1
|
Yuan P, Huang ZH, Yang YH, Bao FC, Sun K, Chao FF, Liu TT, Zhang JJ, Xu JM, Li XN, Li F, Ma T, Li H, Li ZH, Zhang SF, Hu J, Qi Y. A 18F-FDG PET/CT-based deep learning-radiomics-clinical model for prediction of cervical lymph node metastasis in esophageal squamous cell carcinoma. Cancer Imaging 2024; 24:153. [PMID: 39533388 PMCID: PMC11556142 DOI: 10.1186/s40644-024-00799-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND To develop an artificial intelligence (AI)-based model using Radiomics, deep learning (DL) features extracted from 18F-fluorodeoxyglucose (18F-FDG) Positron emission tomography/Computed Tomography (PET/CT) images of tumor and cervical lymph node with clinical feature for predicting cervical lymph node metastasis (CLNM) in patients with esophageal squamous cell carcinoma (ESCC). METHODS The study included 300 ESCC patients from the First Affiliated Hospital of Zhengzhou University who were divided into a training cohort and an internal testing cohort with an 8:2 ratio. Another 111 patients from Shanghai Chest Hospital were included as the external cohort. For each sample, we extracted 428 PET/CT-based Radiomics features from the gross tumor volume (GTV) and cervical lymph node (CLN) delineated layer by layer and 256 PET/CT-based DL features from the maximum cross-section of GTV and CLN images We input these features into seven different machine learning algorithms and ultimately selected logistic regression (LR) as the model classifier. Subsequently, we evaluated seven models (Clinical, Radiomics, Radiomics-Clinical, DL-Clinical, DL-Radiomics, DL-Radiomics-Clinical) using Radiomics features, DL features and clinical feature. RESULTS The DL-Radiomics-Clinical (DRC) model demonstrated higher AUC of 0.955 and 0.916 compared to the other six models in both internal and external testing cohorts respectively. The DRC model achieved the highest accuracy among the seven models in both the internal and external test sets, with scores of 0.951 and 0.892, respectively. CONCLUSIONS Through the combination of Radiomics features and DL features from PET/CT imaging and clinical feature, we developed a predictive model exhibiting exceptional classification capabilities. This model can be considered as a non-invasive method for predication of CLNM in patients with ESCC. It might facilitate decision-making regarding to the extend of lymph node dissection, and to select candidates for postoperative adjuvant therapy.
Collapse
Affiliation(s)
- Ping Yuan
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China.
| | - Zhen-Hao Huang
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Yun-Hai Yang
- Surgical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fei-Chao Bao
- Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Sun
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Fang-Fang Chao
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Ting-Ting Liu
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Jing-Jing Zhang
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Jin-Ming Xu
- Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang province, China
| | - Xiang-Nan Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Feng Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Tao Ma
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Hao Li
- School of Artiffcial Intelligence, Sun Yat-sen University, Zhuhai, Guangdong province, China
| | - Zi-Hao Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Shan-Feng Zhang
- School of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan province, China
| | - Jian Hu
- Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang province, China.
| | - Yu Qi
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China.
| |
Collapse
|
2
|
O'Shea R, Withey SJ, Owczarczyk K, Rookyard C, Gossage J, Godfrey E, Jobling C, Parsons SL, Skipworth RJE, Goh V. Multicentre validation of CT grey-level co-occurrence matrix features for overall survival in primary oesophageal adenocarcinoma. Eur Radiol 2024; 34:6919-6928. [PMID: 38526750 PMCID: PMC11399295 DOI: 10.1007/s00330-024-10666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Personalising management of primary oesophageal adenocarcinoma requires better risk stratification. Lack of independent validation of proposed imaging biomarkers has hampered clinical translation. We aimed to prospectively validate previously identified prognostic grey-level co-occurrence matrix (GLCM) CT features for 3-year overall survival. METHODS Following ethical approval, clinical and contrast-enhanced CT data were acquired from participants from five institutions. Data from three institutions were used for training and two for testing. Survival classifiers were modelled on prespecified variables ('Clinical' model: age, clinical T-stage, clinical N-stage; 'ClinVol' model: clinical features + CT tumour volume; 'ClinRad' model: ClinVol features + GLCM_Correlation and GLCM_Contrast). To reflect current clinical practice, baseline stage was also modelled as a univariate predictor ('Stage'). Discrimination was assessed by area under the receiver operating curve (AUC) analysis; calibration by Brier scores; and clinical relevance by thresholding risk scores to achieve 90% sensitivity for 3-year mortality. RESULTS A total of 162 participants were included (144 male; median 67 years [IQR 59, 72]; training, 95 participants; testing, 67 participants). Median survival was 998 days [IQR 486, 1594]. The ClinRad model yielded the greatest test discrimination (AUC, 0.68 [95% CI 0.54, 0.81]) that outperformed Stage (ΔAUC, 0.12 [95% CI 0.01, 0.23]; p = .04). The Clinical and ClinVol models yielded comparable test discrimination (AUC, 0.66 [95% CI 0.51, 0.80] vs. 0.65 [95% CI 0.50, 0.79]; p > .05). Test sensitivity of 90% was achieved by ClinRad and Stage models only. CONCLUSIONS Compared to Stage, multivariable models of prespecified clinical and radiomic variables yielded improved prediction of 3-year overall survival. CLINICAL RELEVANCE STATEMENT Previously identified radiomic features are prognostic but may not substantially improve risk stratification on their own. KEY POINTS • Better risk stratification is needed in primary oesophageal cancer to personalise management. • Previously identified CT features-GLCM_Correlation and GLCM_Contrast-contain incremental prognostic information to age and clinical stage. • Compared to staging, multivariable clinicoradiomic models improve discrimination of 3-year overall survival.
Collapse
Affiliation(s)
- Robert O'Shea
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Samuel J Withey
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Radiology, Royal Marsden Hospital NHS Trust, Sutton, Surrey, UK
| | - Kasia Owczarczyk
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Clinical Oncology, Guy's & St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Christopher Rookyard
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - James Gossage
- Department of Surgery, Guy's & St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Edmund Godfrey
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Craig Jobling
- Department of Radiology, Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - Simon L Parsons
- Department of Surgery, Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | | | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
- Department of Radiology, Guy's & St Thomas' Hospitals NHS Foundation Trust, Westminster Bridge Road, London, SE1 7EG, UK.
| |
Collapse
|
3
|
Garbarino GM, Polici M, Caruso D, Laghi A, Mercantini P, Pilozzi E, van Berge Henegouwen MI, Gisbertz SS, van Grieken NCT, Berardi E, Costa G. Radiomics in Oesogastric Cancer: Staging and Prediction of Preoperative Treatment Response: A Narrative Review and the Results of Personal Experience. Cancers (Basel) 2024; 16:2664. [PMID: 39123392 PMCID: PMC11311587 DOI: 10.3390/cancers16152664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Oesophageal, gastroesophageal, and gastric malignancies are often diagnosed at locally advanced stage and multimodal therapy is recommended to increase the chances of survival. However, given the significant variation in treatment response, there is a clear imperative to refine patient stratification. The aim of this narrative review was to explore the existing evidence and the potential of radiomics to improve staging and prediction of treatment response of oesogastric cancers. METHODS The references for this review article were identified via MEDLINE (PubMed) and Scopus searches with the terms "radiomics", "texture analysis", "oesophageal cancer", "gastroesophageal junction cancer", "oesophagogastric junction cancer", "gastric cancer", "stomach cancer", "staging", and "treatment response" until May 2024. RESULTS Radiomics proved to be effective in improving disease staging and prediction of treatment response for both oesophageal and gastric cancer with all imaging modalities (TC, MRI, and 18F-FDG PET/CT). The literature data on the application of radiomics to gastroesophageal junction cancer are very scarce. Radiomics models perform better when integrating different imaging modalities compared to a single radiology method and when combining clinical to radiomics features compared to only a radiomics signature. CONCLUSIONS Radiomics shows potential in noninvasive staging and predicting response to preoperative therapy among patients with locally advanced oesogastric cancer. As a future perspective, the incorporation of molecular subgroup analysis to clinical and radiomic features may even increase the effectiveness of these predictive and prognostic models.
Collapse
Affiliation(s)
| | - Michela Polici
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Paolo Mercantini
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Mark I. van Berge Henegouwen
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Suzanne S. Gisbertz
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Nicole C. T. van Grieken
- Department of Pathology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Eva Berardi
- Department of Radiology, San Camillo Hospital, ASL RM 1, 00152 Rome, Italy
| | - Gianluca Costa
- Department of Life Science, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
4
|
Fan L, Yang Z, Chang M, Chen Z, Wen Q. CT-based delta-radiomics nomogram to predict pathological complete response after neoadjuvant chemoradiotherapy in esophageal squamous cell carcinoma patients. J Transl Med 2024; 22:579. [PMID: 38890720 PMCID: PMC11186275 DOI: 10.1186/s12967-024-05392-4] [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: 07/21/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND This study developed a nomogram model using CT-based delta-radiomics features and clinical factors to predict pathological complete response (pCR) in esophageal squamous cell carcinoma (ESCC) patients receiving neoadjuvant chemoradiotherapy (nCRT). METHODS The study retrospectively analyzed 232 ESCC patients who underwent pretreatment and post-treatment CT scans. Patients were divided into training (n = 186) and validation (n = 46) sets through fivefold cross-validation. 837 radiomics features were extracted from regions of interest (ROIs) delineations on CT images before and after nCRT to calculate delta values. The LASSO algorithm selected delta-radiomics features (DRF) based on classification performance. Logistic regression constructed a nomogram incorporating DRFs and clinical factors. Receiver operating characteristic (ROC) and area under the curve (AUC) analyses evaluated nomogram performance for predicting pCR. RESULTS No significant differences existed between the training and validation datasets. The 4-feature delta-radiomics signature (DRS) demonstrated good predictive accuracy for pCR, with α-binormal-based and empirical AUCs of 0.871 and 0.869. T-stage (p = 0.001) and differentiation degree (p = 0.018) were independent predictors of pCR. The nomogram combined the DRS and clinical factors improved the classification performance in the training dataset (AUCαbin = 0.933 and AUCemp = 0.941). The validation set showed similar performance with AUCs of 0.958 and 0.962. CONCLUSIONS The CT-based delta-radiomics nomogram model with clinical factors provided high predictive accuracy for pCR in ESCC patients after nCRT.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Minghui Chang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Zheng Chen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Qiang Wen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
5
|
Fiorino C, Palumbo D, Mori M, Palazzo G, Pellegrini AE, Albarello L, Belardo A, Canevari C, Cossu A, Damascelli A, Elmore U, Mazza E, Pavarini M, Passoni P, Puccetti F, Slim N, Steidler S, Del Vecchio A, Di Muzio NG, Chiti A, Rosati R, De Cobelli F. Early regression index (ERI) on MR images as response predictor in esophageal cancer treated with neoadjuvant chemo-radiotherapy: Interim analysis of the prospective ESCAPE trial. Radiother Oncol 2024; 194:110160. [PMID: 38369025 DOI: 10.1016/j.radonc.2024.110160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE The early regression index (ERI) predicts treatment response in rectal cancer patients. Aim of current study was to prospectively assess tumor response to neoadjuvant chemo-radiotherapy (nCRT) of locally advanced esophageal cancer using ERI, based on MRI. MATERIAL AND METHODS From January 2020 to May 2023, 30 patients with esophageal cancer were enrolled in a prospective study (ESCAPE). PET-MRI was performed: i) before nCRT (tpre); ii) at mid-radiotherapy, tmid; iii) after nCRT, 2-6 weeks before surgery (tpost); nCRT delivered 41.4 Gy/23fr with concurrent carboplatin and paclitaxel. For patients that skipped surgery, complete clinical response (cCR) was assessed if patients showed no local relapse after 18 months; patients with pathological complete response (pCR) or with cCR were considered as complete responders (pCR + cCR). GTV volumes were delineated by two observers (Vpre, Vmid, Vpost) on T2w MRI: ERI and other volume regression parameters at tmid and tpost were tested as predictors of pCR + cCR. RESULTS Complete data of 25 patients were available at the time of the analysis: 3/25 with complete response at imaging refused surgery and 2/3 were cCR; in total, 10/25 patients showed pCR + cCR (pCR = 8/22). Both ERImid and ERIpost classified pCR + cCR patients, with ERImid showing better performance (AUC:0.78, p = 0.014): A two-variable logistic model combining ERImid and Vpre improved performances (AUC:0.93, p < 0.0001). Inter-observer variability in contouring GTV did not affect the results. CONCLUSIONS Despite the limited numbers, interim analysis of ESCAPE study suggests ERI as a potential predictor of complete response after nCRT for esophageal cancer. Further validation on larger populations is warranted.
Collapse
Affiliation(s)
- C Fiorino
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy.
| | - D Palumbo
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - M Mori
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - G Palazzo
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | | | - L Albarello
- Pathology, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Belardo
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - C Canevari
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Cossu
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Damascelli
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - U Elmore
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - E Mazza
- Oncology, IRCCS San Raffaele Hospital, Milano, Italy
| | - M Pavarini
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - P Passoni
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy
| | - F Puccetti
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - N Slim
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy
| | - S Steidler
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Del Vecchio
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - N G Di Muzio
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - A Chiti
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - R Rosati
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - F De Cobelli
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| |
Collapse
|
6
|
Kawahara D, Murakami Y, Awane S, Emoto Y, Iwashita K, Kubota H, Sasaki R, Nagata Y. Radiomics and dosiomics for predicting complete response to definitive chemoradiotherapy patients with oesophageal squamous cell cancer using the hybrid institution model. Eur Radiol 2024; 34:1200-1209. [PMID: 37589902 DOI: 10.1007/s00330-023-10020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/08/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVES To develop a multi-institutional prediction model to estimate the local response to oesophageal squamous cell carcinoma (ESCC) treated with definitive radiotherapy based on radiomics and dosiomics features. METHODS The local responses were categorised into two groups (incomplete and complete). An external validation model and a hybrid model that the patients from two institutions were mixed randomly were proposed. The ESCC patients at stages I-IV who underwent chemoradiotherapy from 2012 to 2017 and had follow-up duration of more than 5 years were included. The patients who received palliative or pre-operable radiotherapy and had no FDG PET images were excluded. The segmentations included the GTV, CTV, and PTV which are used in treatment planning. In addition, shrinkage, expansion, and shell regions were created. Radiomic and dosiomic features were extracted from CT, FDG PET images, and dose distribution. Machine learning-based prediction models were developed using decision tree, support vector machine, k-nearest neighbour (kNN) algorithm, and neural network (NN) classifiers. RESULTS A total of 116 and 26 patients enrolled at Centre 1 and Centre 2, respectively. The external validation model exhibited the highest accuracy with 65.4% for CT-based radiomics, 77.9% for PET-based radiomics, and 72.1% for dosiomics based on the NN classifiers. The hybrid model exhibited the highest accuracy of 84.4% for CT-based radiomics based on the kNN classifier, 86.0% for PET-based radiomics, and 79.0% for dosiomics based on the NN classifiers. CONCLUSION The proposed hybrid model exhibited promising predictive performance for the local response to definitive radiotherapy in ESCC patients. CLINICAL RELEVANCE STATEMENT The prediction of the complete response for oesophageal cancer patients may contribute to improving overall survival. The hybrid model has the potential to improve prediction performance than the external validation model that was conventionally proposed. KEY POINTS • Radiomics and dosiomics used to predict response in patients with oesophageal cancer receiving definitive radiotherapy. • Hybrid model with neural network classifier of PET-based radiomics improved prediction accuracy by 8.1%. • The hybrid model has the potential to improve prediction performance.
Collapse
Affiliation(s)
- Daisuke Kawahara
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan.
| | - Yuji Murakami
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Shota Awane
- School of Medicine, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Yuki Emoto
- Department of Radiation Oncology, Hyogo Cancer Center, 70, Kitaoji-Cho 13, Akashi-Shi, Hyogo, Japan
| | - Kazuma Iwashita
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Hikaru Kubota
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
- Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima, 732-0057, Japan
| |
Collapse
|
7
|
Yang Z, Guan F, Bronk L, Zhao L. Multi-omics approaches for biomarker discovery in predicting the response of esophageal cancer to neoadjuvant therapy: A multidimensional perspective. Pharmacol Ther 2024; 254:108591. [PMID: 38286161 DOI: 10.1016/j.pharmthera.2024.108591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/02/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery has been established as the standard treatment strategy for operable locally advanced esophageal cancer (EC). However, achieving pathologic complete response (pCR) or near pCR to NCRT is significantly associated with a considerable improvement in survival outcomes, while pCR patients may help organ preservation for patients by active surveillance to avoid planned surgery. Thus, there is an urgent need for improved biomarkers to predict EC chemoradiation response in research and clinical settings. Advances in multiple high-throughput technologies such as next-generation sequencing have facilitated the discovery of novel predictive biomarkers, specifically based on multi-omics data, including genomic/transcriptomic sequencings and proteomic/metabolomic mass spectra. The application of multi-omics data has shown the benefits in improving the understanding of underlying mechanisms of NCRT sensitivity/resistance in EC. Particularly, the prominent development of artificial intelligence (AI) has introduced a new direction in cancer research. The integration of multi-omics data has significantly advanced our knowledge of the disease and enabled the identification of valuable biomarkers for predicting treatment response from diverse dimension levels, especially with rapid advances in biotechnological and AI methodologies. Herein, we summarize the current status of research on the use of multi-omics technologies in predicting NCRT response for EC patients. Current limitations, challenges, and future perspectives of these multi-omics platforms will be addressed to assist in experimental designs and clinical use for further integrated analysis.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China
| | - Fada Guan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Lawrence Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China.
| |
Collapse
|
8
|
Liu Y, Wang Y, Wang X, Xue L, Zhang H, Ma Z, Deng H, Yang Z, Sun X, Men Y, Ye F, Men K, Qin J, Bi N, Wang Q, Hui Z. MR radiomics predicts pathological complete response of esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy: a multicenter study. Cancer Imaging 2024; 24:16. [PMID: 38263134 PMCID: PMC10804642 DOI: 10.1186/s40644-024-00659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND More than 40% of patients with resectable esophageal squamous cell cancer (ESCC) achieve pathological complete response (pCR) after neoadjuvant chemoradiotherapy (nCRT), who have favorable prognosis and may benefit from an organ-preservation strategy. Our study aims to develop and validate a machine learning model based on MR radiomics to accurately predict the pCR of ESCC patients after nCRT. METHODS In this retrospective multicenter study, eligible patients with ESCC who underwent baseline MR (T2-weighted imaging) and nCRT plus surgery were enrolled between September 2014 and September 2022 at institution 1 (training set) and between December 2017 and August 2021 at institution 2 (testing set). Models were constructed using machine learning algorithms based on clinical factors and MR radiomics to predict pCR after nCRT. The area under the curve (AUC) and cutoff analysis were used to evaluate model performance. RESULTS A total of 155 patients were enrolled in this study, 82 in the training set and 73 in the testing set. The radiomics model was constructed based on two radiomics features, achieving AUCs of 0.968 (95%CI 0.933-0.992) in the training set and 0.885 (95%CI 0.800-0.958) in the testing set. The cutoff analysis resulted in an accuracy of 82.2% (95%CI 72.6-90.4%), a sensitivity of 75.0% (95%CI 58.3-91.7%), and a specificity of 85.7% (95%CI 75.5-96.0%) in the testing set. CONCLUSION A machine learning model based on MR radiomics was developed and validated to accurately predict pCR after nCRT in patients with ESCC.
Collapse
Affiliation(s)
- Yunsong Liu
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Yi Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China
| | - Xin Wang
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Huan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China
| | - Zeliang Ma
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Heping Deng
- Department of Diagnostic Radiology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, China
| | - Zhaoyang Yang
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Xujie Sun
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Yu Men
- Department of VIP Medical Services & Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, 100021, China
| | - Feng Ye
- Department of Diagnostic Radiology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Kuo Men
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China.
| | - Zhouguang Hui
- Department of VIP Medical Services & Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
9
|
Chen C, Wang J, Pan D, Wang X, Xu Y, Yan J, Wang L, Yang X, Yang M, Liu G. Applications of multi-omics analysis in human diseases. MedComm (Beijing) 2023; 4:e315. [PMID: 37533767 PMCID: PMC10390758 DOI: 10.1002/mco2.315] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 08/04/2023] Open
Abstract
Multi-omics usually refers to the crossover application of multiple high-throughput screening technologies represented by genomics, transcriptomics, single-cell transcriptomics, proteomics and metabolomics, spatial transcriptomics, and so on, which play a great role in promoting the study of human diseases. Most of the current reviews focus on describing the development of multi-omics technologies, data integration, and application to a particular disease; however, few of them provide a comprehensive and systematic introduction of multi-omics. This review outlines the existing technical categories of multi-omics, cautions for experimental design, focuses on the integrated analysis methods of multi-omics, especially the approach of machine learning and deep learning in multi-omics data integration and the corresponding tools, and the application of multi-omics in medical researches (e.g., cancer, neurodegenerative diseases, aging, and drug target discovery) as well as the corresponding open-source analysis tools and databases, and finally, discusses the challenges and future directions of multi-omics integration and application in precision medicine. With the development of high-throughput technologies and data integration algorithms, as important directions of multi-omics for future disease research, single-cell multi-omics and spatial multi-omics also provided a detailed introduction. This review will provide important guidance for researchers, especially who are just entering into multi-omics medical research.
Collapse
Affiliation(s)
- Chongyang Chen
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
- Co‐innovation Center of NeurodegenerationNantong UniversityNantongChina
| | - Jing Wang
- Shenzhen Key Laboratory of Modern ToxicologyShenzhen Medical Key Discipline of Health Toxicology (2020–2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Donghui Pan
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Xinyu Wang
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Yuping Xu
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Junjie Yan
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Lizhen Wang
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern ToxicologyShenzhen Medical Key Discipline of Health Toxicology (2020–2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Min Yang
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Gong‐Ping Liu
- Co‐innovation Center of NeurodegenerationNantong UniversityNantongChina
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
10
|
Yang Z, Gong J, Li J, Sun H, Pan Y, Zhao L. The gap before real clinical application of imaging-based machine-learning and radiomic models for chemoradiation outcome prediction in esophageal cancer: a systematic review and meta-analysis. Int J Surg 2023; 109:2451-2466. [PMID: 37463039 PMCID: PMC10442126 DOI: 10.1097/js9.0000000000000441] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/01/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Due to tumoral heterogeneity and the lack of robust biomarkers, the prediction of chemoradiotherapy response and prognosis in patients with esophageal cancer (EC) is challenging. The goal of this study was to assess the study quality and clinical value of machine learning and radiomic-based quantitative imaging studies for predicting the outcomes of EC patients after chemoradiotherapy. MATERIALS AND METHODS PubMed, Embase, and Cochrane were searched for eligible articles. The methodological quality and risk of bias were evaluated using the Radiomics Quality Score (RQS), Image Biomarkers Standardization Initiative (IBSI) Guideline, and Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD) statement, as well as the modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. A meta-analysis of the evidence focusing on predicting chemoradiotherapy response and outcome in EC patients was implemented. RESULTS Forty-six studies were eligible for qualitative synthesis. The mean RQS score was 9.07, with an adherence rate of 42.52%. The adherence rates of the TRIPOD and IBSI were 61.70 and 43.17%, respectively. Ultimately, 24 studies were included in the meta-analysis, of which 16 studies had a pooled sensitivity, specificity, and area under the curve (AUC) of 0.83 (0.76-0.89), 0.83 (0.79-0.86), and 0.84 (0.81-0.87) in neoadjuvant chemoradiotherapy datasets, as well as 0.84 (0.75-0.93), 0.89 (0.83-0.93), and 0.93 (0.90-0.95) in definitive chemoradiotherapy datasets, respectively. Moreover, radiomics could distinguish patients from the low-risk and high-risk groups with different disease-free survival (DFS) (pooled hazard ratio: 3.43, 95% CI 2.39-4.92) and overall survival (pooled hazard ratio: 2.49, 95% CI 1.91-3.25). The results of subgroup and regression analyses showed that some of the heterogeneity was explained by the combination with clinical factors, sample size, and usage of the deep learning (DL) signature. CONCLUSIONS Noninvasive radiomics offers promising potential for optimizing treatment decision-making in EC patients. However, it is necessary to make scientific advancements in EC radiomics regarding reproducibility, clinical usefulness analysis, and open science categories. Improved model reporting of study objectives, blind assessment, and image processing steps are required to help promote real clinical applications of radiomics in EC research.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital
| | - Jie Gong
- Department of Radiation Oncology, Xijing Hospital
| | - Jie Li
- Department of Radiation Oncology, Xijing Hospital
| | - Hongfei Sun
- Department of Radiation Oncology, Xijing Hospital
| | - Yanglin Pan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, People’s Republic of China
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital
| |
Collapse
|
11
|
Menon N, Guidozzi N, Chidambaram S, Markar SR. Performance of radiomics-based artificial intelligence systems in the diagnosis and prediction of treatment response and survival in esophageal cancer: a systematic review and meta-analysis of diagnostic accuracy. Dis Esophagus 2023; 36:doad034. [PMID: 37236811 PMCID: PMC10789236 DOI: 10.1093/dote/doad034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/04/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Radiomics can interpret radiological images with more detail and in less time compared to the human eye. Some challenges in managing esophageal cancer can be addressed by incorporating radiomics into image interpretation, treatment planning, and predicting response and survival. This systematic review and meta-analysis provides a summary of the evidence of radiomics in esophageal cancer. The systematic review was carried out using Pubmed, MEDLINE, and Ovid EMBASE databases-articles describing radiomics in esophageal cancer were included. A meta-analysis was also performed; 50 studies were included. For the assessment of treatment response using 18F-FDG PET/computed tomography (CT) scans, seven studies (443 patients) were included in the meta-analysis. The pooled sensitivity and specificity were 86.5% (81.1-90.6) and 87.1% (78.0-92.8). For the assessment of treatment response using CT scans, five studies (625 patients) were included in the meta-analysis, with a pooled sensitivity and specificity of 86.7% (81.4-90.7) and 76.1% (69.9-81.4). The remaining 37 studies formed the qualitative review, discussing radiomics in diagnosis, radiotherapy planning, and survival prediction. This review explores the wide-ranging possibilities of radiomics in esophageal cancer management. The sensitivities of 18F-FDG PET/CT scans and CT scans are comparable, but 18F-FDG PET/CT scans have improved specificity for AI-based prediction of treatment response. Models integrating clinical and radiomic features facilitate diagnosis and survival prediction. More research is required into comparing models and conducting large-scale studies to build a robust evidence base.
Collapse
Affiliation(s)
- Nainika Menon
- Department of General Surgery, Oxford University Hospitals, Oxford, UK
| | - Nadia Guidozzi
- Department of General Surgery, University of Witwatersrand, Johannesburg, South Africa
| | - Swathikan Chidambaram
- Academic Surgical Unit, Department of Surgery and Cancer, Imperial College London, St Mary’s Hospital, London, UK
| | - Sheraz Rehan Markar
- Department of General Surgery, Oxford University Hospitals, Oxford, UK
- Nuffield Department of Surgery, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Mirshahvalad SA, Seyedinia SS, Huemer F, Schweighofer-Zwink G, Koch O, Hitzl W, Weiss L, Emannuel K, Greil R, Pirich C, Beheshti M. Prognostic value of [ 18F]FDG PET/CT on treatment response and progression-free survival of gastroesophageal cancer patients undergoing perioperative FLOT chemotherapy. Eur J Radiol 2023; 163:110843. [PMID: 37119707 DOI: 10.1016/j.ejrad.2023.110843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/17/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE To evaluate the prognostic role of [18F]FDG PET/CT metabolic parameters in gastric cancer (GC) and gastroesophageal adenocarcinoma (GEJAC) patients receiving neoadjuvant chemotherapy. METHOD In this retrospective study, 31 patients with biopsy-proven GC or GEJAC were included between August 2016 and March 2020. [18F]FDG PET/CT was performed before the neoadjuvant chemotherapy. Primary tumours' semi-quantitative metabolic parameters were extracted. All patients received a perioperative FLOT regimen thereafter. Post-chemotherapy [18F]FDG PET/CT was performed in most patients (17/31). All patients underwent surgical resection. Histopathology response to treatment and progression-free survival (PFS) were evaluated. Two-sided p-values < 0.05 were considered statistically significant. RESULTS Thirty-one patients (mean age = 62 ± 8), including 21 GC and 10 GEJAC patients, were evaluated. 20/31(65%) patients were histopathology responders to neoadjuvant chemotherapy, including twelve complete and eight partial responders. During the median follow-up of 42.0 months, nine patients experienced recurrence. The median PFS was 60(95% CI:32.9-87.1) months. Pre-neoadjuvant chemotherapy SULpeak was significantly correlated with pathological response to treatment (p-value = 0.03;odds ratio = 16.75). In survival analysis, SUVmax (p-value = 0.01;hazard ratio[HR] = 1.55), SUVmean (p-value = 0.04;HR = 2.73), SULpeak (p-value < 0.001;HR = 1.91) and SULmean (p-value = 0.04;HR = 4.22) in the post-neoadjuvant chemotherapy pre-operative [18F]FDG PET/CT showed significant correlation with PFS. Additionally, aspects of staging were significantly correlated with PFS (p-value = 0.01;HR = 2.21). CONCLUSIONS Pre-neoadjuvant chemotherapy [18F]FDG PET/CT parameters, especially SULpeak, could predict the pathological response to treatment in GC and GEJAC patients. Additionally, in survival analysis, post-chemotherapy metabolic parameters significantly correlated with PFS. Thus, performing [18F]FDG PET/CT before chemotherapy may help to identify patients at risk for inadequate response to perioperative FLOT and, after chemotherapy, may predict clinical outcomes.
Collapse
Affiliation(s)
- Seyed Ali Mirshahvalad
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria; Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Seyedeh Sara Seyedinia
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Gregor Schweighofer-Zwink
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Oliver Koch
- Department of Surgery, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Wolfgang Hitzl
- Biostatistics and Publication of Clinical Trial Studies, Research and Innovation Management (RIM), Paracelsus Medical University, 5020 Salzburg, Austria; Department of Ophthalmology and Optometry, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas Weiss
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Klaus Emannuel
- Department of Surgery, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christian Pirich
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
13
|
Pollard JH, DiCamillo PA, Dundar A, Averill SL, Aswani Y. Gastrointestinal Malignancies. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:407-455. [DOI: 10.1002/9781119603627.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Machine learning approach using 18 F-FDG PET-based radiomics in differentiation of lung adenocarcinoma with bronchoalveolar distribution and infection. Nucl Med Commun 2023; 44:302-308. [PMID: 36756766 DOI: 10.1097/mnm.0000000000001667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
OBJECTIVE In this study, we aimed to evaluate the role of 18F-fluorodeoxyglucose PET/computerized tomography ( 18 F-FDG PET/CT)-based radiomic features in the differentiation of infection and malignancy in consolidating pulmonary lesions and to develop a prediction model based on radiomic features. MATERIAL AND METHODS The images of 106 patients who underwent 18 F-FDG PET/CT of consolidated lesions observed in the lung between January 2015 and July 2020 were evaluated using LIFEx software. The region of interest of the lung lesions was determined and volumetric and textural features were obtained. Clinical and radiomic data were evaluated with machine learning algorithms to build a model. RESULTS There was a significant difference in all standardized uptake value (SUV) parameters and 26 texture features between the infection and cancer groups. The features with a correlation coefficient of less than 0.7 among the significant features were determined as SUV mean , GLZLM_SZE, GLZLM_LZE, GLZLM_SZLGE and GLZLM_ZLNU. These five features were analyzed in the Waikato Environment for Knowledge Analysis program to create a model that could distinguish infection and cancer groups, and the model performance was found to be the highest with logistic regression (area under curve, 0.813; accuracy, 75.7%). The sensitivity and specificity values of the model in distinguishing cancer patients were calculated as 80.6 and 70.6%, respectively. CONCLUSIONS In our study, we created prediction models based on radiomic analysis of 18 F-FDG PET/CT images. Texture analysis with machine learning algorithms is a noninvasive method that can be useful in the differentiation of infection and malignancy in consolidating lung lesions in the clinical setting.
Collapse
|
15
|
Guo H, Tang HT, Hu WL, Wang JJ, Liu PZ, Yang JJ, Hou SL, Zuo YJ, Deng ZQ, Zheng XY, Yan HJ, Jiang KY, Huang H, Zhou HN, Tian D. The application of radiomics in esophageal cancer: Predicting the response after neoadjuvant therapy. Front Oncol 2023; 13:1082960. [PMID: 37091180 PMCID: PMC10117779 DOI: 10.3389/fonc.2023.1082960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Esophageal cancer (EC) is one of the fatal malignant neoplasms worldwide. Neoadjuvant therapy (NAT) combined with surgery has become the standard treatment for locally advanced EC. However, the treatment efficacy for patients with EC who received NAT varies from patient to patient. Currently, the evaluation of efficacy after NAT for EC lacks accurate and uniform criteria. Radiomics is a multi-parameter quantitative approach for developing medical imaging in the era of precision medicine and has provided a novel view of medical images. As a non-invasive image analysis method, radiomics is an inevitable trend in NAT efficacy prediction and prognosis classification of EC by analyzing the high-throughput imaging features of lesions extracted from medical images. In this literature review, we discuss the definition and workflow of radiomics, the advances in efficacy prediction after NAT, and the current application of radiomics for predicting efficacy after NAT.
Collapse
Affiliation(s)
- Hai Guo
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, Sichuan Tianfu New Area People’s Hospital, Chengdu, China
| | - Hong-Tao Tang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Wen-Long Hu
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jun-Jie Wang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Pei-Zhi Liu
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jun-Jie Yang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Sen-Lin Hou
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yu-Jie Zuo
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Zhi-Qiang Deng
- College of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Xiang-Yun Zheng
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Hao-Ji Yan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kai-Yuan Jiang
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Heng Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hai-Ning Zhou
- Department of Thoracic Surgery, Suining Central Hospital, Suining, China
- *Correspondence: Dong Tian, ; Hai-Ning Zhou,
| | - Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Dong Tian, ; Hai-Ning Zhou,
| |
Collapse
|
16
|
Liu W, Zeng C, Wang S, Zhan Y, Huang R, Luo T, Peng G, Wu Y, Qiu Z, Li D, Wu F, Chen C. A combined predicting model for benign esophageal stenosis after simultaneous integrated boost in esophageal squamous cell carcinoma patients (GASTO1072). Front Oncol 2022; 12:1026305. [PMID: 37078004 PMCID: PMC10107369 DOI: 10.3389/fonc.2022.1026305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
PurposeWe aimed to develop a combined predicting model for benign esophageal stenosis (BES) after simultaneous integrated boost (SIB) with concurrent chemotherapy in patients with esophageal squamous cell carcinoma (ESCC).MethodsThis study included 65 patients with EC who underwent SIB with chemotherapy. Esophageal stenosis was evaluated using esophagograms and the severity of eating disorders. Risk factors were investigated using univariate and multivariate analyses. Radiomics features were extracted based on contrast-enhanced CT (CE-CT) before treatment. The least absolute shrinkage and selection operator (LASSO) regression analysis was used for feature selection and radiomics signature construction. The model’s performance was evaluated using Harrell’s concordance index and receiver operating characteristic curves.ResultsThe patients were stratified into low- and high-risk groups according to BES after SIB. The area under the curves of the clinical model, Rad-score, and the combined model were 0.751, 0.820 and 0.864, respectively. In the validation cohort, the AUCs of these three models were 0.854, 0.883 and 0.917, respectively. The Hosmer-Lemeshow test showed that there was no deviation from model fitting for the training cohort (p=0.451) and validation cohort (p=0.481). The C-indexes of the nomogram were 0.864 and 0.958 for the training and validation cohort, respectively. The model combined with Rad-score and clinical factors achieved favorable prediction ability.ConclusionDefinitive chemoradiotherapy could alleviate tumor-inducing esophageal stenosis but result in benign stenosis. We constructed and tested a combined predicting model for benign esophageal stenosis after SIB. The nomogram incorporating both radiomics signature and clinical prognostic factors showed favorable predictive accuracy for BES in ESCC patients who received SIB with chemotherapy.Trial registration number and date of registrationRegistered in www.Clinicaltrial.gov, ID: NCT01670409, August 12, 2012
Collapse
Affiliation(s)
- Weitong Liu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Jieyang People’s Hospital, Jeiyang, China
| | - Chengbing Zeng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Siyan Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yizhou Zhan
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ruihong Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ting Luo
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Shenshan Central Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, China
| | - Guobo Peng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yanxuan Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zihan Qiu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Derui Li
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Fangcai Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Chuangzhen Chen, ; Fangcai Wu,
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Chuangzhen Chen, ; Fangcai Wu,
| |
Collapse
|
17
|
Anconina R, Ortega C, Metser U, Liu ZA, Elimova E, Allen M, Darling GE, Wong R, Taylor K, Yeung J, Chen EX, Swallow CJ, Jang RW, Veit-Haibach P. Combined 18 F-FDG PET/CT Radiomics and Sarcopenia Score in Predicting Relapse-Free Survival and Overall Survival in Patients With Esophagogastric Cancer. Clin Nucl Med 2022; 47:684-691. [PMID: 35543637 DOI: 10.1097/rlu.0000000000004253] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE The aim of this study was to determine if radiomic features combined with sarcopenia measurements on pretreatment 18 F-FDG PET/CT can improve outcome prediction in surgically treated adenocarcinoma esophagogastric cancer patients. PATIENTS AND METHODS One hundred forty-five esophageal adenocarcinoma patients with curative therapeutic intent and available pretreatment 18 F-FDG PET/CT were included. Textural features from PET and CT images were evaluated using LIFEx software ( lifexsoft.org ). Sarcopenia measurements were done by measuring the Skeletal Muscle Index at L3 level on the CT component. Univariable and multivariable analyses were conducted to create a model including the radiomic parameters, clinical features, and Skeletal Muscle Index score to predict patients' outcome. RESULTS In multivariable analysis, we combined clinicopathological parameters including ECOG, surgical T, and N staging along with imaging derived sarcopenia measurements and radiomic features to build a predictor model for relapse-free survival and overall survival. Overall, adding sarcopenic status to the model with clinical features only (likelihood ratio test P = 0.03) and CT feature ( P = 0.0037) improved the model fit for overall survival. Similarly, adding sarcopenic status ( P = 0.051), CT feature ( P = 0.042), and PET feature ( P = 0.011) improved the model fit for relapse-free survival. CONCLUSIONS PET and CT radiomics derived from combined PET/CT integrated with clinicopathological parameters and sarcopenia measurement might improve outcome prediction in patients with nonmetastatic esophagogastric adenocarcinoma.
Collapse
Affiliation(s)
- Reut Anconina
- From the Department of Medical Imaging, Sunnybrook Health Sciences Centre
| | - Claudia Ortega
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network
| | - Ur Metser
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network
| | | | - Elena Elimova
- Medical Oncology, Princess Margaret Cancer Centre, University Health Network
| | - Michael Allen
- Medical Oncology, Princess Margaret Cancer Centre, University Health Network
| | - Gail E Darling
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital, University Health Network
| | | | - Kirsty Taylor
- Medical Oncology, Princess Margaret Cancer Centre, University Health Network
| | - Jonathan Yeung
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital, University Health Network
| | - Eric X Chen
- Medical Oncology, Princess Margaret Cancer Centre, University Health Network
| | - Carol J Swallow
- Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Raymond W Jang
- Medical Oncology, Princess Margaret Cancer Centre, University Health Network
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network
| |
Collapse
|
18
|
Dmytriw AA, Ortega C, Anconina R, Metser U, Liu ZA, Liu Z, Li X, Sananmuang T, Yu E, Joshi S, Waldron J, Huang SH, Bratman S, Hope A, Veit-Haibach P. Nasopharyngeal Carcinoma Radiomic Evaluation with Serial PET/CT: Exploring Features Predictive of Survival in Patients with Long-Term Follow-Up. Cancers (Basel) 2022; 14:3105. [PMID: 35804877 PMCID: PMC9264840 DOI: 10.3390/cancers14133105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
PURPOSE We aim determine the value of PET and CT radiomic parameters on survival with serial follow-up PET/CT in patients with nasopharyngeal carcinoma (NPC) for which curative intent therapy is undertaken. METHODS Patients with NPC and available pre-treatment as well as follow up PET/CT were included from 2005 to 2006 and were followed to 2021. Baseline demographic, radiological and outcome data were collected. Univariable Cox proportional hazard models were used to evaluate features from baseline and follow-up time points, and landmark analyses were performed for each time point. RESULTS Sixty patients were enrolled, and two-hundred and seventy-eight (278) PET/CT were at baseline and during follow-up. Thirty-eight percent (38%) were female, and sixty-two patients were male. All patients underwent curative radiation or chemoradiation therapy. The median follow-up was 11.72 years (1.26-14.86). Five-year and ten-year overall survivals (OSs) were 80.0% and 66.2%, and progression-free survival (PFS) was 90.0% and 74.4%. Time-dependent modelling suggested that, among others, PET gray-level zone length matrix (GLZLM) gray-level non-uniformity (GLNU) (HR 2.74 95% CI 1.06, 7.05) was significantly associated with OS. Landmark analyses suggested that CT parameters were most predictive at 15 month, whereas PET parameters were most predictive at time points 3, 6, 9 and 15 month. CONCLUSIONS This study with long-term follow up data on NPC suggests that mainly PET-derived radiomic features are predictive for OS but not PFS in a time-dependent evaluation. Furthermore, CT radiomic measures may predict OS and PFS best at initial and long-term follow-up time points and PET measures may be more predictive in the interval. These modalities are commonly used in NPC surveillance, and prospective validation should be considered.
Collapse
Affiliation(s)
- Adam A. Dmytriw
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (A.A.D.); (R.A.)
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| | - Claudia Ortega
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| | - Reut Anconina
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada; (A.A.D.); (R.A.)
| | - Ur Metser
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| | - Zhihui A. Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (Z.A.L.); (Z.L.); (X.L.)
| | - Zijin Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (Z.A.L.); (Z.L.); (X.L.)
| | - Xuan Li
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (Z.A.L.); (Z.L.); (X.L.)
| | - Thiparom Sananmuang
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
- Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University,270 Rama VI Road, Ratchathewi, Bangkok 10400, Thailand
| | - Eugene Yu
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| | - Sayali Joshi
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| | - John Waldron
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (J.W.); (S.H.H.); (S.B.); (A.H.)
| | - Shao Hui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (J.W.); (S.H.H.); (S.B.); (A.H.)
| | - Scott Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (J.W.); (S.H.H.); (S.B.); (A.H.)
| | - Andrew Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; (J.W.); (S.H.H.); (S.B.); (A.H.)
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada; (C.O.); (U.M.); (T.S.); (E.Y.); (S.J.)
| |
Collapse
|
19
|
O'Shea RJ, Rookyard C, Withey S, Cook GJR, Tsoka S, Goh V. Radiomic assessment of oesophageal adenocarcinoma: a critical review of 18F-FDG PET/CT, PET/MRI and CT. Insights Imaging 2022; 13:104. [PMID: 35715706 PMCID: PMC9206060 DOI: 10.1186/s13244-022-01245-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/28/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Radiomic models present an avenue to improve oesophageal adenocarcinoma assessment through quantitative medical image analysis. However, model selection is complicated by the abundance of available predictors and the uncertainty of their relevance and reproducibility. This analysis reviews recent research to facilitate precedent-based model selection for prospective validation studies. METHODS This analysis reviews research on 18F-FDG PET/CT, PET/MRI and CT radiomics in oesophageal adenocarcinoma between 2016 and 2021. Model design, testing and reporting are evaluated according to the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD) score and Radiomics Quality Score (RQS). Key results and limitations are analysed to identify opportunities for future research in the area. RESULTS Radiomic models of stage and therapeutic response demonstrated discriminative capacity, though clinical applications require greater sensitivity. Although radiomic models predict survival within institutions, generalisability is limited. Few radiomic features have been recommended independently by multiple studies. CONCLUSIONS Future research must prioritise prospective validation of previously proposed models to further clinical translation.
Collapse
Affiliation(s)
- Robert J O'Shea
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th floor, Becket House, 1 Lambeth Palace Rd, London, SE1 7EU, UK.
| | - Chris Rookyard
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th floor, Becket House, 1 Lambeth Palace Rd, London, SE1 7EU, UK
| | - Sam Withey
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th floor, Becket House, 1 Lambeth Palace Rd, London, SE1 7EU, UK
- King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK
| | - Sophia Tsoka
- Department of Informatics, School of Natural and Mathematical Sciences, King's College London, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th floor, Becket House, 1 Lambeth Palace Rd, London, SE1 7EU, UK
- Department of Radiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
20
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 2, Infradiaphragmatic Cancers, Blood Malignancies, Melanoma and Musculoskeletal Cancers. Diagnostics (Basel) 2022; 12:diagnostics12061330. [PMID: 35741139 PMCID: PMC9222024 DOI: 10.3390/diagnostics12061330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
The objective of this review was to summarize published radiomics studies dealing with infradiaphragmatic cancers, blood malignancies, melanoma, and musculoskeletal cancers, and assess their quality. PubMed database was searched from January 1990 to February 2022 for articles performing radiomics on PET imaging of at least 1 specified tumor type. Exclusion criteria includd: non-oncological studies; supradiaphragmatic tumors; reviews, comments, cases reports; phantom or animal studies; technical articles without a clinically oriented question; studies including <30 patients in the training cohort. The review database contained PMID, first author, year of publication, cancer type, number of patients, study design, independent validation cohort and objective. This database was completed twice by the same person; discrepant results were resolved by a third reading of the articles. A total of 162 studies met inclusion criteria; 61 (37.7%) studies included >100 patients, 13 (8.0%) were prospective and 61 (37.7%) used an independent validation set. The most represented cancers were esophagus, lymphoma, and cervical cancer (n = 24, n = 24 and n = 19 articles, respectively). Most studies focused on 18F-FDG, and prognostic and response to treatment objectives. Although radiomics and artificial intelligence are technically challenging, new contributions and guidelines help improving research quality over the years and pave the way toward personalized medicine.
Collapse
Affiliation(s)
- David Morland
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
- Correspondence:
| | - Elizabeth Katherine Anna Triumbari
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
21
|
Jha AK, Mithun S, Purandare NC, Kumar R, Rangarajan V, Wee L, Dekker A. Radiomics: a quantitative imaging biomarker in precision oncology. Nucl Med Commun 2022; 43:483-493. [PMID: 35131965 DOI: 10.1097/mnm.0000000000001543] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cancer treatment is heading towards precision medicine driven by genetic and biochemical markers. Various genetic and biochemical markers are utilized to render personalized treatment in cancer. In the last decade, noninvasive imaging biomarkers have also been developed to assist personalized decision support systems in oncology. The imaging biomarkers i.e., radiomics is being researched to develop specific digital phenotype of tumor in cancer. Radiomics is a process to extract high throughput data from medical images by using advanced mathematical and statistical algorithms. The radiomics process involves various steps i.e., image generation, segmentation of region of interest (e.g. a tumor), image preprocessing, radiomic feature extraction, feature analysis and selection and finally prediction model development. Radiomics process explores the heterogeneity, irregularity and size parameters of the tumor to calculate thousands of advanced features. Our study investigates the role of radiomics in precision oncology. Radiomics research has witnessed a rapid growth in the last decade with several studies published that show the potential of radiomics in diagnosis and treatment outcome prediction in oncology. Several radiomics based prediction models have been developed and reported in the literature to predict various prediction endpoints i.e., overall survival, progression-free survival and recurrence in various cancer i.e., brain tumor, head and neck cancer, lung cancer and several other cancer types. Radiomics based digital phenotypes have shown promising results in diagnosis and treatment outcome prediction in oncology. In the coming years, radiomics is going to play a significant role in precision oncology.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Sneha Mithun
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Nilendu C Purandare
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Science, New Delhi, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital
- Homi Bhabha National Institute (HBNI), Deemed University, Mumbai
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, The Netherlands
| |
Collapse
|
22
|
Prediction of Non-Response to Neoadjuvant Chemoradiotherapy in Esophageal Cancer Patients with 18F-FDG PET Radiomics Based Machine Learning Classification. Diagnostics (Basel) 2022; 12:diagnostics12051070. [PMID: 35626225 PMCID: PMC9139915 DOI: 10.3390/diagnostics12051070] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Background: Approximately 26% of esophageal cancer (EC) patients do not respond to neoadjuvant chemoradiotherapy (nCRT), emphasizing the need for pre-treatment selection. The aim of this study was to predict non-response using a radiomic model on baseline 18F-FDG PET. Methods: Retrospectively, 143 18F-FDG PET radiomic features were extracted from 199 EC patients (T1N1-3M0/T2–4aN0-3M0) treated between 2009 and 2019. Non-response (n = 57; 29%) was defined as Mandard Tumor Regression Grade 4–5 (n = 44; 22%) or interval progression (n = 13; 7%). Randomly, 139 patients (70%) were allocated to explore all combinations of 24 feature selection strategies and 6 classification methods towards the cross-validated average precision (AP). The predictive value of the best-performing model, i.e AP and area under the ROC curve analysis (AUC), was evaluated on an independent test subset of 60 patients (30%). Results: The best performing model had an AP (mean ± SD) of 0.47 ± 0.06 on the training subset, achieved by a support vector machine classifier trained on five principal components of relevant clinical and radiomic features. The model was externally validated with an AP of 0.66 and an AUC of 0.67. Conclusion: In the present study, the best-performing model on pre-treatment 18F-FDG PET radiomics and clinical features had a small clinical benefit to identify non-responders to nCRT in EC.
Collapse
|
23
|
Deantonio L, Garo ML, Paone G, Valli MC, Cappio S, La Regina D, Cefali M, Palmarocchi MC, Vannelli A, De Dosso S. 18F-FDG PET Radiomics as Predictor of Treatment Response in Oesophageal Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:861638. [PMID: 35371989 PMCID: PMC8965232 DOI: 10.3389/fonc.2022.861638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/16/2022] [Indexed: 12/22/2022] Open
Abstract
The best treatment strategy for oesophageal cancer patients achieving a complete clinical response after neoadjuvant chemoradiation is a burning topic. The available diagnostic tools, such as 18F-FDG PET/CT performed routinely, cannot accurately evaluate the presence or absence of the residual tumour. The emerging field of radiomics may encounter the critical challenge of personalised treatment. Radiomics is based on medical image analysis, executed by extracting information from many image features; it has been shown to provide valuable information for predicting treatment responses in oesophageal cancer. This systematic review with a meta-analysis aims to provide current evidence of 18F-FDG PET-based radiomics in predicting response treatments following neoadjuvant chemoradiotherapy in oesophageal cancer. A comprehensive literature review identified 1160 studies, of which five were finally included in the study. Our findings provided that pooled Area Under the Curve (AUC) of the five selected studies was relatively high at 0.821 (95% CI: 0.737–0.904) and not influenced by the sample size of the studies. Radiomics models exhibited a good performance in predicting pathological complete responses (pCRs). This review further strengthens the great potential of 18F-FDG PET-based radiomics to predict pCRs in oesophageal cancer patients who underwent neoadjuvant chemoradiotherapy. Additionally, our review imparts additional support to prospective studies on 18F-FDG PET radiomics for a tailored treatment strategy of oesophageal cancer patients.
Collapse
Affiliation(s)
- Letizia Deantonio
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland.,University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland
| | | | - Gaetano Paone
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Maria Carla Valli
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Stefano Cappio
- Clinic for Radiology, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Davide La Regina
- Department of Surgery, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland
| | - Marco Cefali
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Maria Celeste Palmarocchi
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | | | - Sara De Dosso
- University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland.,Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| |
Collapse
|
24
|
Fusco R, Granata V, Grazzini G, Pradella S, Borgheresi A, Bruno A, Palumbo P, Bruno F, Grassi R, Giovagnoni A, Grassi R, Miele V, Barile A. Radiomics in medical imaging: pitfalls and challenges in clinical management. Jpn J Radiol 2022; 40:919-929. [PMID: 35344132 DOI: 10.1007/s11604-022-01271-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND Radiomics and radiogenomics are two words that recur often in language of radiologists, nuclear doctors and medical physicists especially in oncology field. Radiomics is the technique of medical images analysis to extract quantitative data that are not detected by human eye. METHODS This article is a narrative review on Radiomics in Medical Imaging. In particular, the review exposes the process, the limitations related to radiomics, and future prospects are discussed. RESULTS Several studies showed that radiomics is very promising. However, there were some critical issues: poor standardization and generalization of radiomics results, data-quality control, repeatability, reproducibility, database balancing and issues related to model overfitting. CONCLUSIONS Radiomics procedure should made considered all pitfalls and challenges to obtain robust and reproducible results that could be generalized in other patients cohort.
Collapse
Affiliation(s)
| | - Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli", Naples, Italy.
| | - Giulia Grazzini
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Silvia Pradella
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Alessandra Borgheresi
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Alessandra Bruno
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Pierpaolo Palumbo
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Diagnostic Imaging, Area of Cardiovascular and Interventional Imaging, Abruzzo Health Unit 1, 67100, L'Aquila, Italy
| | - Federico Bruno
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Roberta Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Division of Radiology, "Università Degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Andrea Giovagnoni
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Roberto Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Division of Radiology, "Università Degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Vittorio Miele
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Antonio Barile
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| |
Collapse
|
25
|
Jiang W, de Jong JM, van Hillegersberg R, Read M. Predicting Response to Neoadjuvant Therapy in Oesophageal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14040996. [PMID: 35205743 PMCID: PMC8869950 DOI: 10.3390/cancers14040996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 12/20/2022] Open
Abstract
(1) Background: Oesophageal cancers are often late-presenting and have a poor 5-year survival rate. The standard treatment of oesophageal adenocarcinomas involves neoadjuvant chemotherapy with or without radiotherapy followed by surgery. However, less than one third of patients respond to neoadjuvant therapy, thereby unnecessarily exposing patients to toxicity and deconditioning. Hence, there is an urgent need for biomarkers to predict response to neoadjuvant therapy. This review explores the current biomarker landscape. (2) Methods: MEDLINE, EMBASE and ClinicalTrial databases were searched with key words relating to “predictive biomarker”, “neoadjuvant therapy” and “oesophageal adenocarcinoma” and screened as per the inclusion and exclusion criteria. All peer-reviewed full-text articles and conference abstracts were included. (3) Results: The search yielded 548 results of which 71 full-texts, conference abstracts and clinical trials were eligible for review. A total of 242 duplicates were removed, 191 articles were screened out, and 44 articles were excluded. (4) Discussion: Biomarkers were discussed in seven categories including imaging, epigenetic, genetic, protein, immunologic, blood and serum-based with remaining studies grouped in a miscellaneous category. (5) Conclusion: Although promising markers and novel methods have emerged, current biomarkers lack sufficient evidence to support clinical application. Novel approaches have been recommended to assess predictive potential more efficiently.
Collapse
Affiliation(s)
- William Jiang
- Upper Gastrointestinal Surgery Department, St Vincent’s Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia
- Correspondence: (W.J.); (M.R.)
| | - Jelske M. de Jong
- Gastrointestinal Oncology Department, The University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (J.M.d.J.); (R.v.H.)
| | - Richard van Hillegersberg
- Gastrointestinal Oncology Department, The University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (J.M.d.J.); (R.v.H.)
| | - Matthew Read
- Upper Gastrointestinal Surgery Department, St Vincent’s Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia
- Correspondence: (W.J.); (M.R.)
| |
Collapse
|
26
|
Methodological quality of machine learning-based quantitative imaging analysis studies in esophageal cancer: a systematic review of clinical outcome prediction after concurrent chemoradiotherapy. Eur J Nucl Med Mol Imaging 2021; 49:2462-2481. [PMID: 34939174 PMCID: PMC9206619 DOI: 10.1007/s00259-021-05658-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/12/2021] [Indexed: 10/24/2022]
Abstract
PURPOSE Studies based on machine learning-based quantitative imaging techniques have gained much interest in cancer research. The aim of this review is to critically appraise the existing machine learning-based quantitative imaging analysis studies predicting outcomes of esophageal cancer after concurrent chemoradiotherapy in accordance with PRISMA guidelines. METHODS A systematic review was conducted in accordance with PRISMA guidelines. The citation search was performed via PubMed and Embase Ovid databases for literature published before April 2021. From each full-text article, study characteristics and model information were summarized. We proposed an appraisal matrix with 13 items to assess the methodological quality of each study based on recommended best-practices pertaining to quality. RESULTS Out of 244 identified records, 37 studies met the inclusion criteria. Study endpoints included prognosis, treatment response, and toxicity after concurrent chemoradiotherapy with reported discrimination metrics in validation datasets between 0.6 and 0.9, with wide variation in quality. A total of 30 studies published within the last 5 years were evaluated for methodological quality and we found 11 studies with at least 6 "good" item ratings. CONCLUSION A substantial number of studies lacked prospective registration, external validation, model calibration, and support for use in clinic. To further improve the predictive power of machine learning-based models and translate into real clinical applications in cancer research, appropriate methodologies, prospective registration, and multi-institution validation are recommended.
Collapse
|
27
|
Chidambaram S, Sounderajah V, Maynard N, Markar SR. Diagnostic Performance of Artificial Intelligence-Centred Systems in the Diagnosis and Postoperative Surveillance of Upper Gastrointestinal Malignancies Using Computed Tomography Imaging: A Systematic Review and Meta-Analysis of Diagnostic Accuracy. Ann Surg Oncol 2021; 29:1977-1990. [PMID: 34762214 PMCID: PMC8810479 DOI: 10.1245/s10434-021-10882-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022]
Abstract
Background Upper gastrointestinal cancers are aggressive malignancies with poor prognosis, even following multimodality therapy. As such, they require timely and accurate diagnostic and surveillance strategies; however, such radiological workflows necessitate considerable expertise and resource to maintain. In order to lessen the workload upon already stretched health systems, there has been increasing focus on the development and use of artificial intelligence (AI)-centred diagnostic systems. This systematic review summarizes the clinical applicability and diagnostic performance of AI-centred systems in the diagnosis and surveillance of esophagogastric cancers. Methods A systematic review was performed using the MEDLINE, EMBASE, Cochrane Review, and Scopus databases. Articles on the use of AI and radiomics for the diagnosis and surveillance of patients with esophageal cancer were evaluated, and quality assessment of studies was performed using the QUADAS-2 tool. A meta-analysis was performed to assess the diagnostic accuracy of sequencing methodologies. Results Thirty-six studies that described the use of AI were included in the qualitative synthesis and six studies involving 1352 patients were included in the quantitative analysis. Of these six studies, four studies assessed the utility of AI in gastric cancer diagnosis, one study assessed its utility for diagnosing esophageal cancer, and one study assessed its utility for surveillance. The pooled sensitivity and specificity were 73.4% (64.6–80.7) and 89.7% (82.7–94.1), respectively. Conclusions AI systems have shown promise in diagnosing and monitoring esophageal and gastric cancer, particularly when combined with existing diagnostic methods. Further work is needed to further develop systems of greater accuracy and greater consideration of the clinical workflows that they aim to integrate within.
Collapse
Affiliation(s)
| | - Viknesh Sounderajah
- Department of Surgery and Cancer, Imperial College London, London, UK.,Institute of Global Health Innovation, Imperial College London, London, UK
| | - Nick Maynard
- Department of Surgery, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Sheraz R Markar
- Department of Surgery and Cancer, Imperial College London, London, UK. .,Department of Surgery, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, UK. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Xue C, Yuan J, Lo GG, Chang ATY, Poon DMC, Wong OL, Zhou Y, Chu WCW. Radiomics feature reliability assessed by intraclass correlation coefficient: a systematic review. Quant Imaging Med Surg 2021; 11:4431-4460. [PMID: 34603997 DOI: 10.21037/qims-21-86] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022]
Abstract
Radiomics research is rapidly growing in recent years, but more concerns on radiomics reliability are also raised. This review attempts to update and overview the current status of radiomics reliability research in the ever expanding medical literature from the perspective of a single reliability metric of intraclass correlation coefficient (ICC). To conduct this systematic review, Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. After literature search and selection, a total of 481 radiomics studies using CT, PET, or MRI, covering a wide range of subject and disease types, were included for review. In these highly heterogeneous studies, feature reliability to image segmentation was much more investigated than reliability to other factors, such as image acquisition, reconstruction, post-processing, and feature quantification. The reported ICCs also suggested high radiomics feature reliability to image segmentation. Image acquisition was found to introduce much more feature variability than image segmentation, in particular for MRI, based on the reported ICC values. Image post-processing and feature quantification yielded different levels of radiomics reliability and might be used to mitigate image acquisition-induced variability. Some common flaws and pitfalls in ICC use were identified, and suggestions on better ICC use were given. Due to the extremely high study heterogeneities and possible risks of bias, the degree of radiomics feature reliability that has been achieved could not yet be safely synthesized or derived in this review. More future researches on radiomics reliability are warranted.
Collapse
Affiliation(s)
- Cindy Xue
- Medical Physics and Research Department, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China.,Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Jing Yuan
- Medical Physics and Research Department, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Gladys G Lo
- Department of Diagnostic & Interventional Radiology, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Amy T Y Chang
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Oi Lei Wong
- Medical Physics and Research Department, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Yihang Zhou
- Medical Physics and Research Department, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong, China
| | - Winnie C W Chu
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
29
|
Karahan Şen NP, Aksu A, Çapa Kaya G. A different overview of staging PET/CT images in patients with esophageal cancer: the role of textural analysis with machine learning methods. Ann Nucl Med 2021; 35:1030-1037. [PMID: 34106428 DOI: 10.1007/s12149-021-01638-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study evaluates the ability of several machine learning (ML) algorithms, developed using volumetric and texture data extracted from baseline 18F-FDG PET/CT studies performed initial staging of patient with esophageal cancer (EC), to predict survival and histopathology. METHODS The initial staging 18F-FDG PET/CT images obtained on newly diagnosed EC patients between January 2008 and June 2019 were evaluated using LIFEx software. A region of interest (ROI) of the primary tumor was created and volumetric and textural features were obtained. A significant relationship between these features and pathological subtypes, 1-year, and 5-year survival was investigated. Due to the nonhomogeneity of the data, nonparametric test (The Mann-Whitney U test) was used for each feature, in pairwise comparisons of independent variables. A p value of < 0.05 was considered significant. Receiver operating curve (ROC) analysis was performed for features with p < 0.05. Correlation between the significant features was evaluated with Spearman correlation test; features with correlation coefficient < 0.8 were evaluated with several ML algorithms. RESULTS In predicting survival in a 1-year follow-up J48 was obtained as the most successful algorithm (AUC: 0.581, PRC: 0.565, MCC: 0.258, acc: 64.29%). 5-year survival results were more promising than 1-year survival results with (AUC: 0.820, PRC: 0.860, MCC: 271, acc: 81.36%) by logistic regression. It is revealed that the most successful algorithm was naive bayes (AUC: 0.680 PRC: 0.776, MCC: 0.298, acc: 82.66%) in the histopathological discrimination. CONCLUSION Texture analysis with ML algorithms could be predictive of overall survival and discriminating histopathological subtypes of EC.
Collapse
Affiliation(s)
- Nazlı Pınar Karahan Şen
- Department of Nuclear Medicine, Dokuz Eylul University Faculty of Medicine, İnciraltı mah. Mithatpaşa cad. no:1606 Balçova, Izmir, Turkey.
| | - Ayşegül Aksu
- Başakşehir Çam ve Sakura City Hospital, Department of Nuclear Medicine, Istanbul, Turkey
| | - Gamze Çapa Kaya
- Department of Nuclear Medicine, Dokuz Eylul University Faculty of Medicine, İnciraltı mah. Mithatpaşa cad. no:1606 Balçova, Izmir, Turkey
| |
Collapse
|
30
|
Nardone V, Boldrini L, Grassi R, Franceschini D, Morelli I, Becherini C, Loi M, Greto D, Desideri I. Radiomics in the Setting of Neoadjuvant Radiotherapy: A New Approach for Tailored Treatment. Cancers (Basel) 2021; 13:3590. [PMID: 34298803 PMCID: PMC8303203 DOI: 10.3390/cancers13143590] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Neoadjuvant radiotherapy is currently used mainly in locally advanced rectal cancer and sarcoma and in a subset of non-small cell lung cancer and esophageal cancer, whereas in other diseases it is under investigation. The evaluation of the efficacy of the induction strategy is made possible by performing imaging investigations before and after the neoadjuvant therapy and is usually challenging. In the last decade, texture analysis (TA) has been developed to help the radiologist to quantify and identify the parameters related to tumor heterogeneity, which cannot be appreciated by the naked eye. The aim of this narrative is to review the impact of TA on the prediction of response to neoadjuvant radiotherapy and or chemoradiotherapy. MATERIALS AND METHODS Key references were derived from a PubMed query. Hand searching and ClinicalTrials.gov were also used. RESULTS This paper contains a narrative report and a critical discussion of radiomics approaches in different fields of neoadjuvant radiotherapy, including esophageal cancer, lung cancer, sarcoma, and rectal cancer. CONCLUSIONS Radiomics can shed a light on the setting of neoadjuvant therapies that can be used to tailor subsequent approaches or even to avoid surgery in the future. At the same, these results need to be validated in prospective and multicenter trials.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (R.G.)
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, 20122 Milan, Italy
| | - Luca Boldrini
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Roberta Grassi
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (R.G.)
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, 20122 Milan, Italy
| | - Davide Franceschini
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy;
| | - Ilaria Morelli
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Carlotta Becherini
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Mauro Loi
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (M.L.); (D.G.); (I.D.)
| | - Daniela Greto
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (M.L.); (D.G.); (I.D.)
| | - Isacco Desideri
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (M.L.); (D.G.); (I.D.)
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| |
Collapse
|
31
|
Kao YS, Hsu Y. A Meta-Analysis for Using Radiomics to Predict Complete Pathological Response in Esophageal Cancer Patients Receiving Neoadjuvant Chemoradiation. In Vivo 2021; 35:1857-1863. [PMID: 33910873 DOI: 10.21873/invivo.12448] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Preservation of organ function is important in cancer treatment. The 'watch-and-wait' strategy is an important approach in management of esophageal cancer. However, clinical imaging cannot accurately evaluate the presence or absence of residual tumor after neoadjuvant chemoradiation. As a result, using radiomics to predict complete pathological response in esophageal cancer has gained in popularity in recent years. Given that the characteristics of patients and sites vary considerably, a meta-analysis is needed to investigate the predictive power of radiomics in esophageal cancer. PATIENTS AND METHODS PRISMA guidelines were used to conduct this study. PubMed, Cochrane, and Embase were searched for literature review. The quality of the selected studies was evaluated by the radiomics quality score. I2 score and Cochran's Q test were used to evaluate heterogeneity between studies. A funnel plot was used for evaluation of publication bias. RESULTS A total of seven articles were collected for this meta-analysis. The pooled area under the receiver operating characteristics curve of the seven selected articles for predicting pathological complete response in eosphageal cancer patient was quite high, achieving a pooled value of 0.813 (95% confidence intervaI=0.761-0.866). The radiomics quality score ranged from -2 to 16 (maximum score: 36 points). Three out of the seven studies used machine learning algorithms, while the others used traditional biostatistics methods. One of the seven studies used morphology class features, while four studies used first-order features, and five used second-order features. CONCLUSION Using radiomics to predict complete pathological response after neoadjuvant chemoradiotherapy in esophageal cancer is feasible. In the future, prospective, multicenter studies should be carried out for predicting pathological complete response in patients with esophageal cancer.
Collapse
Affiliation(s)
- Yung-Shuo Kao
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan, R.O.C.;
| | - Yen Hsu
- Department of Family Medicine, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| |
Collapse
|
32
|
Xie CY, Pang CL, Chan B, Wong EYY, Dou Q, Vardhanabhuti V. Machine Learning and Radiomics Applications in Esophageal Cancers Using Non-Invasive Imaging Methods-A Critical Review of Literature. Cancers (Basel) 2021; 13:2469. [PMID: 34069367 PMCID: PMC8158761 DOI: 10.3390/cancers13102469] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer (EC) is of public health significance as one of the leading causes of cancer death worldwide. Accurate staging, treatment planning and prognostication in EC patients are of vital importance. Recent advances in machine learning (ML) techniques demonstrate their potential to provide novel quantitative imaging markers in medical imaging. Radiomics approaches that could quantify medical images into high-dimensional data have been shown to improve the imaging-based classification system in characterizing the heterogeneity of primary tumors and lymph nodes in EC patients. In this review, we aim to provide a comprehensive summary of the evidence of the most recent developments in ML application in imaging pertinent to EC patient care. According to the published results, ML models evaluating treatment response and lymph node metastasis achieve reliable predictions, ranging from acceptable to outstanding in their validation groups. Patients stratified by ML models in different risk groups have a significant or borderline significant difference in survival outcomes. Prospective large multi-center studies are suggested to improve the generalizability of ML techniques with standardized imaging protocols and harmonization between different centers.
Collapse
Affiliation(s)
- Chen-Yi Xie
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Chun-Lap Pang
- Department of Radiology, The Christies’ Hospital, Manchester M20 4BX, UK;
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester M15 6FH, UK
| | - Benjamin Chan
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Emily Yuen-Yuen Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Qi Dou
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China;
| | - Varut Vardhanabhuti
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
33
|
Li Y, Liu J, Li HX, Cai XW, Li ZG, Ye XD, Teng HH, Fu XL, Yu W. Radiomics Signature Facilitates Organ-Saving Strategy in Patients With Esophageal Squamous Cell Cancer Receiving Neoadjuvant Chemoradiotherapy. Front Oncol 2021; 10:615167. [PMID: 33680935 PMCID: PMC7933499 DOI: 10.3389/fonc.2020.615167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
After neoadjuvant chemoradiotherapy (NCRT) in locally advanced esophageal squamous cell cancer (ESCC), roughly 40% of the patients may achieve pathologic complete response (pCR). Those patients may benefit from organ-saving strategy if the probability of pCR could be correctly identified before esophagectomy. A reliable approach to predict pathological response allows future studies to investigate individualized treatment plans.
Collapse
Affiliation(s)
- Yue Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-Xuan Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xu-Wei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Dan Ye
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hao-Hua Teng
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Long Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Zhang C, Shi Z, Kalendralis P, Whybra P, Parkinson C, Berbee M, Spezi E, Roberts A, Christian A, Lewis W, Crosby T, Dekker A, Wee L, Foley KG. Prediction of lymph node metastases using pre-treatment PET radiomics of the primary tumour in esophageal adenocarcinoma: an external validation study. Br J Radiol 2020; 94:20201042. [PMID: 33264032 DOI: 10.1259/bjr.20201042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To improve clinical lymph node staging (cN-stage) in oesophageal adenocarcinoma by developing and externally validating three prediction models; one with clinical variables only, one with positron emission tomography (PET) radiomics only, and a combined clinical and radiomics model. METHODS Consecutive patients with fluorodeoxyglucose (FDG) avid tumours treated with neoadjuvant therapy between 2010 and 2016 in two international centres (n = 130 and n = 60, respectively) were included. Four clinical variables (age, gender, clinical T-stage and tumour regression grade) and PET radiomics from the primary tumour were used for model development. Diagnostic accuracy, area under curve (AUC), discrimination and calibration were calculated for each model. The prognostic significance was also assessed. RESULTS The incidence of lymph node metastases was 58% in both cohorts. The areas under the curve of the clinical, radiomics and combined models were 0.79, 0.69 and 0.82 in the developmental cohort, and 0.65, 0.63 and 0.69 in the external validation cohort, with good calibration demonstrated. The area under the curve of current cN-stage in development and validation cohorts was 0.60 and 0.66, respectively. For overall survival, the combined clinical and radiomics model achieved the best discrimination performance in the external validation cohort (X2 = 6.08, df = 1, p = 0.01). CONCLUSION Accurate diagnosis of lymph node metastases is crucial for prognosis and guiding treatment decisions. Despite finding improved predictive performance in the development cohort, the models using PET radiomics derived from the primary tumour were not fully replicated in an external validation cohort. ADVANCES IN KNOWLEDGE This international study attempted to externally validate a new prediction model for lymph node metastases using PET radiomics. A model combining clinical variables and PET radiomics improved discrimination of lymph node metastases, but these results were not externally replicated.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Zhenwei Shi
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Petros Kalendralis
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Phil Whybra
- School of Engineering, Cardiff University, Cardiff, UK
| | | | - Maaike Berbee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Ashley Roberts
- Department of Radiology, University Hospital of Wales, Cardiff, UK
| | - Adam Christian
- Department of Pathology, University Hospital of Wales, Cardiff, UK
| | - Wyn Lewis
- Department of Upper GI Surgery, University Hospital of Wales, Cardiff, UK
| | - Tom Crosby
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff, UK
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kieran G Foley
- Department of Radiology, Velindre Cancer Centre, Cardiff, UK
| |
Collapse
|
35
|
Rishi A, Zhang GG, Yuan Z, Sim AJ, Song EY, Moros EG, Tomaszewski MR, Latifi K, Pimiento JM, Fontaine JP, Mehta R, Harrison LB, Hoffe SE, Frakes JM. Pretreatment CT and 18 F-FDG PET-based radiomic model predicting pathological complete response and loco-regional control following neoadjuvant chemoradiation in oesophageal cancer. J Med Imaging Radiat Oncol 2020; 65:102-111. [PMID: 33258556 DOI: 10.1111/1754-9485.13128] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/21/2020] [Indexed: 01/12/2023]
Abstract
INTRODUCTION To develop a radiomic-based model to predict pathological complete response (pCR) and outcome following neoadjuvant chemoradiotherapy (NACRT) in oesophageal cancer. METHODS We analysed 68 patients with oesophageal cancer treated with NACRT followed by esophagectomy, who had staging 18F-fluorodeoxyglucose (18 F-FDG) positron emission tomography (PET) and computed tomography (CT) scans performed at our institution. An in-house data-chjmirocterization algorithm was used to extract 3D-radiomic features from the segmented primary disease. Prediction models were constructed and internally validated. Composite feature, Fc = α * FPET + (1 - α) * FCT , 0 ≤ α ≤ 1, was constructed for each corresponding CT and PET feature. Loco-regional control (LRC), recurrence-free survival (RFS), metastasis-free survival (MFS) and overall survival (OS) were estimated by Kaplan-Meier analysis, and compared using log-rank test. RESULTS Median follow-up was 59 months. pCR was achieved in 34 (50%) patients. Five-year RFS, LRC, MFS and OS were 67.1%, 88.5%, 75.6% and 57.6%, respectively. Tumour Regression Grade (TRG) 0-1 indicative of complete response or minimal residual disease was significantly associated with improved 5-year LRC [93.7% vs 71.8%; P = 0.020; HR 0.19, 95% CI 0.04-0.85]. Four sepjmirote pCR predictive models were built for CT alone, PET alone, CT+PET and composite. CT, PET and CT+PET models had AUC 0.73 ± 0.08, 0.66 ± 0.08 and 0.77 ± 0.07, respectively. The composite model resulted in an improvement of pCR predicting power with AUC 0.87 ± 0.06. Stratifying patients with a low versus high radiomic score showed clinically relevant improvement in 5-year LRC favouring low-score group (91.1% vs. 80%, 95% CI 0.09-1.77, P = 0.2). CONCLUSION The composite CT/PET radiomics model was highly predictive of pCR following NACRT. Validation in larger data sets is warranted to determine whether the model can predict clinical outcomes.
Collapse
Affiliation(s)
- Anupam Rishi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Geoffrey G Zhang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Zhigang Yuan
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Austin J Sim
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Ethan Y Song
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Eduardo G Moros
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Michal R Tomaszewski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Kujtim Latifi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jose M Pimiento
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jacques-Pierre Fontaine
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Louis B Harrison
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Sjmiroh E Hoffe
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jessica M Frakes
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
36
|
Zhang Y, Li X, Lv Y, Gu X. Review of Value of CT Texture Analysis and Machine Learning in Differentiating Fat-Poor Renal Angiomyolipoma from Renal Cell Carcinoma. Tomography 2020; 6:325-332. [PMID: 33364422 PMCID: PMC7744193 DOI: 10.18383/j.tom.2020.00039] [Citation(s) in RCA: 5] [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] [Indexed: 12/15/2022] Open
Abstract
The diagnosis of patients with suspected angiomyolipoma relies on the detection of abundant macroscopic intralesional fat, which is always of no use to differentiate fat-poor angiomyolipoma (fp-AML) from renal cell carcinoma and diagnosis of fp-AML excessively depends on individual experience. Texture analysis was proven to be a potentially useful biomarker for distinguishing between benign and malignant tumors because of its capability of providing objective and quantitative assessment of lesions by analyzing features that are not visible to the human eye. This review aimed to summarize the literature on the use of texture analysis to diagnose patients with fat-poor angiomyolipoma vs those with renal cell carcinoma and to evaluate its current application, limitations, and future challenges in order to avoid unnecessary surgical resection.
Collapse
Affiliation(s)
- Yuhan Zhang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China; and
| | - Xu Li
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China; and
| | - Yang Lv
- Department of Anesthesia, The Second Hospital of Jilin University, Changchun, China
| | - Xinquan Gu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China; and
| |
Collapse
|
37
|
Wang HH, de Heer EC, Hulshoff JB, Kats-Ugurlu G, Burgerhof JGM, van Etten B, Plukker JTM, Hospers GAP. Effect of Extending the Original CROSS Criteria on Tumor Response to Neoadjuvant Chemoradiotherapy in Esophageal Cancer Patients: A National Multicenter Cohort Analysis. Ann Surg Oncol 2020; 28:3951-3960. [PMID: 33249520 PMCID: PMC8184698 DOI: 10.1245/s10434-020-09372-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022]
Abstract
Background Extending the original criteria of the Chemoradiotherapy for Oesophageal Cancer followed by Surgery Study (CROSS) in daily practice may increase the treatment outcome of esophageal cancer (EC) patients. This retrospective national cohort study assessed the impact on the pathologic complete response (pCR) rate and surgical outcome. Patients and Methods Data from EC patients treated between 2009 and 2017 were collected from the national Dutch Upper Gastrointestinal Cancer Audit database. Patients had locally advanced EC (cT1/N+ or cT2-4a/N0-3/M0) and were treated according to the CROSS regimen. CROSS (n = 1942) and the extended CROSS (e-CROSS; n = 1359) represent patients fulfilling the original or extended CROSS criteria, respectively. The primary outcome was total pCR (ypT0N0), while secondary outcomes were local esophageal pCR (ypT0), surgical radicality, and postoperative morbidity and mortality. Results Overall, CROSS and e-CROSS did not differ in total or local pCR rate, although a trend was observed (23.2% vs. 20.4%, p = 0.052; and 26.7% vs. 23.8%, p = 0.061). When stratifying by histology, the pCR rate was higher in the CROSS group compared with e-CROSS in squamous cell carcinomas (48.2% vs. 33.3%, p = 0.000) but not in adenocarcinomas (16.8% vs. 16.9%, p = 0.908). Surgical radicality did not differ between groups. Postoperative mortality (3.2% vs. 4.6%, p = 0.037) and morbidity (58.3% vs. 61.8%, p = 0.048) were higher in e-CROSS. Conclusion Extending the CROSS inclusion criteria for neoadjuvant chemoradiotherapy in routine clinical practice of EC patients had no impact on the pCR rate and on radicality, but was associated with increased postoperative mortality and morbidity. Importantly, effects differed between histological subtypes. Hence, in future studies, we should carefully reconsider who will benefit most in the real-world setting. Electronic supplementary material The online version of this article (10.1245/s10434-020-09372-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helena Hong Wang
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ellen C de Heer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Binne Hulshoff
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gursah Kats-Ugurlu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johannes G M Burgerhof
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Boudewijn van Etten
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Th M Plukker
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | | |
Collapse
|
38
|
Simoni N, Rossi G, Benetti G, Zuffante M, Micera R, Pavarana M, Guariglia S, Zivelonghi E, Mengardo V, Weindelmayer J, Giacopuzzi S, de Manzoni G, Cavedon C, Mazzarotto R. 18F-FDG PET/CT Metrics Are Correlated to the Pathological Response in Esophageal Cancer Patients Treated With Induction Chemotherapy Followed by Neoadjuvant Chemo-Radiotherapy. Front Oncol 2020; 10:599907. [PMID: 33330097 PMCID: PMC7729075 DOI: 10.3389/fonc.2020.599907] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/27/2020] [Indexed: 12/04/2022] Open
Abstract
Background and Objective The aim of this study was to assess the ability of Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography (18F-FDG PET/CT) to provide functional information useful in predicting pathological response to an intensive neoadjuvant chemo-radiotherapy (nCRT) protocol for both esophageal squamous cell carcinoma (SCC) and adenocarcinoma (ADC) patients. Material and Methods Esophageal carcinoma (EC) patients, treated in our Center between 2014 and 2018, were retrospectively reviewed. The nCRT protocol schedule consisted of an induction phase of weekly administered docetaxel, cisplatin, and 5-fluorouracil (TCF) for 3 weeks, followed by a concomitant phase of weekly TCF for 5 weeks with concurrent radiotherapy (50–50.4 Gy in 25–28 fractions). Three 18F-FDG PET/CT scans were performed: before (PET1) and after (PET2) induction chemotherapy (IC), and prior to surgery (PET3). Correlation between PET parameters [maximum and mean standardized uptake value (SUVmax and SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG)], radiomic features and tumor regression grade (TGR) was investigated. Results Fifty-four patients (35 ADC, 19 SCC; 48 cT3/4; 52 cN+) were eligible for the analysis. Pathological response to nCRT was classified as major (TRG1-2, 41/54, 75.9%) or non-response (TRG3-4, 13/54, 24.1%). A major response was statistically correlated with SCC subtype (p = 0.02) and smaller tumor length (p = 0.03). MTV and TLG measured prior to IC (PET1) were correlated to TRG1-2 response (p = 0.02 and p = 0.02, respectively). After IC (PET2), SUVmean and TLG correlated with major response (p = 0.03 and p = 0.04, respectively). No significance was detected when relative changes of metabolic parameters between PET1 and PET2 were evaluated. At textural quantitative analysis, three independent radiomic features extracted from PET1 images ([JointEnergy and InverseDifferenceNormalized of GLCM and LowGrayLevelZoneEmphasis of GLSZM) were statistically correlated with major response (p < 0.0002). Conclusions 18F-FDG PET/CT traditional metrics and textural features seem to predict pathologic response (TRG) in EC patients treated with induction chemotherapy followed by neoadjuvant chemo-radiotherapy. Further investigations are necessary in order to obtain a reliable predictive model to be used in the clinical practice.
Collapse
Affiliation(s)
- Nicola Simoni
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Gabriella Rossi
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Giulio Benetti
- Department of Medical Physics, University of Verona Hospital Trust, Verona, Italy
| | - Michele Zuffante
- Department of Nuclear Medicine, University of Verona Hospital Trust, Verona, Italy
| | - Renato Micera
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Michele Pavarana
- Department of Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Stefania Guariglia
- Department of Medical Physics, University of Verona Hospital Trust, Verona, Italy
| | - Emanuele Zivelonghi
- Department of Medical Physics, University of Verona Hospital Trust, Verona, Italy
| | - Valentina Mengardo
- Department of General and Upper G.I. Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Jacopo Weindelmayer
- Department of General and Upper G.I. Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Simone Giacopuzzi
- Department of General and Upper G.I. Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Giovanni de Manzoni
- Department of General and Upper G.I. Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Carlo Cavedon
- Department of Medical Physics, University of Verona Hospital Trust, Verona, Italy
| | - Renzo Mazzarotto
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona, Italy
| |
Collapse
|
39
|
Beukinga RJ, Wang D, Karrenbeld A, Dijksterhuis WPM, Faber H, Burgerhof JGM, Mul VEM, Slart RHJA, Coppes RP, Plukker JTM. Addition of HER2 and CD44 to 18F-FDG PET-based clinico-radiomic models enhances prediction of neoadjuvant chemoradiotherapy response in esophageal cancer. Eur Radiol 2020; 31:3306-3314. [PMID: 33151397 PMCID: PMC8043921 DOI: 10.1007/s00330-020-07439-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/21/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To assess the complementary value of human epidermal growth factor receptor 2 (HER2)-related biological tumor markers to clinico-radiomic models in predicting complete response to neoadjuvant chemoradiotherapy (NCRT) in esophageal cancer patients. METHODS Expression of HER2 was assessed by immunohistochemistry in pre-treatment tumor biopsies of 96 patients with locally advanced esophageal cancer. Five other potentially active HER2-related biological tumor markers in esophageal cancer were examined in a sub-analysis on 43 patients. Patients received at least four of the five cycles of chemotherapy and full radiotherapy regimen followed by esophagectomy. Three reference clinico-radiomic models based on 18F-FDG PET were constructed to predict pathologic response, which was categorized into complete versus incomplete (Mandard tumor regression grade 1 vs. 2-5). The complementary value of the biological tumor markers was evaluated by internal validation through bootstrapping. RESULTS Pathologic examination revealed 21 (22%) complete and 75 (78%) incomplete responders. HER2 and cluster of differentiation 44 (CD44), analyzed in the sub-analysis, were univariably associated with pathologic response. Incorporation of HER2 and CD44 into the reference models improved the overall performance (R2s of 0.221, 0.270, and 0.225) and discrimination AUCs of 0.759, 0.857, and 0.816. All models exhibited moderate to good calibration. The remaining studied biological tumor markers did not yield model improvement. CONCLUSIONS Incorporation of HER2 and CD44 into clinico-radiomic prediction models improved NCRT response prediction in esophageal cancer. These biological tumor markers are promising in initial response evaluation. KEY POINTS • A multimodality approach, integrating independent genomic and radiomic information, is promising to improve prediction of γpCR in patients with esophageal cancer. • HER2 and CD44 are potential biological tumor markers in the initial work-up of patients with esophageal cancer. • Prediction models combining 18F-FDG PET radiomic features with HER2 and CD44 may be useful in the decision to omit surgery after neoadjuvant chemoradiotherapy in patients with esophageal cancer.
Collapse
Affiliation(s)
- Roelof J Beukinga
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.
| | - Da Wang
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arend Karrenbeld
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Willemieke P M Dijksterhuis
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hette Faber
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johannes G M Burgerhof
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Véronique E M Mul
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.,Faculty of Science and Technology, Department of Biomedical Photonic Imaging, University of Twente, Enschede, The Netherlands
| | - Robert P Coppes
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Th M Plukker
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
40
|
Harada K, Patnana M, Wang X, Iwatsuki M, Murphy MAB, Zhao M, Das P, Minsky BD, Weston B, Lee JH, Bhutani MS, Estrella JS, Shanbhag N, Ikoma N, Badgwell BD, Ajani JA. Low metabolic activity in primary gastric adenocarcinoma is associated with resistance to chemoradiation and the presence of signet ring cells. Surg Today 2020; 50:1223-1231. [PMID: 32409870 PMCID: PMC9396945 DOI: 10.1007/s00595-020-02018-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/17/2020] [Indexed: 12/14/2022]
Abstract
PURPOSES Preoperative chemoradiation is a potential treatment option for localized gastric adenocarcinoma (GAC). Currently, the response to chemoradiation cannot be predicted. We analyzed the pretreatment maximum standardized uptake value (SUVmax) and total lesion glycolysis (TLG) on positron emission tomography/computed tomography as potential predictors of the response to chemoradiation. METHODS We analyzed the SUVmax and TLG data from 59 GAC patients who received preoperative chemoradiation. We used logistic regression models to predict a pathologic complete response (pCR) and Kaplan-Meier curves to determine overall survival among patients with high and low SUVmax or TLG. RESULTS Twenty-nine patients (49%) had Siewert type III adenocarcinoma and 30 (51%) had tumors located in the lower stomach. Forty-one patients had poorly differentiated GAC, and 26 had signet ring cells. The median SUVmax was 7.3 (0-28.2) and the median TLG was 56.6 (0-1881.5). Patients with signet ring cells had a low pCR rate, as well as a low SUVmax and TLG. In the multivariable logistic regression model, high SUVmax was a predictor of pCR (odds ratio = 11.1, 95% confidence interval = 2.12-50.0, p = 0.004). Overall survival was not associated with the SUVmax (log-rank p = 0.69) or TLG (log-rank p = 0.85) CONCLUSION: A high SUVmax was associated with sensitivity to chemoradiation and pCR in GAC, and signet ring cells seemed to confer resistance.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Madhavi Patnana
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Wang
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Masaaki Iwatsuki
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Mariela A Blum Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Meina Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Weston
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey H Lee
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manoop S Bhutani
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeannelyn S Estrella
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Namita Shanbhag
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Elsherif SB, Andreou S, Virarkar M, Soule E, Gopireddy DR, Bhosale PR, Lall C. Role of precision imaging in esophageal cancer. J Thorac Dis 2020; 12:5159-5176. [PMID: 33145093 PMCID: PMC7578477 DOI: 10.21037/jtd.2019.08.15] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Esophageal cancer is a major cause of morbidity and mortality worldwide. Recent advancements in the management of esophageal cancer have allowed for earlier detection, improved ability to monitor progression, and superior treatment options. These innovations allow treatment teams to formulate more customized management plans and have led to an increase in patient survival rates. For example, in order for the most effective management plan to be constructed, accurate staging must be performed to determine tumor resectability. This article reviews the multimodality imaging approach involved in making a diagnosis, staging, evaluating treatment response and detecting recurrence in esophageal cancer.
Collapse
Affiliation(s)
- Sherif B Elsherif
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA.,Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonia Andreou
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Mayur Virarkar
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erik Soule
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | | | - Priya R Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chandana Lall
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
42
|
Characterization of FDG PET Images Using Texture Analysis in Tumors of the Gastro-Intestinal Tract: A Review. Biomedicines 2020; 8:biomedicines8090304. [PMID: 32846986 PMCID: PMC7556033 DOI: 10.3390/biomedicines8090304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
Radiomics or textural feature extraction obtained from positron emission tomography (PET) images through complex mathematical models of the spatial relationship between multiple image voxels is currently emerging as a new tool for assessing intra-tumoral heterogeneity in medical imaging. In this paper, available literature on texture analysis using FDG PET imaging in patients suffering from tumors of the gastro-intestinal tract is reviewed. While texture analysis of FDG PET images appears clinically promising, due to the lack of technical specifications, a large variability in the implemented methodology used for texture analysis and lack of statistical robustness, at present, no firm conclusions can be drawn regarding the predictive or prognostic value of FDG PET texture analysis derived indices in patients suffering from gastro-enterologic tumors. In order to move forward in this field, a harmonized image acquisition and processing protocol as well as a harmonized protocol for texture analysis of tumor volumes, allowing multi-center studies excluding statistical biases should be considered. Furthermore, the complementary and additional value of CT-imaging, as part of the PET/CT imaging technique, warrants exploration.
Collapse
|
43
|
Li XF, Wang Q, Duan SF, Yao B, Liu CY. Heterogeneity of T3 stage esophageal squamous cell carcinoma in different parts based on enhanced CT radiomics. Medicine (Baltimore) 2020; 99:e21470. [PMID: 32769880 PMCID: PMC7593053 DOI: 10.1097/md.0000000000021470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Esophageal cancer is a common malignant tumor of the digestive system with a high incidence and a poor prognosis. At the present, CT-based radiomics is providing more and more valuable information. However, the heterogeneity of the study and the poor repeatability of the texture feature parameters have limited its wider clinical application. In the present study, we focused on comparing the differences in the texture features of T3 stage esophageal squamous cell carcinoma at different locations and normal esophageal wall, aiming to provide some pieces of useful information for future research on esophageal squamous cell carcinoma.Fifty seven cases with throat CT imaging, including esophageal cancer contrast enhanced CT and conventional CT of healthy control group. The texture characteristics in control group and tumor group among different parts were compared. Using Univariable analysis, we compared the difference and conducted receiver-operator curve analysis to evaluate the performance of tumor grade diagnosis model.53 radiomic features were significantly different in control group and so as 93 features for tumor group. The upper section was the mostly different from the other 2 sections. Run-length matrix (RLM) features in tumor group accounted for the highest proportion, only Surface Volume Ratio was different.There are differences in the texture features of the tube wall in different parts of the esophagus of healthy adults, and this difference is more obvious in pT3 stage esophageal squamous cell carcinoma. In the future radiomics study of esophageal squamous cell carcinoma, we need to pay attention to this to avoid affecting the accuracy of the results.
Collapse
Affiliation(s)
| | - Qiang Wang
- Department of Radiotherapy, Xuzhou Cancer Hospital, Xuzhou, Jiangsu Province
| | | | | | | |
Collapse
|
44
|
Harada K, Wu CC, Wang X, Mizrak Kaya D, Amlashi FG, Iwatsuki M, Blum Murphy MA, Maru DM, Weston B, Lee JH, Rogers JE, Thomas I, Shanbhag N, Bhutani MS, Hofstetter WL, Nguyen QN, Ajani JA. Total Lesion Glycolysis Assessment Identifies a Patient Fraction With a High Cure Rate Among Esophageal Adenocarcinoma Patients Treated With Definitive Chemoradiation. Ann Surg 2020; 272:311-318. [PMID: 32675544 DOI: 10.1097/sla.0000000000003228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE We aimed to determine whether tumor metabolism could be prognostic of cure in L-EAC patients who receive definitive chemoradiation. SUMMARY BACKGROUND DATA Patients with inoperable localized esophageal adenocarcinoma (L-EAC) often receive definitive chemoradiation; however, biomarkers and/or imaging variables to prognosticate cure are missing. METHODS Two hundred sixty-six patients with L-EAC who had chemoradiation but not surgery were analyzed from the prospectively maintained EAC databases in the Department of Gastrointestinal Medical Oncology at The University of Texas MD Anderson Cancer Center (Texas, USA) between March 2002 and April 2015. Maximum standardized uptake value (SUVmax) and total lesion glycolysis (TLG) from the positron emission tomography data were evaluated. RESULTS Of 266 patients, 253 (95%) were men; the median age was 67 years (range 20-91 yrs) and 153 had poorly differentiated L-EAC. The median SUVmax was 10.3 (range 0-87) and the median TLG was 85.7 (range 0-3227). Both SUVmax and TLG were higher among those with: tumors >5 cm in length, high clinical stage, and high tumor and node categories by TNM staging (all P < 0.0001). Of 234 patients evaluable for cure, 60 (25.6%) achieved cure. In the multivariable logistic regression model, low TLG (but not low SUVmax) was associated with cure (continuous TLG value: odds ratio 0.70, 95% confidence interval (CI) 0.54-0.92). TLG was quantified into 4 quartile categorical variables; first quartile (Q1; <32), second quartile (Q2; 32.0-85.6), third quartile (Q3; 85.6-228.4), and fourth quartile (Q4; >228.4); the cure rate was only 10.3% in Q4 and 5.1% in Q3 but increased to 28.8% in Q2, and 58.6% in Q1. The cross-validation resulted in an average accuracy of prediction score of 0.81 (95% CI, 0.75-0.86). CONCLUSIONS In this cross-validated model, 59% of patients in the 1st quartile were cured following definitive chemoradiation. Baseline TLG could be pursued as one of the tools for esophageal preservation.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Carol C Wu
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dilsa Mizrak Kaya
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fatemeh G Amlashi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Masaaki Iwatsuki
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Mariela A Blum Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dipen M Maru
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian Weston
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffrey H Lee
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jane E Rogers
- Department of Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Irene Thomas
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Namita Shanbhag
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Manoop S Bhutani
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
45
|
Dercle L, Henry T, Carré A, Paragios N, Deutsch E, Robert C. Reinventing radiation therapy with machine learning and imaging bio-markers (radiomics): State-of-the-art, challenges and perspectives. Methods 2020; 188:44-60. [PMID: 32697964 DOI: 10.1016/j.ymeth.2020.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy is a pivotal cancer treatment that has significantly progressed over the last decade due to numerous technological breakthroughs. Imaging is now playing a critical role on deployment of the clinical workflow, both for treatment planning and treatment delivery. Machine-learning analysis of predefined features extracted from medical images, i.e. radiomics, has emerged as a promising clinical tool for a wide range of clinical problems addressing drug development, clinical diagnosis, treatment selection and implementation as well as prognosis. Radiomics denotes a paradigm shift redefining medical images as a quantitative asset for data-driven precision medicine. The adoption of machine-learning in a clinical setting and in particular of radiomics features requires the selection of robust, representative and clinically interpretable biomarkers that are properly evaluated on a representative clinical data set. To be clinically relevant, radiomics must not only improve patients' management with great accuracy but also be reproducible and generalizable. Hence, this review explores the existing literature and exposes its potential technical caveats, such as the lack of quality control, standardization, sufficient sample size, type of data collection, and external validation. Based upon the analysis of 165 original research studies based on PET, CT-scan, and MRI, this review provides an overview of new concepts, and hypotheses generating findings that should be validated. In particular, it describes evolving research trends to enhance several clinical tasks such as prognostication, treatment planning, response assessment, prediction of recurrence/relapse, and prediction of toxicity. Perspectives regarding the implementation of an AI-based radiotherapy workflow are presented.
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, USA
| | - Theophraste Henry
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alexandre Carré
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Eric Deutsch
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charlotte Robert
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
46
|
Bibault JE, Xing L, Giraud P, El Ayachy R, Giraud N, Decazes P, Burgun A, Giraud P. Radiomics: A primer for the radiation oncologist. Cancer Radiother 2020; 24:403-410. [PMID: 32265157 DOI: 10.1016/j.canrad.2020.01.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Radiomics are a set of methods used to leverage medical imaging and extract quantitative features that can characterize a patient's phenotype. All modalities can be used with several different software packages. Specific informatics methods can then be used to create meaningful predictive models. In this review, we will explain the major steps of a radiomics analysis pipeline and then present the studies published in the context of radiation therapy. METHODS A literature review was performed on Medline using the search engine PubMed. The search strategy included the search terms "radiotherapy", "radiation oncology" and "radiomics". The search was conducted in July 2019 and reference lists of selected articles were hand searched for relevance to this review. RESULTS A typical radiomics workflow always includes five steps: imaging and segmenting, data curation and preparation, feature extraction, exploration and selection and finally modeling. In radiation oncology, radiomics studies have been published to explore different clinical outcome in lung (n=5), head and neck (n=5), esophageal (n=3), rectal (n=3), pancreatic (n=2) cancer and brain metastases (n=2). The quality of these retrospective studies is heterogeneous and their results have not been translated to the clinic. CONCLUSION Radiomics has a great potential to predict clinical outcome and better personalize treatment. But the field is still young and constantly evolving. Improvement in bias reduction techniques and multicenter studies will hopefully allow more robust and generalizable models.
Collapse
Affiliation(s)
- J-E Bibault
- Radiation Oncology Department, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France; Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France.
| | - L Xing
- Laboratory of Artificial Intelligence in Medicine and Biomedical Physics, Stanford University School of Medicine, 875 Blake Wilbur Drive, 94305-5847 Stanford, CA, USA
| | - P Giraud
- Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - R El Ayachy
- Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - N Giraud
- Radiation Oncology Department, CHU de Bordeaux, hôpital Haut-Lévêque, avenue Magellan, 33600 Pessac, France
| | - P Decazes
- Nuclear Medicine Department, centre Henri-Becquerel, 1, rue d'Amiens, 76038 Rouen, France; Quantif, EA 4108, université de Rouen, avenue de l'Université, 76801 Saint-Étienne-du-Rouvray, France
| | - A Burgun
- Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France; Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France; Biomedical Informatics and Public Health Department, hôpital européen Georges-Pompidou, Assistance publique-hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France
| | - P Giraud
- Radiation Oncology Department, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France
| |
Collapse
|
47
|
Shen LF, Zhou SH, Yu Q. Predicting response to radiotherapy in tumors with PET/CT: when and how? Transl Cancer Res 2020; 9:2972-2981. [PMID: 35117653 PMCID: PMC8798842 DOI: 10.21037/tcr.2020.03.16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/25/2020] [Indexed: 11/11/2022]
Abstract
Radiotherapy is one of the main methods for tumor treatment, with the improved radiotherapy delivery technique to combat cancer, there is a growing interest for finding effective and feasible ways to predict tumor radiosensitivity. Based on a series of changes in metabolism, microvessel density, hypoxic microenvironment, and cytokines of tumors after radiotherapy, a variety of radiosensitivity detection methods have been studied. Among the detection methods, positron emission tomography-computed tomography (PET/CT) is a feasible tool for response evaluation following definitive radiotherapy for cancers with a high negative predictive value. The prognostic or predictive value of PET/CT is currently being studied widely. However, there are many unresolved issues, such as the optimal probe of PET/CT for radiosensitivity prediction, the selection of the most useful PET/CT parameters and their optimal cut-offs such as total lesion glycolysis (TLG), metabolic tumor volume (MTV) and standardized uptake value (SUV), and the optimal timing of PET/CT pre-treatment, during or following RT. Different radiosensitivity of tumors, modes of radiotherapy action and fraction scheduling may complicate the appropriate choice. In this study, we will discuss the diverse methods for evaluating radiosensitivity, and will also focus on the selection of the optimal probe, timing, cut-offs and parameters of PET/CT for evaluating the radiotherapy response.
Collapse
Affiliation(s)
- Li-Fang Shen
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qi Yu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
48
|
Li Y, Han G, Wu X, Li Z, Zhao K, Zhang Z, Liu Z, Liang C. Normalization of multicenter CT radiomics by a generative adversarial network method. Phys Med Biol 2020; 66. [PMID: 32209747 DOI: 10.1088/1361-6560/ab8319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/25/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE To reduce the variability of radiomics features caused by computed tomography (CT) imaging protocols through using a generative adversarial network (GAN) method. MATERIAL AND METHODS In this study, we defined a set of images acquired with a certain imaging protocol as a domain, and a total of 4 domains (A, B, C, and T [target]) from 3 different scanners were included. In dataset#1, 60 patinets for each domain were collected. Datasets#2 and #3 included 40 slices of spleen for each of the domains. In dataset#4, the slices of 3 colorectal cancer groups (n = 28, 38, and 32) were separately retrieved from 3 different scanners, and each group contained short-term and long-term survivors. 77 features were extracted for evaluation by comparing features distributions. First, we trained the GAN model on dataset#1 to learn how to normalize images from domains A, B, and C to T. Next, by comparing feature distributions between normalized images of the different domains, we identified the appropriate model and assessed it , in dataset #2 and dataset#3, respectively. Finally, to investigate whether our proposed method could facilitate multicenter radiomics analysis, we built the lasso classifier to distinguish short-term from long-term survivors based on a certain group in dataset#4, and validate it in another two groups, which formed a cross-validation between groups in dataset#4. RESULTS After normalization, the percentage of aligned features between domains A vs T, B vs T, and C vs T increased from 10.4 %, 18.2%, and 50.1% to 93.5%, 89.6%, and 77.9%, respectively. In the cross-validation results, average improvement of the area under the receiver operating characteristic curve achieved 11% (3%-32%). CONCLUSION Our proposed GAN-based normalization method could reduce the variability of radiomics features caused by different CT imaging protocols and facilitate multicenter radiomics analysis.
Collapse
Affiliation(s)
- Yajun Li
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | - Guoqiang Han
- College of Electronic and Information Engineering, South China University of Technology, Guangzhou, CHINA
| | - Xiaomei Wu
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | - Zhenhui Li
- Yunnan Cancer Hospital, Kunming, Yunnan, CHINA
| | - Ke Zhao
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | | | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, CHINA
| | - Changhong Liang
- Radiology, Guangdong General Hospital, Guangzhou, 510080, CHINA
| |
Collapse
|
49
|
Borggreve AS, Goense L, van Rossum PSN, Heethuis SE, van Hillegersberg R, Lagendijk JJW, Lam MGEH, van Lier ALHMW, Mook S, Ruurda JP, van Vulpen M, Voncken FEM, Aleman BMP, Bartels-Rutten A, Ma J, Fang P, Musall BC, Lin SH, Meijer GJ. Preoperative Prediction of Pathologic Response to Neoadjuvant Chemoradiotherapy in Patients With Esophageal Cancer Using 18F-FDG PET/CT and DW-MRI: A Prospective Multicenter Study. Int J Radiat Oncol Biol Phys 2020; 106:998-1009. [PMID: 31987972 DOI: 10.1016/j.ijrobp.2019.12.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/06/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Accurate preoperative prediction of pathologic response to neoadjuvant chemoradiotherapy (nCRT) in patients with esophageal cancer could enable omission of esophagectomy in patients with a pathologic complete response (pCR). This study aimed to evaluate the individual and combined value of 18F-fluorodeoxyglucose positron emission tomography with integrated computed tomography (18F-FDG PET/CT) and diffusion-weighted magnetic resonance imaging (DW-MRI) during and after nCRT to predict pathologic response in patients with esophageal cancer. METHODS AND MATERIALS In this multicenter prospective study, patients scheduled to receive nCRT followed by esophagectomy for esophageal cancer underwent 18F-FDG PET/CT and DW-MRI scanning before the start of nCRT, during nCRT, and before esophagectomy. Response to nCRT was based on histopathologic evaluation of the resection specimen. Relative changes in 18F-FDG PET/CT and DW-MRI parameters were compared between patients with pCR and non-pCR groups. Multivariable ridge regression analyses with bootstrapped c-indices were performed to evaluate the individual and combined value of 18F-FDG PET/CT and DW-MRI. RESULTS pCR was found in 26.1% of 69 patients. Relative changes in 18F-FDG PET/CT parameters after nCRT (Δ standardized uptake value [SUV]mean,postP = .016, and Δ total lesion glycolysis postP = .024), as well as changes in DW-MRI parameters during nCRT (Δ apparent diffusion coefficient [ADC]duringP = .008) were significantly different between pCR and non-pCR. A c-statistic of 0.84 was obtained for a model with ΔADCduring, ΔSUVmean,post, and histology in classifying patients as pCR (versus 0.82 for ΔADCduring and 0.79 for ΔSUVmean,post alone). CONCLUSIONS Changes on 18F-FDG PET/CT after nCRT and early changes on DW-MRI during nCRT can help identify pCR to nCRT in esophageal cancer. Moreover, 18F-FDG PET/CT and DW-MRI might be of complementary value in the assessment of pCR.
Collapse
Affiliation(s)
- Alicia S Borggreve
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lucas Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Peter S N van Rossum
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Sophie E Heethuis
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | | | - Jan J W Lagendijk
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Marnix G E H Lam
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Astrid L H M W van Lier
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Stella Mook
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | | | - Francine E M Voncken
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Berthe M P Aleman
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Annemarieke Bartels-Rutten
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Penny Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin C Musall
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands.
| |
Collapse
|
50
|
Nomori H, Shiraishi A, O’uchi T, Honma K, Shoji K, Misawa M, Sugimura H, Oyama Y. Positron Emission Tomography in T3/T4 Non-Small Cell Lung Cancer After Induction Chemoradiotherapy. Ann Thorac Surg 2020; 109:255-261. [DOI: 10.1016/j.athoracsur.2019.06.089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/14/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
|