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

Open Access 01.12.2024 | Research

A chest CT-based nomogram for predicting survival in acute myeloid leukemia

verfasst von: Xiaoping Yi, Huien Zhan, Jun Lyu, Juan Du, Min Dai, Min Zhao, Yu Zhang, Cheng Zhou, Xin Xu, Yi Fan, Lin Li, Baoxia Dong, Xinya Jiang, Zeyu Xiao, Jihao Zhou, Minyi Zhao, Jian Zhang, Yan Fu, Tingting Chen, Yang Xu, Jie Tian, Qifa Liu, Hui Zeng

Erschienen in: BMC Cancer | Ausgabe 1/2024

Abstract

Background

The identification of survival predictors is crucial for early intervention to improve outcome in acute myeloid leukemia (AML). This study aim to identify chest computed tomography (CT)-derived features to predict prognosis for acute myeloid leukemia (AML).

Methods

952 patients with pathologically-confirmed AML were retrospectively enrolled between 2010 and 2020. CT-derived features (including body composition and subcutaneous fat features), were obtained from the initial chest CT images and were used to build models to predict the prognosis. A CT-derived MSF nomogram was constructed using multivariate Cox regression incorporating CT-based features. The performance of the prediction models was assessed with discrimination, calibration, decision curves and improvements.

Results

Three CT-derived features, including myosarcopenia, spleen_CTV, and SF_CTV (MSF) were identified as the independent predictors for prognosis in AML (P < 0.01). A CT-MSF nomogram showed a performance with AUCs of 0.717, 0.794, 0.796 and 0.792 for predicting the 1-, 2-, 3-, and 5-year overall survival (OS) probabilities in the validation cohort, which were significantly higher than the ELN risk model. Moreover, a new MSN stratification system (MSF nomogram plus ELN risk model) could stratify patients into new high, intermediate and low risk group. Patients with high MSN risk may benefit from intensive treatment (P = 0.0011).

Conclusions

In summary, the chest CT-MSF nomogram, integrating myosarcopenia, spleen_CTV, and SF_CTV features, could be used to predict prognosis of AML.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-024-12188-8 .
Xiaoping Yi, Huien Zhan, Jun Lyu and Juan Du contributed equally to this work.
The original version of this article was revised: The equal contributions statement has been corrected to inlcude author Xiaoping Yi
A correction to this article is available online at https://​doi.​org/​10.​1186/​s12885-024-12323-5.

Publisher’s Note

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

Introduction

Acute myeloid leukemia (AML) is the common hematological malignancy comprising approximately 15% of leukemia cases. AML is driven by a series of genetic and epigenetic events, with induction chemotherapy and hematopoietic stem cell transplantation remaining the standard treatments [1, 2]. Despite a relatively high response rate, the prognosis of AML patients varies. The risk factors for AML with poor prognosis include clinical demographics, blood cell counts, serum oncological indicators, protein expression, and gene profiles [3, 4]. However, there is still an urgent need for new noninvasive, objective and time-efficient methods to identify those with poor prognosis and aid choice of therapy.
Chest computed tomography (CT) is the most widely used imaging method for routinely assessing lung conditions in all malignant diseases, including AML. However, it is challenging to predict prognostic status based on the traditional radiological evaluation of CT images in AML. CT based body composition evaluation, such as of myosarcopenia, refers to the computational extraction and analysis of imaging features from clinically acquired radiological images. Sarcopenia (loss of lean muscle mass) is an important reason for longer hospital stays and premature mortality in patients with nonmalignant disease [5]. Recently, the preliminary prognostic impact of sarcopenia has also been highlighted in AML [68] and acute lymphoblastic leukemia patients [9], demonstrating the potential for imaging-based prognostic prediction in leukemia. Notably, these studies were all conducted based on abdominal CT imaging. Unfortunately, AML patients do not routinely undergo abdominal CT scans unless they have coexisting abdominal diseases. Instead, a pretreatment chest CT is performed for AML patients prior to hospitalization in many institutions. Nevertheless, whether it is feasible to quantify CT-derived features based on pretreatment chest CT for the prognosis of AML patients is still unknown.

Methods

Patients

Through an evaluation of our institutional medical database from January 2010 to February 2020, data were retrospectively collected for de novo AML diagnosed in accordance with WHO criteria in ten participating hospitals. Details of the patient recruitment process and exclusion criteria are shown in Fig. 1. All 952 patients were randomly allocated into a training cohort (476 patients) and a validation cohort (476 patients). The general information of patients in these two cohorts is presented in Table 1.
Table 1
Clinical and body composition status data of 952 leukemia cases
Characteristics
All samples (n = 952)
Training (n = 476)
Validation (n = 476)
P Value
Sex (n, %)
   
0.696
Female
438 (46.01)
222 (46.6)
216 (45.4)
 
Male
514 (53.99)
254 (53.4)
260 (54.6)
 
Subtype (n, %)
   
0.547
M0
12 (1.26)
7 (1.50)
5 (1.10)
 
M1
56 (5.88)
23 (4.80)
33 (6.90)
 
M2
465 (48.84)
234 (49.20)
231 (48.50)
 
M3
36 (3.78)
21 (4.40)
15 (3.20)
 
M4
217 (22.79)
114 (23.90)
103 (21.60)
 
M5
161 (16.91)
74 (15.50)
87 (18.30)
 
M6
5 (0.53)
3 (0.60)
2 (0.40)
 
Treatment (n, %)
   
0.105
Standard-dose
762 (80.04)
371 (77.90)
391 (82.10)
 
Low-intensity
190 (19.96)
105 (22.10)
85 (17.90)
 
ELN 2022 Risk (n, %)
   
0.743
Adverse
398 (41.81)
202 (42.44)
196 (41.18)
 
Intermediate
385 (40.44)
194 (40.76)
191 (40.12)
 
Favorable
169 (17.75)
80 (16.8)
89 (18.70)
 
BMT
   
0.81
Non- Transplantation
755 (79.31)
379 (79.60)
376 (79.00)
 
Transplantation
197 (20.69)
97 (20.40)
100 (21.00)
 
Age (median, range)
45 (32, 55)
46 (33, 56)
44 (32, 55)
0.649
BMI
22 (19.98, 24.23)
22.03 (20.1, 24.30)
21.97 (19.82, 24.11)
0.306
SP (mmHg)
115.50 (107.00, 125.00)
115.0 (107.0, 124.75)
116.0 (107.0, 125.00)
0.812
DP (mmHg)
72 (65, 79)
71 (65, 78)
72 (66, 79)
0.587
WBC (×109/L)
16.39 (5.51, 46.29)
16.42 (4.93, 45.29)
16.32 (5.88, 48.68)
0.646
HGB(g/L)
74.00 (62.00, 92.00)
73.50 (62.0, 91.75)
74.00 (61.00,92.00)
0.904
PLT (×109/L)
43.00 (22.00, 80.00)
43.50 (21.00, 86.75)
42.00(23.00, 76.75)
0.924
Neu (×109/L)
2.10 (0.57, 7.79)
2.00 (0.56, 7.60)
2.1 ( 0.55, 8.72)
0.95
Lym (×109/L)
3.63 (1.40, 10.16)
3.53 (1.37, 10.48)
3.79 (1.40, 9.36)
0.928
Mono (×109/L)
4.11 (0.5, 19.4)
3.83 (0.50, 19.40)
4.34 (0.51, 19.55)
0.871
NLR (%)
0.64 (0.19, 1.76)
0.61 (0.19, 1.76)
0.65 (0.18, 1.80)
0.977
LMR (%)
0.91 (0.34, 4.76)
0.89 (0.34, 4.80)
0.93 (0.34, 4.55)
0.922
PLR (%)
10.93 (3.68, 38.97)
10.96 (3.63, 41.20)
10.85 (3.72, 35.79)
0.749
RDW (fl.)
60.40 (50.70, 88.20)
60.70 (50.80, 88.28)
59.95 (50.63, 88.15)
0.548
MPV (fl.)
10.30 (9.20, 11.90)
10.20 (9.04, 11.80)
10.50 (9.30, 12.00)
0.133
Albumin (g/L)
37.20 (33.30, 41.00)
36.85 (33.05, 40.60)
37.60 (3.40, 41.68)
0.121
Globulin (g/L)
28.50 (24.60, 33.20)
28.40 (24.65, 32.88)
28.50 (24.45, 33.28)
0.59
AGR
1.31 (1.07, 1.57)
1.30 (1.07, 1.56)
1.30 (1.07, 1.58)
0.521
HDL (mmol/L)
0.84 (0.71, 0.97)
0.85 (0.72, 0.98)
0.83 (0.68, 0.97)
0.074
LDL (mmol/L)
1.92 (1.50, 2.50)
1.93 (1.52, 2.52)
1.91 (1.48, 2.47)
0.527
TG (mmol/L)
1.46 (1.07, 2.06)
1.46 (1.08, 2.03)
1.43 (1.02, 2.12)
0.934
TC (mmol/L)
3.32 (2.75, 4.15)
3.34 (2.77, 4.14)
3.31 (2.70, 4.18)
0.882
BUN(mmol/L)
4.34 (3.30, 5.59)
4.38 (3.38, 5.60)
4.30 (3.25, 5.51)
0.277
Scr (umol/L)
71.00 (59.00,86.00)
73.00 (60.00, 87.00)
70.00 (58.00, 85.00)
0.085
BG
5.67 (5.10, 6.76)
5.67 (5.10, 6.75)
5.67 (5.10, 6.77)
0.937
LDH(U/L)
511.00 (296.80, 862.00)
519.10 (307.00, 872.00)
497.50 (285.53, 846.00)
0.395
BM(blast)
0.61 (0.39, 0.79)
0.61 (0.41, 0.77)
0.61 (0.37, 0.81)
0.989
Note—Unless otherwise indicated, data are numbers of patients, and data in parentheses are
percentages. # age is presented as median (minimum ∼ maximum)

Acquisition and retrieval procedure of CT images and radiological and body composition feature extraction

CT image acquisition, the image retrieval procedure, the algorithms for radiological and body composition feature extraction, and intra-observer (reader 1 twice) and inter-observer (reader 1 vs. reader 2) reproducibility evaluation were performed as previously described [10]. Briefly, CT-derived features, including skeletal muscle index (SMI), skeletal muscle radiation attenuation (SM-RA), liver CT value (liver_CTV) (Housfield units, HU), visceral or subcutaneous fat index (VFI or SFI), spleen CT value (spleen_CTV), and subcutaneous fat CT value (SF_CTV) evaluated by reader 1 were obtained from the initial chest CT images at the level of the fourth thoracic vertebra (T4) and were used to build models to predict the prognosis of AML.

Development of an individualized prediction model

The multivariable Cox regression method, which is suitable for the regression of medical data, was performed based on the candidate clinical and radiological predictors. Briefly, LASSO regression and clinical experience was used to screen for correlation factors [11]. Then, a prognostic model for predicting the 1-, 2-, 3-, and 5-year OS probabilities was developed by Cox regression. A predictive model was also constructed by using an ensemble machine learning method or deep learning algorithm. Details of machine learning and deep learning methods can be found in the supplementary methods.

Performance, validation and clinical use of the nomogram

The calibration of the nomogram was evaluated by calibration curves, and the diagnostic efficiency was quantified with Harrell’s C-index.
The performance of the nomogram was tested in the validation cohort with a series of indicators [12]. The concordance index (C-index) and the area under the time-dependent receiver operating characteristic (ROC) curve (AUC) were used to evaluate the differentiation ability of the new model. The performance of the new nomogram was further supplemented with two more indicators (net reclassification improvement [NRI] and integrated discrimination improvement [IDI]) to increase the accuracy and comprehensiveness of the comparisons. The consistency between the survival probabilities predicted using the nomogram and the actual result was evaluated by drawing calibration plots. Finally, decision-curve analysis (DCA) was performed to evaluate the clinical validity of the model. The total scores on the nomogram were divided into high-, and low- risk groups by X-Tile software. The MSN risk groups were identified by combining the CT-MSF nomogram risk and the ELN risk.

Statistical analysis

All of the statistical analysis were performed using IBM SPSS Statistics software (version 27.0, SPSS, Chicago, IL, USA), R software (version 4.0.3; http://​www.​Rproject.​org) and X-tile software (version 3.6.1; http://​tissuearray.​org/​). A bilateral probability value of p < 0.05 was considered indicative of statistical significance. *, P < 0.05. **, P < 0.01. ***, P < 0.001.

Data availability

The data and code of this study are available from the corresponding author upon request.

Results

Enrollment, characteristics and CT-derived features of the AML patients

A total of 952 AML patients at ten different hospitals (H1-10), were retrospectively included in this study. These patients were randomly assigned into the training cohort and validation cohort (Fig. 1A). Characteristics of these patients were presented in Fig. 1B; Table 1. No significant difference was observed in any parameters. Image pre-processing and analysis, prediction model construction process were performed as shown in Fig. 1C.

Establishment and evaluation of the CT-MSF model

To identify prognostic factors for AML, we performed LASSO regression analysis with clinicopathological characteristics and CT-derived features as variables. The LASSO regression analysis indicated that myosarcopenia, bone marrow transplantation (BMT), risk, BMT, spleen CT value (spleen_CTV), subcutaneous fat CT value (SF_CTV), age, red blood cell distribution width (RDW), high-density lipoprotein (HDL) and triglyceride (TG) were independent predictors for overall survival (OS) (Fig. 2A, supplementary Table 1). By multivariate Cox regression analysis, we established a new model integrating myosarcopenia, spleen_CTV and SF_CTV (CT-MSF). The CT-MSF nomogram showed a performance with a C index of 0.735 and 0.690 for the training and validation cohort, which were significantly higher than the C index of 0.557 and 0.557 for the ELN risk model (P < 0.05). Moreover, this predictive model achieved AUCs of 0.717, 0.794, 0.796 and 0.792 for predicting the 1-, 2-, 3- and 5-year OS probabilities in the validation cohort, respectively. When compared to the traditional ELN risk model (blue), the prediction performance of CT-MSF model (red) had significantly better predictive performance (Fig. 2B-E). Interestingly, the CT-MSF nomogram (cyan) added more benefit than the ELN risk model (purple) (Figure S1). The calibration curves of the nomogram for the probability of OS showed good agreement between prediction and observation in the validation cohort (Figure S2). The net reclassification improvement (NRI) and integrated discrimination improvement (IDI) also indicated that the CT-MSF model achieved satisfactory efficiency (supplementary Table 2). Machine learning and deep learning models were also constructed but did not show better performance than the traditional Cox method included above (data not shown). Collectively, we have identified independent predictors and developed a CT-MSF nomogram to predict prognosis of AML with high accuracy.
We next assessed whether the CT-MSF model could be used to predict overall survival by applying the model to the whole dataset. Based on the scores generated by our model, all cases were classified into the CT-MSF high-risk and CT-MSF low-risk groups (CT-MSF risk), by using the X-title method. As expected, patients within the high-risk group had shorter survival than that of the low-risk patients (p < 0.0001, log-rank test) (Figure S3A, supplementary Table 3). We also observed that the CT-MSF high-risk patients demonstrated higher cumulative hazard than the low-risk group (Figure S3B).

Stratification of AML patients by a combination of CT-MSF and ELN model

Next, we plotted the overall survival of this cohort with the combination of risk subgroups determined by the CT-MSF model and the ELN risk model to stratify these 952 patients. Interestingly, the survival curves fell mainly into 3 new groups. The CT-MSF low-risk and ELN low- and intermediate-risk groups formed a new MSN low-risk group with better survival. The CT-MSF low-risk/ELN high-risk group composed the new MSN intermediate-risk group, which had a medium survival time. Notably, all the CT-MSF high-risk group patients fell into the new MSF-ELN (MSN) high-risk group and had worse survival, regardless of their ELN risk (Fig. 3). Taken together, these results indicate that our CT-MSF model could further stratify patients when combined with the ELN risk model.
To clarify whether the effect of intensive and low-intensity treatment could be predicted by this new MSN risk score, we conducted prognostic analysis of patients who underwent different treatment choice. The results showed that the overall survival of patients with low and intermediate MSN risk was not affected by the choice of chemotherapy (Figure S4A and B). In contrast, among the MSN high-risk patients, the survival time of patients receiving intensive treatment was significantly longer than the low-intensity group (P <0.05, Figure S4C). Our data indicated that the new MSN stratification system combining the CT-MSF model and ELN risk model aid in choosing the therapy.

Discussion

Our findings provide preliminary data to support the inclusion of CT-derived body composition parameters, sarcopenia for example, as biomarkers of prognosis in AML. Recent studies have reported that sarcopenia had adverse implications, including the increased severity of the disease, longer hospital stays, more complications, earlier postoperative recurrence, and poor prognosis [68]. Our findings were generally in line with these prior studies. Moreover, our model included Spleen_CTV and SF_CTV in addition to sarcopenic features. Consistently, these parameters are known to be related to prognosis in AML. A lower Spleen_CTV reflects decreased spleen density due to multiple complex reasons, including systemic inflammation, abnormal fat deposition, tumor cell infiltration, and spleen tissue cell damage [13]. The increased SF_CTV may be caused by various reasons, with systemic inflammation as the most common contributor. Therefore, myosarcopenia, spleen-CTV and SF-CTV can be used to predict the overall treatment response, representing multiscale biological characteristics associated with metabolic status, gene status, and various pathological conditions, which could potentially predict prognosis in patients with AML.
Our model could predict prognosis better than ELN risk model alone, as evidenced by our AUC values, C-index, NRI, IDI as well as DCA in the validation cohorts. We speculate that the improved model performance in our study may be due to our research strategy. First, we enrolled 952 patients from ten hospitals and pretreatment chest CT images is commonly available in all hospitals in China. Moreover, the CT images used in our cohort were obtained from different scanners with the same non-enhanced scanning protocol, which largely considered imaging variability. Finally, in addition to Cox regression, we also tried the other algorithms including ensemble machine learning classification method and deep learning to select the best method. Our study showed that the Cox model with optimization algorithm has better expressiveness and is not inferior to all the rest models in performance.
In addition to CT-derived features, our MSF nomogram also includes clinical characteristics that yet to be included in prognostic prediction models of AML, including RDW, HDL and TG. RDW is traditionally considered as a marker of the differential diagnosis of anemia, and it has been reported as a prognostic factor in AML [14]. Increased RDW is related to oxidative stress, poor nutritional status and older age and may also suggest a proinflammatory state [15]. HDL and TG are both indicators of lipid metabolism and HDL levels have been implied to be associated with the pathogenesis of AML [16]. Dysregulated lipid metabolism has been reported to be involved in the pathogenesis of AML, and several key enzymes involved in lipid synthesis have been studied and explored as the targets to treat cancers, HMGCR for example [1618]. Consistently, these three traits serve as independent factors and important contributing factors in our nomogram.
However, there were limitations to this study. First, though we adopted a multicenter design in the present study, potential selection bias was unavoidable. In addition, the retrospective nature limits our exploration of the imaging and pathological correlation. Moreover, our sample size was still modest. In the future, a larger independent, prospective, multicenter study is needed for validation.

Conclusions

In summary, we presented an MSF nomogram utilizing chest CT images to predict prognosis in patients with AML. The combination of the MSF nomogram and ELN risk yielded an MSN stratification system that could stratify AML patients. Moreover, this MSN stratification system could be used to assist in the choice of therapy to achieve better outcomes. The information from the current study could be used to assist clinicians in selecting optimal therapies for personalized treatment of AML.

Acknowledgements

We thank Dr. Yangqiu Li (Jinan University) and Dr. Xiaoxia Hu (Shanghai Ruijin Hospital) for critical reading the manuscript. We also thank all participating hospital staff for their efforts in assisting data collection and analysis.

Declarations

This multicenter retrospective study was approved by the institutional review board of the First Affiliated Hospital of Jinan University and the institutional review boards of all other involved hospitals (IRB#: 201801002112). The requirement for written informed consent was waived.
All authors have read and approved the manuscript and agree with submission to BMC Cancer.

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.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Wang M, Lindberg J, Klevebring D, Nilsson C, Mer AS, Rantalainen M, Lehmann S, Gronberg H. Validation of risk stratification models in acute myeloid leukemia using sequencing-based molecular profiling. Leukemia. 2017;31(10):2029–36.CrossRefPubMedPubMedCentral Wang M, Lindberg J, Klevebring D, Nilsson C, Mer AS, Rantalainen M, Lehmann S, Gronberg H. Validation of risk stratification models in acute myeloid leukemia using sequencing-based molecular profiling. Leukemia. 2017;31(10):2029–36.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Pastore F, Dufour A, Benthaus T, Metzeler KH, Maharry KS, Schneider S, Ksienzyk B, Mellert G, Zellmeier E, Kakadia PM, et al. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2014;32(15):1586–94.CrossRefPubMedPubMedCentral Pastore F, Dufour A, Benthaus T, Metzeler KH, Maharry KS, Schneider S, Ksienzyk B, Mellert G, Zellmeier E, Kakadia PM, et al. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2014;32(15):1586–94.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Itzykson R, Fournier E, Berthon C, Rollig C, Braun T, Marceau-Renaut A, Pautas C, Nibourel O, Lemasle E, Micol JB, et al. Genetic identification of patients with AML older than 60 years achieving long-term survival with intensive chemotherapy. Blood. 2021;138(7):507–19.CrossRefPubMed Itzykson R, Fournier E, Berthon C, Rollig C, Braun T, Marceau-Renaut A, Pautas C, Nibourel O, Lemasle E, Micol JB, et al. Genetic identification of patients with AML older than 60 years achieving long-term survival with intensive chemotherapy. Blood. 2021;138(7):507–19.CrossRefPubMed
4.
Zurück zum Zitat Short NJ, Macaron W, Kadia T, Dinardo C, Issa GC, Daver N, Wang S, Jorgensen J, Nguyen D, Bidikian A, et al. Clinical outcomes and impact of therapeutic intervention in patients with acute myeloid leukemia who experience measurable residual disease (MRD) recurrence following MRD-negative remission. Am J Hematol. 2022;97(11):E408–11.CrossRefPubMed Short NJ, Macaron W, Kadia T, Dinardo C, Issa GC, Daver N, Wang S, Jorgensen J, Nguyen D, Bidikian A, et al. Clinical outcomes and impact of therapeutic intervention in patients with acute myeloid leukemia who experience measurable residual disease (MRD) recurrence following MRD-negative remission. Am J Hematol. 2022;97(11):E408–11.CrossRefPubMed
5.
Zurück zum Zitat Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB, Baracos VE. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31(12):1539–47.CrossRefPubMed Martin L, Birdsell L, Macdonald N, Reiman T, Clandinin MT, McCargar LJ, Murphy R, Ghosh S, Sawyer MB, Baracos VE. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol. 2013;31(12):1539–47.CrossRefPubMed
6.
Zurück zum Zitat Nakamura N, Ninomiya S, Matsumoto T, Nakamura H, Kitagawa J, Shiraki M, Hara T, Shimizu M, Tsurumi H. Prognostic impact of skeletal muscle assessed by computed tomography in patients with acute myeloid leukemia. Ann Hematol. 2019;98(2):351–9.CrossRefPubMed Nakamura N, Ninomiya S, Matsumoto T, Nakamura H, Kitagawa J, Shiraki M, Hara T, Shimizu M, Tsurumi H. Prognostic impact of skeletal muscle assessed by computed tomography in patients with acute myeloid leukemia. Ann Hematol. 2019;98(2):351–9.CrossRefPubMed
7.
Zurück zum Zitat Jung J, Lee E, Shim H, Park JH, Eom HS, Lee H. Prediction of clinical outcomes through assessment of Sarcopenia and Adipopenia using computed tomography in adult patients with acute myeloid leukemia. Int J Hematol. 2021;114(1):44–52.CrossRefPubMed Jung J, Lee E, Shim H, Park JH, Eom HS, Lee H. Prediction of clinical outcomes through assessment of Sarcopenia and Adipopenia using computed tomography in adult patients with acute myeloid leukemia. Int J Hematol. 2021;114(1):44–52.CrossRefPubMed
8.
Zurück zum Zitat Sun Q, Cui J, Liu W, Li J, Hong M, Qian S. The Prognostic Value of Sarcopenia in Acute myeloid leukemia patients and the development and validation of a Novel Nomogram for Predicting Survival. Front Oncol. 2022;12:828939.CrossRefPubMedPubMedCentral Sun Q, Cui J, Liu W, Li J, Hong M, Qian S. The Prognostic Value of Sarcopenia in Acute myeloid leukemia patients and the development and validation of a Novel Nomogram for Predicting Survival. Front Oncol. 2022;12:828939.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Suzuki D, Kobayashi R, Sano H, Hori D, Kobayashi K. Sarcopenia after induction therapy in childhood acute lymphoblastic leukemia: its clinical significance. Int J Hematol. 2018;107(4):486–9.CrossRefPubMed Suzuki D, Kobayashi R, Sano H, Hori D, Kobayashi K. Sarcopenia after induction therapy in childhood acute lymphoblastic leukemia: its clinical significance. Int J Hematol. 2018;107(4):486–9.CrossRefPubMed
10.
Zurück zum Zitat Yi X, Chen Q, Yang J, Jiang D, Zhu L, Liu H, Pang P, Zeng F, Chen C, Gong G, et al. CT-Based Sarcopenic Nomogram for Predicting Progressive Disease in Advanced Non-small-cell Lung Cancer. Front Oncol. 2021;11:643941.CrossRefPubMedPubMedCentral Yi X, Chen Q, Yang J, Jiang D, Zhu L, Liu H, Pang P, Zeng F, Chen C, Gong G, et al. CT-Based Sarcopenic Nomogram for Predicting Progressive Disease in Advanced Non-small-cell Lung Cancer. Front Oncol. 2021;11:643941.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Xu F, Zhang L, Wang Z, Han D, Li C, Zheng S, Yin H, Lyu J. A New Scoring System for Predicting In-hospital death in patients having liver cirrhosis with esophageal varices. Front Med (Lausanne). 2021;8:678646.CrossRefPubMed Xu F, Zhang L, Wang Z, Han D, Li C, Zheng S, Yin H, Lyu J. A New Scoring System for Predicting In-hospital death in patients having liver cirrhosis with esophageal varices. Front Med (Lausanne). 2021;8:678646.CrossRefPubMed
12.
Zurück zum Zitat Li C, Yang J, Xu F, Han D, Zheng S, Kaaya RE, Wang S, Lyu J. A prognostic nomogram for the cancer-specific survival of patients with upper-tract urothelial carcinoma based on the Surveillance, Epidemiology, and end results database. BMC Cancer. 2020;20(1):534.CrossRefPubMedPubMedCentral Li C, Yang J, Xu F, Han D, Zheng S, Kaaya RE, Wang S, Lyu J. A prognostic nomogram for the cancer-specific survival of patients with upper-tract urothelial carcinoma based on the Surveillance, Epidemiology, and end results database. BMC Cancer. 2020;20(1):534.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Seban RD, Rouzier R, Latouche A, Deleval N, Guinebretiere JM, Buvat I, Bidard FC, Champion L. Total metabolic tumor volume and spleen metabolism on baseline [18F]-FDG PET/CT as independent prognostic biomarkers of recurrence in resected breast cancer. Eur J Nucl Med Mol Imaging. 2021;48(11):3560–70.CrossRefPubMed Seban RD, Rouzier R, Latouche A, Deleval N, Guinebretiere JM, Buvat I, Bidard FC, Champion L. Total metabolic tumor volume and spleen metabolism on baseline [18F]-FDG PET/CT as independent prognostic biomarkers of recurrence in resected breast cancer. Eur J Nucl Med Mol Imaging. 2021;48(11):3560–70.CrossRefPubMed
14.
Zurück zum Zitat Vucinic V, Ruhnke L, Sockel K, Rohnert MA, Backhaus D, Brauer D, Franke GN, Niederwieser D, Bornhauser M, Rollig C, et al. The diagnostic red blood cell distribution width as a prognostic factor in acute myeloid leukemia. Blood Adv. 2021;5(24):5584–7.CrossRefPubMedPubMedCentral Vucinic V, Ruhnke L, Sockel K, Rohnert MA, Backhaus D, Brauer D, Franke GN, Niederwieser D, Bornhauser M, Rollig C, et al. The diagnostic red blood cell distribution width as a prognostic factor in acute myeloid leukemia. Blood Adv. 2021;5(24):5584–7.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Salvagno GL, Sanchis-Gomar F, Picanza A, Lippi G. Red blood cell distribution width: a simple parameter with multiple clinical applications. Crit Rev Clin Lab Sci. 2015;52(2):86–105.CrossRefPubMed Salvagno GL, Sanchis-Gomar F, Picanza A, Lippi G. Red blood cell distribution width: a simple parameter with multiple clinical applications. Crit Rev Clin Lab Sci. 2015;52(2):86–105.CrossRefPubMed
16.
Zurück zum Zitat Pabst T, Kortz L, Fiedler GM, Ceglarek U, Idle JR, Beyoglu D. The plasma lipidome in acute myeloid leukemia at diagnosis in relation to clinical disease features. BBA Clin. 2017;7:105–14.CrossRefPubMedPubMedCentral Pabst T, Kortz L, Fiedler GM, Ceglarek U, Idle JR, Beyoglu D. The plasma lipidome in acute myeloid leukemia at diagnosis in relation to clinical disease features. BBA Clin. 2017;7:105–14.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Pandyra AA, Mullen PJ, Goard CA, Ericson E, Sharma P, Kalkat M, Yu R, Pong JT, Brown KR, Hart T, et al. Genome-wide RNAi analysis reveals that simultaneous inhibition of specific mevalonate pathway genes potentiates tumor cell death. Oncotarget. 2015;6(29):26909–21.CrossRefPubMedPubMedCentral Pandyra AA, Mullen PJ, Goard CA, Ericson E, Sharma P, Kalkat M, Yu R, Pong JT, Brown KR, Hart T, et al. Genome-wide RNAi analysis reveals that simultaneous inhibition of specific mevalonate pathway genes potentiates tumor cell death. Oncotarget. 2015;6(29):26909–21.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat van Besien H, Sassano A, Altman JK, Platanias LC. Antileukemic properties of 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors. Leuk Lymphoma. 2013;54(12):2601–5.CrossRefPubMed van Besien H, Sassano A, Altman JK, Platanias LC. Antileukemic properties of 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors. Leuk Lymphoma. 2013;54(12):2601–5.CrossRefPubMed
Metadaten
Titel
A chest CT-based nomogram for predicting survival in acute myeloid leukemia
verfasst von
Xiaoping Yi
Huien Zhan
Jun Lyu
Juan Du
Min Dai
Min Zhao
Yu Zhang
Cheng Zhou
Xin Xu
Yi Fan
Lin Li
Baoxia Dong
Xinya Jiang
Zeyu Xiao
Jihao Zhou
Minyi Zhao
Jian Zhang
Yan Fu
Tingting Chen
Yang Xu
Jie Tian
Qifa Liu
Hui Zeng
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2024
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-12188-8

Weitere Artikel der Ausgabe 1/2024

BMC Cancer 1/2024 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

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