Comparison of malignancy‐prediction efficiency between contrast and non‐contract CT‐based radiomics features in gastrointestinal stromal tumors: A multicenter study

This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset fo...

Full description

Saved in:
Bibliographic Details
Published inClinical and translational medicine Vol. 10; no. 3; pp. e291 - n/a
Main Authors Zhang, Qing‐Wei, Zhou, Xiao‐Xuan, Zhang, Ran‐Ying, Chen, Shuang‐Li, Liu, Qiang, Wang, Jian, Zhang, Yan, Lin, Jiang, Xu, Jian‐Rong, Gao, Yun‐Jie, Ge, Zhi‐Zheng
Format Journal Article
LanguageEnglish
Published United States John Wiley and Sons Inc 01.07.2020
Wiley
Subjects
Online AccessGet full text

Cover

Loading…
Abstract This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE‐RS and radiomics signature from contrast‐enhanced CT (CE‐RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE‐RS. The AUC values were comparable between NE‐RS and CE‐RS in the training (.965 vs .936; P = .251), internal validation (.967 vs .960; P = .801), and external validation (.941 vs .899; P = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE‐RS. With 0.185 selected as the cutoff of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high‐malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE‐RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high‐malignancy potential by 5.9% (P = .025), 2.5% (P = .317), 10.5% (P = .008) for the training set, internal validation set, and external validation set, respectively. The NE‐RS had comparable prediction efficiency in the diagnosis of high‐risk GISTs to CE‐RS. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs. A dataset for 370 GIST patients was collected from four centers and was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance (mRMR) algorithm, and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. The area under the curve (AUC) values were comparable between radiomics signatures from non‐enhanced CT (NE‐RS) and radiomics signatures from enhanced CT (CE‐RS) in the three cohorts in diagnosis of high malignancy potential of GISTs. With 0.185 selected as the cut‐off of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy for diagnosis high‐malignancy potential GIST was 89%, 85% for the internal validation and external validation cohort. Compared with only NE‐RS, the radiomics model with combination of radiomic signature and tumor size, had increased accuracy of 91%, 89% for the internal validation and external validation cohort. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
AbstractList Abstract This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE‐RS and radiomics signature from contrast‐enhanced CT (CE‐RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE‐RS. The AUC values were comparable between NE‐RS and CE‐RS in the training (.965 vs .936; P  = .251), internal validation (.967 vs .960; P  = .801), and external validation (.941 vs .899; P  = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE‐RS. With 0.185 selected as the cutoff of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high‐malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE‐RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high‐malignancy potential by 5.9% ( P  = .025), 2.5% ( P  = .317), 10.5% ( P  = .008) for the training set, internal validation set, and external validation set, respectively. The NE‐RS had comparable prediction efficiency in the diagnosis of high‐risk GISTs to CE‐RS. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE-RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast-enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum-redundancy maximum-relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE-RS and radiomics signature from contrast-enhanced CT (CE-RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE-RS. The AUC values were comparable between NE-RS and CE-RS in the training (.965 vs .936; P = .251), internal validation (.967 vs .960; P = .801), and external validation (.941 vs .899; P = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE-RS. With 0.185 selected as the cutoff of NE-RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high-malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE-RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high-malignancy potential by 5.9% (P = .025), 2.5% (P = .317), 10.5% (P = .008) for the training set, internal validation set, and external validation set, respectively. The NE-RS had comparable prediction efficiency in the diagnosis of high-risk GISTs to CE-RS. The NE-RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE‐RS and radiomics signature from contrast‐enhanced CT (CE‐RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE‐RS. The AUC values were comparable between NE‐RS and CE‐RS in the training (.965 vs .936; P  = .251), internal validation (.967 vs .960; P  = .801), and external validation (.941 vs .899; P  = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE‐RS. With 0.185 selected as the cutoff of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high‐malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE‐RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high‐malignancy potential by 5.9% ( P  = .025), 2.5% ( P  = .317), 10.5% ( P  = .008) for the training set, internal validation set, and external validation set, respectively. The NE‐RS had comparable prediction efficiency in the diagnosis of high‐risk GISTs to CE‐RS. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs. A dataset for 370 GIST patients was collected from four centers and was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance (mRMR) algorithm, and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. The area under the curve (AUC) values were comparable between radiomics signatures from non‐enhanced CT (NE‐RS) and radiomics signatures from enhanced CT (CE‐RS) in the three cohorts in diagnosis of high malignancy potential of GISTs. With 0.185 selected as the cut‐off of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy for diagnosis high‐malignancy potential GIST was 89%, 85% for the internal validation and external validation cohort. Compared with only NE‐RS, the radiomics model with combination of radiomic signature and tumor size, had increased accuracy of 91%, 89% for the internal validation and external validation cohort. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE‐RS and radiomics signature from contrast‐enhanced CT (CE‐RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE‐RS. The AUC values were comparable between NE‐RS and CE‐RS in the training (.965 vs .936; P = .251), internal validation (.967 vs .960; P = .801), and external validation (.941 vs .899; P = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE‐RS. With 0.185 selected as the cutoff of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high‐malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE‐RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high‐malignancy potential by 5.9% (P = .025), 2.5% (P = .317), 10.5% (P = .008) for the training set, internal validation set, and external validation set, respectively. The NE‐RS had comparable prediction efficiency in the diagnosis of high‐risk GISTs to CE‐RS. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs. A dataset for 370 GIST patients was collected from four centers and was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance (mRMR) algorithm, and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. The area under the curve (AUC) values were comparable between radiomics signatures from non‐enhanced CT (NE‐RS) and radiomics signatures from enhanced CT (CE‐RS) in the three cohorts in diagnosis of high malignancy potential of GISTs. With 0.185 selected as the cut‐off of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy for diagnosis high‐malignancy potential GIST was 89%, 85% for the internal validation and external validation cohort. Compared with only NE‐RS, the radiomics model with combination of radiomic signature and tumor size, had increased accuracy of 91%, 89% for the internal validation and external validation cohort. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
Abstract This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the malignancy degree of gastrointestinal stromal tumors (GISTs) and the comparison of these signatures with those from contrast‐enhanced CT. A dataset for 370 GIST patients was collected from four centers. This dataset was divided into cohorts for training, as well as internal and external validation. The minimum‐redundancy maximum‐relevance algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm were used to filter unstable features. (a) NE‐RS and radiomics signature from contrast‐enhanced CT (CE‐RS) were built and compared for the prediction of malignancy potential of GIST based on the area under the receiver operating characteristic curve (AUC). (b) The radiomics model was also developed with both the tumor size and NE‐RS. The AUC values were comparable between NE‐RS and CE‐RS in the training (.965 vs .936; P = .251), internal validation (.967 vs .960; P = .801), and external validation (.941 vs .899; P = .173) cohorts in diagnosis of high malignancy potential of GISTs. We next focused on the NE‐RS. With 0.185 selected as the cutoff of NE‐RS for diagnosis of the malignancy potential of GISTs, accuracy, sensitivity, and specificity for diagnosis high‐malignancy potential GIST was 90.0%, 88.2%, and 92.3%, respectively, in the training cohort. For the internal validation set, the corresponding metrics are 89.1%, 94.9%, and 80.0%, respectively. The corresponding metrics for the external cohort are 84.6%, 76.1%, and 91.0%, respectively. Compared with only NE‐RS, the radiomics model increased the sensitivity in the diagnosis of GIST with high‐malignancy potential by 5.9% (P = .025), 2.5% (P = .317), 10.5% (P = .008) for the training set, internal validation set, and external validation set, respectively. The NE‐RS had comparable prediction efficiency in the diagnosis of high‐risk GISTs to CE‐RS. The NE‐RS and radiomics model both had excellent accuracy in predicting malignancy potential of GISTs.
Author Wang, Jian
Zhang, Qing‐Wei
Lin, Jiang
Chen, Shuang‐Li
Liu, Qiang
Zhang, Yan
Zhang, Ran‐Ying
Xu, Jian‐Rong
Zhou, Xiao‐Xuan
Gao, Yun‐Jie
Ge, Zhi‐Zheng
AuthorAffiliation 3 Department of Radiology Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging Fenglin Road 180 Shanghai 200032 China
2 Department of Radiology Sir Run Run Shaw Hospital (SRRSH), School of Medicine, Zhejiang University Hangzhou China
4 Department of Radiology The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
5 Department of Pathology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
6 Department of radiology Tongde Hospital of Zhejiang Province Hangzhou China
7 Department of Radiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
1 Division of Gastroenterology and Hepatology Key Laboratory of Gastroenterology and Hepatology Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease Shanghai China
AuthorAffiliation_xml – name: 3 Department of Radiology Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging Fenglin Road 180 Shanghai 200032 China
– name: 6 Department of radiology Tongde Hospital of Zhejiang Province Hangzhou China
– name: 1 Division of Gastroenterology and Hepatology Key Laboratory of Gastroenterology and Hepatology Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease Shanghai China
– name: 5 Department of Pathology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
– name: 7 Department of Radiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
– name: 2 Department of Radiology Sir Run Run Shaw Hospital (SRRSH), School of Medicine, Zhejiang University Hangzhou China
– name: 4 Department of Radiology The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
Author_xml – sequence: 1
  givenname: Qing‐Wei
  orcidid: 0000-0001-6908-049X
  surname: Zhang
  fullname: Zhang, Qing‐Wei
  organization: Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease
– sequence: 2
  givenname: Xiao‐Xuan
  surname: Zhou
  fullname: Zhou, Xiao‐Xuan
  organization: Sir Run Run Shaw Hospital (SRRSH), School of Medicine, Zhejiang University
– sequence: 3
  givenname: Ran‐Ying
  surname: Zhang
  fullname: Zhang, Ran‐Ying
  organization: Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging
– sequence: 4
  givenname: Shuang‐Li
  surname: Chen
  fullname: Chen, Shuang‐Li
  organization: The First Affiliated Hospital of Wenzhou Medical University
– sequence: 5
  givenname: Qiang
  surname: Liu
  fullname: Liu, Qiang
  organization: Shanghai Jiao Tong University
– sequence: 6
  givenname: Jian
  surname: Wang
  fullname: Wang, Jian
  organization: Tongde Hospital of Zhejiang Province
– sequence: 7
  givenname: Yan
  surname: Zhang
  fullname: Zhang, Yan
  organization: Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease
– sequence: 8
  givenname: Jiang
  surname: Lin
  fullname: Lin, Jiang
  email: lin.jiang@zs-hospital.sh.cn
  organization: Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging
– sequence: 9
  givenname: Jian‐Rong
  surname: Xu
  fullname: Xu, Jian‐Rong
  email: renjixujr@163.com
  organization: Shanghai Jiao Tong University
– sequence: 10
  givenname: Yun‐Jie
  surname: Gao
  fullname: Gao, Yun‐Jie
  email: gao_y_j2003@aliyun.com
  organization: Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease
– sequence: 11
  givenname: Zhi‐Zheng
  orcidid: 0000-0001-5817-5530
  surname: Ge
  fullname: Ge, Zhi‐Zheng
  email: zhizhengge@aliyun.com
  organization: Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease
BackLink https://www.ncbi.nlm.nih.gov/pubmed/32634272$$D View this record in MEDLINE/PubMed
BookMark eNp1ksFu1DAQhiNUREupeAPkG0hoi-3EScyhUhUVqFTEZTlbs_Z4cZXYi-1Q7Y1H4Dn6WDwJXrZU7QFf7Pnnn89ja55XBz54rKqXjJ4ySvk7nSd-KtmT6ohTyhas5u3Bg_NhdZLSNS2rb6Ts-LPqsKh1wzt-VN0OYdpAdCl4EiyZYHRrD15vf__8tYlonM6upNBapx0Wnaww3yB6ooPPEVIm4A0pHZWCvaQzGZYlWkFCQyIYFyanE7EIeY6YiPNkXQpjcD5jys7DSHZhuZzkeQoxvSfnZJrH7DQWSyzZ2WxfVE8tjAlP7vbj6uuHi-XwaXH15ePlcH610KJu2AJr3vW07ZnQgJbqrhGUtxKE0UYaUXd9j7URPa0bK7hGJqhEaVsB0NAO2vq4utxzTYBrtYlugrhVAZz6K4S4VhBLayMqKYWFbqVpi30j0PaWyraV2BohYKV5YZ3tWZt5NaHZPSfC-Aj6OOPdN7UOP1TXsL6nXQG8uQPE8H0uv6UmlzSOI3gMc1K84awRXStpsb7eW3UMKUW099cwqnaDonaDoiQrzlcPu7r3_RuLYni7N9y4Ebf_46hh-ZkX3B9K2tCf
CitedBy_id crossref_primary_10_1259_bjro_20220055
crossref_primary_10_4251_wjgo_v16_i4_1296
crossref_primary_10_1002_mp_17276
crossref_primary_10_1007_s00432_023_05545_w
crossref_primary_10_1186_s12967_023_04520_w
crossref_primary_10_3389_fonc_2022_905036
crossref_primary_10_1007_s10396_023_01373_0
crossref_primary_10_3390_diagnostics12112722
crossref_primary_10_1007_s00432_023_05170_7
crossref_primary_10_1007_s00330_023_10249_3
crossref_primary_10_3389_fonc_2022_966743
crossref_primary_10_1007_s00261_021_03026_7
crossref_primary_10_3389_fonc_2021_737302
crossref_primary_10_1007_s11604_023_01441_y
Cites_doi 10.7150/jca.29443
10.1007/s00330-018-5629-2
10.1093/bioinformatics/btt383
10.1007/s00330-015-4172-7
10.1053/j.semdp.2006.09.001
10.6004/jnccn.2016.0078
10.1016/j.vgie.2019.03.010
10.18637/jss.v033.i01
10.1093/annonc/mdx034
10.1186/s40169-020-0263-4
10.1038/nrclinonc.2017.141
10.1037/0033-2909.86.2.420
10.1111/j.1443-1661.2010.01032.x
10.5946/ce.2015.096
10.1111/den.12149
10.18637/jss.v061.i08
10.1016/j.humpath.2008.06.025
10.1158/0008-5472.CAN-17-0339
10.1007/s10147-008-0798-7
10.1002/cncr.30239
10.1016/j.tranon.2019.06.005
10.1007/s00261-015-0438-4
ContentType Journal Article
Copyright 2020 The Authors. published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics
2020 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.
Copyright_xml – notice: 2020 The Authors. published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics
– notice: 2020 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.
DBID 24P
WIN
NPM
AAYXX
CITATION
7X8
5PM
DOA
DOI 10.1002/ctm2.91
DatabaseName Wiley Online Library Open Access
Wiley Online Library Free Content
PubMed
CrossRef
MEDLINE - Academic
PubMed Central (Full Participant titles)
DOAJ Directory of Open Access Journals
DatabaseTitle PubMed
CrossRef
MEDLINE - Academic
DatabaseTitleList CrossRef
PubMed



Database_xml – sequence: 1
  dbid: DOA
  name: DOAJ Directory of Open Access Journals
  url: https://www.doaj.org/
  sourceTypes: Open Website
– sequence: 2
  dbid: 24P
  name: Wiley Online Library Open Access
  url: https://authorservices.wiley.com/open-science/open-access/browse-journals.html
  sourceTypes: Publisher
– sequence: 3
  dbid: NPM
  name: PubMed
  url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed
  sourceTypes: Index Database
DeliveryMethod fulltext_linktorsrc
Discipline Medicine
DocumentTitleAlternate ZHANG et al
EISSN 2001-1326
EndPage n/a
ExternalDocumentID oai_doaj_org_article_995fa7bc06e845ef8f09669e6d55abc2
10_1002_ctm2_91
32634272
CTM291
Genre shortCommunication
Journal Article
GrantInformation_xml – fundername: Shanghai Municipal Key Clinical Specialty: shslczdzk05902
– fundername: Shanghai Municipal Education Commission: Gaofeng Clinical Medicine grant support: DLY201501
– fundername: Cross medical research fund of translational medicine, Shanghai Jiao Tong University:ZH2018ZDA06
– fundername: Shanghai Municipal Commission of Health and Family Planning:2018YQ29
– fundername: National Natural Science Foundation of China
  funderid: 81670505; 81772519
– fundername: three‐year action plan for Shin Kang of Shanghai: 16CR3113B
– fundername: National Natural Science Foundation of China
  grantid: 81772519
– fundername: three-year action plan for Shin Kang of Shanghai: 16CR3113B
– fundername: National Natural Science Foundation of China
  grantid: 81670505
– fundername: ;
  grantid: 81670505; 81772519
GroupedDBID -A0
0R~
1OC
24P
2VQ
3V.
4.4
53G
5VS
7X7
8AO
8FI
8FJ
AAHHS
AAKKN
AAYZJ
ABDBF
ABEEZ
ABUWG
ACACY
ACCFJ
ACGFS
ACULB
ACXQS
ADBBV
ADINQ
ADRAZ
ADZOD
AEEZP
AEQDE
AFGXO
AFKRA
AHBXF
AHBYD
AHMBA
AHSBF
AHYZX
AIWBW
AJBDE
ALIPV
ALMA_UNASSIGNED_HOLDINGS
ALUQN
AMKLP
AMTXH
AN0
AOIJS
BAPOH
BAWUL
BCNDV
BENPR
BFQNJ
BNQBC
BPHCQ
BVXVI
C24
C6C
CCPQU
DIK
EBS
EJD
FYUFA
GROUPED_DOAJ
GX1
H13
HMCUK
HYE
IAO
KQ8
M48
M~E
OK1
PIMPY
PQQKQ
PROAC
RBZ
RNS
RPM
RSV
SMD
SOJ
UKHRP
WIN
ITC
NPM
AAYXX
CITATION
7X8
5PM
ID FETCH-LOGICAL-c5341-e327806815caef0c7450269a5dcd9d53788e3d58034f52ce1509e9f65aa407a63
IEDL.DBID RPM
ISSN 2001-1326
IngestDate Thu Jul 04 21:08:38 EDT 2024
Tue Sep 17 21:00:49 EDT 2024
Fri Aug 16 21:21:44 EDT 2024
Thu Sep 26 16:29:57 EDT 2024
Sat Sep 28 08:26:12 EDT 2024
Sat Aug 24 01:05:59 EDT 2024
IsDoiOpenAccess true
IsOpenAccess true
IsPeerReviewed true
IsScholarly true
Issue 3
Keywords radiomics signature
prediction
gastrointestinal stromal tumor
malignant potential
Language English
License Attribution
2020 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.
This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
LinkModel DirectLink
MergedId FETCHMERGED-LOGICAL-c5341-e327806815caef0c7450269a5dcd9d53788e3d58034f52ce1509e9f65aa407a63
Notes Qing‐Wei Zhang, Xiao‐Xuan Zhou, Ran‐Ying Zhang, and Shuang‐Li Chen contributed equally to this work.
ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ORCID 0000-0001-5817-5530
0000-0001-6908-049X
OpenAccessLink https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7418807/
PMID 32634272
PQID 2421457690
PQPubID 23479
PageCount 7
ParticipantIDs doaj_primary_oai_doaj_org_article_995fa7bc06e845ef8f09669e6d55abc2
pubmedcentral_primary_oai_pubmedcentral_nih_gov_7418807
proquest_miscellaneous_2421457690
crossref_primary_10_1002_ctm2_91
pubmed_primary_32634272
wiley_primary_10_1002_ctm2_91_CTM291
PublicationCentury 2000
PublicationDate July 2020
PublicationDateYYYYMMDD 2020-07-01
PublicationDate_xml – month: 07
  year: 2020
  text: July 2020
PublicationDecade 2020
PublicationPlace United States
PublicationPlace_xml – name: United States
– name: Hoboken
PublicationTitle Clinical and translational medicine
PublicationTitleAlternate Clin Transl Med
PublicationYear 2020
Publisher John Wiley and Sons Inc
Wiley
Publisher_xml – name: John Wiley and Sons Inc
– name: Wiley
References 2013; 29
2010; 33
2010; 22
2019; 4
2013; 25
2006; 23
2017; 14
2017; 28
2015; 40
2019; 10
2017; 77
2019; 12
2008; 39
2020; 9
2016; 122
2019; 29
2008; 13
1979; 86
2014; 61
2016; 49
2016; 26
2016; 14
e_1_2_9_20_1
e_1_2_9_11_1
e_1_2_9_22_1
e_1_2_9_10_1
e_1_2_9_21_1
e_1_2_9_13_1
e_1_2_9_12_1
e_1_2_9_23_1
e_1_2_9_8_1
Committee AT (e_1_2_9_4_1) 2019; 4
e_1_2_9_7_1
e_1_2_9_6_1
e_1_2_9_5_1
e_1_2_9_2_1
Pang T (e_1_2_9_3_1) 2019; 10
e_1_2_9_9_1
e_1_2_9_15_1
e_1_2_9_14_1
e_1_2_9_17_1
e_1_2_9_16_1
e_1_2_9_19_1
e_1_2_9_18_1
References_xml – volume: 12
  start-page: 1229
  issue: 9
  year: 2019
  end-page: 1236
  article-title: Building CT radiomics‐based models for preoperatively predicting malignant potential and mitotic count of gastrointestinal stromal tumors
  publication-title: Transl Oncol
– volume: 13
  start-page: 416
  issue: 5
  year: 2008
  end-page: 430
  article-title: Clinical practice guidelines for gastrointestinal stromal tumor (GIST) in Japan: English version
  publication-title: Int J Clin Oncol
– volume: 39
  start-page: 1411
  issue: 10
  year: 2008
  end-page: 1419
  article-title: Risk stratification of patients diagnosed with gastrointestinal stromal tumor
  publication-title: Hum Pathol
– volume: 28
  start-page: 1191
  issue: 6
  year: 2017
  end-page: 1206
  article-title: Promises and challenges for the implementation of computational medical imaging (radiomics) in oncology
  publication-title: Ann Oncol
– volume: 9
  start-page: 12
  issue: 1
  year: 2020
  article-title: Personalized CT‐based radiomics nomogram preoperative predicting Ki‐67 expression in gastrointestinal stromal tumors: a multicenter development and validation cohort
  publication-title: Clin Transl Med
– volume: 61
  start-page: 1
  issue: 8
  year: 2014
  end-page: 36
  article-title: OptimalCutpoints: an R package for selecting optimal cutpoints in diagnostic tests
  publication-title: J Stat Softw
– volume: 14
  start-page: 758
  issue: 6
  year: 2016
  end-page: 786
  article-title: Soft tissue sarcoma, version 2.2016, NCCN clinical practice guidelines in oncology
  publication-title: J Natl Compr Canc Netw
– volume: 122
  start-page: 3110
  issue: 20
  year: 2016
  end-page: 3118
  article-title: Diagnostic and treatment strategy for small gastrointestinal stromal tumors
  publication-title: Cancer
– volume: 4
  start-page: 343
  issue: 8
  year: 2019
  end-page: 350
  article-title: ASGE guideline for endoscopic full‐thickness resection and submucosal tunnel endoscopic resection
  publication-title: VideoGIE
– volume: 23
  start-page: 70
  issue: 2
  year: 2006
  end-page: 83
  article-title: Gastrointestinal stromal tumors: pathology and prognosis at different sites
  publication-title: Semin Diagn Pathol
– volume: 25
  start-page: 479
  issue: 5
  year: 2013
  end-page: 489
  article-title: Submucosal tumors: comprehensive guide for the diagnosis and therapy of gastrointestinal submucosal tumors
  publication-title: Dig Endosc
– volume: 10
  start-page: 4132
  issue: 17
  year: 2019
  end-page: 4141
  article-title: Comparison of safety and outcomes between endoscopic and surgical resections of small (≤5 cm) primary gastric gastrointestinal stromal tumors
  publication-title: J Cancer
– volume: 49
  start-page: 235
  issue: 3
  year: 2016
  end-page: 240
  article-title: Current guidelines in the management of upper gastrointestinal subepithelial tumors
  publication-title: Clin Endosc
– volume: 14
  start-page: 749
  issue: 12
  year: 2017
  end-page: 762
  article-title: Radiomics: the bridge between medical imaging and personalized medicine
  publication-title: Nat Rev Clin Oncol
– volume: 29
  start-page: 1074
  issue: 3
  year: 2019
  end-page: 1082
  article-title: Radiomics nomogram for predicting the malignant potential of gastrointestinal stromal tumours preoperatively
  publication-title: Eur Radiol
– volume: 86
  start-page: 420
  issue: 2
  year: 1979
  article-title: Intraclass correlations: uses in assessing rater reliability
  publication-title: Psychol Bull
– volume: 33
  start-page: 1
  issue: 1
  year: 2010
  end-page: 22
  article-title: Regularization paths for generalized linear models via coordinate descent
  publication-title: J Stat Softw
– volume: 40
  start-page: 2331
  issue: 7
  year: 2015
  end-page: 2337
  article-title: CT textural analysis of hepatic metastatic colorectal cancer: pre‐treatment tumor heterogeneity correlates with pathology and clinical outcomes
  publication-title: Abdom Imaging
– volume: 29
  start-page: 2365
  issue: 18
  year: 2013
  end-page: 2368
  article-title: mRMRe: an R package for parallelized mRMR ensemble feature selection
  publication-title: Bioinformatics
– volume: 26
  start-page: 3086
  issue: 9
  year: 2016
  end-page: 3093
  article-title: Predictive features of CT for risk stratifications in patients with primary gastrointestinal stromal tumour
  publication-title: Eur Radiol
– volume: 22
  start-page: 354
  issue: 4
  year: 2010
  end-page: 356
  article-title: Small gastrointestinal stromal tumor of the stomach showing rapid growth and early metastasis to the liver
  publication-title: Dig Endosc
– volume: 77
  start-page: e104
  issue: 21
  year: 2017
  end-page: e107
  article-title: Computational radiomics system to decode the radiographic phenotype
  publication-title: Cancer Res
– volume: 10
  start-page: 4132
  issue: 17
  year: 2019
  ident: e_1_2_9_3_1
  article-title: Comparison of safety and outcomes between endoscopic and surgical resections of small (≤5 cm) primary gastric gastrointestinal stromal tumors
  publication-title: J Cancer
  doi: 10.7150/jca.29443
  contributor:
    fullname: Pang T
– ident: e_1_2_9_11_1
  doi: 10.1007/s00330-018-5629-2
– ident: e_1_2_9_15_1
  doi: 10.1093/bioinformatics/btt383
– ident: e_1_2_9_7_1
  doi: 10.1007/s00330-015-4172-7
– ident: e_1_2_9_6_1
  doi: 10.1053/j.semdp.2006.09.001
– ident: e_1_2_9_18_1
  doi: 10.6004/jnccn.2016.0078
– volume: 4
  start-page: 343
  issue: 8
  year: 2019
  ident: e_1_2_9_4_1
  article-title: ASGE guideline for endoscopic full‐thickness resection and submucosal tunnel endoscopic resection
  publication-title: VideoGIE
  doi: 10.1016/j.vgie.2019.03.010
  contributor:
    fullname: Committee AT
– ident: e_1_2_9_16_1
  doi: 10.18637/jss.v033.i01
– ident: e_1_2_9_9_1
  doi: 10.1093/annonc/mdx034
– ident: e_1_2_9_10_1
  doi: 10.1186/s40169-020-0263-4
– ident: e_1_2_9_8_1
  doi: 10.1038/nrclinonc.2017.141
– ident: e_1_2_9_14_1
  doi: 10.1037/0033-2909.86.2.420
– ident: e_1_2_9_20_1
  doi: 10.1111/j.1443-1661.2010.01032.x
– ident: e_1_2_9_21_1
  doi: 10.5946/ce.2015.096
– ident: e_1_2_9_2_1
  doi: 10.1111/den.12149
– ident: e_1_2_9_17_1
  doi: 10.18637/jss.v061.i08
– ident: e_1_2_9_5_1
  doi: 10.1016/j.humpath.2008.06.025
– ident: e_1_2_9_13_1
  doi: 10.1158/0008-5472.CAN-17-0339
– ident: e_1_2_9_22_1
  doi: 10.1007/s10147-008-0798-7
– ident: e_1_2_9_19_1
  doi: 10.1002/cncr.30239
– ident: e_1_2_9_12_1
  doi: 10.1016/j.tranon.2019.06.005
– ident: e_1_2_9_23_1
  doi: 10.1007/s00261-015-0438-4
SSID ssj0000849972
Score 2.2711368
Snippet This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the...
This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE-RS) to preoperatively predict the...
Abstract This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the...
Abstract This work seeks the development and validation of radiomics signatures from nonenhanced computed tomography (CT, NE‐RS) to preoperatively predict the...
SourceID doaj
pubmedcentral
proquest
crossref
pubmed
wiley
SourceType Open Website
Open Access Repository
Aggregation Database
Index Database
Publisher
StartPage e291
SubjectTerms gastrointestinal stromal tumor
malignant potential
prediction
radiomics signature
Short Communication
SummonAdditionalLinks – databaseName: DOAJ Directory of Open Access Journals
  dbid: DOA
  link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwrV3NbtQwELZQD4gL4p9QQIPUa2h-7DjmVlZUFdJy2kq9RU48LnvYpEqyB248As_Rx-JJmHHS1a4AceEYO47_xp5v4pnPQpx4Qg2kOWyMaGQs66yOjU5dbCQZK9opTBwHOC-_FBeX8vOVutq76ot9wiZ64GngTo1R3uq6SQospUJfegLdhcHCKWXrZtp9U7VnTIU9uJQcETpFyTLL6GkzbrL3Jj1QP4Gl_0_Q8ncPyX3kGlTP-SPxcMaMcDa19bG4h-0TcX85n4o_FbeL3WWC0HnYELS-Zh6Nbz-__7jp-TUefcDAFsGhljB7Z0FwVLfDCLZ10HYtFZiSmhEWK3piJeegt27N0csDeAxEoAOsW7imgn3HfBO0TXAD-ZEqh3G76frhA5xBcFfk3mEPgcj2mbg8_7RaXMTzHQxxo0jBxZhnukyKMlWNRZ80Wiqy2oxVrnHGKWajx9ypMsmlV1mDhC8NGl8oa8lUtEX-XBxR8_GlgMxK5hz1mgC9pK-ZknYT6WQqa0Ql60jA3dRUNxPVRjWRKmcVz15l0kh85CnbZTM3dkggialmian-JTGReHc34RWtJT4gsS1226Hi43FJBphJIvFiEoBdVQRzc5lpKq0PROOgLYc57fpr4OtmgqAy0ZE4CUL0t85Vi9UyM-mr_9HHY_Eg4x8Dwa_4tTga-y2-IfQ01m_DQvkFIaAhOQ
  priority: 102
  providerName: Directory of Open Access Journals
– databaseName: Scholars Portal Open Access Journals
  dbid: M48
  link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwdV1La9wwEBYhhZBL6bvuCxVyVeqVJdsqlJIuDaGwPe1Cbka2RtuFrp3YXkj-T35oZ2TvkqXJ0fJDsmek-caa-YaxE4-oAS2HFQBGCVXKUphs4oRR6KxkTkPsKMF59ju9WKhfl_rygG2rdI4fsHvQtaN6Uov27-nN9e13nPDfRgLRL1W_lqeUwf5EqkSRms9GjB-W41xRcigVmaPwIfS-0iF59v69x-wI2xMlM7lnoAKP_0Pg8_8YyvvYNhin82fs6Ygq-dmgBs_ZAdQv2NFs3Dd_ye6mu3KDvPF8jeB7SUwbt-KqpYtIOhwCmwSlYvIxeouHQHbb9dzWjtdNLYaGqufTuSAD6Hhr3YoymzvuIZCEdnxV8yXe1DbERYFLCA2NDrFb3m_WTdt95Wc8hDLSe0HLA8ntK7Y4_zmfXoixPoOoNBo_AYnM8jjNJ7qy4OMqUxo9OmO1q5xxmpjqIXE6jxPltawAsacB41NtLbqRNk1es0McOrxlXFpFfKQ-Q7Cv8Gkmx5VGOTVRJYBWZcT4VijF1UDDUQyEy7IgERZmErEfJKzdaeLNDg1NuyzGaVgYo73NyipOIVcafO7RhUsNpE5rW1YyYp-3oi5wntHmia2h2XQFbZ0rdM5MHLE3g-h3XW1VJ2LZnlLsjWX_TL36E7i8iTwoj7OInQT1eezliul8Js3k3aNdv2fHkv4EhEDiD-ywbzfwEeFSX34K0-EfwPIVrA
  priority: 102
  providerName: Scholars Portal
– databaseName: Wiley Online Library Open Access
  dbid: 24P
  link: http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV3NjtQwDI7QIiEuiH_Kn4y017KdNGkbbsuI1QppEIdZaW9V2jjDHKZdtZ0DNx6B5-CxeBLstFt2BEgcm8ZtItuxndhfhDj25DWQ5bAxolGxqmQVm3zhYqMoWMmdxsRxgfPqU3Z-oT5e6ssbV32N-BDzhhtrRlivWcFt1Z_8Bg2th518y3Xrt8mpKVigpfo8b68kheKSUL5ajpOGKObKxpJZpj6ZaA9sUYDs_5uf-We65E03Ntihs_vi3uRAwunI8QfiFjYPxZ3VdET-SPxYzjcLQuthR372hkE1vv789v2q427MCsAAHcF1lzClakHIWrf9ALZx0LQNEYxN9QDLNT2xxXPQWbflUuYePAZU0B62DWyIsGsZfILWDB4gP9LPYdjv2q5_B6cQchd5dthBQLV9LC7OPqyX5_F0IUNca7J2MaYyL5KsWOjaok_qXGkK4YzVrnbGaYamx9TpIkmV17JGcjYNGp9paylutFn6RBzR8PGZAGkVA5D6nLx7RV8zBS0tyqmFqhC1qiIB16wpr0bcjXJEWJYlc680i0i8Z5bNrxkoOzS03aac9K40RnubV3WSYaE0-sJTzJYZzJzWtqplJN5cM7wkxeLTEttgu-9LPitXFI2ZJBJPRwGYf0XSlCqZE3V-IBoHYzl802y_BPBuRgsqkjwSx0GI_jW5crleSbN4_n_dXoi7kvcBQhrxS3E0dHt8Rc7SUL0OavELWYwYwA
  priority: 102
  providerName: Wiley-Blackwell
Title Comparison of malignancy‐prediction efficiency between contrast and non‐contract CT‐based radiomics features in gastrointestinal stromal tumors: A multicenter study
URI https://onlinelibrary.wiley.com/doi/abs/10.1002%2Fctm2.91
https://www.ncbi.nlm.nih.gov/pubmed/32634272
https://search.proquest.com/docview/2421457690
https://pubmed.ncbi.nlm.nih.gov/PMC7418807
https://doaj.org/article/995fa7bc06e845ef8f09669e6d55abc2
Volume 10
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV3NbpwwELayOVS9VP0v_Vm5Uq7sssYG3FuKGkWVqHLYSLkhY5vtSgVWwB5y6yP0OfJYeZLOGFhl1fbSC5L581gzeL7BM58JOSsBNYDnUL61kvu8YIUv45XxJYdgJTbCBgYLnLNv0eU1_3ojbk6ImGphXNK-LraL-ke1qLffXW7lrtLLKU9seZWlyLiSBPFyRmZxGD4I0d30m3AsBh0KZJFgdKn7ii0k7gkDWCXkLGZHTshx9f8NYP6ZJ_kQvzoHdPGUPBmRIz0fJHxGTmz9nDzKxrXxF-QuPWwpSJuSVgCwN8imcXv_89euxdtQB9Q6zggsuKRjjhZ16eqq66mqDa2bGh4YTumepmtooasztFVmizXMHS2towPt6LamG3iwbZB1AiYLFBCb0Dnt91XTdp_oOXVJizg621JHZ_uSXF98WaeX_rgTg68FuDnfhixOgihZCa1sGeiYC4jdpBJGG2kEctLb0IgkCHkpmLaAMqWVZSSUgoBRReErcgri2zeEMsWRebSMAdZzeJtMYE7hhq94Ya3ghUfopJp8NxBu5AO1MstRkblceeQzquxwGRmy3Ymm3eSjneRSilLFhQ4im3Bhy6SEYC2SNjJCqEIzj3ycFJ7DF4XLJKq2zb7LcZGcQxgmA4-8Hgzg0NVkQB6Jj0zjSJbjK2DEjrV7NFqPnDkj-tfg8nSdMbl6-98dvCOPGf4TcCnF78lp3-7tBwBOfTEnM8av5u63Axwznszdp_MbsNkjqQ
link.rule.ids 230,315,733,786,790,870,891,2115,2236,11589,24346,27957,27958,46087,46511,53827,53829
linkProvider National Library of Medicine
linkToHtml http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV3NjtQwDI7QIgEXxD_lN0h7LdtJk7bhtoxYDbCz4jAr7S1KG2eYw7SrtnPgxiPwHDwWT4KddgsjQOLYJG4SOY7txP7C2KFHqwE1h40BtIxlKcpY5zMXa4nOSu4UJI4SnJdn2eJcfrhQF2NUJeXCDPgQ04EbSUbYr0nA6UD66BdqaNVvxWtKXL9O73mTUAr5aTpfSQpJOaH0thxFDaHTlQ05s0R9NNLuKaOA2f83Q_PPeMnf7digiE7usNujBcmPB5bfZdegvsduLMc78vvs-3x6WpA3nm_R0F4TqsaXH1-_XbbUjHjBIWBHUOIlH2O1eAhbt13Pbe143dRIMBRVPZ-v8ItUnuOtdRvKZe64hwAL2vFNzddI2DaEPoGbBg2QPrFz3u-2Tdu94cc8BC_S7KDlAdb2ATs_ebeaL-LxRYa4UqjuYkhFXiRZMVOVBZ9UuVTow2mrXOW0U4RND6lTRZJKr0QFaG1q0D5T1qLjaLP0ITvA4cNjxoWVhEDqczTvJf5NF7i3SCdnsgRQsowYv2KNuRyAN8wAsSwMcc_oWcTeEsumakLKDgVNuzaj4Bmtlbd5WSUZFFKBLzw6bZmGzClly0pE7NUVww1KFl2X2BqaXWfoslyiO6aTiD0aFsDUFa6mVIocqfO9pbE3lv2aevM5oHcTXFCR5BE7DIvoX5Mz89VS6NmT_2v2kt1crJan5vT92cen7JagQ4EQU_yMHfTtDp6j5dSXL4KI_ARGnxwh
linkToPdf http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV3NbtQwELZQkSouiH_Cr5F6DU0cO4m5lYVV-dmqh63UW-TE42UPm6yS7IEbj8Bz8Fg8CTNOGroCJI6xPbGt8Xhm7JnPjB05tBpQc5gQQMtQlqIMdRbbUEt0VjKrILKU4Lw4S08v5MdLdXntqa8BH2I6cCPJ8Ps1CfjWuuPfoKFVvxGvKW_9pkyjnPwuIc-n45Uol5QSSk_LUdAQ-lzpkDJL1Mcj7Z4u8pD9f7Mz_wyXvG7Gej00v8NujwYkPxk4fpfdgPoeO1yMV-T32Y_Z9LIgbxzfoJ29IlCNrz-_fd-21IxYwcFDR1DeJR9DtbiPWjddz01ted3USDAUVT2fLfGLNJ7lrbFrSmXuuAOPCtrxdc1XSNg2BD6BewYNkD6xc97vNk3bveEn3Mcu0uyg5R7V9gG7mL9fzk7D8UGGsFKo7UJIRJZHaR6ryoCLqkwqdOG0Ubay2iqCpofEqjxKpFOiAjQ2NWiXKmPQbzRp8pAd4PDhMePCSAIgdRla9xL_pnPcWqSVsSwBlCwDxq9YU2wH3I1iQFgWBXGv0HHA3hLLpmoCyvYFTbsqRrkrtFbOZGUVpZBLBS536LOlGlKrlCkrEbBXVwwvULDotsTU0Oy6gu7KJXpjOgrYo2EBTF3hakqkyJA621sae2PZr6nXXzx4N6EF5VEWsCO_iP41uWK2XAgdP_m_Zi_Z4fm7efH5w9mnp-yWoCMBH1H8jB307Q6eo93Uly-8hPwClHwbUw
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Comparison+of+malignancy-prediction+efficiency+between+contrast+and+non-contract+CT-based+radiomics+features+in+gastrointestinal+stromal+tumors%3A+A+multicenter+study&rft.jtitle=Clinical+and+translational+medicine&rft.au=Zhang%2C+Qing-Wei&rft.au=Zhou%2C+Xiao-Xuan&rft.au=Zhang%2C+Ran-Ying&rft.au=Chen%2C+Shuang-Li&rft.date=2020-07-01&rft.issn=2001-1326&rft.eissn=2001-1326&rft.volume=10&rft.issue=3&rft.spage=e291&rft_id=info:doi/10.1002%2Fctm2.91&rft_id=info%3Apmid%2F32634272&rft.externalDocID=32634272
thumbnail_l http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=2001-1326&client=summon
thumbnail_m http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=2001-1326&client=summon
thumbnail_s http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=2001-1326&client=summon