(4):460.PubMedCrossRef da Silva GL, Valle S, Pinto FJ, Almeida AG. Leukaemic myocardial infiltration presenting as acute heart failure. Eur Heart J Cardiovasc Imaging. 2015;16(4):460.PubMedCrossRef
16.
Zurück zum Zitat Chang K, Kim DY, Lee KH, Huh J, Kang JW, Shin DY, et al. An isolated cardiac relapse after allogeneic hematopoietic stem cell transplantation for acute lymphoblastic leukemia. Korean J Intern Med. 2017;32(4):753–7.PubMedCrossRef Chang K, Kim DY, Lee KH, Huh J, Kang JW, Shin DY, et al. An isolated cardiac relapse after allogeneic hematopoietic stem cell transplantation for acute lymphoblastic leukemia. Korean J Intern Med. 2017;32(4):753–7.PubMedCrossRef
17.
Zurück zum Zitat Zhang W, Azibani F, Okello E, Kayima J, Sinabulya I, Leeta J, et al. Clinical characterization, cardiovascular risk factor profile and cardiac strain analysis in a Uganda cancer population: the SATRACD study. PLoS ONE. 2021;16(4):e0249717.PubMedPubMedCentralCrossRef Zhang W, Azibani F, Okello E, Kayima J, Sinabulya I, Leeta J, et al. Clinical characterization, cardiovascular risk factor profile and cardiac strain analysis in a Uganda cancer population: the SATRACD study. PLoS ONE. 2021;16(4):e0249717.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat De Lazzari M, Fedrigo M, Perazzolo Marra M, Calabrò F, Tarantini G, D’Amore EG, et al. Relapsing Leuk Infiltrating Heart Circ Heart Fail. 2015;8(6):1133–4.CrossRef De Lazzari M, Fedrigo M, Perazzolo Marra M, Calabrò F, Tarantini G, D’Amore EG, et al. Relapsing Leuk Infiltrating Heart Circ Heart Fail. 2015;8(6):1133–4.CrossRef
19.
Zurück zum Zitat Lancellotti P, Nkomo VT, Badano LP, Bergler-Klein J, Bogaert J, Davin L, et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26(9):1013–32.PubMedCrossRef Lancellotti P, Nkomo VT, Badano LP, Bergler-Klein J, Bogaert J, Davin L, et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26(9):1013–32.PubMedCrossRef
20.
Zurück zum Zitat Xiao W, Ma L, Shang Y, Yang F, Tan Y, Chen G, et al. Cardiac-related lesions in newly diagnosed patients with Acute Leukemia: A Chinese Population-based real-world study. Front Med (Lausanne). 2022;9:844350.PubMedCrossRef Xiao W, Ma L, Shang Y, Yang F, Tan Y, Chen G, et al. Cardiac-related lesions in newly diagnosed patients with Acute Leukemia: A Chinese Population-based real-world study. Front Med (Lausanne). 2022;9:844350.PubMedCrossRef
21.
Zurück zum Zitat Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405.PubMedCrossRef Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405.PubMedCrossRef
22.
Zurück zum Zitat Kim IC, Song JE, Lee HJ, Park JH, Hyun M, Lee JY, et al. The implication of Cardiac Injury score on In-hospital mortality of Coronavirus Disease 2019. J Korean Med Sci. 2020;35(39):e349.PubMedPubMedCentralCrossRef Kim IC, Song JE, Lee HJ, Park JH, Hyun M, Lee JY, et al. The implication of Cardiac Injury score on In-hospital mortality of Coronavirus Disease 2019. J Korean Med Sci. 2020;35(39):e349.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Shi S, Qin M, Shen B, Cai Y, Liu T, Yang F, et al. Association of Cardiac Injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5(7):802–10.PubMedPubMedCentralCrossRef Shi S, Qin M, Shen B, Cai Y, Liu T, Yang F, et al. Association of Cardiac Injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5(7):802–10.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat DeFilippis AP, Chapman AR, Mills NL, de Lemos JA, Arbab-Zadeh A, Newby LK, et al. Assessment and treatment of patients with type 2 myocardial infarction and Acute Nonischemic Myocardial Injury. Circulation. 2019;140(20):1661–78.PubMedPubMedCentralCrossRef DeFilippis AP, Chapman AR, Mills NL, de Lemos JA, Arbab-Zadeh A, Newby LK, et al. Assessment and treatment of patients with type 2 myocardial infarction and Acute Nonischemic Myocardial Injury. Circulation. 2019;140(20):1661–78.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Tyebally S, Chen D, Bhattacharyya S, Mughrabi A, Hussain Z, Manisty C, et al. Cardiac tumors: JACC CardioOncology State-of-the-art review. JACC CardioOncol. 2020;2(2):293–311.PubMedPubMedCentralCrossRef Tyebally S, Chen D, Bhattacharyya S, Mughrabi A, Hussain Z, Manisty C, et al. Cardiac tumors: JACC CardioOncology State-of-the-art review. JACC CardioOncol. 2020;2(2):293–311.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Banke A, Schou M, Videbaek L, Møller JE, Torp-Pedersen C, Gustafsson F, et al. Incidence of cancer in patients with chronic heart failure: a long-term follow-up study. Eur J Heart Fail. 2016;18(3):260–6.PubMedCrossRef Banke A, Schou M, Videbaek L, Møller JE, Torp-Pedersen C, Gustafsson F, et al. Incidence of cancer in patients with chronic heart failure: a long-term follow-up study. Eur J Heart Fail. 2016;18(3):260–6.PubMedCrossRef
27.
Zurück zum Zitat Assuncao BMBL, Handschumacher MD, Brunner AM, Yucel E, Bartko PE, Cheng K-H, et al. Acute Leukemia is Associated with Cardiac alterations before Chemotherapy. J Am Soc Echocardiogr. 2017;30(11):1111–8.PubMedCrossRef Assuncao BMBL, Handschumacher MD, Brunner AM, Yucel E, Bartko PE, Cheng K-H, et al. Acute Leukemia is Associated with Cardiac alterations before Chemotherapy. J Am Soc Echocardiogr. 2017;30(11):1111–8.PubMedCrossRef
28.
Zurück zum Zitat Pavlovic M, Dordevic A, Haßfeld S, Cuspidi C, Düngen HD, Tadic M. Left ventricular mechanics in patients with hematological malignancies before initiation of chemo- and radiotherapy. Int J Cardiovasc Imaging. 2021;37(3):881–7.PubMedCrossRef Pavlovic M, Dordevic A, Haßfeld S, Cuspidi C, Düngen HD, Tadic M. Left ventricular mechanics in patients with hematological malignancies before initiation of chemo- and radiotherapy. Int J Cardiovasc Imaging. 2021;37(3):881–7.PubMedCrossRef
29.
Zurück zum Zitat Pavo N, Raderer M, Hülsmann M, Neuhold S, Adlbrecht C, Strunk G, et al. Cardiovascular biomarkers in patients with cancer and their association with all-cause mortality. Heart. 2015;101(23):1874–80.PubMedCrossRef Pavo N, Raderer M, Hülsmann M, Neuhold S, Adlbrecht C, Strunk G, et al. Cardiovascular biomarkers in patients with cancer and their association with all-cause mortality. Heart. 2015;101(23):1874–80.PubMedCrossRef
31.
Zurück zum Zitat Barcos M, Lane W, Gomez GA, Han T, Freeman A, Preisler H, et al. An autopsy study of 1206 acute and chronic leukemias (1958 to 1982). Cancer. 1987;60(4):827–37.PubMedCrossRef Barcos M, Lane W, Gomez GA, Han T, Freeman A, Preisler H, et al. An autopsy study of 1206 acute and chronic leukemias (1958 to 1982). Cancer. 1987;60(4):827–37.PubMedCrossRef
33.
Zurück zum Zitat Leptidis J, Aloizos S, Chlorokostas P, Gourgiotis S. Fatal cardiac tamponade as the first manifestation of acute myeloid leukemia. Am J Emerg Med. 2014;32(10):1294e1–2.CrossRef Leptidis J, Aloizos S, Chlorokostas P, Gourgiotis S. Fatal cardiac tamponade as the first manifestation of acute myeloid leukemia. Am J Emerg Med. 2014;32(10):1294e1–2.CrossRef
34.
Zurück zum Zitat Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): a focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev. 2018;43:8–15.PubMedCrossRef Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): a focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev. 2018;43:8–15.PubMedCrossRef
36.
Zurück zum Zitat Nishida K, Otsu K. Inflammation and metabolic cardiomyopathy. Cardiovasc Res. 2017;113(4):389–98.PubMedCrossRef Nishida K, Otsu K. Inflammation and metabolic cardiomyopathy. Cardiovasc Res. 2017;113(4):389–98.PubMedCrossRef
37.
Zurück zum Zitat Ameri P, Canepa M, Anker MS, Belenkov Y, Bergler-Klein J, Cohen-Solal A, et al. Cancer diagnosis in patients with heart failure: epidemiology, clinical implications and gaps in knowledge. Eur J Heart Fail. 2018;20(5):879–87.PubMedCrossRef Ameri P, Canepa M, Anker MS, Belenkov Y, Bergler-Klein J, Cohen-Solal A, et al. Cancer diagnosis in patients with heart failure: epidemiology, clinical implications and gaps in knowledge. Eur J Heart Fail. 2018;20(5):879–87.PubMedCrossRef
38.
Zurück zum Zitat Negishi K, Negishi T, Haluska BA, Hare JL, Plana JC, Marwick TH. Use of speckle strain to assess left ventricular responses to cardiotoxic chemotherapy and cardioprotection. Eur Heart J Cardiovasc Imaging. 2014;15(3):324–31.PubMedCrossRef Negishi K, Negishi T, Haluska BA, Hare JL, Plana JC, Marwick TH. Use of speckle strain to assess left ventricular responses to cardiotoxic chemotherapy and cardioprotection. Eur Heart J Cardiovasc Imaging. 2014;15(3):324–31.PubMedCrossRef
39.
Zurück zum Zitat Esmaeilzadeh M, Urzua Fresno CM, Somerset E, Shalmon T, Amir E, Fan CS, et al. A combined Echocardiography Approach for the diagnosis of Cancer Therapy-Related Cardiac Dysfunction in Women with early-stage breast Cancer. JAMA Cardiol. 2022;7(3):330–40.PubMedPubMedCentralCrossRef Esmaeilzadeh M, Urzua Fresno CM, Somerset E, Shalmon T, Amir E, Fan CS, et al. A combined Echocardiography Approach for the diagnosis of Cancer Therapy-Related Cardiac Dysfunction in Women with early-stage breast Cancer. JAMA Cardiol. 2022;7(3):330–40.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol. 2020;31(2):171–90.PubMedCrossRef Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol. 2020;31(2):171–90.PubMedCrossRef
41.
Zurück zum Zitat Horacek JM, Tichy M, Pudil R, Jebavy L. Glycogen phosphorylase BB could be a new circulating biomarker for detection of anthracycline cardiotoxicity. Ann Oncol. 2008;19(9):1656–7.PubMedCrossRef Horacek JM, Tichy M, Pudil R, Jebavy L. Glycogen phosphorylase BB could be a new circulating biomarker for detection of anthracycline cardiotoxicity. Ann Oncol. 2008;19(9):1656–7.PubMedCrossRef
42.
Zurück zum Zitat Fichtlscherer S, Zeiher AM, Dimmeler S. Circulating microRNAs: biomarkers or mediators of cardiovascular diseases? Arterioscler Thromb Vasc Biol. 2011;31(11):2383–90.PubMedCrossRef Fichtlscherer S, Zeiher AM, Dimmeler S. Circulating microRNAs: biomarkers or mediators of cardiovascular diseases? Arterioscler Thromb Vasc Biol. 2011;31(11):2383–90.PubMedCrossRef
43.
Zurück zum Zitat Curigliano G, Cardinale D, Dent S, Criscitiello C, Aseyev O, Lenihan D, et al. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J Clin. 2016;66(4):309–25.PubMedCrossRef Curigliano G, Cardinale D, Dent S, Criscitiello C, Aseyev O, Lenihan D, et al. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J Clin. 2016;66(4):309–25.PubMedCrossRef
45.
Zurück zum Zitat Preoperative N-T. Pro–B-Type natriuretic peptide and Cardiovascular events after noncardiac surgery. Ann Intern Med. 2020;172(2):96–104.CrossRef Preoperative N-T. Pro–B-Type natriuretic peptide and Cardiovascular events after noncardiac surgery. Ann Intern Med. 2020;172(2):96–104.CrossRef
47.
Zurück zum Zitat Xian J, Shao H, Chen X, Zhang S, Quan J, Zou Q, et al. Nucleophosmin mutants promote adhesion, Migration and Invasion of Human Leukemia THP-1 cells through MMPs Up-regulation via Ras/ERK MAPK signaling. Int J Biol Sci. 2016;12(2):144–55.PubMedPubMedCentralCrossRef Xian J, Shao H, Chen X, Zhang S, Quan J, Zou Q, et al. Nucleophosmin mutants promote adhesion, Migration and Invasion of Human Leukemia THP-1 cells through MMPs Up-regulation via Ras/ERK MAPK signaling. Int J Biol Sci. 2016;12(2):144–55.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Shroff GS, Truong MT, Carter BW, Benveniste MF, Kanagal-Shamanna R, Rauch G, et al. Leukemic Involv Thorax Radiographics. 2019;39(1):44–61. Shroff GS, Truong MT, Carter BW, Benveniste MF, Kanagal-Shamanna R, Rauch G, et al. Leukemic Involv Thorax Radiographics. 2019;39(1):44–61.
49.
Zurück zum Zitat Choi JW, Pai SH. Erythropoietic activities in acute leukemia and in malignant lymphoma with or without bone marrow involvement. Ann Clin Lab Sci. 2003;33(4):407–10.PubMed Choi JW, Pai SH. Erythropoietic activities in acute leukemia and in malignant lymphoma with or without bone marrow involvement. Ann Clin Lab Sci. 2003;33(4):407–10.PubMed
50.
Zurück zum Zitat Mahdi A, Cortese-Krott MM, Kelm M, Li N, Pernow J. Novel perspectives on redox signaling in red blood cells and platelets in cardiovascular disease. Free Radic Biol Med. 2021;168:95–109.PubMedCrossRef Mahdi A, Cortese-Krott MM, Kelm M, Li N, Pernow J. Novel perspectives on redox signaling in red blood cells and platelets in cardiovascular disease. Free Radic Biol Med. 2021;168:95–109.PubMedCrossRef
51.
Zurück zum Zitat Mahdi A, Collado A, Tengbom J, Jiao T, Wodaje T, Johansson N, et al. Erythrocytes induce vascular dysfunction in COVID-19. JACC: Basic to Translational Science. 2022;7(3):193–204.PubMedPubMedCentral Mahdi A, Collado A, Tengbom J, Jiao T, Wodaje T, Johansson N, et al. Erythrocytes induce vascular dysfunction in COVID-19. JACC: Basic to Translational Science. 2022;7(3):193–204.PubMedPubMedCentral
52.
Zurück zum Zitat Wang Y, Lai X, Wang J, Xu Y, Zhang X, Zhu X, et al. TMBcat: a multi-endpoint p-value criterion on different discrepancy metrics for superiorly inferring tumor mutation burden thresholds. Front Immunol. 2022;13:995180.PubMedPubMedCentralCrossRef Wang Y, Lai X, Wang J, Xu Y, Zhang X, Zhu X, et al. TMBcat: a multi-endpoint p-value criterion on different discrepancy metrics for superiorly inferring tumor mutation burden thresholds. Front Immunol. 2022;13:995180.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Wang Y, Wang J, Fang W, Xiao X, Wang Q, Zhao J, et al. TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits. Front Immunol. 2023;14:1151755.PubMedPubMedCentralCrossRef Wang Y, Wang J, Fang W, Xiao X, Wang Q, Zhao J, et al. TMBserval: a statistical explainable learning model reveals weighted tumor mutation burden better categorizing therapeutic benefits. Front Immunol. 2023;14:1151755.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Liu R, Lai X, Wang J, Zhang X, Zhu X, Lai PBS, et al. A non-linear ensemble model-based surgical risk calculator for mixed data from multiple surgical fields. BMC Med Inform Decis Mak. 2021;21(Suppl 2):88.PubMedPubMedCentralCrossRef Liu R, Lai X, Wang J, Zhang X, Zhu X, Lai PBS, et al. A non-linear ensemble model-based surgical risk calculator for mixed data from multiple surgical fields. BMC Med Inform Decis Mak. 2021;21(Suppl 2):88.PubMedPubMedCentralCrossRef
Metadaten
Titel
Development of a risk assessment model for cardiac injury in patients newly diagnosed with acute myeloid leukemia based on a multicenter, real-world analysis in China
verfasst von
Linlu Ma
Qian Wang
Xinqi Li
Yufeng Shang
Nan Zhang
Jinxian Wu
Yuxing Liang
Guopeng Chen
Yuxin Tan
Xiaoyan Liu
Guolin Yuan
Fuling Zhou
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2024
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-11847-0

Weitere Artikel der Ausgabe 1/2024

BMC Cancer 1/2024 Zur Ausgabe

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.