1
|
Ma J, Nie X, Kong X, Xiao L, Liu H, Shi S, Wu Y, Li N, Hu L, Li X. MRI T2WI-based radiomics combined with KRAS gene mutation constructed models for predicting liver metastasis in rectal cancer. BMC Med Imaging 2024; 24:262. [PMID: 39367333 PMCID: PMC11453062 DOI: 10.1186/s12880-024-01439-6] [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: 05/31/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND The study aimed to identify the optimal model for predicting rectal cancer liver metastasis (RCLM). This involved constructing various prediction models to aid clinicians in early diagnosis and precise decision-making. METHODS A retrospective analysis was conducted on 193 patients diagnosed with rectal adenocarcinoma were randomly divided into training set (n = 136) and validation set (n = 57) at a ratio of 7:3. The predictive performance of three models was internally validated by 10-fold cross-validation in the training set. Delineation of the tumor region of interest (ROI) was performed, followed by the extraction of radiomics features from the ROI. The least absolute shrinkage and selection operator (LASSO) regression algorithm and multivariate Cox analysis were employed to reduce the dimensionality of radiomics features and identify significant features. Logistic regression was employed to construct three prediction models: clinical, radiomics, and combined models (radiomics + clinical). The predictive performance of each model was assessed and compared. RESULTS KRAS mutation emerged as an independent predictor of liver metastasis, yielding an odds ratio (OR) of 8.296 (95%CI: 3.471-19.830; p < 0.001). 5 radiomics features will be used to construct radiomics model. The combined model was built by integrating radiomics model with clinical model. In both the training set (AUC:0.842, 95%CI: 0.778-0.907) and the validation set (AUC: 0.805; 95%CI: 0.692-0.918), the AUCs for the combined model surpassed those of the radiomics and clinical models. CONCLUSIONS Our study reveals that KRAS mutation stands as an independent predictor of RCLM. The radiomics features based on MR play a crucial role in the evaluation of RCLM. The combined model exhibits superior performance in the prediction of liver metastasis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jiaqi Ma
- Department of Magnetic Resonance Imaging Diagnostic, The 2nd Affiliated Hospital of Harbin Medical University, Baojian Road, Nangang District, Harbin, 150086, China
| | - Xinsheng Nie
- Medical Imaging Center, the Xinjiang Production and Construction Corps Tenth Division Beitun Hospital, Beitun, 836099, China
| | - Xiangjiang Kong
- Medical Imaging Center, the Xinjiang Production and Construction Corps Tenth Division Beitun Hospital, Beitun, 836099, China
| | - Lingqing Xiao
- Medical Imaging Center, the Xinjiang Production and Construction Corps Tenth Division Beitun Hospital, Beitun, 836099, China
| | - Han Liu
- Department of Magnetic Resonance Imaging Diagnostic, The 2nd Affiliated Hospital of Harbin Medical University, Baojian Road, Nangang District, Harbin, 150086, China
| | - Shengming Shi
- Department of Magnetic Resonance Imaging Diagnostic, The 2nd Affiliated Hospital of Harbin Medical University, Baojian Road, Nangang District, Harbin, 150086, China
| | - Yupeng Wu
- Department of Magnetic Resonance Imaging Diagnostic, The 2nd Affiliated Hospital of Harbin Medical University, Baojian Road, Nangang District, Harbin, 150086, China
| | - Na Li
- Medical Imaging Center, the Xinjiang Production and Construction Corps Tenth Division Beitun Hospital, Beitun, 836099, China
| | - Linlin Hu
- Medical Imaging Center, the Xinjiang Production and Construction Corps Tenth Division Beitun Hospital, Beitun, 836099, China
| | - Xiaofu Li
- Department of Magnetic Resonance Imaging Diagnostic, The 2nd Affiliated Hospital of Harbin Medical University, Baojian Road, Nangang District, Harbin, 150086, China.
| |
Collapse
|
2
|
Li M, Yuan Y, Zhou H, Feng F, Xu G. A multicenter study: predicting KRAS mutation and prognosis in colorectal cancer through a CT-based radiomics nomogram. Abdom Radiol (NY) 2024; 49:1816-1828. [PMID: 38393357 DOI: 10.1007/s00261-024-04218-7] [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: 11/23/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE To establish a CT-based radiomics nomogram for preoperative prediction of KRAS mutation and prognostic stratification in colorectal cancer (CRC) patients. METHODS In a retrospective analysis, 408 patients with confirmed CRC were included, comprising 168 cases in the training set, 111 cases in the internal validation set, and 129 cases in the external validation set. Radiomics features extracted from the primary tumors were meticulously screened to identify those closely associated with KRAS mutation. Subsequently, a radiomics nomogram was constructed by integrating these radiomics features with clinically significant parameters. The diagnostic performance was assessed through the area under the receiver operating characteristic curve (AUC). Lastly, the prognostic significance of the nomogram was explored, and Kaplan-Meier analysis was employed to depict survival curves for the high-risk and low-risk groups. RESULTS A radiomics model was constructed using 19 radiomics features significantly associated with KRAS mutation. Furthermore, a nomogram was developed by integrating these radiomics features with two clinically significant parameters (age, tumor location). The nomogram achieved AUCs of 0.834, 0.813, and 0.811 in the training set, internal validation set, and external validation set, respectively. Additionally, the nomogram effectively stratified patients into high-risk (KRAS mutation) and low-risk (KRAS wild-type) groups, demonstrating a significant difference in overall survival (P < 0.001). Patients categorized in the high-risk group exhibited inferior overall survival in contrast to those classified in the low-risk group. CONCLUSIONS The CT-based radiomics nomogram demonstrates the capability to effectively predict KRAS mutation in CRC patients and stratify their prognosis preoperatively.
Collapse
Affiliation(s)
- Manman Li
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Yiwen Yuan
- Department of Translational Medical Center, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Hui Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Guodong Xu
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
3
|
Granata V, Fusco R, De Muzio F, Brunese MC, Setola SV, Ottaiano A, Cardone C, Avallone A, Patrone R, Pradella S, Miele V, Tatangelo F, Cutolo C, Maggialetti N, Caruso D, Izzo F, Petrillo A. Radiomics and machine learning analysis by computed tomography and magnetic resonance imaging in colorectal liver metastases prognostic assessment. LA RADIOLOGIA MEDICA 2023; 128:1310-1332. [PMID: 37697033 DOI: 10.1007/s11547-023-01710-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE The aim of this study was the evaluation radiomics analysis efficacy performed using computed tomography (CT) and magnetic resonance imaging in the prediction of colorectal liver metastases patterns linked to patient prognosis: tumor growth front; grade; tumor budding; mucinous type. Moreover, the prediction of liver recurrence was also evaluated. METHODS The retrospective study included an internal and validation dataset; the first was composed by 119 liver metastases from 49 patients while the second consisted to 28 patients with single lesion. Radiomic features were extracted using PyRadiomics. Univariate and multivariate approaches including machine learning algorithms were employed. RESULTS The best predictor to identify tumor growth was the Wavelet_HLH_glcm_MaximumProbability with an accuracy of 84% and to detect recurrence the best predictor was wavelet_HLH_ngtdm_Complexity with an accuracy of 90%, both extracted by T1-weigthed arterial phase sequence. The best predictor to detect tumor budding was the wavelet_LLH_glcm_Imc1 with an accuracy of 88% and to identify mucinous type was wavelet_LLH_glcm_JointEntropy with an accuracy of 92%, both calculated on T2-weigthed sequence. An increase statistically significant of accuracy (90%) was obtained using a linear weighted combination of 15 predictors extracted by T2-weigthed images to detect tumor front growth. An increase statistically significant of accuracy at 93% was obtained using a linear weighted combination of 11 predictors by the T1-weigthed arterial phase sequence to classify tumor budding. An increase statistically significant of accuracy at 97% was obtained using a linear weighted combination of 16 predictors extracted on CT to detect recurrence. An increase statistically significant of accuracy was obtained in the tumor budding identification considering a K-nearest neighbors and the 11 significant features extracted T1-weigthed arterial phase sequence. CONCLUSIONS The results confirmed the Radiomics capacity to recognize clinical and histopathological prognostic features that should influence the choice of treatments in colorectal liver metastases patients to obtain a more personalized therapy.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy.
| | | | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Maria Chiara Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Alessandro Ottaiano
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Claudia Cardone
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Antonio Avallone
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Renato Patrone
- Division of Hepatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Silvia Pradella
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
- SIRM Foundation, Italian Society of Medical and Interventional Radiology (SIRM), 20122, Milan, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
- SIRM Foundation, Italian Society of Medical and Interventional Radiology (SIRM), 20122, Milan, Italy
| | - Fabiana Tatangelo
- Division of Pathological Anatomy and Cytopathology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084, Salerno, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Radiology Unit-Sant'Andrea University Hospital, Sapienza-University of Rome, 00189, Rome, Italy
| | - Francesco Izzo
- Division of Hepatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| |
Collapse
|
4
|
Mendes Serrão E, Klug M, Moloney BM, Jhaveri A, Lo Gullo R, Pinker K, Luker G, Haider MA, Shinagare AB, Liu X. Current Status of Cancer Genomics and Imaging Phenotypes: What Radiologists Need to Know. Radiol Imaging Cancer 2023; 5:e220153. [PMID: 37921555 DOI: 10.1148/rycan.220153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology. Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
- Eva Mendes Serrão
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Maximiliano Klug
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Brian M Moloney
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Aaditeya Jhaveri
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Roberto Lo Gullo
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Katja Pinker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Gary Luker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Masoom A Haider
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Atul B Shinagare
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Xiaoyang Liu
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| |
Collapse
|
5
|
Tan Q, Wang Q, Jin S, Zhou F, Zou X. Network Evolution Model-based prediction of tumor mutation burden from radiomic-clinical features in endometrial cancers. BMC Cancer 2023; 23:712. [PMID: 37525139 PMCID: PMC10388464 DOI: 10.1186/s12885-023-11118-4] [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: 03/13/2023] [Accepted: 06/27/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Endometrial Cancer (EC) is one of the most prevalent malignancies that affect the female population globally. In the context of immunotherapy, Tumor Mutation Burden (TMB) in the DNA polymerase epsilon (POLE) subtype of this cancer holds promise as a viable therapeutic target. METHODS We devised a method known as NEM-TIE to forecast the TMB status of patients with endometrial cancer. This approach utilized a combination of the Network Evolution Model, Transfer Information Entropy, Clique Percolation (CP) methodology, and Support Vector Machine (SVM) classification. To construct the Network Evolution Model, we employed an adjacency matrix that utilized transfer information entropy to assess the information gain between nodes of radiomic-clinical features. Subsequently, using the CP algorithm, we unearthed potentially pivotal modules in the Network Evolution Model. Finally, the SVM classifier extracted essential features from the module set. RESULTS Upon analyzing the importance of modules, we discovered that the dependence count energy in tumor volumes-of-interest holds immense significance in distinguishing TMB statuses among patients with endometrial cancer. Using the 13 radiomic-clinical features extracted via NEM-TIE, we demonstrated that the area under the receiver operating characteristic curve (AUROC) in the test set is 0.98 (95% confidence interval: 0.95-1.00), surpassing the performance of existing techniques such as the mRMR and Laplacian methods. CONCLUSIONS Our study proposed the NEM-TIE method as a means to identify the TMB status of patients with endometrial cancer. The integration of radiomic-clinical data utilizing the NEM-TIE method may offer a novel technology for supplementary diagnosis.
Collapse
Affiliation(s)
- Qing Tan
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, 430072, China
| | - Qian Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China
| | - Suoqin Jin
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, 430072, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China.
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430072, China.
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
6
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Patrone R, Ottaiano A, Nasti G, Silvestro L, Cassata A, Grassi F, Avallone A, Izzo F, Petrillo A. Colorectal liver metastases patients prognostic assessment: prospects and limits of radiomics and radiogenomics. Infect Agent Cancer 2023; 18:18. [PMID: 36927442 PMCID: PMC10018963 DOI: 10.1186/s13027-023-00495-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
In this narrative review, we reported un up-to-date on the role of radiomics to assess prognostic features, which can impact on the liver metastases patient treatment choice. In the liver metastases patients, the possibility to assess mutational status (RAS or MSI), the tumor growth pattern and the histological subtype (NOS or mucinous) allows a better treatment selection to avoid unnecessary therapies. However, today, the detection of these features require an invasive approach. Recently, radiomics analysis application has improved rapidly, with a consequent growing interest in the oncological field. Radiomics analysis allows the textural characteristics assessment, which are correlated to biological data. This approach is captivating since it should allow to extract biological data from the radiological images, without invasive approach, so that to reduce costs and time, avoiding any risk for the patients. Several studies showed the ability of Radiomics to identify mutational status, tumor growth pattern and histological type in colorectal liver metastases. Although, radiomics analysis in a non-invasive and repeatable way, however features as the poor standardization and generalization of clinical studies results limit the translation of this analysis into clinical practice. Clear limits are data-quality control, reproducibility, repeatability, generalizability of results, and issues related to model overfitting.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy.
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, Napoli, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, Milan, 20122, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari "Aldo Moro", Bari, 70124, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Alessandro Ottaiano
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Guglielmo Nasti
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Lucrezia Silvestro
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Antonio Cassata
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesca Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, 80138, Italy
| | - Antonio Avallone
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| |
Collapse
|
7
|
Crimì F, Zanon C, Cabrelle G, Luong KD, Albertoni L, Bao QR, Borsetto M, Baratella E, Capelli G, Spolverato G, Fassan M, Pucciarelli S, Quaia E. Contrast-Enhanced CT Texture Analysis in Colon Cancer: Correlation with Genetic Markers. Tomography 2022; 8:2193-2201. [PMID: 36136880 PMCID: PMC9498512 DOI: 10.3390/tomography8050184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The purpose of the study was to determine whether contrast-enhanced CT texture features relate to, and can predict, the presence of specific genetic mutations involved in CRC carcinogenesis. Materials and methods: This retrospective study analyzed the pre-operative CT in the venous phase of patients with CRC, who underwent testing for mutations in the KRAS, NRAS, BRAF, and MSI genes. Using a specific software based on CT images of each patient, for each slice including the tumor a region of interest was manually drawn along the margin, obtaining the volume of interest. A total of 56 texture parameters were extracted that were compared between the wild-type gene group and the mutated gene group. A p-value of <0.05 was considered statistically significant. Results: The study included 47 patients with stage III-IV CRC. Statistically significant differences between the MSS group and the MSI group were found in four parameters: GLRLM RLNU (area under the curve (AUC) 0.72, sensitivity (SE) 77.8%, specificity (SP) 65.8%), GLZLM SZHGE (AUC 0.79, SE 88.9%, SP 65.8%), GLZLM GLNU (AUC 0.74, SE 88.9%, SP 60.5%), and GLZLM ZLNU (AUC 0.77, SE 88.9%, SP 65.8%). Conclusions: The findings support the potential role of the CT texture analysis in detecting MSI in CRC based on pre-treatment CT scans.
Collapse
Affiliation(s)
- Filippo Crimì
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
| | - Chiara Zanon
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
| | - Giulio Cabrelle
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
| | - Kim Duyen Luong
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
| | - Laura Albertoni
- Pathology and Cytopathology Unit, Department of Medicine, University of Padova, 35128 Padua, Italy
| | - Quoc Riccardo Bao
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy
| | - Marta Borsetto
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy
| | - Elisa Baratella
- Department of Radiology, Cattinara Hospital, University of Trieste, 34127 Trieste, Italy
| | - Giulia Capelli
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy
| | - Matteo Fassan
- Pathology and Cytopathology Unit, Department of Medicine, University of Padova, 35128 Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgical, Oncological, and Gastroenterological Sciences (DiSCOG), University of Padova, 35128 Padova, Italy
| | - Emilio Quaia
- Institute of Radiology, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy
| |
Collapse
|
8
|
Zhang G, Chen L, Liu A, Pan X, Shu J, Han Y, Huan Y, Zhang J. Comparable Performance of Deep Learning-Based to Manual-Based Tumor Segmentation in KRAS/NRAS/BRAF Mutation Prediction With MR-Based Radiomics in Rectal Cancer. Front Oncol 2021; 11:696706. [PMID: 34395262 PMCID: PMC8358773 DOI: 10.3389/fonc.2021.696706] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022] Open
Abstract
Radiomic features extracted from segmented tumor regions have shown great power in gene mutation prediction, while deep learning–based (DL-based) segmentation helps to address the inherent limitations of manual segmentation. We therefore investigated whether deep learning–based segmentation is feasible in predicting KRAS/NRAS/BRAF mutations of rectal cancer using MR-based radiomics. In this study, we proposed DL-based segmentation models with 3D V-net architecture. One hundred and eight patients’ images (T2WI and DWI) were collected for training, and another 94 patients’ images were collected for validation. We evaluated the DL-based segmentation manner and compared it with the manual-based segmentation manner through comparing the gene prediction performance of six radiomics-based models on the test set. The performance of the DL-based segmentation was evaluated by Dice coefficients, which are 0.878 ± 0.214 and 0.955 ± 0.055 for T2WI and DWI, respectively. The performance of the radiomics-based model in gene prediction based on DL-segmented VOI was evaluated by AUCs (0.714 for T2WI, 0.816 for DWI, and 0.887 for T2WI+DWI), which were comparable to that of corresponding manual-based VOI (0.637 for T2WI, P=0.188; 0.872 for DWI, P=0.181; and 0.906 for T2WI+DWI, P=0.676). The results showed that 3D V-Net architecture could conduct reliable rectal cancer segmentation on T2WI and DWI images. All-relevant radiomics-based models presented similar performances in KRAS/NRAS/BRAF prediction between the two segmentation manners.
Collapse
Affiliation(s)
- Guangwen Zhang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Chen
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Aie Liu
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xianpan Pan
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Jun Shu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ye Han
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Huan
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinsong Zhang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Katabathina VS, Marji H, Khanna L, Ramani N, Yedururi S, Dasyam A, Menias CO, Prasad SR. Decoding Genes: Current Update on Radiogenomics of Select Abdominal Malignancies. Radiographics 2021; 40:1600-1626. [PMID: 33001791 DOI: 10.1148/rg.2020200042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Technologic advances in chromosomal analysis and DNA sequencing have enabled genome-wide analysis of cancer cells, yielding considerable data on the genetic basis of malignancies. Evolving knowledge of tumor genetics and oncologic pathways has led to a better understanding of histopathologic features, tumor classification, tumor biologic characteristics, and imaging findings and discovery of targeted therapeutic agents. Radiogenomics is a rapidly evolving field of imaging research aimed at correlating imaging features with gene mutations and gene expression patterns, and it may provide surrogate imaging biomarkers that may supplant genetic tests and be used to predict treatment response and prognosis and guide personalized treatment options. Multidetector CT, multiparametric MRI, and PET with use of multiple radiotracers are some of the imaging techniques commonly used to assess radiogenomic associations. Select abdominal malignancies demonstrate characteristic imaging features that correspond to gene mutations. Recent advances have enabled us to understand the genetics of steatotic and nonsteatotic hepatocellular adenomas, a plethora of morphologic-molecular subtypes of hepatic malignancies, a variety of clear cell and non-clear cell renal cell carcinomas, a myriad of hereditary and sporadic exocrine and neuroendocrine tumors of the pancreas, and the development of targeted therapeutic agents for gastrointestinal stromal tumors based on characteristic KIT gene mutations. Mutations associated with aggressive phenotypes of these malignancies can sometimes be predicted on the basis of their imaging characteristics. Radiologists should be familiar with the genetics and pathogenesis of common cancers that have associated imaging biomarkers, which can help them be integral members of the cancer management team and guide clinicians and pathologists. Online supplemental material is available for this article. ©RSNA, 2020 See discussion on this article by Luna (pp 1627-1630).
Collapse
Affiliation(s)
- Venkata S Katabathina
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Haneen Marji
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Lokesh Khanna
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Nisha Ramani
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Sireesha Yedururi
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Anil Dasyam
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Christine O Menias
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| | - Srinivasa R Prasad
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., H.M., L.K.); Departments of Radiology (S.Y., S.R.P.) and Pathology (N.R.), University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.D.); and Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M.)
| |
Collapse
|
10
|
Du B, Wang S, Cui Y, Liu G, Li X, Li Y. Can 18F-FDG PET/CT predict EGFR status in patients with non-small cell lung cancer? A systematic review and meta-analysis. BMJ Open 2021; 11:e044313. [PMID: 34103313 PMCID: PMC8190055 DOI: 10.1136/bmjopen-2020-044313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES This study aimed to explore the diagnostic significance of 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/CT for predicting the presence of epidermal growth factor receptor (EGFR) mutations in patients with non-small cell lung cancer (NSCLC). DESIGN A systematic review and meta-analysis. DATA SOURCES The PubMed, EMBASE and Cochrane library databases were searched from the earliest available date to December 2020. ELIGIBILITY CRITERIA FOR SELECTING STUDIES The review included primary studies that compared the mean maximum of standard uptake value (SUVmax) between wild-type and mutant EGFR, and evaluated the diagnostic value of 18F-FDG PET/CT using SUVmax for prediction of EGFR status in patients with NSCLC. DATA EXTRACTION AND SYNTHESIS The main analysis was to assess the sensitivity and specificity, the positive diagnostic likelihood ratio (DLR+) and DLR-, as well as the diagnostic OR (DOR) of SUVmax in prediction of EGFR mutations. Each data point of the summary receiver operator characteristic (SROC) graph was derived from a separate study. A random effects model was used for statistical analysis of the data, and then diagnostic performance for prediction was further assessed. RESULTS Across 15 studies (3574 patients), the pooled sensitivity for 18F-FDG PET/CT was 0.70 (95% CI 0.60 to 0.79) with a pooled specificity of 0.59 (95% CI 0.52 to 0.66). The overall DLR+ was 1.74 (95% CI 1.49 to 2.03) and DLR- was 0.50 (95% CI 0.38 to 0.65). The pooled DOR was 3.50 (95% CI 2.37 to 5.17). The area under the SROC curve was 0.68 (95% CI 0.64 to 0.72). The likelihood ratio scatter plot based on average sensitivity and specificity was in the lower right quadrant. CONCLUSION Meta-analysis results showed 18F-FDG PET/CT had low pooled sensitivity and specificity. The low DOR and the likelihood ratio scatter plot indicated that 18F-FDG PET/CT should be used with caution when predicting EGFR mutations in patients with NSCLC.
Collapse
Affiliation(s)
- Bulin Du
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Shu Wang
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yan Cui
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Guanghui Liu
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xuena Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Zhang Z, Shen L, Wang Y, Wang J, Zhang H, Xia F, Wan J, Zhang Z. MRI Radiomics Signature as a Potential Biomarker for Predicting KRAS Status in Locally Advanced Rectal Cancer Patients. Front Oncol 2021; 11:614052. [PMID: 34026605 PMCID: PMC8138318 DOI: 10.3389/fonc.2021.614052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/18/2021] [Indexed: 12/21/2022] Open
Abstract
Background and Purpose Locally advanced rectal cancer (LARC) is a heterogeneous disease with little information about KRAS status and image features. The purpose of this study was to analyze the association between T2 magnetic resonance imaging (MRI) radiomics features and KRAS status in LARC patients. Material and Methods Eighty-three patients with KRAS status information and T2 MRI images between 2012.05 and 2019.09 were included. Least absolute shrinkage and selection operator (LASSO) regression was performed to assess the associations between features and gene status. The patients were divided 7:3 into training and validation sets. The C-index and the average area under the receiver operator characteristic curve (AUC) were used for performance evaluation. Results The clinical characteristics of 83 patients in the KRAS mutant and wild-type cohorts were balanced. Forty-two (50.6%) patients had KRAS mutations, and 41 (49.4%) patients had wild-type KRAS. A total of 253 radiomics features were extracted from the T2-MRI images of LARC patients. One radiomic feature named X.LL_scaled_std, a standard deviation value of scaled wavelet-transformed low-pass channel filter, was selected from 253 features (P=0.019). The radiomics-based C-index values were 0.801 (95% CI: 0.772-0.830) and 0.703 (95% CI: 0.620-0.786) in the training and validation sets, respectively. Conclusion Radiomics features could differentiate KRAS status in LARC patients based on T2-MRI images. Further validation in a larger dataset is necessary in the future.
Collapse
Affiliation(s)
- ZhiYuan Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - LiJun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Jiazhou Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - JueFeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| |
Collapse
|
12
|
Damilakis E, Mavroudis D, Sfakianaki M, Souglakos J. Immunotherapy in Metastatic Colorectal Cancer: Could the Latest Developments Hold the Key to Improving Patient Survival? Cancers (Basel) 2020; 12:E889. [PMID: 32268531 PMCID: PMC7225960 DOI: 10.3390/cancers12040889] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has considerably increased the number of anticancer agents in many tumor types including metastatic colorectal cancer (mCRC). Anti-PD-1 (programmed death 1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint inhibitors (ICI) have been shown to benefit the mCRC patients with mismatch repair deficiency (dMMR) or high microsatellite instability (MSI-H). However, ICI is not effective in mismatch repair proficient (pMMR) colorectal tumors, which constitute a large population of patients. Several clinical trials evaluating the efficacy of immunotherapy combined with chemotherapy, radiation therapy, or other agents are currently ongoing to extend the benefit of immunotherapy to pMMR mCRC cases. In dMMR patients, MSI testing through immunohistochemistry and/or polymerase chain reaction can be used to identify patients that will benefit from immunotherapy. Next-generation sequencing has the ability to detect MSI-H using a low amount of nucleic acids and its application in clinical practice is currently being explored. Preliminary data suggest that radiomics is capable of discriminating MSI from microsatellite stable mCRC and may play a role as an imaging biomarker in the future. Tumor mutational burden, neoantigen burden, tumor-infiltrating lymphocytes, immunoscore, and gastrointestinal microbiome are promising biomarkers that require further investigation and validation.
Collapse
Affiliation(s)
- Emmanouil Damilakis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
| | - Dimitrios Mavroudis
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - John Souglakos
- Department of Medical Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece; (D.M.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| |
Collapse
|
13
|
Liu Y, Carpenter AB, Pirozzi CJ, Yuan H, Waitkus MS, Zhou Z, Hansen L, Seywald M, Odion R, Greer PK, Hawk T, Chin BB, Vaidyanathan G, Zalutsky MR, Yan H, Vo-Dinh T. Non-invasive sensitive brain tumor detection using dual-modality bioimaging nanoprobe. NANOTECHNOLOGY 2019; 30:275101. [PMID: 30856613 PMCID: PMC6948110 DOI: 10.1088/1361-6528/ab0e9c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Despite decades of efforts, non-invasive sensitive detection of small malignant brain tumors still remains challenging. Here we report a dual-modality 124I-labeled gold nanostar (124I-GNS) probe for sensitive brain tumor imaging with positron emission tomography (PET) and subcellular tracking with two-photon photoluminescence (TPL) and electron microscopy (EM). Experiment results showed that the developed nanoprobe has potential to reach sub-millimeter intracranial brain tumor detection using PET scan, which is superior to any currently available non-invasive imaging modality. Microscopic examination using TPL and EM further confirmed that GNS nanoparticles permeated the brain tumor leaky vasculature and accumulated inside brain tumor cells following systemic administration. Selective brain tumor targeting by enhanced permeability and retention effect and ultrasensitive imaging render 124I-GNS nanoprobe promise for future brain tumor-related preclinical and translational applications.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | - Hsiangkuo Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Matthew S. Waitkus
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Landon Hansen
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Michelle Seywald
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ren Odion
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Paula K. Greer
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Thomas Hawk
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Bennett B. Chin
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | | | - Michael R. Zalutsky
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Hai Yan
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Tuan Vo-Dinh
- Department of Chemistry, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Fitzpatrick Institute for Photonics, Duke University, Durham, NC, USA
| |
Collapse
|