1
|
Xue N, Wen X, Wang Q, Shen Y, Qu Y, Xu Q, Chen S, Chen J. Establishing and validating models integrated with hematological biomarkers and clinical characteristics for the prognosis of non-esophageal squamous cell carcinoma patients. Ann Med 2025; 57:2483985. [PMID: 40152751 PMCID: PMC11956093 DOI: 10.1080/07853890.2025.2483985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This study aimed to construct a novel model and validate its predictive power in non-esophageal squamous cell carcinoma (NESCC) patients. METHODS This retrospective study included 151 patients between October 2006 and September 2016. The LASSO Cox and Random Survival Forest (RSF) models were developed with the help of hematological biomarkers and clinical characteristics. The concordance index (C-index) was used to assess the prognostic power of the LASSO Cox model, RSF model, and TNM staging. Based on the risk scores of the LASSO Cox and RSF models, we divided patients into low-risk and high-risk subgroups. RESULTS We constructed two models in NESCC patients according to LASSO Cox regression and RSF models. The RSF model reached a C-index of 0.841 (95% CI: 0.792-0.889) in the primary cohort and 0.880 (95% CI: 0.830-0.930) in the validation cohort, which was higher than the C-index of the LASSO Cox model 0.656 (95% CI: 0.580-0.732) and 0.632 (95% CI: 0.542-0.720) in the two cohorts. The integrated C/D area under the ROC curve (AUC) values for the LASSO Cox and RSF models were 0.701 and 0.861, respectively. In both two models, Kaplan-Meier survival analysis and the estimated restricted mean survival time (RMST) values indicated that the low-risk subgroup had a better prognostic outcome than the high-risk subgroup (p < 0.05). CONCLUSIONS The RSF model has better prediction power than the LASSO Cox and the TNM staging models. It has a guiding value for the choice of individualized treatment in patients with NESCC.
Collapse
Affiliation(s)
- Ning Xue
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Xiaoyan Wen
- Central Sterilization Supply Department, The Guanghua Stomatological College of Sun Yat-sen University, Hospital of Stomatology, SunYat-sen University, Guangzhou, P. R. China
| | - Qian Wang
- Department of radiation oncology, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Yong Shen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Yuanye Qu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Qingxia Xu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Shulin Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Research Center for Translational Medicine, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jing Chen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| |
Collapse
|
2
|
Lee SW, Choi MH, Lee YJ, Choi M. Comparison of Muscle and Fat Measurements Between True Noncontrast and Virtual Noncontrast Images From Three Phases of Dual-Energy Dynamic Liver CT. Acad Radiol 2025:S1076-6332(25)00413-1. [PMID: 40393827 DOI: 10.1016/j.acra.2025.04.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/22/2025]
Abstract
RATIONALE AND OBJECTIVES To determine whether virtual noncontrast (VNC) images derived from three-phase dynamic dual-energy CT (DECT) imaging can reliably substitute true noncontrast (TNC) images in assessing muscle and fat with automatic segmentation software. MATERIALS AND METHODS The data from 476 dynamic liver DECT examinations performed between April 2019 and December 2020 were retrospectively analyzed. VNC images were generated from arterial (VNCa), portal-venous (VNCp), and delayed (VNCd) phase images. Automated software measured muscle, visceral fat (VF), and subcutaneous fat (SF) areas. Sarcopenia was defined using muscle-related indices. Differences in muscle and fat measurements, as well as sarcopenia prevalence, between TNC and VNC images were assessed using paired t tests and McNemar tests, respectively. RESULTS The average age of the 476 patients (307 men) was 58.4±12.5years. Muscle density and area differed significantly between the TNC and VNC images; TNC images showed higher mean muscle density and smaller skeletal muscle area (SMA) than all VNC images (P<0.001). VF and SF attenuations were significantly lower on TNC images than on all VNC images (P<0.001). The proportions of sarcopenic patients did not differ significantly between TNC and VNCp or VNCd, regardless of the muscle index used. CONCLUSION Despite significant differences in muscle and fat attenuations between TNC and VNC images, VNCp and VNCd may be acceptable alternatives for measuring muscle and fat areas. However, TNC and VNC images should not be used interchangeably for assessing tissue attenuation or specific muscle components such as LAMA or NAMA.
Collapse
Affiliation(s)
- Sheen-Woo Lee
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea (S.W.L., M.H.C., Y.J.L.)
| | - Moon Hyung Choi
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea (S.W.L., M.H.C., Y.J.L.).
| | - Young Joon Lee
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea (S.W.L., M.H.C., Y.J.L.)
| | - Maria Choi
- Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (M.C.)
| |
Collapse
|
3
|
Yi J, Marcinkiewicz AM, Shanbhag A, Miller RJH, Geers J, Zhang W, Killekar A, Manral N, Lemley M, Buchwald M, Kwiecinski J, Zhou J, Kavanagh PB, Liang JX, Builoff V, Ruddy TD, Einstein AJ, Feher A, Miller EJ, Sinusas AJ, Berman DS, Dey D, Slomka PJ. AI-based volumetric six-tissue body composition quantification from CT cardiac attenuation scans for mortality prediction: a multicentre study. Lancet Digit Health 2025:100862. [PMID: 40382274 DOI: 10.1016/j.landig.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/09/2024] [Accepted: 02/06/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND CT attenuation correction (CTAC) scans are routinely obtained during cardiac perfusion imaging, but currently only used for attenuation correction and visual calcium estimation. We aimed to develop a novel artificial intelligence (AI)-based approach to obtain volumetric measurements of chest body composition from CTAC scans and to evaluate these measures for all-cause mortality risk stratification. METHODS We applied AI-based segmentation and image-processing techniques on CTAC scans from a large international image-based registry at four sites (Yale University, University of Calgary, Columbia University, and University of Ottawa), to define the chest rib cage and multiple tissues. Volumetric measures of bone, skeletal muscle, subcutaneous adipose tissue, intramuscular adipose tissue (IMAT), visceral adipose tissue (VAT), and epicardial adipose tissue (EAT) were quantified between automatically identified T5 and T11 vertebrae. The independent prognostic value of volumetric attenuation and indexed volumes were evaluated for predicting all-cause mortality, adjusting for established risk factors and 18 other body composition measures via Cox regression models and Kaplan-Meier curves. FINDINGS The end-to-end processing time was less than 2 min per scan with no user interaction. Between 2009 and 2021, we included 11 305 participants from four sites participating in the REFINE SPECT registry, who underwent single-photon emission computed tomography cardiac scans. After excluding patients who had incomplete T5-T11 scan coverage, missing clinical data, or who had been used for EAT model training, the final study group comprised 9918 patients. 5451 (55%) of 9918 participants were male and 4467 (45%) of 9918 participants were female. Median follow-up time was 2·48 years (IQR 1·46-3·65), during which 610 (6%) patients died. High VAT, EAT, and IMAT attenuation were associated with an increased all-cause mortality risk (adjusted hazard ratio 2·39, 95% CI 1·92-2·96; p<0·0001, 1·55, 1·26-1·90; p<0·0001, and 1·30, 1·06-1·60; p=0·012, respectively). Patients with high bone attenuation were at reduced risk of death (0·77, 0·62-0·95; p=0·016). Likewise, high skeletal muscle volume index was associated with a reduced risk of death (0·56, 0·44-0·71; p<0·0001). INTERPRETATION CTAC scans obtained routinely during cardiac perfusion imaging contain important volumetric body composition biomarkers that can be automatically measured and offer important additional prognostic value. FUNDING The National Heart, Lung, and Blood Institute, National Institutes of Health.
Collapse
Affiliation(s)
- Jirong Yi
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna M Marcinkiewicz
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Center of Radiological Diagnostics, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Aakash Shanbhag
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Signal and Image Processing Institute, Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, USA
| | - Robert J H Miller
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - Jolien Geers
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Cardiology, Centrum voor Hart-en Vaatziekten (CHVZ), Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wenhao Zhang
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aditya Killekar
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nipun Manral
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mark Lemley
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mikolaj Buchwald
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | - Jianhang Zhou
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul B Kavanagh
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joanna X Liang
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Valerie Builoff
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Terrence D Ruddy
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Andrew J Einstein
- Division of Cardiology, Department of Medicine, and Department of Radiology, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
| | - Attila Feher
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel S Berman
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Damini Dey
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Piotr J Slomka
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences, and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Luo Y, Liu J, Huang J, Ma L, Li Z. The Ratio of Visceral to Subcutaneous Adipose Tissue Is Associated With Postoperative Anastomotic Leakage in Patients With Rectal Cancer With Gender Differences in Opposite Direction. Cancer Med 2025; 14:e70933. [PMID: 40346009 PMCID: PMC12062873 DOI: 10.1002/cam4.70933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Anastomotic leakage (AL) is a severe postoperative complication in colorectal cancer and exerts negative impacts on patients' outcomes. Studies have found that body composition measured by CT images was associated with increased overall postoperative complications in colorectal cancer; however, few focused on postoperative AL in rectal cancer. This study aimed to explore the association between body composition parameters measured by CT images and postoperative AL in patients with rectal cancer, with an emphasis on subgroup analysis by gender. METHODS From February 2014 to January 2020, a total of 444 patients with rectal adenocarcinoma who underwent radical proctectomy were included. Out of all patients, 21 developed AL after surgery. Body composition parameters, including the areas, mean CT values, height-normalized indices of subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), intramuscular adipose tissue (IMAT) and skeletal muscle (SM) were derived from preoperative contrast-enhanced arterial phase CT images at the third lumbar level. The ratio of visceral to subcutaneous adipose tissue (VSR) was calculated. Clinical and body composition parameters were compared between the AL group and the non-AL group in all patients and separately in different genders. RESULTS Body composition parameters were not significantly different in the AL group and the non-AL group in all patients. However, most body composition parameters were significantly different between male and female patients. After separately analyzing by gender, VSR was significantly associated with postoperative AL in male and female. After multivariate regression, VSR remained an independent predictor for AL (OR: 0.1, p = 0.041 for male and OR: 39.1, p = 0.045 for female). CONCLUSION The VSR measured by CT images is an independent predictor for postoperative AL in patients with rectal cancer; however, it shows gender differences in opposite directions, serving as a protective factor in males, whereas as a risk factor in females.
Collapse
Affiliation(s)
- Yan Luo
- Department of RadiologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jian Liu
- Department of RadiologyWuhan Hospital of Traditional Chinese MedicineWuhanChina
| | - Jiong Huang
- Department of RadiologyThe Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan UniversityWuhanChina
| | - Liya Ma
- Department of RadiologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhen Li
- Department of RadiologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
5
|
Lortie J, Ufearo D, Hetzel S, Pickhardt PJ, Szczykutowicz TP, Kuchnia AJ. Validating a Practical Correction for Intravenous Contrast on Computed Tomography-Based Muscle Density. J Comput Assist Tomogr 2025; 49:480-485. [PMID: 39761492 PMCID: PMC12071502 DOI: 10.1097/rct.0000000000001682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/06/2024] [Indexed: 05/15/2025]
Abstract
OBJECTIVE Computed tomography (CT) measured muscle density is prognostic of health outcomes. However, the use of intravenous contrast obscures prognoses by artificially increasing CT muscle density. We previously established a correction to equalize contrast and noncontrast muscle density measurements. While this correction was validated internally, the objective of this study was to obtain external validation using different patient cohorts, muscle regions, and CT series. METHODS CT images from 109 patients with kidney tumors who received abdominal CT scans with a multiphase intravenous contrast protocol were analyzed. Paraspinal muscle density measurements taken during noncontrast, venous phase, and delayed phase contrast scans were collected. An a priori correction of -7.5 Hounsfield units (HU) was applied to muscle measurements. Equivalence testing was utilized to determine statistical similarity. RESULTS In the sample of 109 patients (mean age: 63 years [SD: 14.3]; 41.3% female), densities in smaller regions of interest within the paraspinal muscles and the entire paraspinal muscle density (PS) in venous and delayed phase contrast scans were higher than in noncontrast. Equivalence testing showed that average corrected contrast and noncontrast muscle densities were within 3 HU for both muscle measures for the total patient sample, and for a majority of male and female subsamples. The correction is suitable for regions of interests of venous contrast (90% CI: -1.90, -0.69 HU) and delayed contrast scans (90% CI: 0.075, 1.29 HU) and within the PS measures of venous contrast (90% CI: -2.04, -0.94 HU) and delayed contrast scans (90% CI: -0.11, 0.89 HU). CONCLUSIONS The previously established correction for contrast of -7.5 HU was applied in a new patient population, axial muscle region, muscle measurement size, and expanded on previously studied contrast phases. The correction produced contrast-corrected muscle densities that were statistically equivalent to noncontrast muscle densities. The simplicity of the correction gives clinicians a tool that seamlessly integrates into practice or research to improve harmonization of data between contrast and noncontrast scans.
Collapse
Affiliation(s)
- Jevin Lortie
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Deborah Ufearo
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Scott Hetzel
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI
| | | | - Timothy P. Szczykutowicz
- Department of Radiology, University of Wisconsin-Madison, Madison, WI
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI
| | - Adam J. Kuchnia
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
6
|
Surov A, Thormann M, Wienke A, Ricke J, Seidensticker M. Different cutoff values of the skeletal muscle mass and myosteatosis result in different clinical impact on overall survival in oncology. A subanalysis of a clinical trial. J Cancer Res Clin Oncol 2025; 151:141. [PMID: 40240716 PMCID: PMC12003470 DOI: 10.1007/s00432-025-06190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Body composition analysis, particularly the assessment of sarcopenia and myosteatosis, has emerged as a potential prognostic tool in oncology. However, the clinical implication of body composition parameters remains inconsistent, largely due to the variability in cutoff values used across studies. This study examines the influence on prevalence and prognostic influence of different cutoff values for sarcopenia and myosteatosis in patients in a standardized cohort from a large clinical trial (SORAMIC). METHODS This study included 179 patients with unresectable liver cancer from the palliative arm of the SORAMIC trial. Skeletal muscle index (SMI) was calculated by measuring the cross-sectional area of skeletal muscle at the third lumbar vertebra (L3) on baseline CT scans. We then applied 14 published cutoff definitions for sarcopenia (SMI) and 7 for myosteatosis (muscle attenuation) to determine their prevalence in this cohort. Cox regression models were used to analyze the relationship between sarcopenia, myosteatosis, and OS. RESULTS The prevalence of sarcopenia ranged from 8.9% (Van der Werf et al.) to 69.8% (Lanic et al.). Overall, 3 of the 14 cutoffs [Van Vledder et al. (HR = 1.53, p = 0.03), Coelen et al. (HR = 1.46, p = 0.03), and Derstine et al. (HR = 1.47, p = 0.04)] showed a relevant association with OS. Other cut off values were not associated with OS. The prevalence of myosteatosis varied between 10.1% (Nachit et al.) and 53.1% (Zhang et al.). One of the 7 cutoffs (Chu et al.) demonstrated a relevant association with OS (HR = 1.53, p = 0.03). CONCLUSION The large variability in prevalence and prognostic impact observed across different cutoff definitions underscores the urgent need for standardized, cancer-specific cutoff values for SMI and muscle attenuation. Establishing uniform criteria will enhance the reliability and clinical applicability of body composition metrics as prognostic tools in oncology. Further research should focus on refining these cutoffs and validating them across diverse cancer populations.
Collapse
Affiliation(s)
- Alexey Surov
- Department of Radiology, Neuroradiology and Nuclear Medicine, Johannes Wesling University Hospital, Ruhr University Bochum, Bochum, Germany
| | - Maximilian Thormann
- Department of Nuclear Medicine, Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- Department of Radiology, University Hospital Magdeburg, Magdeburg, Germany.
| | - Andreas Wienke
- Institute of Medical Epidemiology, Biometry and Informatics, University of Halle, Halle, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Tagliafico AS, Benenati S, Porto I, Martinoli C, Ameri P. Opportunistic prognostication by computerized tomography (CT) in the emergency department: analysis on 1920 patients and creation of a simple and fast scoring system. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-01986-0. [PMID: 40167933 DOI: 10.1007/s11547-025-01986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE To use simple CT measurements of musculoskeletal and cardiovascular systems to create a CT-based score to predict mortality in patients admitted to the Emergency Department (ED). METHODS The study received IRB approval. Non-contrast abdominal CT of > 18 year old patients admitted to the ER between January 2019 and January 2020 were evaluated by a team of twelve radiologists to calculate: (1) diameter of the infrarenal aorta in millimeter; (2) cross sectional area and composition (Hounsfield units) of the psoas muscle at the third lumbar vertebra (LV); (3) bone density, as quantified at the first lumbar vertebra (LV); (4) presence or absence of dilated abdominal aorta. Thirty-day all-cause mortality (ACM) was determined through hospital and electronic records. RESULTS N = 1920 unique patients were evaluated. The mean age was 65 ± 19 years and 46% were female. Death occurred in 7.9% of patients by 30 days from admission. The derivation dataset comprised 1462 patients. At multivariable analysis, age (OR 1.02, 95% CI: 1.007-1.04, p = 0.005), psoas cross sectional area (OR 0.99, 95% CI: 0.997-0.999, p < 0.001) and density (OR 0.96, 95% CI: 0.95-0.98, p < 0.001), and dilated infrarenal aorta (OR 1.85, 95% CI: 1-3.28, p = 0.04) were predictors of the outcome. We accordingly derived a 4-item risk score. In the derivation dataset, the score yielded moderate-high discrimination, with an AUC of 0.73 and excellent diagnostic agreement. In the validation dataset (N = 458), discrimination was high (AUC = 0.83). CONCLUSION Simple measurements gathered during a standard CT may allow determining the risk of mortality in the heterogeneous patient population admitted to the ED in a cost- and time-effective manner.
Collapse
Affiliation(s)
- Alberto Stefano Tagliafico
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy.
- Department of Radiology, IRCCS Policlinico San Martino Hospital, Via Pastore 1, 16132, Genova, Italy.
| | - Stefano Benenati
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Italo Porto
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Carlo Martinoli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Department of Radiology, IRCCS Policlinico San Martino Hospital, Via Pastore 1, 16132, Genova, Italy
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
8
|
Cai W, Zhu Y, Li D, Hu M, Teng Z, Cong R, Chen Z, Sun X, Ma X, Zhao X. Baseline Body Composition and 3D-Extracellular Volume Fraction for Predicting Pancreatic Fistula after Distal Pancreatectomy in Pancreatic Body and/or Tail Adenocarcinoma. Acad Radiol 2025; 32:2027-2040. [PMID: 39537519 DOI: 10.1016/j.acra.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024]
Abstract
RATIONALE AND OBJECTIVES Clinically relevant postoperative pancreatic fistula (CR-POPF) is a threatening complication in body and/or tail pancreatic ductal adenocarcinoma (PDAC) receiving distal pancreatectomy (DP) and is difficult to predict preoperatively. We aimed to identify the role of baseline CT-based body composition analysis and extracellular volume (ECV) map in predicting the risk of CR-POPF preoperatively. MATERIALS AND METHODS A total of 329 resectable PDAC patients were enrolled and underwent multiphasic contrast-enhanced CT. Body composition indicators were calculated, and ECV maps were generated through multiphasic CT images. The differences in clinical variables and quantitative parameters between CR-POPF and non-CR-POPF patients were compared. Correlations between ECV fraction and pancreatic fibrosis stage were analyzed. Multivariate logistic regression was performed to screen the independent predictors and develop prediction models for CR-POPF. Receiver operating characteristic curve was utilized to evaluate the predictive performance. RESULTS Among 329 patients, 19.76% (65/329) developed CR-POPF. Albumin, pancreatic texture, and intraoperative blood loss were used to build the clinical model with an AUC of 0.764. ECV fraction and total muscle ratio (TMR) were chosen to build the radiological model with an AUC of 0.872. A combined nomogram integrated with albumin, ECV fraction, and TMR could significantly improve the discrimination ability to an AUC of 0.924 (Delong test, all p < 0.05). The ECV fraction showed high positive correlation with histological fibrosis grade (Spearman ρ = 0.81). CONCLUSION CT-based body composition analysis and ECV exhibited great potential for predicting CR-POPF in body and/or tail PDAC after DP. The combined nomogram could further improve the predictive performance.
Collapse
Affiliation(s)
- Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Mancang Hu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Ze Teng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (X.S.).
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| |
Collapse
|
9
|
Miller RJH, Yi J, Shanbhag A, Marcinkiewicz A, Patel KK, Lemley M, Ramirez G, Geers J, Chareonthaitawee P, Wopperer S, Berman DS, Di Carli M, Dey D, Slomka PJ. Deep learning-quantified body composition from positron emission tomography/computed tomography and cardiovascular outcomes: a multicentre study. Eur Heart J 2025:ehaf131. [PMID: 40159388 DOI: 10.1093/eurheartj/ehaf131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND AIMS Positron emission tomography (PET)/computed tomography (CT) myocardial perfusion imaging (MPI) is a vital diagnostic tool, especially in patients with cardiometabolic syndrome. Low-dose CT scans are routinely performed with PET for attenuation correction and potentially contain valuable data about body tissue composition. Deep learning and image processing were combined to automatically quantify skeletal muscle (SM), bone and adipose tissue from these scans and then evaluate their associations with death or myocardial infarction (MI). METHODS In PET MPI from three sites, deep learning quantified SM, bone, epicardial adipose tissue (EAT), subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), and intermuscular adipose tissue (IMAT). Sex-specific thresholds for abnormal values were established. Associations with death or MI were evaluated using unadjusted and multivariable models adjusted for clinical and imaging factors. RESULTS This study included 10 085 patients, with median age 68 (interquartile range 59-76) and 5767 (57%) male. Body tissue segmentations were completed in 102 ± 4 s. Higher VAT density was associated with an increased risk of death or MI in both unadjusted [hazard ratio (HR) 1.40, 95% confidence interval (CI) 1.37-1.43] and adjusted (HR 1.24, 95% CI 1.19-1.28) analyses, with similar findings for IMAT, SAT, and EAT. Patients with elevated VAT density and reduced myocardial flow reserve had a significantly increased risk of death or MI (adjusted HR 2.49, 95% CI 2.23-2.77). CONCLUSIONS Volumetric body tissue composition can be obtained rapidly and automatically from standard cardiac PET/CT. This new information provides a detailed, quantitative assessment of sarcopenia and cardiometabolic health for physicians.
Collapse
Affiliation(s)
- Robert J H Miller
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
- Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - Jirong Yi
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| | - Aakash Shanbhag
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
- Signal and Image Processing Institute, Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, USA
| | - Anna Marcinkiewicz
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
- Center of Radiological Diagnostics, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Krishna K Patel
- Department of Medicine (Cardiology) and Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark Lemley
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| | - Giselle Ramirez
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| | - Jolien Geers
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
- Department of Cardiology, Centrum voor Hart-en Vaatziekten (CHVZ), Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Samuel Wopperer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Daniel S Berman
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| | - Marcelo Di Carli
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Damini Dey
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| | - Piotr J Slomka
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, 6500 Wilshire Blvd, Suite 420, Los Angeles, CA 90048, USA
| |
Collapse
|
10
|
Xu K, Wang X, Zhou C, Zuo J, Zeng C, Zhou P, Zhang L, Gao X, Wang X. Synergic value of 3D CT-derived body composition and triglyceride glucose body mass for survival prognostic modeling in unresectable pancreatic cancer. Front Nutr 2025; 12:1499188. [PMID: 40177184 PMCID: PMC11961436 DOI: 10.3389/fnut.2025.1499188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Personalized and accurate survival risk prognostication remains a significant challenge in advanced pancreatic ductal adenocarcinoma (PDAC), despite extensive research on prognostic and predictive markers. Patients with PDAC are prone to muscle loss, fat consumption, and malnutrition, which is associated with inferior outcomes. This study investigated the use of three-dimensional (3D) anthropometric parameters derived from computed tomography (CT) scans and triglyceride glucose-body mass index (TyG-BMI) in relation to overall survival (OS) outcomes in advanced PDAC patients. Additionally, a predictive model for 1 year OS was developed based on body components and hematological indicators. Methods A retrospective analysis was conducted on 303 patients with locally advanced PDAC or synchronous metastases undergoing first-line chemotherapy, all of whom had undergone pretreatment abdomen-pelvis CT scans. Automatic 3D measurements of subcutaneous and visceral fat volume, skeletal muscle volume, and skeletal muscle density (SMD) were assessed at the L3 vertebral level by an artificial intelligence assisted diagnosis system (HY Medical). Various indicators including TyG-BMI, nutritional indicators [geriatric nutritional risk index (GNRI) and prealbumin], and inflammation indicators [(C-reactive protein (CRP) and neutrophil to lymphocyte ratio (NLR)] were also recorded. All patients underwent follow-up for at least 1 year and a dynamic nomogram for personalized survival prediction was constructed. Results We included 211 advanced PDAC patients [mean (standard deviation) age, 63.4 ± 11.2 years; 89 women (42.2) %)]. Factors such as low skeletal muscle index (SMI) (P = 0.011), high visceral to subcutaneous adipose tissue area ratio (VSR) (P < 0.001), high visceral fat index (VFI) (P < 0.001), low TyG-BMI (P = 0.004), and low prealbumin (P = 0.001) were identified as independent risk factors associated with 1 year OS. The area under the curve of the established dynamic nomogram was 0.846 and the calibration curve showed good consistency. High-risk patients (> 211.9 points calculated using the nomogram) had significantly reduced survival rates. Conclusion In this study, the proposed nomogram model (with web-based tool) enabled individualized prognostication of OS and could help to guide risk-adapted nutritional treatment for patients with unresectable PDAC or synchronous metastases.
Collapse
Affiliation(s)
- Kangjing Xu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinbo Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pinwen Zhou
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
11
|
Xie B, Liu B, Chen X, Chuan F, Liao K, Mei M, Li R, Zhou B. ALM adjusted by BMI or weight predicts adverse health outcomes in middle-aged and elderly patients with type 2 diabetes. Sci Rep 2025; 15:7963. [PMID: 40055426 PMCID: PMC11889084 DOI: 10.1038/s41598-025-92860-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 03/03/2025] [Indexed: 03/12/2025] Open
Abstract
The role of skeletal muscle in the prognosis of patients with Type 2 Diabetes Mellitus (T2DM) remains unclear. This study aimed to systematically evaluate the impact of different muscle-mass adjustment standards on adverse health outcomes in middle-aged and elderly T2DM patients. Retrospective cohort study. A total of 1,818 T2DM patients aged 50 years or older were included in this study. The cohort comprised 45.7% females, with a median age of 63 years. Variables closely correlated with total lean mass (TLM) and appendicular lean mass (ALM) were selected as adjustment indicators. The primary composite endpoints were all-cause mortality, cardiovascular disease (CVD), and fragility fractures. Cox proportional hazards models were used to estimate the risk associated with each indicator, and phenotypic characteristics of high-risk patients were evaluated. During a median follow-up of 63 months, 436 patients reached the primary endpoint. ALM/BMI and ALM/weight were negatively correlated with adverse outcomes in both sexes, even after adjusting for confounding factors (males: ALM/BMI (hazard ratio [HR] = 0.998, 95% confidence interval [CI] = 0.996-0.999, P = 0.005) and ALM/weight (HR = 0.924, 95% CI = 0.864-0.987, P = 0.020); females: ALM/BMI (HR = 0.998, 95% CI = 0.996-1.000, P = 0.030) and ALM/weight (HR = 0.917, 95% CI = 0.860-0.978, P = 0.008), respectively). Individuals with lower ALM/BMI and ALM/weight have poorer metabolic status, greater fat accumulation, more complications, and a lower muscle-to-fat ratio. Our findings demonstrate that both ALM/BMI and ALM/weight can predict adverse health outcomes, suggesting their potential as practical, clinically relevant markers for sarcopenia in T2DM.
Collapse
Affiliation(s)
- Bo Xie
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
- Department of Cardiovascular Medicine, Zhuzhou Central Hospital, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Bin Liu
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Xue Chen
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
| | - Fengning Chuan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
| | - Kun Liao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
| | - Mei Mei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
| | - Rong Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China
| | - Bo Zhou
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
12
|
Wang M, Chen Z, Yu T, You L, Peng Y, Chen H, Zhang P, Shi Z, Fang X, Jia L, Xia Z, Ji C, Tang H, Gao C. Low Skeletal Muscle Density Assessed by Abdominal Computerized Tomography Predicts Outcome in Children With Chronic Kidney Disease. J Ren Nutr 2025; 35:281-288. [PMID: 39549931 DOI: 10.1053/j.jrn.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVES Skeletal muscle loss and abnormal fat distribution are predictors of poor clinical outcomes in adults with chronic kidney disease (CKD). However, the relationship between body composition (muscle mass and adipose tissue) and prognosis in children with CKD has not been well elucidated. METHODS The retrospective single-center study enrolled children with CKD and healthy group who underwent an abdominal computerized tomography examination and compared the body composition of the third lumbar spine (L3) between the 2 groups. We defined the primary outcome as hemodialysis, peritoneal dialysis, kidney transplantation, or death. Logistic regression analysis was applied to assess the connection between low skeletal muscle density (SMD) and clinical and demographic variables. Multivariate Cox regression analysis was used to evaluate the risk factors for progression to the primary outcome. Kaplan-Meier survival analysis was performed to compare the effect of different body composition on event-free survival rate. RESULTS Thirty-two patients with CKD [estimated glomerular filtration rate: 14.89 (8.86, 29.88) (mL/min/1.73 m2)] and 66 heathy subjects [estimated glomerular filtration rate: 135.72 (121.70, 161.29) (mL/min/1.73 m2)] were recruited in our study. From the assessment of body composition assessed by computerized tomography, skeletal muscle area, SMD, and skeletal muscle index in the CKD group was lower than those in the healthy group (P < .05). On the other hand, visceral fat area and visceral fat index in the CKD group were significantly higher than those in the healthy group (P < .05). In logistic regression analysis, triglyceride (odds ratio: 8.635, 95% confidence interval (CI): 1.153-64.687) was independently associated with low SMD. After adjusting clinical data and body composition, high serum albumin (hazard ratio: 0.873, 95% CI: 0.798-0.955) and high SMD (hazard ratio: 0.895, 95% CI: 0.822-0.974) were protective factors for delaying renal failure. Based on the Kaplan-Meier analysis, only the group with low SMD had lower event-free survival in comparison to the reference group (P < .05). CONCLUSIONS These findings suggest that there is significant skeletal muscle loss and decrease in SMD in CKD children. Notably, low SMD is indicative of poor prognosis in CKD children.
Collapse
Affiliation(s)
- Meiqiu Wang
- Department of Pediatrics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zijian Chen
- Department of Radiology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tingting Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Lianghui You
- Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yingchao Peng
- Department of Pediatrics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Huangyu Chen
- Department of Information, Jinling Hospital, Nanjing, China
| | - Pei Zhang
- Department of Pediatrics, Jinling Hospital, Nanjing, China
| | - Zhuo Shi
- Department of Pediatrics, Jinling Hospital, Nanjing, China
| | - Xiang Fang
- Department of Pediatrics, Jinling Hospital, Nanjing, China
| | - LiLi Jia
- Department of Pediatrics, Jinling Hospital, Nanjing, China
| | - Zhengkun Xia
- Department of Pediatrics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Chenbo Ji
- Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China; Nanjing Medical Key Laboratory of Female Fertility Preservation and Restoration, Nanjing, China.
| | - Hao Tang
- Department of Radiology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China.
| | - Chunlin Gao
- Department of Pediatrics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
13
|
Yatim K, Ribas GT, Elton DC, Rockenbach MABC, Al Jurdi A, Pickhardt PJ, Garrett JW, Dreyer KJ, Bizzo BC, Riella LV. Applying Artificial Intelligence to Quantify Body Composition on Abdominal CTs and Better Predict Kidney Transplantation Wait-List Mortality. J Am Coll Radiol 2025; 22:332-341. [PMID: 40044312 DOI: 10.1016/j.jacr.2025.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Prekidney transplant evaluation routinely includes abdominal CT for presurgical vascular assessment. A wealth of body composition data are available from these CT examinations, but they remain an underused source of data, often missing from prognostication models, as these measurements require organ segmentation not routinely performed clinically by radiologists. We hypothesize that artificial intelligence facilitates accurate extraction of abdominal CT body composition data, allowing better prediction of outcomes. METHODS We conducted a retrospective, single-center observational study of kidney transplant candidates wait-listed between January 1, 2007, and December 31, 2017, with available CT data. Validated deep learning models quantified body composition including fat, aortic calcification, bone density, and muscle mass. Logistic regression was used to compare body composition data to Expected Post-Transplant Survival Score (EPTS) as a predictor of 5-year wait-list mortality. RESULTS In all, 899 patients were followed for a median 943 days (interquartile range 320-1,697). Of 899, 589 (65.5%) were men and 680 of 899 (75.6%) were White, non-Hispanic. Of 899, 167 patients (18.6%) died while on the waiting list. Myosteatosis (defined as the lowest tertile of muscle attenuation) and increased total aortic and abdominal calcification were associated with increased 5-year wait-list mortality. Logistic regression showed that imaging parameters performed similarly to EPTS at predicting 5-year wait-list mortality (area under receiver operating characteristic curve 0.70 [0.64-0.75] versus 0.67 [0.62-0.72], respectively), and combining body composition parameters with EPTS led to a slight improved survival prediction (area under receiver operating characteristic curve = 0.72, 95% confidence interval 0.66-0.76). CONCLUSIONS Fully automated quantification of body composition in kidney transplant candidates is feasible. Myosteatosis and atherosclerosis are associated with 5-year wait-list mortality.
Collapse
Affiliation(s)
- Karim Yatim
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts
| | - Guilherme T Ribas
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel C Elton
- Mass General Brigham AI, Mass General Brigham, Boston, Massachusetts
| | - Marcio A B C Rockenbach
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Mass General Brigham AI, Mass General Brigham, Boston, Massachusetts
| | - Ayman Al Jurdi
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts; Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - John W Garrett
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Keith J Dreyer
- Mass General Brigham AI, Mass General Brigham, Boston, Massachusetts
| | - Bernardo C Bizzo
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Mass General Brigham AI, Mass General Brigham, Boston, Massachusetts
| | - Leonardo V Riella
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts; Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
14
|
Salhöfer L, Bonella F, Meetschen M, Umutlu L, Forsting M, Schaarschmidt BM, Opitz MK, Kleesiek J, Hosch R, Koitka S, Parmar V, Nensa F, Haubold J. Automated 3D-Body Composition Analysis as a Predictor of Survival in Patients With Idiopathic Pulmonary Fibrosis. J Thorac Imaging 2025; 40:e0803. [PMID: 39183570 PMCID: PMC11837968 DOI: 10.1097/rti.0000000000000803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is the most common interstitial lung disease, with a median survival time of 2 to 5 years. The focus of this study is to establish a novel imaging biomarker. MATERIALS AND METHODS In this study, 79 patients (19% female) with a median age of 70 years were studied retrospectively. Fully automated body composition analysis (BCA) features (bone, muscle, total adipose tissue, intermuscular, and intramuscular adipose tissue) were combined into Sarcopenia, Fat, and Myosteatosis indices and compared between patients with a survival of more or less than 2 years. In addition, we divided the cohort at the median (high=≥ median, low= RESULTS A high Sarcopenia and Fat index and low Myosteatosis index were associated with longer median survival (35 vs. 16 mo for high vs. low Sarcopenia index, P =0.066; 44 vs. 14 mo for high vs. low Fat index, P <0.001; and 33 vs. 14 mo for low vs. high Myosteatosis index, P =0.0056) and better 5-year survival rates (34.0% vs. 23.6% for high vs. low Sarcopenia index; 47.3% vs. 9.2% for high vs. low Fat index; and 11.2% vs. 42.7% for high vs. low Myosteatosis index). Adjusted multivariate Cox regression showed a significant impact of the Fat (HR=0.71, P =0.01) and Myosteatosis (HR=1.12, P =0.005) on overall survival. CONCLUSION The fully automated BCA provides biomarkers with a predictive value for the overall survival in patients with IPF.
Collapse
Affiliation(s)
- Luca Salhöfer
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Francesco Bonella
- Department of Pneumology, Center for Interstitial and Rare Lung Diseases, University Hospital Essen, Essen, Germany
| | - Mathias Meetschen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | | | - Marcel Klaus Opitz
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Jens Kleesiek
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Rene Hosch
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Sven Koitka
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Vicky Parmar
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Felix Nensa
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
15
|
Pickhardt PJ, Kattan MW, Lee MH, Pooler BD, Pyrros A, Liu D, Zea R, Summers RM, Garrett JW. Biological age model using explainable automated CT-based cardiometabolic biomarkers for phenotypic prediction of longevity. Nat Commun 2025; 16:1432. [PMID: 39920106 PMCID: PMC11806064 DOI: 10.1038/s41467-025-56741-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
We derive and test a CT-based biological age model for predicting longevity, using an automated pipeline of explainable AI algorithms that quantifies skeletal muscle, abdominal fat, aortic calcification, bone density, and solid abdominal organs. We apply these AI tools to abdominal CT scans from 123,281 adults (mean age, 53.6 years; 47% women; median follow-up, 5.3 years). The final weighted CT biomarker selection was based on the index of prediction accuracy. The CT model significantly outperforms standard demographic data for predicting longevity (IPA = 29.2 vs. 21.7; 10-year AUC = 0.880 vs. 0.779; p < 0.001). Age- and sex-corrected survival hazard ratio for the highest-vs-lowest risk quartile was 8.73 (95% CI,8.14-9.36) for the CT biological age model, and increased to 24.79 after excluding cancer diagnoses within 5 years of CT. Muscle density, aortic plaque burden, visceral fat density, and bone density contributed the most. Here we show a personalized phenotypic CT biological age model that can be opportunistically-derived, regardless of clinical indication, to better inform risk assessment.
Collapse
Affiliation(s)
- Perry J Pickhardt
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA.
- The Department of Medical Physics, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA.
| | - Michael W Kattan
- The Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Matthew H Lee
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - B Dustin Pooler
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Ayis Pyrros
- Department of Radiology, Duly Health and Care, Downers Grove, IL, USA
- Department of Biomedical and Health Information Sciences, University of Illinois-Chicago, Chicago, IL, USA
| | - Daniel Liu
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Ryan Zea
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - John W Garrett
- The Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
- The Department of Medical Physics, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| |
Collapse
|
16
|
Kim H, Baek S, Han S, Kim GM, Sohn J, Rhee Y, Hong N, Kim MH. Low Skeletal Muscle Radiodensity Predicts Response to CDK4/6 Inhibitors Plus Aromatase Inhibitors in Advanced Breast Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e13666. [PMID: 39686815 DOI: 10.1002/jcsm.13666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Recent evidence indicates that a dysregulated host metabolism influences treatment outcomes in patients with breast cancer. We investigated the association of computed tomography (CT)-derived body composition indices with therapeutic responses in patients with hormone receptor-positive, HER2-negative advanced breast cancer (ABC) on endocrine plus CDK4/6 inhibitor (CDK4/6i) treatment. METHODS The study involved a retrospective cohort of patients with ABC at the Yonsei Cancer Center who received CDK4/6i and aromatase inhibitors as first-line therapy between January 2017 and October 2020. Body composition parameters were estimated from the non-enhanced CT images of the third lumbar spine by commercialized deep learning software. Patients with low skeletal muscle radiodensity (SMD) were defined as patients with SMD of low tertile (≤ 28.7 Hounsfield Units). The primary outcome was progression-free survival (PFS). RESULTS Among the 247 female participants (median age, 53 years; mean body mass index [BMI], 23.7 kg/m2), 45.7% had disease progression or death during a median follow-up of 36.4 months. After adjusting for age and visceral metastasis, SMD was the only independent predictor among body composition parameters for worse PFS (adjusted hazard ratio [HR] = 1.20 per standard deviation decrement, 95% CI: 1.01-1.42, p = 0.041), whereas BMI, muscle area, and fat area were not. Participants with low SMD had a higher risk of progression than those without (PFS, 27.2 vs. 51.1 months, p = 0.009; adjusted HR 1.84, 95% CI: 1.22-2.76, p = 0.003). Strong associations between low SMD and poor PFS were observed in groups with pre-menopause status (HR, 3.04 vs. 1.19 in post-menopause; 95% CI: 1.54-5.99, p for interaction < 0.05) and without visceral metastases (HR, 2.95 vs. 1.19 in with visceral metastases; 95% CI: 1.59-5.49, p for interaction < 0.05). CONCLUSIONS CT-defined low SMD predicts poor treatment outcomes in patients with ABC undergoing first-line treatment with aromatase inhibitors and CDK4/6i.
Collapse
Affiliation(s)
- Hyunwook Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Seungjin Baek
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sookyeong Han
- Endocrine Research Institute, Severance Hospital, Seoul, South Korea
| | - Gun Min Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Yumie Rhee
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Namki Hong
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Hwan Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
17
|
Salhöfer L, Jost G, Meetschen M, van Landeghem D, Forsting M, Bos D, Bojahr C, Hosch R, Nensa F, Pietsch H, Haubold J. The Impact of Radiation Dose on CT-Based Body Composition Analysis: A Large-Animal Study. J Cachexia Sarcopenia Muscle 2025; 16:e13741. [PMID: 39980200 PMCID: PMC11842463 DOI: 10.1002/jcsm.13741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND CT-based body composition analysis (BCA) enables the extraction of biomarkers from routine CT data. The influence of body composition on the prognosis of different patient groups has been highlighted in recent years. Typically, the segmentation of muscle and fat compartments is performed with a thresholding-based subsegmentation, which might be influenced by the image noise as a function of radiation dose. This study was performed to investigate the impact of the radiation dose on a fully automated, volumetric CT-based BCA. METHODS In this animal study, 20 Göttingen minipigs were subjected to CT scans on six occasions under five different dose settings with gradations compared to the control given in % from volumetric CT dose index (CTDIvol) of the control (5%, 10%, 20%, 40%, control [10.01 mGy]). A database with full dose (FD) and quarter dose (QD) CT scans from The Cancer Imaging Archive served as a human validation cohort. A previously open-source published and validated BCA network was applied to each scan. The following features were extracted as volumes (mL): bone, muscle, subcutaneous adipose tissue (SAT), intermuscular and intramuscular adipose tissue (IMAT), visceral adipose tissue (VAT) and total adipose tissue (TAT). Statistical significance was assessed by a one-way ANOVA with Tukey's multiple comparisons or Kruskal-Wallis with Dunn's post-hoc tests. The correlation between feature volumes in the dose gradations and the control group was analysed using the Spearman or Pearson method. RESULTS All BCA features remained consistent up to the 10% group and showed no significant differences compared with the control. In the lowest dose group (5%), there were significant differences concerning the muscle (5% = 1295 mL [211 mL], control = 1338 mL [248 mL]; p = 0.032) and VAT volumetry (5% = 353 mL [208 mL], control = 312 mL [204 mL]; p = 0.026) with median differences of -3.13% (muscle) and 12.3% (VAT), respectively. Significant and strong positive correlations were observed between all low-dose groups and the control (r > 0.977, p < 0.001). The human validation analysis yielded constant volumes for every BCA feature with a strong positive correlation (r > 0.933, p < 0.001). CONCLUSIONS Fully automated BCA maintains consistent results in various low-dose settings. Significant deviations are only observed after more than 90% dose reduction in the lowest dose settings (5%), which are currently not used in the clinical routine. This large-animal study demonstrates the consistency of fully automated BCA in different dose settings and may therefore facilitate its integration into the clinical routine.
Collapse
Affiliation(s)
- Luca Salhöfer
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| | - Gregor Jost
- MR and CT Contrast Media ResearchBayer AGBerlinGermany
| | - Mathias Meetschen
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| | - Daniel van Landeghem
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
| | - Michael Forsting
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
| | - Denise Bos
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
| | - Christian Bojahr
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| | - Rene Hosch
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| | - Felix Nensa
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| | | | - Johannes Haubold
- Institute of Interventional and Diagnostic Radiology and NeuroradiologyUniversity Hospital EssenEssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital EssenEssenGermany
| |
Collapse
|
18
|
Hittalamani IM. Augmenting the MELD Score with Machine Learning-Derived Body Composition Metrics on Abdominal CT to Predict 90-Day Mortality Post-TIPSS. Cardiovasc Intervent Radiol 2025; 48:231-232. [PMID: 39681742 DOI: 10.1007/s00270-024-03936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024]
Affiliation(s)
- Iranna Mallappa Hittalamani
- Department of Interventional Radiology, 2Nd Floor, OPD 28A, KLEs Dr Prabhakar Kore, Hospital and Medical Research Centre, Jawaharlal Nehru Medical College, Belagavi, Karnataka, India.
| |
Collapse
|
19
|
Cabini RF, Cozzi A, Leu S, Thelen B, Krause R, Del Grande F, Pizzagalli DU, Rizzo SMR. CompositIA: an open-source automated quantification tool for body composition scores from thoraco-abdominal CT scans. Eur Radiol Exp 2025; 9:12. [PMID: 39881078 PMCID: PMC11780042 DOI: 10.1186/s41747-025-00552-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Body composition scores allow for quantifying the volume and physical properties of specific tissues. However, their manual calculation is time-consuming and prone to human error. This study aims to develop and validate CompositIA, an automated, open-source pipeline for quantifying body composition scores from thoraco-abdominal computed tomography (CT) scans. METHODS A retrospective dataset of 205 contrast-enhanced thoraco-abdominal CT examinations was used for training, while 54 scans from a publicly available dataset were used for independent testing. Two radiology residents performed manual segmentation, identifying the centers of the L1 and L3 vertebrae and segmenting the corresponding axial slices. MultiResUNet was used to identify CT slices intersecting the L1 and L3 vertebrae, and its performance was evaluated using the mean absolute error (MAE). Two U-nets were used to segment the axial slices, with performance evaluated through the volumetric Dice similarity coefficient (vDSC). CompositIA's performance in quantifying body composition indices was assessed using mean percentage relative error (PRE), regression, and Bland-Altman analyses. RESULTS On the independent dataset, CompositIA achieved a MAE of about 5 mm in detecting slices intersecting the L1 and L3 vertebrae, with a MAE < 10 mm in at least 85% of cases and a vDSC greater than 0.85 in segmenting axial slices. Regression and Bland-Altman analyses demonstrated a strong linear relationship and good agreement between automated and manual scores (p values < 0.001 for all indices), with mean PREs ranging from 5.13% to 15.18%. CONCLUSION CompositIA facilitated the automated quantification of body composition scores, achieving high precision in independent testing. RELEVANCE STATEMENT CompositIA is an automated, open-source pipeline for quantifying body composition indices from CT scans, simplifying clinical assessments, and expanding their applicability. KEY POINTS Manual body composition assessment from CTs is time-consuming and prone to errors. CompositIA was trained on 205 CT scans and tested on 54 scans. CompositIA demonstrated mean percentage relative errors under 15% compared to manual indices. CompositIA simplifies body composition assessment through an artificial intelligence-driven and open-source pipeline.
Collapse
Affiliation(s)
- Raffaella Fiamma Cabini
- Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
- International Center of Advanced Computing in Medicine (ICAM), Pavia, Italy
| | - Andrea Cozzi
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Svenja Leu
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Benedikt Thelen
- Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
| | - Rolf Krause
- Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
- International Center of Advanced Computing in Medicine (ICAM), Pavia, Italy
| | - Filippo Del Grande
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Diego Ulisse Pizzagalli
- Euler Institute, Università della Svizzera italiana, Lugano, Switzerland.
- International Center of Advanced Computing in Medicine (ICAM), Pavia, Italy.
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland.
| | - Stefania Maria Rita Rizzo
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| |
Collapse
|
20
|
Styczen H, Maus V, Weiss D, Goertz L, Hosch R, Rubbert C, Beck N, Holtkamp M, Salhöfer L, Schubert R, Deuschl C, Nensa F, Haubold J. Impact of imaging biomarkers from body composition analysis on outcome of endovascularly treated acute ischemic stroke patients. J Neurointerv Surg 2025:jnis-2024-022275. [PMID: 39327046 DOI: 10.1136/jnis-2024-022275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND We investigate the association of imaging biomarkers extracted from fully automated body composition analysis (BCA) of computed tomography (CT) angiography images of endovascularly treated acute ischemic stroke (AIS) patients regarding angiographic and clinical outcome. METHODS Retrospective analysis of AIS patients treated with mechanical thrombectomy (MT) at three tertiary care-centers between March 2019-January 2022. Baseline demographics, angiographic outcome and clinical outcome evaluated by the modified Rankin Scale (mRS) at discharge were noted. Multiple tissues, such as muscle, bone, and adipose tissue were acquired with a deep-learning-based, fully automated BCA from CT images of the supra-aortic angiography. RESULTS A total of 290 stroke patients who underwent MT due to cerebral vessel occlusion in the anterior circulation were included in the study. In the univariate analyses, among all BCA markers, only the lower sarcopenia marker was associated with a poor outcome (P=0.007). It remained an independent predictor for an unfavorable outcome in a logistic regression analysis (OR 0.6, 95% CI 0.3 to 0.9, P=0.044). Fat index (total adipose tissue/bone) and myosteatosis index (inter- and intramuscular adipose tissue/total adipose tissue*100) did not affect clinical outcomes. CONCLUSION Acute ischemic stroke patients with a lower sarcopenia marker are at risk for an unfavorable outcome. Imaging biomarkers extracted from BCA can be easily obtained from existing CT images, making it readily available at the beginning of treatment. However, further research is necessary to determine whether sarcopenia provides additional value beyond established outcome predictors. Understanding its role could lead to optimized, individualized treatment plans for post-stroke patients, potentially improving recovery outcomes.
Collapse
Affiliation(s)
- Hanna Styczen
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Volker Maus
- Department of Radiology, Neuroradiology and Nuclear Medicine, Knappschaftskrankenhaus Langendreer, Ruhr-University Bochum, Bochum, Germany
- Klinikum Aschaffenburg-Alzenau, Institute for Radiology and Neuroradiology, Aschaffenburg, Germany
| | - Daniel Weiss
- Department of Diagnostic and Interventional Radiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Lukas Goertz
- Department of Diagnostic and Interventional Radiology, University Hospital of Cologne, Cologne, Germany
| | - René Hosch
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Essen, Germany
| | - Christian Rubbert
- Department of Diagnostic and Interventional Radiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Nikolas Beck
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Mathias Holtkamp
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Essen, Germany
| | - Luca Salhöfer
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Essen, Germany
| | - Rosa Schubert
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Cornelius Deuschl
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Felix Nensa
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen, Essen, Germany
| |
Collapse
|
21
|
Vieira FT, Cai Y, Gonzalez MC, Goodpaster BH, Prado CM, Haqq AM. Poor muscle quality: A hidden and detrimental health condition in obesity. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09941-0. [PMID: 39833502 DOI: 10.1007/s11154-025-09941-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Poor muscle quality (MQ) is a hidden health condition in obesity, commonly disregarded and underdiagnosed, associated with poor health-related outcomes. This narrative review provides an in-depth exploration of MQ in obesity, including definitions, available assessment methods and challenges, pathophysiology, association with health outcomes, and potential interventions. MQ is a broad term that can include imaging, histological, functional, or metabolic assessments, evaluating beyond muscle quantity. MQ assessment is highly heterogeneous and requires further standardization. Common definitions of MQ include 1) muscle-specific strength (or functional MQ), the ratio between muscle strength and muscle quantity, and 2) muscle composition (or morphological MQ), mainly evaluating muscle fat infiltration. An individual with obesity might still have normal or higher muscle quantity despite having poor MQ, and techniques for direct measurements are needed. However, the use of body composition and physical function assessments is still limited in clinical practice. Thus, more accessible techniques for assessing strength, muscle mass, and composition should be further explored. Obesity leads to adipocyte dysfunction, generating a low-grade chronic inflammatory state, which leads to mitochondrial dysfunction. Adipocyte and mitochondrial dysfunction result in metabolic dysfunction manifesting clinically as insulin resistance, dyslipidemia, and fat infiltration into organs such as muscle, which in excess is termed myosteatosis. Myosteatosis decreases muscle cell function and insulin sensitivity, creating a vicious cycle of inflammation and metabolic derangements. Myosteatosis increases the risk of poor muscle function, systemic metabolic complications, and mortality, presenting prognostic potential. Interventions shown to improve MQ include nutrition, physical activity/exercise, pharmacology, and metabolic and bariatric surgery.
Collapse
Affiliation(s)
- Flavio T Vieira
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Yuanjun Cai
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - M Cristina Gonzalez
- Postgraduate Program in Nutrition and Food, Federal University of Pelotas, Pelotas, Rio Grande Do Sul, Brazil
| | | | - Carla M Prado
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Andrea M Haqq
- Human Nutrition Research Unit, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
22
|
Tang H, Wang R, Hu N, Wang J, Wei Z, Gao X, Xie C, Qiu Y, Chen X. The association between computed tomography-based osteosarcopenia and osteoporotic vertebral fractures: a longitudinal study. J Endocrinol Invest 2025; 48:109-119. [PMID: 38890220 DOI: 10.1007/s40618-024-02415-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE Osteoporosis and sarcopenia usually coexist in older population. The concept of osteosarcopenia has been proposed in recent years. However, studies on the relationship between osteosarcopenia and the risk of fracture are rare, and the association is unclear at present. This study aimed to investigate the association between osteosarcopenia evaluated based on chest computed tomography (CT) and osteoporotic vertebral fracture (OVF). METHODS This study recruited 7906 individuals aged 50 years and older who did not have OVFs and underwent chest CT for physical examination between July 2016 and September 2019. Subjects were followed up annually until June 2023. Osteosarcopenia was defined by a low muscle area of the erector spinae (< 25.4 cm2) and the bone attenuation (Hounsfield unit, HU < 135). Genant's grades were used to define OVFs. Control subjects were selected by Propensity Score Matching at a ratio 20:1. Cox proportional hazards models were used to assess the associations between osteosarcopenia and OVFs. RESULTS Of the 7906 participants included, 95 had a new OVF within a median follow-up of 3 years. A total of 1900 control subjects were matched. Individuals in the osteosarcopenia group had a higher prevalence of spinal fractures than those in normal group (16.4% vs. 0.4%, P < 0.001). Osteosarcopenia was independently associated with OVF (adjusted hazard ratio (aHR): 12.67, 95% confidence interval (CI) 3.79-42.40) and severe OVF (aHR = 14.07, 95% CI 1.84-107.66). Similar trends were observed in males, females and those subjects aged older than 60 years. Osteosarcopenia had good predictive efficacy for OVF (area under the curve = 0.836). A nomogram was also developed for clinical application. CONCLUSION Osteosarcopenia assessed based on chest CT was associated with OVF, and osteosarcopenia has good performance for vertebral fracture prediction.
Collapse
Affiliation(s)
- H Tang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - R Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - N Hu
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - J Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - Z Wei
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - X Gao
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China
| | - C Xie
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Y Qiu
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China.
| | - X Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, China.
| |
Collapse
|
23
|
Lee MH, Zea R, Garrett JW, Summers RM, Pickhardt PJ. AI-based abdominal CT measurements of orthotopic and ectopic fat predict mortality and cardiometabolic disease risk in adults. Eur Radiol 2025; 35:520-531. [PMID: 38995381 DOI: 10.1007/s00330-024-10935-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/27/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVES To evaluate the utility of CT-based abdominal fat measures for predicting the risk of death and cardiometabolic disease in an asymptomatic adult screening population. METHODS Fully automated AI tools quantifying abdominal adipose tissue (L3 level visceral [VAT] and subcutaneous [SAT] fat area, visceral-to-subcutaneous fat ratio [VSR], VAT attenuation), muscle attenuation (L3 level), and liver attenuation were applied to non-contrast CT scans in asymptomatic adults undergoing CT colonography (CTC). Longitudinal follow-up documented subsequent deaths, cardiovascular events, and diabetes. ROC and time-to-event analyses were performed to generate AUCs and hazard ratios (HR) binned by octile. RESULTS A total of 9223 adults (mean age, 57 years; 4071:5152 M:F) underwent screening CTC from April 2004 to December 2016. 549 patients died on follow-up (median, nine years). Fat measures outperformed BMI for predicting mortality risk-5-year AUCs for muscle attenuation, VSR, and BMI were 0.721, 0.661, and 0.499, respectively. Higher visceral, muscle, and liver fat were associated with increased mortality risk-VSR > 1.53, HR = 3.1; muscle attenuation < 15 HU, HR = 5.4; liver attenuation < 45 HU, HR = 2.3. Higher VAT area and VSR were associated with increased cardiovascular event and diabetes risk-VSR > 1.59, HR = 2.6 for cardiovascular event; VAT area > 291 cm2, HR = 6.3 for diabetes (p < 0.001). A U-shaped association was observed for SAT with a higher risk of death for very low and very high SAT. CONCLUSION Fully automated CT-based measures of abdominal fat are predictive of mortality and cardiometabolic disease risk in asymptomatic adults and uncover trends that are not reflected in anthropomorphic measures. CLINICAL RELEVANCE STATEMENT Fully automated CT-based measures of abdominal fat soundly outperform anthropometric measures for mortality and cardiometabolic risk prediction in asymptomatic patients. KEY POINTS Abdominal fat depots associated with metabolic dysregulation and cardiovascular disease can be derived from abdominal CT. Fully automated AI body composition tools can measure factors associated with increased mortality and cardiometabolic risk. CT-based abdominal fat measures uncover trends in mortality and cardiometabolic risk not captured by BMI in asymptomatic outpatients.
Collapse
Affiliation(s)
- Matthew H Lee
- Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine & Public Health, 600 Highland Ave, Madison, WI, 53792, USA.
| | - Ryan Zea
- Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine & Public Health, 600 Highland Ave, Madison, WI, 53792, USA
| | - John W Garrett
- Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine & Public Health, 600 Highland Ave, Madison, WI, 53792, USA
| | - Ronald M Summers
- National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Perry J Pickhardt
- Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine & Public Health, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
24
|
Nelson LW, Lee MH, Garrett JW, Pickhardt SG, Warner JD, Summers RM, Pickhardt PJ. Intrapatient Changes in CT-Based Body Composition After Initiation of Semaglutide (Glucagon-Like Peptide-1 Receptor Agonist) Therapy. AJR Am J Roentgenol 2024:1-10. [PMID: 39230989 DOI: 10.2214/ajr.24.31805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
BACKGROUND. The long-acting glucagon-like peptide-1 receptor agonist semaglutide is used to treat type 2 diabetes or obesity in adults. Clinical trials have observed associations of semaglutide with weight loss, improved control of diabetes, and cardiovascular risk reduction. OBJECTIVE. The purpose of this study was to evaluate intrapatient changes in body composition after initiation of semaglutide therapy by applying an automated suite of CT-based artificial intelligence (AI) body composition tools. METHODS. This retrospective study included adult patients who were receiving semaglutide treatment and who, between January 2016 and November 2023, underwent abdominopelvic CT within both 5 years before and 5 years after initiation of semaglutide. An automated suite of previously validated CT-based AI body composition tools was applied to scans obtained before semaglutide initiation (hereafter, presemaglutide scans) and scans obtained after semaglutide initiation (hereafter, postsemaglutide scans) to quantify visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) area, skeletal muscle area and attenuation, intermuscular adipose tissue (IMAT) area, liver volume and attenuation, and trabecular bone mineral density (BMD). Patients with weight loss of 5 kg or more and those with weight gain of 5 kg or more between the scans were compared. RESULTS. The study included 241 patients (151 women and 90 men; mean age, 60.4 ± 12.4 [SD] years). In the weight-loss group (n = 67), the postsemaglutide scan, compared with the presemaglutide scan, showed a decrease in VAT area (309.4 vs 341.1 cm2, p < .001), SAT area (371.4 vs 410.7 cm2, p < .001), muscle area (179.2 vs 193.0, p < 0.001), and liver volume (2379.0 vs 2578 HU, p = .009) and an increase in liver attenuation (74.5 vs 67.6 HU, p = .03). In the weight-gain group (n = 48), the postsemaglutide scan, compared with the presemaglutide scan, showed an increase in VAT area (334.0 vs 312.8, p = .002), SAT area (485.8 vs 448.8 cm2, p = .01), and IMAT area (48.4 vs 37.6, p = .009) and a decrease in muscle attenuation (5.9 vs 13.1, p < .001). Other comparisons were not statistically significant (p > .05). CONCLUSION. Patients using semaglutide who lost versus gained weight showed distinct patterns of changes in CT-based body composition measures. Those with weight loss had overall favorable shifts in measures related to cardiometabolic risk. A decrease in muscle attenuation in those with weight gain is consistent with decreased muscle quality. CLINICAL IMPACT. Among patients using semaglutide, automated CT-based AI tools provide biomarkers of changes in body composition beyond those that are evident by standard clinical measures.
Collapse
Affiliation(s)
- Leslie W Nelson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Matthew H Lee
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - John W Garrett
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Silas G Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Joshua D Warner
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, MD
| | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
25
|
Jung M, Raghu VK, Reisert M, Rieder H, Rospleszcz S, Pischon T, Niendorf T, Kauczor HU, Völzke H, Bülow R, Russe MF, Schlett CL, Lu MT, Bamberg F, Weiss J. Deep learning-based body composition analysis from whole-body magnetic resonance imaging to predict all-cause mortality in a large western population. EBioMedicine 2024; 110:105467. [PMID: 39622188 DOI: 10.1016/j.ebiom.2024.105467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Manually extracted imaging-based body composition measures from a single-slice area (A) have shown associations with clinical outcomes in patients with cardiometabolic disease and cancer. With advances in artificial intelligence, fully automated volumetric (V) segmentation approaches are now possible, but it is unknown whether these measures carry prognostic value to predict mortality in the general population. Here, we developed and tested a deep learning framework to automatically quantify volumetric body composition measures from whole-body magnetic resonance imaging (MRI) and investigated their prognostic value to predict mortality in a large Western population. METHODS The framework was developed using data from two large Western European population-based cohort studies, the UK Biobank (UKBB) and the German National Cohort (NAKO). Body composition was defined as (i) subcutaneous adipose tissue (SAT), (ii) visceral adipose tissue (VAT), (iii) skeletal muscle (SM), SM fat fraction (SMFF), and (iv) intramuscular adipose tissue (IMAT). The prognostic value of the body composition measures was assessed in the UKBB using Cox regression analysis. Additionally, we extracted body composition areas for every level of the thoracic and lumbar spine (i) to compare the proposed volumetric whole-body approach to the currently established single-slice area approach on the height of the L3 vertebra and (ii) to investigate the correlation between volumetric and single slice area body composition measures on the level of each vertebral body. FINDINGS In 36,317 UKBB participants (mean age 65.1 ± 7.8 years, age range 45-84 years; 51.7% female; 1.7% [634/36,471] all-cause deaths; median follow-up 4.8 years), Cox regression revealed an independent association between VSM (adjusted hazard ratio [aHR]: 0.88, 95% confidence interval [CI] [0.81-0.91], p = 0.00023), VSMFF (aHR: 1.06, 95% CI [1.02-1.10], p = 0.0043), and VIMAT (aHR: 1.19, 95% CI [1.05-1.35], p = 0.0056) and mortality after adjustment for demographics (age, sex, BMI, race) and cardiometabolic risk factors (alcohol consumption, smoking status, hypertension, diabetes, history of cancer, blood serum markers). This association was attenuated when using traditional single-slice area measures. Highest correlation coefficients (R) between volumetric and single-slice area body composition measures were located at vertebra L5 for SAT (R = 0.820) and SMFF (R = 0.947), at L3 for VAT (R = 0.892), SM (R = 0.944), and at L4 for IMAT (R = 0.546) (all p < 0.0001). A similar pattern was found in 23,725 NAKO participants (mean age 53.9 ± 8.3 years, age range 40-75; 44.9% female). INTERPRETATION Automated volumetric body composition assessment from whole-body MRI predicted mortality in a large Western population beyond traditional clinical risk factors. Single slice areas were highly correlated with volumetric body composition measures but their association with mortality attenuated after multivariable adjustment. As volumetric body composition measures are increasingly accessible using automated techniques, identifying high-risk individuals may help to improve personalised prevention and lifestyle interventions. FUNDING This project was conducted using data from the German National Cohort (NAKO) (www.nako.de). The NAKO is funded by the Federal Ministry of Education and Research (BMBF) [project funding reference numbers: 01ER1301A/B/C, 01ER1511D, and 01ER1801A/B/C/D], federal states of Germany and the Helmholtz Association, the participating universities and the institutes of the Leibniz Association. This research has been conducted using the UK Biobank Resource under Application Number 80337. MJ was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)-518480401. VKR was funded by American Heart Association Career Development Award 935176 and National Heart, Lung, and Blood Institute-K01HL168231.
Collapse
Affiliation(s)
- Matthias Jung
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Vineet K Raghu
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Marco Reisert
- Medical Physics, Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany; Department of Stereotactic and Functional Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany.
| | - Hanna Rieder
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Susanne Rospleszcz
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, 13125, Germany.
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, 13125, Germany.
| | - Hans-Ulrich Kauczor
- Department of Diagnostic and Interventional Radiology, Member of the German Center of Lung Research, University Hospital Heidelberg, Heidelberg, 69120, Germany.
| | - Henry Völzke
- Institute for Community Medicine, Ernst Moritz Arndt University, Greifswald, 17489, Germany.
| | - Robin Bülow
- Institute for Diagnostic Radiology and Neuroradiology, University Medicine, Ernst Moritz Arndt University Greifswald, Greifswald, 17475, Germany.
| | - Maximilian F Russe
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Michael T Lu
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Jakob Weiss
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
26
|
Winder C, Clark M, Frood R, Smith L, Bulpitt A, Cook G, Scarsbrook A. Automated extraction of body composition metrics from abdominal CT or MR imaging: A scoping review. Eur J Radiol 2024; 181:111764. [PMID: 39368243 DOI: 10.1016/j.ejrad.2024.111764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE To review methodological approaches for automated segmentation of subcutaneous adipose tissue, visceral adipose tissue, and skeletal muscle from abdominal cross-sectional imaging for body composition analysis. METHOD Four databases were searched for publications describing automated segmentation of subcutaneous adipose tissue, visceral adipose tissue, and/or skeletal muscle from abdominal CT or MR imaging between 2019 and 2023. Included reports were evaluated to assess how imaging modality, cohort size, vertebral level, model dimensionality, and use of a volume or single slice affected segmentation accuracy and/or clinical utility. Exclusion criteria included reports not in English language, manual or semi-automated segmentation methods, reports prior to 2019 or solely of paediatric patients, and those not describing the use of abdominal CT or MR. RESULTS After exclusions, 172 reports were included in the review. CT imaging was utilised approximately four times as often as MRI, and segmentation accuracy did not significantly differ between the two modalities. Cohort size had no significant effect on segmentation accuracy. There was little evidence to refute the current practice of extracting body composition metrics from the third lumbar vertebral level. There was no clear benefit of using a 3D model to perform segmentation over a 2D approach. CONCLUSION Automated segmentation of intra-abdominal soft tissues for body composition analysis is an intense area of research activity. Segmentation accuracy is not affected by cross-sectional imaging modality. Extracting metrics from a single slice at the third lumbar vertebral level is a common approach, however, extracting metrics from a volumetric slab surrounding this level may increase the resilience of the technique, which is important for clinical translation. A paucity of publicly available datasets led to most reports using different data sources, preventing direct comparison of segmentation techniques. Future efforts should prioritise creating a standardised dataset to facilitate benchmarking of different algorithms and subsequent clinical adoption.
Collapse
Affiliation(s)
- Christopher Winder
- UKRI CDT in AI for Medical Diagnosis and Care, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK; School of Computing, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK.
| | - Matthew Clark
- Department of Radiology, St.James University Hospital, Beckett St, Harehills, LS9 7TF, Leeds, UK.
| | - Russell Frood
- Department of Radiology, St.James University Hospital, Beckett St, Harehills, LS9 7TF, Leeds, UK; CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK.
| | - Lesley Smith
- CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK.
| | - Andrew Bulpitt
- School of Computing, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK.
| | - Gordon Cook
- CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK; Leeds Cancer Centre, St. James University Hospital, Beckett St, Harehills, LS9 7TF, Leeds, UK.
| | - Andrew Scarsbrook
- Department of Radiology, St.James University Hospital, Beckett St, Harehills, LS9 7TF, Leeds, UK; Leeds Cancer Centre, St. James University Hospital, Beckett St, Harehills, LS9 7TF, Leeds, UK; Leeds Institute of Medical Research, University of Leeds, Woodhouse, LS2 9JT, Leeds, UK.
| |
Collapse
|
27
|
Chen HB, Miao Q, Liu YS, Lou XY, Zhang LD, Tan XD, Liang KK. The prognostic value of myosteatosis in pancreatic cancer: A systematic review and meta-analysis. Clin Nutr 2024; 43:116-123. [PMID: 39442392 DOI: 10.1016/j.clnu.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND AIMS The phenomenon of myosteatosis, characterized by the accumulation of ectopic fat within and surrounding skeletal muscle, has been identified as a potential adverse factor in the prognosis of individuals with cancer. This systematic review and meta-analysis sought to examine the association between myosteatosis and survival rates as well as postoperative complications in patients diagnosed with pancreatic cancer (PC). METHODS A systematic search was conducted on Web of Science, Embase, and Pubmed until March 25, 2024, to identify pertinent articles assessing the prognostic significance of myosteatosis in patients with PC, utilizing the search terms: myosteatosis, PC, and prognosis. The selected studies were utilized to investigate the prognostic impact of myosteatosis on the survival of PC patients. Forest plots and pooled effects models were employed to present the findings of this meta-analysis. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale (NOS). A total of 565 studies were initially identified from the three databases, with 14 retrospective cohort studies ultimately included in the final quantitative analysis. RESULTS The meta-analysis revealed a significant association between myosteatosis and both overall survival (OS) [Hazard Ratio (HR): 1.55, 95 % Confidence Interval (CI): 1.40-1.72, P < 0.001, I2 = 0.0 %] and recurrence-free survival (RFS) (HR 1.48, 95 % CI: 1.17-1.86, P = 0.001, I2 = 0.0 %) in patients diagnosed with PC. Subgroup analyses revealed that myosteatosis continued to be a negative prognostic factor in PC across various treatment modalities, patient populations, and myosteatosis assessment methods. Additionally, myosteatosis was identified as a risk factor for postoperative complications, with a pooled odds ratio of 2.20 (95 % CI: 1.45-3.35, P < 0.001, I2 = 37.5 %). All included studies achieved NOS scores of 6 or higher, indicating a relatively high level of methodological quality. CONCLUSION These results suggest that myosteatosis is significantly associated with both survival outcomes and postoperative complications in patients with PC.
Collapse
Affiliation(s)
- Hong-Bo Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Qi Miao
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110002, China
| | - Ya-Shu Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xin-Yu Lou
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Lu-Dan Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ke-Ke Liang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
28
|
Waite S, Davenport MS, Graber ML, Banja JD, Sheppard B, Bruno MA. Opportunity and Opportunism in Artificial Intelligence-Powered Data Extraction: A Value-Centered Approach. AJR Am J Roentgenol 2024; 223:e2431686. [PMID: 39291941 DOI: 10.2214/ajr.24.31686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Radiologists' traditional role in the diagnostic process is to respond to specific clinical questions and reduce uncertainty enough to permit treatment decisions to be made. This charge is rapidly evolving due to forces such as artificial intelligence (AI), big data (opportunistic imaging, imaging prognostication), and advanced diagnostic technologies. A new modernistic paradigm is emerging whereby radiologists, in conjunction with computer algorithms, will be tasked with extracting as much information from imaging data as possible, often without a specific clinical question being posed and independent of any stated clinical need. In addition, AI algorithms are increasingly able to predict long-term outcomes using data from seemingly normal examinations, enabling AI-assisted prognostication. As these algorithms become a standard component of radiology practice, the sheer amount of information they demand will increase the need for streamlined workflows, communication, and data management techniques. In addition, the provision of such information raises reimbursement, liability, and access issues. Guidelines will be needed to ensure that all patients have access to the benefits of this new technology and guarantee that mined data do not inadvertently create harm. In this Review, we discuss the challenges and opportunities relevant to radiologists in this changing landscape, with an emphasis on ensuring that radiologists provide high-value care.
Collapse
Affiliation(s)
- Stephen Waite
- Departments of Radiology and Internal Medicine, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203
| | - Matthew S Davenport
- Departments of Radiology and Urology, Ronald Weiser Center for Prostate Cancer, Michigan Medicine, Ann Arbor, MI
| | - Mark L Graber
- Department of Internal Medicine, Stony Brook University, Stony Brook, NY
| | - John D Banja
- Department of Rehabilitation Medicine and Center for Ethics, Emory University, Atlanta, GA
| | | | - Michael A Bruno
- Departments of Radiology and Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA
| |
Collapse
|
29
|
Chima RS, Glushko T, Park MA, Hodul P, Davis EW, Martin K, Qayyum A, Permuth JB, Jeong D. Effect of Intravenous Contrast on CT Body Composition Measurements in Patients with Intraductal Papillary Mucinous Neoplasm. Diagnostics (Basel) 2024; 14:2593. [PMID: 39594259 PMCID: PMC11592622 DOI: 10.3390/diagnostics14222593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The effect of differing post-contrast phases on CT body composition measurements is not yet known. METHODS A fully automated AI-based body composition analysis using DAFS was performed on a retrospective cohort of 278 subjects undergoing pre-treatment triple-phase CT for pancreatic intraductal papillary mucinous neoplasm. The CT contrast phases included noncontrast (NON), arterial (ART), and venous (VEN) phases. The software selected a single axial CT image at mid-L3 on each phase for body compartment segmentation. The areas (cm2) were calculated for skeletal muscle (SM), intermuscular adipose tissue (IMAT), visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT). The mean Hounsfield units of skeletal muscle (SMHU) within the segmented regions were calculated. Bland-Altman and Chi Square analyses were performed. RESULTS SM-NON had a lower percentage of bias [LOA] than SM-ART, -0.7 [-7.6, 6.2], and SM-VEN, -0.3 [-7.6, 7.0]; VAT-NON had a higher percentage of bias than ART, 3.4 [-18.2, 25.0], and VEN, 5.8 [-15.0, 26.6]; and this value was lower for SAT-NON than ART, -0.4 [-14.9, 14.2], and VEN, -0.5 [-14.3, 13.4]; and higher for IMAT-NON than ART, 5.9 [-17.9, 29.7], and VEN, 9.5 [-17.0, 36.1]. The bias in SMHU NON [LOA] was lower than that in ART, -3.8 HU [-9.8, 2.1], and VEN, -7.8 HU [-14.8, -0.8]. CONCLUSIONS IV contrast affects the voxel HU of fat and muscle, impacting CT analysis of body composition. We noted a relatively smaller bias in the SM, VAT, and SAT areas across the contrast phases. However, SMHU and IMAT experienced larger bias. During threshold risk stratification for CT-based measurements of SMHU and IMAT, the IV contrast phase should be taken into consideration.
Collapse
Affiliation(s)
- Ranjit S. Chima
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA (D.J.)
| | - Tetiana Glushko
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA (D.J.)
| | - Margaret A. Park
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Pamela Hodul
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Evan W. Davis
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Katelyn Martin
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Aliya Qayyum
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA (D.J.)
| | - Jennifer B. Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Daniel Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA (D.J.)
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
30
|
Mironchuk O, Chang AL, Rahmani F, Portell K, Nunez E, Nigogosyan Z, Ma D, Popuri K, Chow VTY, Beg MF, Luo J, Ippolito JE. Volumetric body composition analysis of the Cancer Genome Atlas reveals novel body composition traits and molecular markers Associated with Renal Carcinoma outcomes. Sci Rep 2024; 14:27022. [PMID: 39505904 PMCID: PMC11541764 DOI: 10.1038/s41598-024-76280-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Clinically, the body mass index remains the most frequently used metric of overall obesity, although it is flawed by its inability to account for different adipose (i.e., visceral, subcutaneous, and inter/intramuscular) compartments, as well as muscle mass. Numerous prior studies have demonstrated linkages between specific adipose or muscle compartments to outcomes of multiple diseases. Although there are no universally accepted standards for body composition measurement, many studies use a single slice at the L3 vertebral level. In this study, we use computed tomography (CT) studies from patients in The Cancer Genome Atlas (TCGA) to compare current L3-based techniques with volumetric techniques, demonstrating potential limitations with level-based approaches for assessing outcomes. In addition, we identify gene expression signatures in normal kidney that correlate with fat and muscle body composition traits that can be used to predict sex-specific outcomes in renal cell carcinoma.
Collapse
Affiliation(s)
| | - Andrew L Chang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Kaitlyn Portell
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Elena Nunez
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Zack Nigogosyan
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA
| | - Da Ma
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Siteman Cancer Center Biostatistics and Qualitative Research Shared Resource, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, Mail Stop Code: 8131, 4559 Scott Ave, St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Bernal-Contreras KD, Berrospe-Alfaro M, de Cárdenas-Rojo RL, Ramos-Ostos MH, Uribe M, López-Méndez I, Juárez-Hernández E. Body composition differences in patients with Metabolic Dysfunction-Associated Steatotic Liver Disease. Front Nutr 2024; 11:1490277. [PMID: 39564205 PMCID: PMC11575703 DOI: 10.3389/fnut.2024.1490277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024] Open
Abstract
Background Although body composition (BC) has been associated with Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), there is little evidence of differences in BC in patients with MASLD regarding body mass index (BMI). The aim of this study was to determine differences in BC in terms of BMI and metabolic comorbidities in patients with MASLD. Materials and methods It is a cross-sectional study with patients who attended the check-up unit. Liver steatosis was evaluated by controlled attenuation parameter, and patients were classified into five groups according to BMI, presence of MASLD, and metabolic characteristics: <25 kg/m2 non-MASLD; <25 kg/m2-MASLD; Overweight-MASLD; Metabolically Healthy Obese (MHO)-MASLD; and Metabolically Unhealthy Obese (MUO)-MASLD. BC was assessed by bioelectrical impedance and a Bioimpedance Vectorial Analysis (BIVA) was carried out. Differences in BC were analyzed by a One-Way ANOVA test. Univariate and multivariate analyses were performed for factors associated with abnormal BC. Results A total of 316 patients were included. 59% (n = 189) were male, with a mean age of 49 ± 10 years. Fat% significantly higher according to BMI was not different between BMI <25 kg/m2-MASLD and Overweight-MASLD groups. Skeletal muscle mass (SMM) was significantly lower in obesity groups with respect to overweight and normal weight groups (p < 0.05); however, no differences were observed in the post-hoc analysis. Extracellular Water/Intracellular Water ratio was significantly higher in the MHO-MASLD group and MUO-MASLD group compared with the BMI <25 kg/m2 non-MASLD group and with the BMI <25 kg/m2-MASLD group. Abnormal Waist Circumference (WC) and liver steatosis were independent factors associated with abnormal BC. Conclusion BC in MASLD patients varies according to BMI increase; changes could be explained by loss of SMM and not necessarily by the presence of metabolic abnormalities. High WC and the presence of steatosis are independent factors associated with altered BC.
Collapse
Affiliation(s)
| | | | | | - Martha H Ramos-Ostos
- Integral Diagnosis and Treatment Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
| | - Misael Uribe
- Gastroenterology and Obesity Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
| | - Iván López-Méndez
- Hepatology and Transplants Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
| | - Eva Juárez-Hernández
- Translational Research Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
| |
Collapse
|
32
|
Huang Y, Zhu Y, Xia W, Xie H, Yu H, Chen L, Shi L, Yu R. Computed tomography-based body composition indicative of diabetes after hypertriglyceridemic acute pancreatitis. Diabetes Res Clin Pract 2024; 217:111862. [PMID: 39299391 DOI: 10.1016/j.diabres.2024.111862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Post‑acute pancreatitis prediabetes/diabetes mellitus (PPDM‑A) is one of the common sequelae of acute pancreatitis (AP). The aim of our study was to build a machine learning (ML)-based prediction model for PPDM-A in hypertriglyceridemic acute pancreatitis (HTGP). METHODS We retrospectively enrolled 165 patients for our study. Demographic and laboratory data and body composition were collected. Multivariate logistic regression was applied to select features for ML. Support vector machine (SVM), linear discriminant analysis (LDA), and logistic regression (LR) were used to develop prediction models for PPDM-A. RESULTS 65 patients were diagnosed with PPDM-A, and 100 patients were diagnosed with non-PPDM-A. Of the 84 body composition-related parameters, 15 were significant in discriminating between the PPDM-A and non-PPDM-A groups. Using clinical indicators and body composition parameters to develop ML models, we found that the SVM model presented the best predictive ability, obtaining the best AUC=0.796 in the training cohort, and the LDA and LR model showing an AUC of 0.783 and 0.745, respectively. CONCLUSIONS The association between body composition and PPDM-A provides insight into the potential pathogenesis of PPDM-A. Our model is feasible for reliably predicting PPDM-A in the early stages of AP and enables early intervention in patients with potential PPDM-A.
Collapse
Affiliation(s)
- Yingbao Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zhu
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weizhi Xia
- Department of Radiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huanhuan Xie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huajun Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifang Chen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liuzhi Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Risheng Yu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
33
|
Salhöfer L, Bonella F, Meetschen M, Umutlu L, Forsting M, Schaarschmidt BM, Opitz M, Beck N, Zensen S, Hosch R, Parmar V, Nensa F, Haubold J. CT-based body composition analysis and pulmonary fat attenuation volume as biomarkers to predict overall survival in patients with non-specific interstitial pneumonia. Eur Radiol Exp 2024; 8:114. [PMID: 39400764 PMCID: PMC11473462 DOI: 10.1186/s41747-024-00519-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Non-specific interstitial pneumonia (NSIP) is an interstitial lung disease that can result in end-stage fibrosis. We investigated the influence of body composition and pulmonary fat attenuation volume (CTpfav) on overall survival (OS) in NSIP patients. METHODS In this retrospective single-center study, 71 NSIP patients with a median age of 65 years (interquartile range 21.5), 39 females (55%), who had a computed tomography from August 2009 to February 2018, were included, of whom 38 (54%) died during follow-up. Body composition analysis was performed using an open-source nnU-Net-based framework. Features were combined into: Sarcopenia (muscle/bone); Fat (total adipose tissue/bone); Myosteatosis (inter-/intra-muscular adipose tissue/total adipose tissue); Mediastinal (mediastinal adipose tissue/bone); and Pulmonary fat index (CTpfav/lung volume). Kaplan-Meier analysis with a log-rank test and multivariate Cox regression were used for survival analyses. RESULTS Patients with a higher (> median) Sarcopenia and lower (< median) Mediastinal Fat index had a significantly better survival probability (2-year survival rate: 83% versus 71% for high versus low Sarcopenia index, p = 0.023; 83% versus 72% for low versus high Mediastinal fat index, p = 0.006). In univariate analysis, individuals with a higher Pulmonary fat index exhibited significantly worse survival probability (2-year survival rate: 61% versus 94% for high versus low, p = 0.003). Additionally, it was an independent risk predictor for death (hazard ratio 2.37, 95% confidence interval 1.03-5.48, p = 0.043). CONCLUSION Fully automated body composition analysis offers interesting perspectives in patients with NSIP. Pulmonary fat index was an independent predictor of OS. RELEVANCE STATEMENT The Pulmonary fat index is an independent predictor of OS in patients with NSIP and demonstrates the potential of fully automated, deep-learning-driven body composition analysis as a biomarker for prognosis estimation. KEY POINTS This is the first study assessing the potential of CT-based body composition analysis in patients with non-specific interstitial pneumonia (NSIP). A single-center analysis of 71 patients with board-certified diagnosis of NSIP is presented Indices related to muscle, mediastinal fat, and pulmonary fat attenuation volume were significantly associated with survival at univariate analysis. CT pulmonary fat attenuation volume, normalized by lung volume, resulted as an independent predictor for death.
Collapse
Affiliation(s)
- Luca Salhöfer
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany.
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Mathias Meetschen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Benedikt M Schaarschmidt
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Marcel Opitz
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Nikolas Beck
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sebastian Zensen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - René Hosch
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Vicky Parmar
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Felix Nensa
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
34
|
Nault JC, Calderaro J, Ronot M. Integration of new technologies in the multidisciplinary approach to primary liver tumours: The next-generation tumour board. J Hepatol 2024; 81:756-762. [PMID: 38871125 DOI: 10.1016/j.jhep.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
Primary liver tumours, including benign liver tumours, hepatocellular carcinoma and cholangiocarcinoma, present a multifaceted challenge, necessitating a collaborative approach, as evidenced by the role of the multidisciplinary tumour board (MDTB). The approach to managing primary liver tumours involves specialised teams, including surgeons, radiologists, oncologists, pathologists, hepatologists, and radiation oncologists, coming together to propose individualised treatment plans. The evolving landscape of primary liver cancer treatment introduces complexities, particularly with the expanding array of systemic and locoregional therapies, alongside the potential integration of molecular biology and artificial intelligence (AI) into MDTBs in the future. Precision medicine demands collaboration across disciplines, challenging traditional frameworks. In the next decade, we anticipate the convergence of AI, molecular biology, pathology, and advanced imaging, requiring adaptability in MDTB structure to incorporate these cutting-edge technologies. Navigating this evolution also requires a focus on enhancing basic, translational, and clinical research, as well as boosting clinical trials through an upgraded use of MDTBs as hubs for scientific collaboration and raising literacy about AI and new technologies. In this review, we will delineate the current unmet needs in the clinical management of primary liver cancers, discuss our perspective on the future role of MDTBs in primary liver cancers ("next generation" MDTBs), and unravel the potential power and limitations of novel technologies that may shape the multidisciplinary care landscape for primary liver cancers in the coming decade.
Collapse
Affiliation(s)
- Jean-Charles Nault
- Liver unit, Hôpital Avicenne, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bobigny, France; Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, team « Functional Genomics of Solid Tumors », F-75006 Paris, France.
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France; Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France; MINT-Hep, Mondor Integrative Hepatology, Créteil, France
| | - Maxime Ronot
- Université de Paris, INSERM U1149 "Centre de Recherche sur l'inflammation", CRI, Paris, France; Department of Radiology, AP-HP, Hôpital Beaujon APHP.Nord, Clichy, France
| |
Collapse
|
35
|
Chung YE. Deep learning assisted biomarker development in patients with chronic hepatitis B: Editorial on "Prognostic role of computed tomography analysis using deep learning algorithm in patients with chronic hepatitis B viral infection". Clin Mol Hepatol 2024; 30:669-672. [PMID: 39038960 PMCID: PMC11540355 DOI: 10.3350/cmh.2024.0563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 07/24/2024] Open
Affiliation(s)
- Yong Eun Chung
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Warner JD, Blake GM, Garrett JW, Lee MH, Nelson LW, Summers RM, Pickhardt PJ. Correlation of HbA1c levels with CT-based body composition biomarkers in diabetes mellitus and metabolic syndrome. Sci Rep 2024; 14:21875. [PMID: 39300115 DOI: 10.1038/s41598-024-72702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes mellitus and metabolic syndrome are closely linked with visceral body composition, but clinical assessment is limited to external measurements and laboratory values including hemoglobin A1c (HbA1c). Modern deep learning and AI algorithms allow automated extraction of biomarkers for organ size, density, and body composition from routine computed tomography (CT) exams. Comparing visceral CT biomarkers across groups with differing glycemic control revealed significant, progressive CT biomarker changes with increasing HbA1c. For example, in the unenhanced female cohort, mean changes between normal and poorly-controlled diabetes showed: 53% increase in visceral adipose tissue area, 22% increase in kidney volume, 24% increase in liver volume, 6% decrease in liver density (hepatic steatosis), 16% increase in skeletal muscle area, and 21% decrease in skeletal muscle density (myosteatosis) (all p < 0.001). The multisystem changes of metabolic syndrome can be objectively and retrospectively measured using automated CT biomarkers, with implications for diabetes, metabolic syndrome, and GLP-1 agonists.
Collapse
Affiliation(s)
- Joshua D Warner
- The Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Glen M Blake
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - John W Garrett
- The Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Matthew H Lee
- The Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Leslie W Nelson
- The Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Perry J Pickhardt
- The Departments of Radiology and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI, 53792-3252, USA.
| |
Collapse
|
37
|
Cho SW, Baek S, Han S, Kim CO, Kim HC, Rhee Y, Hong N. Metabolic phenotyping with computed tomography deep learning for metabolic syndrome, osteoporosis and sarcopenia predicts mortality in adults. J Cachexia Sarcopenia Muscle 2024; 15:1418-1429. [PMID: 38649795 PMCID: PMC11294037 DOI: 10.1002/jcsm.13487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 03/06/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Computed tomography (CT) body compositions reflect age-related metabolic derangements. We aimed to develop a multi-outcome deep learning model using CT multi-level body composition parameters to detect metabolic syndrome (MS), osteoporosis and sarcopenia by identifying metabolic clusters simultaneously. We also investigated the prognostic value of metabolic phenotyping by CT model for long-term mortality. METHODS The derivation set (n = 516; 75% train set, 25% internal test set) was constructed using age- and sex-stratified random sampling from two community-based cohorts. Data from participants in the individual health assessment programme (n = 380) were used as the external test set 1. Semi-automatic quantification of body compositions at multiple levels of abdominal CT scans was performed to train a multi-layer perceptron (MLP)-based multi-label classification model. External test set 2 to test the prognostic value of the model output for mortality was built using data from individuals who underwent abdominal CT in a tertiary-level institution (n = 10 141). RESULTS The mean ages of the derivation and external sets were 62.8 and 59.7 years, respectively, without difference in sex distribution (women 50%) or body mass index (BMI; 23.9 kg/m2). Skeletal muscle density (SMD) and bone density (BD) showed a more linear decrement across age than skeletal muscle area. Alternatively, an increase in visceral fat area (VFA) was observed in both men and women. Hierarchical clustering based on multi-level CT body composition parameters revealed three distinctive phenotype clusters: normal, MS and osteosarcopenia clusters. The L3 CT-parameter-based model, with or without clinical variables (age, sex and BMI), outperformed clinical model predictions of all outcomes (area under the receiver operating characteristic curve: MS, 0.76 vs. 0.55; osteoporosis, 0.90 vs. 0.79; sarcopenia, 0.85 vs. 0.81 in external test set 1; P < 0.05 for all). VFA contributed the most to the MS predictions, whereas SMD, BD and subcutaneous fat area were features of high importance for detecting osteoporosis and sarcopenia. In external test set 2 (mean age 63.5 years, women 79%; median follow-up 4.9 years), a total of 907 individuals (8.9%) died during follow-up. Among model-predicted metabolic phenotypes, sarcopenia alone (adjusted hazard ratio [aHR] 1.55), MS + sarcopenia (aHR 1.65), osteoporosis + sarcopenia (aHR 1.83) and all three combined (aHR 1.87) remained robust predictors of mortality after adjustment for age, sex and comorbidities. CONCLUSIONS A CT body composition-based MLP model detected MS, osteoporosis and sarcopenia simultaneously in community-dwelling and hospitalized adults. Metabolic phenotypes predicted by the CT MLP model were associated with long-term mortality, independent of covariates.
Collapse
Affiliation(s)
- Sang Wouk Cho
- Department of Internal Medicine, Endocrine Research InstituteSeverance Hospital, Yonsei University College of MedicineSeoulSouth Korea
- Institue for Innovation in Digital Healthcare (IIDH)Yonsei University Health SystemSeoulSouth Korea
| | - Seungjin Baek
- Department of Internal Medicine, Endocrine Research InstituteSeverance Hospital, Yonsei University College of MedicineSeoulSouth Korea
| | - Sookyeong Han
- Department of Internal Medicine, Endocrine Research InstituteSeverance Hospital, Yonsei University College of MedicineSeoulSouth Korea
- Institue for Innovation in Digital Healthcare (IIDH)Yonsei University Health SystemSeoulSouth Korea
| | - Chang Oh Kim
- Division of Geriatric Medicine, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Hyeon Chang Kim
- Institue for Innovation in Digital Healthcare (IIDH)Yonsei University Health SystemSeoulSouth Korea
- Department of Preventive MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Yumie Rhee
- Department of Internal Medicine, Endocrine Research InstituteSeverance Hospital, Yonsei University College of MedicineSeoulSouth Korea
- Institue for Innovation in Digital Healthcare (IIDH)Yonsei University Health SystemSeoulSouth Korea
| | - Namki Hong
- Department of Internal Medicine, Endocrine Research InstituteSeverance Hospital, Yonsei University College of MedicineSeoulSouth Korea
- Institue for Innovation in Digital Healthcare (IIDH)Yonsei University Health SystemSeoulSouth Korea
| |
Collapse
|
38
|
Pickhardt PJ. Harnessing the Value of Incidental Tissue and Organ Data at Body CT. Radiology 2024; 312:e241349. [PMID: 39105643 DOI: 10.1148/radiol.241349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Affiliation(s)
- Perry J Pickhardt
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| |
Collapse
|
39
|
Chang Y, Yoon SH, Kwon R, Kang J, Kim YH, Kim JM, Chung HJ, Choi J, Jung HS, Lim GY, Ahn J, Wild SH, Byrne CD, Ryu S. Automated Comprehensive CT Assessment of the Risk of Diabetes and Associated Cardiometabolic Conditions. Radiology 2024; 312:e233410. [PMID: 39105639 DOI: 10.1148/radiol.233410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Background CT performed for various clinical indications has the potential to predict cardiometabolic diseases. However, the predictive ability of individual CT parameters remains underexplored. Purpose To evaluate the ability of automated CT-derived markers to predict diabetes and associated cardiometabolic comorbidities. Materials and Methods This retrospective study included Korean adults (age ≥ 25 years) who underwent health screening with fluorine 18 fluorodeoxyglucose PET/CT between January 2012 and December 2015. Fully automated CT markers included visceral and subcutaneous fat, muscle, bone density, liver fat, all normalized to height (in meters squared), and aortic calcification. Predictive performance was assessed with area under the receiver operating characteristic curve (AUC) and Harrell C-index in the cross-sectional and survival analyses, respectively. Results The cross-sectional and cohort analyses included 32166 (mean age, 45 years ± 6 [SD], 28833 men) and 27 298 adults (mean age, 44 years ± 5 [SD], 24 820 men), respectively. Diabetes prevalence and incidence was 6% at baseline and 9% during the 7.3-year median follow-up, respectively. Visceral fat index showed the highest predictive performance for prevalent and incident diabetes, yielding AUC of 0.70 (95% CI: 0.68, 0.71) for men and 0.82 (95% CI: 0.78, 0.85) for women and C-index of 0.68 (95% CI: 0.67, 0.69) for men and 0.82 (95% CI: 0.77, 0.86) for women, respectively. Combining visceral fat, muscle area, liver fat fraction, and aortic calcification improved predictive performance, yielding C-indexes of 0.69 (95% CI: 0.68, 0.71) for men and 0.83 (95% CI: 0.78, 0.87) for women. The AUC for visceral fat index in identifying metabolic syndrome was 0.81 (95% CI: 0.80, 0.81) for men and 0.90 (95% CI: 0.88, 0.91) for women. CT-derived markers also identified US-diagnosed fatty liver, coronary artery calcium scores greater than 100, sarcopenia, and osteoporosis, with AUCs ranging from 0.80 to 0.95. Conclusion Automated multiorgan CT analysis identified individuals at high risk of diabetes and other cardiometabolic comorbidities. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Pickhardt in this issue.
Collapse
Affiliation(s)
- Yoosoo Chang
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Soon Ho Yoon
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Ria Kwon
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Jeonggyu Kang
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Young Hwan Kim
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Jong-Min Kim
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Han-Jae Chung
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - JunHyeok Choi
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Hyun-Suk Jung
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Ga-Young Lim
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Jiin Ahn
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Sarah H Wild
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Christopher D Byrne
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| | - Seungho Ryu
- From the Center for Cohort Studies (Y.C., R.K., J.K., J.H.C., H.S.J., G.Y.L., J.A., S.R.) and Department of Occupational and Environmental Medicine (Y.C., S.R.), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, Republic of Korea; Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea (Y.C., S.R.); Department of Radiology, Seoul National University Hospital, Seoul National College of Medicine, Seoul, Republic of Korea (S.H.Y.); Research & Science Division, MEDICAL IP, Seoul, Republic of Korea (J.M.K., H.J.C.); Institute of Medical Research, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (R.K., G.Y.L.); Department of Nuclear Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea (Y.H.K.); Usher Institute, University of Edinburgh, Edinburgh, United Kingdom (S.H.W.); Department of Nutrition and Metabolism, University of Southampton Faculty of Medicine, Southampton, United Kingdom (C.D.B.); and National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom (C.D.B.)
| |
Collapse
|
40
|
Yi J, Michalowska AM, Shanbhag A, Miller RJH, Geers J, Zhang W, Killekar A, Manral N, Lemley M, Buchwald M, Kwiecinski J, Zhou J, Kavanagh PB, Liang JX, Builoff V, Ruddy TD, Einstein AJ, Feher A, Miller EJ, Sinusas AJ, Berman DS, Dey D, Slomka PJ. AI-based volumetric six-tissue body composition quantification from CT cardiac attenuation scans enhances mortality prediction: multicenter study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.30.24311224. [PMID: 39132480 PMCID: PMC11312626 DOI: 10.1101/2024.07.30.24311224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Computed tomography attenuation correction (CTAC) scans are routinely obtained during cardiac perfusion imaging, but currently only utilized for attenuation correction and visual calcium estimation. We aimed to develop a novel artificial intelligence (AI)-based approach to obtain volumetric measurements of chest body composition from CTAC scans and evaluate these measures for all-cause mortality (ACM) risk stratification. Methods We applied AI-based segmentation and image-processing techniques on CTAC scans from a large international image-based registry (four sites), to define chest rib cage and multiple tissues. Volumetric measures of bone, skeletal muscle (SM), subcutaneous, intramuscular (IMAT), visceral (VAT), and epicardial (EAT) adipose tissues were quantified between automatically-identified T5 and T11 vertebrae. The independent prognostic value of volumetric attenuation, and indexed volumes were evaluated for predicting ACM, adjusting for established risk factors and 18 other body compositions measures via Cox regression models and Kaplan-Meier curves. Findings End-to-end processing time was <2 minutes/scan with no user interaction. Of 9918 patients studied, 5451(55%) were male. During median 2.5 years follow-up, 610 (6.2%) patients died. High VAT, EAT and IMAT attenuation were associated with increased ACM risk (adjusted hazard ratio (HR) [95% confidence interval] for VAT: 2.39 [1.92, 2.96], p<0.0001; EAT: 1.55 [1.26, 1.90], p<0.0001; IMAT: 1.30 [1.06, 1.60], p=0.0124). Patients with high bone attenuation were at lower risk of death as compared to subjects with lower bone attenuation (adjusted HR 0.77 [0.62, 0.95], p=0.0159). Likewise, high SM volume index was associated with a lower risk of death (adjusted HR 0.56 [0.44, 0.71], p<0.0001). Interpretations CTAC scans obtained routinely during cardiac perfusion imaging contain important volumetric body composition biomarkers which can be automatically measured and offer important additional prognostic value.
Collapse
Affiliation(s)
- Jirong Yi
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna M Michalowska
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center of Radiological Diagnostics, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Aakash Shanbhag
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Signal and Image Processing Institute, Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, USA
| | - Robert J H Miller
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - Jolien Geers
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Cardiology, Centrum voor Hart-en Vaatziekten (CHVZ), Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wenhao Zhang
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aditya Killekar
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nipun Manral
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mark Lemley
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mikolaj Buchwald
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | - Jianhang Zhou
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul B Kavanagh
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joanna X Liang
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Valerie Builoff
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Terrence D Ruddy
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Andrew J Einstein
- Division of Cardiology, Department of Medicine, and Department of Radiology, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, New York, United States
| | - Attila Feher
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Daniel S Berman
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Damini Dey
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Piotr J Slomka
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
41
|
Zeng F, Jiang W, Chang X, Yang F, Luo X, Liu R, Lei Y, Li J, Pan C, Huang X, Sun H, Lan Y. Sarcopenia is associated with short- and long-term mortality in patients with acute-on-chronic liver failure. J Cachexia Sarcopenia Muscle 2024; 15:1473-1482. [PMID: 38965993 PMCID: PMC11294047 DOI: 10.1002/jcsm.13501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND While sarcopenia is recognized as a predictor of mortality in cirrhosis, its influence on acute-on-chronic liver failure (ACLF) remains uncertain. Despite multiple studies examining the impact of sarcopenia on short-term mortality in patients with ACLF, the sample size of these studies was limited, and their outcomes were inconsistent. Therefore, this study aimed to explore the impact of sarcopenia on both short- and long-term mortality in patients with ACLF. METHODS This retrospective cohort study included 414 patients with ACLF that were treated between January 2016 and September 2022. Sarcopenia was diagnosed based on the measurement of the skeletal muscle index at the third lumbar vertebra (L3-SMI). Subsequently, the patients were divided into sarcopenia and non-sarcopenia groups. We analysed the basic clinical data of the two groups. Multivariate Cox proportional analysis was used to analyse short-term (28 days) and long-term (1 year and overall) mortality rates. RESULTS A total of 414 patients were included, with a mean age of 52.88 ± 13.41 years. Among them, 318 (76.8%) were male, and 239 (57.7%) had sarcopenia. A total of 280 (67.6%) patients died during the study period. Among them, 153 patients died within 28 days (37%) and 209 patients died within 1 year (50.5%). We found that the 28-day, 1-year and overall mortality rates in the sarcopenia group were significantly higher than those in the non-sarcopenia group (37% vs. 22.3%, P < 0.01; 50.5% vs. 34.9%, P < 0.01; and 67.6% vs. 53.1%, P < 0.01, respectively). Multivariate Cox regression analysis revealed that sarcopenia was significantly associated with increased mortality. The hazard ratios for sarcopenia were 2.05 (95% confidence interval [CI] 1.41-3.00, P < 0.01) for 28-day mortality, 1.81 (95% CI 1.29-2.54, P < 0.01) for 1-year mortality and 1.82 (95% CI 1.30-2.55, P < 0.01) for overall mortality. In addition, muscle density and international normalized ratio were associated with short- and long-term mortality. CONCLUSIONS Sarcopenia is associated with both short- and long-term mortality in patients with ACLF. Therefore, regular monitoring for sarcopenia is important for these patients.
Collapse
Affiliation(s)
- Fan Zeng
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Wei Jiang
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
- Clinical Medicine School of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Xiujun Chang
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
- Clinical Medicine School of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Fuxun Yang
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Xiaoxiu Luo
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Rongan Liu
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Yu Lei
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Jiajia Li
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Chun Pan
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Xiaobo Huang
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| | - Huaiqiang Sun
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yunping Lan
- Department of Intensive Care UnitSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalChengduChina
| |
Collapse
|
42
|
Choi I, Choi J, Yong HS, Yang Z. Deep learning-based respiratory muscle segmentation as a potential imaging biomarker for respiratory function assessment. PLoS One 2024; 19:e0306789. [PMID: 39058719 PMCID: PMC11280157 DOI: 10.1371/journal.pone.0306789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Respiratory diseases significantly affect respiratory function, making them a considerable contributor to global mortality. The respiratory muscles play an important role in disease prognosis; as such, quantitative analysis of the respiratory muscles is crucial to assess the status of the respiratory system and the quality of life in patients. In this study, we aimed to develop an automated approach for the segmentation and classification of three types of respiratory muscles from computed tomography (CT) images using artificial intelligence. With a dataset of approximately 600,000 thoracic CT images from 3,200 individuals, we trained the model using the Attention U-Net architecture, optimized for detailed and focused segmentation. Subsequently, we calculated the volumes and densities from the muscle masks segmented by our model and performed correlation analysis with pulmonary function test (PFT) parameters. The segmentation models for muscle tissue and respiratory muscles obtained dice scores of 0.9823 and 0.9688, respectively. The classification model, achieving a generalized dice score of 0.9900, also demonstrated high accuracy in classifying thoracic region muscle types, as evidenced by its F1 scores: 0.9793 for the pectoralis muscle, 0.9975 for the erector spinae muscle, and 0.9839 for the intercostal muscle. In the correlation analysis, the volume of the respiratory muscles showed a strong correlation with PFT parameters, suggesting that respiratory muscle volume may serve as a potential novel biomarker for respiratory function. Although muscle density showed a weaker correlation with the PFT parameters, it has a potential significance in medical research.
Collapse
Affiliation(s)
- Insung Choi
- Department of Integrative Medicine, Major in Digital Healthcare, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Radiology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Juwhan Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Hwan Seok Yong
- Department of Radiology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Zepa Yang
- Department of Radiology, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
43
|
Swartz AZ, Robles ME, Park S, Esfandiari H, Bradshaw M, Koethe JR, Silver HJ. Cardiometabolic Characteristics of Obesity Phenotypes in Persons With HIV. Open Forum Infect Dis 2024; 11:ofae376. [PMID: 39035569 PMCID: PMC11259191 DOI: 10.1093/ofid/ofae376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024] Open
Abstract
Background In the general population, it is established that adipose tissue depots pose various risks for cardiometabolic diseases. The interaction among obesity, HIV, and antiretroviral treatment promotes even greater risk for persons with HIV (PWH). As obesity is a heterogeneous condition, determining the specific obesity phenotypes present and their characteristics is critical to personalize care in PWH. Methods Visceral, sarcopenic, myosteatotic, hepatosteatotic, and metabolically healthy obesity phenotypes were determined by pre-established cut points after segmentation of computed tomography scans at the L3 vertebra. Multivariable linear regression modeling included anthropometrics, clinical biomarkers, and inflammatory factors while controlling for age, sex, race, and body mass index (BMI). Results Of 187 PWH, 86% were male, and the mean ± SD age and BMI were 51.2 ± 12.3 years and 32.6 ± 6.3 kg/m2. Overall, 59% had visceral obesity, 11% sarcopenic obesity, 25% myosteatotic obesity, 9% hepatosteatotic obesity, and 32% metabolically healthy obesity. The strongest predictor of visceral obesity was an elevated triglyceride:high-density lipoprotein (HDL) ratio. Increased subcutaneous fat, waist circumference, and HDL cholesterol were predictors of sarcopenic obesity. Diabetes status and elevated interleukin 6, waist circumference, and HDL cholesterol predicted myosteatotic obesity. An increased CD4+ count and a decreased visceral:subcutaneous adipose tissue ratio predicted hepatosteatotic obesity, though accounting for only 28% of its variability. Participants with metabolically healthy obesity were on average 10 years younger, had higher HDL, lower triglyceride:HDL ratio, and reduced CD4+ counts. Conclusions These findings show that discrete obesity phenotypes are highly prevalent in PWH and convey specific risk factors that measuring BMI alone does not capture. These clinically relevant findings can be used in risk stratification and optimization of personalized treatment regimens. This study is registered at ClinicalTrials.gov (NCT04451980).
Collapse
Affiliation(s)
- Alison Z Swartz
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Michelle E Robles
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Seungweon Park
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Helia Esfandiari
- College of Arts and Sciences, University of Tennessee, Knoxville, Tennessee, USA
| | - Marques Bradshaw
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - John R Koethe
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heidi J Silver
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
44
|
Moeller AR, Garrett JW, Summers RM, Pickhardt PJ. Adjusting for the effect of IV contrast on automated CT body composition measures during the portal venous phase. Abdom Radiol (NY) 2024; 49:2543-2551. [PMID: 38744704 DOI: 10.1007/s00261-024-04376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Fully-automated CT-based algorithms for quantifying numerous biomarkers have been validated for unenhanced abdominal scans. There is great interest in optimizing the documentation and reporting of biophysical measures present on all CT scans for the purposes of opportunistic screening and risk profiling. The purpose of this study was to determine and adjust the effect of intravenous (IV) contrast on these automated body composition measures at routine portal venous phase post-contrast imaging. METHODS Final study cohort consisted of 1,612 older adults (mean age, 68.0 years; 594 women) all imaged utilizing a uniform CT urothelial protocol consisting of pre-contrast, portal venous, and delayed excretory phases. Fully-automated CT-based algorithms for quantifying numerous biomarkers, including muscle and fat area and density, bone mineral density, and solid organ volume were applied to pre-contrast and portal venous phases. The effect of IV contrast upon these body composition measures was analyzed. Regression analyses, including square of the Pearson correlation coefficient (r2), were performed for each comparison. RESULTS We found that simple, linear relationships can be derived to determine non-contrast equivalent values from the post-contrast CT biomeasures. Excellent positive linear correlation (r2 = 0.91-0.99) between pre- and post-contrast values was observed for all automated soft tissue measures, whereas moderate positive linear correlation was observed for bone attenuation (r2 = 0.58-0.76). In general, the area- and volume-based measurement require less adjustment than attenuation-based measures, as expected. CONCLUSION Fully-automated quantitative CT-biomarker measures at portal venous phase abdominal CT can be adjusted to a non-contrast equivalent using simple, linear relationships.
Collapse
Affiliation(s)
- Alexander R Moeller
- Department of Radiology, University of Wisconsin School of Medicine & Public Health, E3/311 Clinical Science Center 600 Highland Ave., Madison, WI, 53792-3252, USA
| | - John W Garrett
- Department of Radiology, University of Wisconsin School of Medicine & Public Health, E3/311 Clinical Science Center 600 Highland Ave., Madison, WI, 53792-3252, USA
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine & Public Health, E3/311 Clinical Science Center 600 Highland Ave., Madison, WI, 53792-3252, USA.
| |
Collapse
|
45
|
Chang YY, Cheng B. Prognostic impact of myosteatosis in patients with colorectal cancer undergoing curative surgery: an updated systematic review and meta-analysis. Front Oncol 2024; 14:1388001. [PMID: 38962266 PMCID: PMC11219791 DOI: 10.3389/fonc.2024.1388001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/16/2024] [Indexed: 07/05/2024] Open
Abstract
Background Colorectal cancer (CRC) is a global health concern, and identifying prognostic factors can improve outcomes. Myosteatosis is fat infiltration into muscles and is a potential predictor of the survival of patients with CRC. Methods This systematic review and meta-analysis aimed to assess the prognostic role of myosteatosis in CRC. PubMed, Embase, and Cochrane CENTRAL were searched up to 1 August 2023, for relevant studies, using combinations of the keywords CRC, myosteatosis, skeletal muscle fat infiltration, and low skeletal muscle radiodensity. Case-control, prospective, and retrospective cohort studies examining the association between myosteatosis and CRC outcomes after curative intent surgery were eligible for inclusion. Primary outcomes were overall survival (OS), disease-free survival (DFS), and cancer-specific survival (CSS). Results A total of 10 studies with a total of 9,203 patients were included. The pooled hazard ratio (HR) for OS (myosteatosis vs. no myosteatosis) was 1.52 [95% confidence interval (CI), 1.38-1.67); for CSS, 1.67 (95% CI, 1.40-1.99); and for DFS, 1.89 (95% CI, 1.35-2.65). Conclusion In patients with CRC undergoing curative intent surgery, myosteatosis is associated with worse OS, CSS, and DFS. These findings underscore the importance of evaluating myosteatosis in patients with CRC to improve outcomes.
Collapse
Affiliation(s)
- Yu-Yao Chang
- Division of Colon and Rectal Surgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Bill Cheng
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
46
|
Pickhardt PJ. Abdominal CT-Based Body Composition Biomarkers for Phenotypic Biologic Aging. Mayo Clin Proc 2024; 99:858-860. [PMID: 38839185 DOI: 10.1016/j.mayocp.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/19/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine & Public Health, Madison, WI.
| |
Collapse
|
47
|
Quint EE, Liu Y, Shafaat O, Ghildayal N, Crosby H, Kamireddy A, Pol RA, Orandi BJ, Segev DL, Weiss CR, McAdams-DeMarco MA. Abdominal computed tomography measurements of body composition and waitlist mortality in kidney transplant candidates. Am J Transplant 2024; 24:591-605. [PMID: 37949413 PMCID: PMC10982050 DOI: 10.1016/j.ajt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/10/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Body mass index is often used to determine kidney transplant (KT) candidacy. However, this measure of body composition (BC) has several limitations, including the inability to accurately capture dry weight. Objective computed tomography (CT)-based measures may improve pre-KT risk stratification and capture physiological aging more accurately. We quantified the association between CT-based BC measurements and waitlist mortality in a retrospective study of 828 KT candidates (2010-2022) with clinically obtained CT scans using adjusted competing risk regression. In total, 42.5% of candidates had myopenia, 11.4% had myopenic obesity (MO), 68.8% had myosteatosis, 24.8% had sarcopenia (probable = 11.2%, confirmed = 10.5%, and severe = 3.1%), and 8.6% had sarcopenic obesity. Myopenia, MO, and sarcopenic obesity were not associated with mortality. Patients with myosteatosis (adjusted subhazard ratio [aSHR] = 1.62, 95% confidence interval [CI]: 1.07-2.45; after confounder adjustment) or sarcopenia (probable: aSHR = 1.78, 95% CI: 1.10-2.88; confirmed: aSHR = 1.68, 95% CI: 1.01-2.82; and severe: aSHR = 2.51, 95% CI: 1.12-5.66; after full adjustment) were at increased risk of mortality. When stratified by age, MO (aSHR = 2.21, 95% CI: 1.28-3.83; P interaction = .005) and myosteatosis (aSHR = 1.95, 95% CI: 1.18-3.21; P interaction = .038) were associated with elevated risk only among candidates <65 years. MO was only associated with waitlist mortality among frail candidates (adjusted hazard ratio = 2.54, 95% CI: 1.28-5.05; P interaction = .021). Transplant centers should consider using BC metrics in addition to body mass index when a CT scan is available to improve pre-KT risk stratification at KT evaluation.
Collapse
Affiliation(s)
- Evelien E Quint
- Division of Transplant Surgery, Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Yi Liu
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Omid Shafaat
- Division of Vascular and Interventional Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nidhi Ghildayal
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Helen Crosby
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Arun Kamireddy
- Division of Vascular and Interventional Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert A Pol
- Division of Transplant Surgery, Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Babak J Orandi
- Division of Endocrinology, Joan & Sanford Weill Medical College of Cornell University, New York, NY, USA
| | - Dorry L Segev
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA; Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA
| | - Clifford R Weiss
- Division of Vascular and Interventional Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mara A McAdams-DeMarco
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA; Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
48
|
Pickhardt PJ. Invited Commentary: Metabolic Syndrome: The Urgent Need for an Imaging-based Definition. Radiographics 2024; 44:e230230. [PMID: 38329902 DOI: 10.1148/rg.230230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Affiliation(s)
- Perry J Pickhardt
- From the Department of Radiology, The University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| |
Collapse
|
49
|
Soares BLDM, Soares NMDM, Burgos MGPDA, de Arruda IKG. Nutritional status and changes in muscle and adipose tissue determined by computed tomography as predictors of mortality in hospitalized patients. Radiol Bras 2024; 57:e20240026. [PMID: 39540014 PMCID: PMC11559939 DOI: 10.1590/0100-3984.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/08/2024] [Accepted: 07/28/2024] [Indexed: 11/16/2024] Open
Abstract
The aim of the present study was to investigate whether nutritional status and changes in muscle and adipose tissue determined by computed tomography are predictors of mortality in hospitalized patients. This was a prospective cohort study involving patients ≥ 20 years of age hospitalized in a public hospital. Sociodemographic and clinical variables were collected from electronic medical records. Nutritional variables were determined. All patients were followed prospectively until the hospital outcome, which could be discharge or death. Body composition was defined from computed tomography images, with the identification of myopenia, myosteatosis, and myopenic obesity. The sample comprised 542 patients. The mortality rate was 10.7% (95% CI: 6.4-15.0%). The independent predictors of mortality were malnutrition, assessed with the subjective global assessment (hazard ratio: 4.18; 95% CI: 1.01-17.22; p = 0.047), and myopenic obesity (hazard ratio: 2.82; 95% CI: 1.11-7.20; p = 0.029). The findings of the present study add to the limited evidence in the literature that body composition is associated with outcomes in hospitalized patients.
Collapse
Affiliation(s)
- Bruna Lúcia de Mendonça Soares
- Graduate Program in Nutrition, Department of Nutrition,
Universidade Federal de Pernambuco (UFPE), Recife, PE, Brazil
- Hospital da Restauração Governador Paulo Guerra,
Recife, PE, Brazil
| | | | | | - Ilma Kruze Grande de Arruda
- Graduate Program in Nutrition, Department of Nutrition,
Universidade Federal de Pernambuco (UFPE), Recife, PE, Brazil
| |
Collapse
|
50
|
Spytek M, Krzyziński M, Langbein SH, Baniecki H, Wright MN, Biecek P. survex: an R package for explaining machine learning survival models. Bioinformatics 2023; 39:btad723. [PMID: 38039146 PMCID: PMC11025379 DOI: 10.1093/bioinformatics/btad723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/10/2023] [Accepted: 11/29/2023] [Indexed: 12/03/2023] Open
Abstract
SUMMARY Due to their flexibility and superior performance, machine learning models frequently complement and outperform traditional statistical survival models. However, their widespread adoption is hindered by a lack of user-friendly tools to explain their internal operations and prediction rationales. To tackle this issue, we introduce the survex R package, which provides a cohesive framework for explaining any survival model by applying explainable artificial intelligence techniques. The capabilities of the proposed software encompass understanding and diagnosing survival models, which can lead to their improvement. By revealing insights into the decision-making process, such as variable effects and importances, survex enables the assessment of model reliability and the detection of biases. Thus, transparency and responsibility may be promoted in sensitive areas, such as biomedical research and healthcare applications. AVAILABILITY AND IMPLEMENTATION survex is available under the GPL3 public license at https://github.com/modeloriented/survex and on CRAN with documentation available at https://modeloriented.github.io/survex.
Collapse
Affiliation(s)
- Mikołaj Spytek
- MI2.AI, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Mateusz Krzyziński
- MI2.AI, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Sophie Hanna Langbein
- Leibniz Institute for Prevention Research and Epidemiology—BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| | - Hubert Baniecki
- MI2.AI, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- MI2.AI, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Marvin N Wright
- Leibniz Institute for Prevention Research and Epidemiology—BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Przemysław Biecek
- MI2.AI, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- MI2.AI, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| |
Collapse
|