1
|
Guerrisi A, Miseo L, Falcone I, Messina C, Ungania S, Elia F, Desiderio F, Valenti F, Cantisani V, Soriani A, Caterino M. Quantitative ultrasound radiomics analysis to evaluate lymph nodes in patients with cancer: a systematic review. ULTRASCHALL IN DER MEDIZIN (STUTTGART, GERMANY : 1980) 2024; 45:586-596. [PMID: 38663433 DOI: 10.1055/a-2275-8342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
This systematic review aims to evaluate the role of ultrasound (US) radiomics in assessing lymphadenopathy in patients with cancer and the ability of radiomics to predict metastatic lymph node involvement. A systematic literature search was performed in the PubMed (MEDLINE), Cochrane Central Register of Controlled Trials (CENTRAL), and EMBASE (Ovid) databases up to June 13, 2023. 42 articles were included in which the lymph node mass was assessed with a US exam, and the analysis was performed using radiomics methods. From the survey of the selected articles, experimental evidence suggests that radiomics features extracted from US images can be a useful tool for predicting and characterizing lymphadenopathy in patients with breast, head and neck, and cervical cancer. This noninvasive and effective method allows the extraction of important information beyond mere morphological characteristics, extracting features that may be related to lymph node involvement. Future studies are needed to investigate the role of US-radiomics in other types of cancers, such as melanoma.
Collapse
Affiliation(s)
- Antonio Guerrisi
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| | - Ludovica Miseo
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| | - Italia Falcone
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Claudia Messina
- Library, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| | - Sara Ungania
- Medical Physics and Expert Systems Laboratory, Department of Research and Advanced Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Fulvia Elia
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| | - Flora Desiderio
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| | - Fabio Valenti
- UOC Oncological Translational Research, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Vito Cantisani
- Department of Radiology, "Sapienza" University of Rome, Roma, Italy
| | - Antonella Soriani
- Medical Physics and Expert Systems Laboratory, Department of Research and Advanced Technologies, IRCCS-Regina Elena National Cancer Institute, Roma, Italy
| | - Mauro Caterino
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Roma, Italy
| |
Collapse
|
2
|
Ismail M, Hanifa MAM, Mahidin EIM, Manan HA, Yahya N. Cone beam computed tomography (CBCT) and megavoltage computed tomography (MVCT)-based radiomics in head and neck cancers: a systematic review and radiomics quality score assessment. Quant Imaging Med Surg 2024; 14:6963-6977. [PMID: 39281127 PMCID: PMC11400681 DOI: 10.21037/qims-24-334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/27/2024] [Indexed: 09/18/2024]
Abstract
Background Cone beam computed tomography (CBCT) and megavoltage computed tomography (MVCT)-based images demonstrate measurable radiomics features that are potentially prognostic. This study aims to systematically synthesize the current research applying radiomics in head and neck cancers for outcome prediction and to assess the radiomics quality score (RQS) of the studies. Methods A systematic search was performed to identify available studies on PubMed, Web of Science, and Scopus databases. Studies related to radiomics in oncology/radiotherapy fields and based on predefined Patient, Intervention, Comparator, Outcome, and Study design (PICOS) criteria were included. The methodological quality of the included study was evaluated independently by two reviewers according to the RQS. The Mann-Whitney U test was performed according to subgroups. The P values <0.05 were considered statistically significant. Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 reporting guidelines were adhered to. Results From a total of 743 identified studies, six original studies were eligible for inclusion in the systematic review (median =97 patients). The intraclass correlation coefficient (ICC) for inter-reviewer on total RQS was excellent with 0.99 [95% confidence interval (CI) of 0.946< ICC <0.999]. There were no significant differences in the analyses between each RQS domain and subgroup components (P always >0.05). Numerically higher RQS domains score for publication year ≤2022 than 2023 and number of patients > median than ≤ median but not statistically significant. Conclusions The number of radiomics studies involving CBCT and MVCT is still very limited. Self-reported RQS assessments should be encouraged for all radiomics studies.
Collapse
Affiliation(s)
- Mahayu Ismail
- Faculty of Health Sciences, National University of Malaysia, Jalan Raja Muda Aziz, Kuala Lumpur, Malaysia
- Department of Radiotherapy and Oncology, Hospital Kuala Lumpur, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Mohd Ariff Mohamed Hanifa
- Department of Radiotherapy and Oncology, Hospital Kuala Lumpur, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Eznal Izwadi Mohd Mahidin
- Department of Radiotherapy and Oncology, Hospital Kuala Lumpur, Jalan Pahang, Kuala Lumpur, Malaysia
| | - Hanani Abdul Manan
- Functional Image Processing Laboratory, Department of Radiology, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Noorazrul Yahya
- Faculty of Health Sciences, National University of Malaysia, Jalan Raja Muda Aziz, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Alabi RO, Elmusrati M, Leivo I, Almangush A, Mäkitie AA. Artificial Intelligence-Driven Radiomics in Head and Neck Cancer: Current Status and Future Prospects. Int J Med Inform 2024; 188:105464. [PMID: 38728812 DOI: 10.1016/j.ijmedinf.2024.105464] [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: 10/15/2023] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Radiomics is a rapidly growing field used to leverage medical radiological images by extracting quantitative features. These are supposed to characterize a patient's phenotype, and when combined with artificial intelligence techniques, to improve the accuracy of diagnostic models and clinical outcome prediction. OBJECTIVES This review aims at examining the application areas of artificial intelligence-based radiomics (AI-based radiomics) for the management of head and neck cancer (HNC). It further explores the workflow of AI-based radiomics for personalized and precision oncology in HNC. Finally, it examines the current challenges of AI-based radiomics in daily clinical oncology and offers possible solutions to these challenges. METHODS Comprehensive electronic databases (PubMed, Medline via Ovid, Scopus, Web of Science, CINAHL, and Cochrane Library) were searched following the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. The quality of included studies and their risk of biases were evaluated using the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD)and Prediction Model Risk of Bias Assessment Tool (PROBAST). RESULTS Out of the 659 search hits retrieved, 45 fulfilled the inclusion criteria. Our review revealed that the application of AI-based radiomics model as an ancillary tool for improved decision-making in HNC management includes radiomics-based cancer diagnosis and radiomics-based cancer prognosis. The radiomics-based cancer diagnosis includes tumor staging, tumor grading, and classification of malignant and benign tumors. Similarly, radiomics-based cancer prognosis includes prediction for treatment response, recurrence, metastasis, and survival. In addition, the challenges in the implementation of these models for clinical evaluations include data imbalance, feature engineering (extraction and selection), model generalizability, multi-modal fusion, and model interpretability. CONCLUSION Considering the highly subjective and interobserver variability that is peculiar to the interpretation of medical images by expert clinicians, AI-based radiomics seeks to offer potentially useful quantitative information, which is not visible to the human eye or unintentionally often remain ignored during clinical imaging practice. By enabling the extraction of this type of information, AI-based radiomics has the potential to revolutionize HNC oncology, providing a platform for more personalized, higher quality, and cost-effective care for HNC patients.
Collapse
Affiliation(s)
- Rasheed Omobolaji Alabi
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland.
| | - Mohammed Elmusrati
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Ilmo Leivo
- University of Turku, Institute of Biomedicine, Pathology, Turku, Finland
| | - Alhadi Almangush
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; University of Turku, Institute of Biomedicine, Pathology, Turku, Finland; Department of Pathology, University of Helsinki, Helsinki, Finland; Faculty of Dentistry, Misurata University, Misurata, Libya
| | - Antti A Mäkitie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
4
|
Dasgupta A, DiCenzo D, Sannachi L, Gandhi S, Pezo RC, Eisen A, Warner E, Wright FC, Look-Hong N, Sadeghi-Naini A, Curpen B, Kolios MC, Trudeau M, Czarnota GJ. Quantitative ultrasound radiomics guided adaptive neoadjuvant chemotherapy in breast cancer: early results from a randomized feasibility study. Front Oncol 2024; 14:1273437. [PMID: 38706611 PMCID: PMC11066296 DOI: 10.3389/fonc.2024.1273437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Background In patients with locally advanced breast cancer (LABC) receiving neoadjuvant chemotherapy (NAC), quantitative ultrasound (QUS) radiomics can predict final responses early within 4 of 16-18 weeks of treatment. The current study was planned to study the feasibility of a QUS-radiomics model-guided adaptive chemotherapy. Methods The phase 2 open-label randomized controlled trial included patients with LABC planned for NAC. Patients were randomly allocated in 1:1 ratio to a standard arm or experimental arm stratified by hormonal receptor status. All patients were planned for standard anthracycline and taxane-based NAC as decided by their medical oncologist. Patients underwent QUS imaging using a clinical ultrasound device before the initiation of NAC and after the 1st and 4th weeks of treatment. A support vector machine-based radiomics model developed from an earlier cohort of patients was used to predict treatment response at the 4th week of NAC. In the standard arm, patients continued to receive planned chemotherapy with the treating oncologists blinded to results. In the experimental arm, the QUS-based prediction was conveyed to the responsible oncologist, and any changes to the planned chemotherapy for predicted non-responders were made by the responsible oncologist. All patients underwent surgery following NAC, and the final response was evaluated based on histopathological examination. Results Between June 2018 and July 2021, 60 patients were accrued in the study arm, with 28 patients in each arm available for final analysis. In patients without a change in chemotherapy regimen (53 of 56 patients total), the QUS-radiomics model at week 4 of NAC that was used demonstrated an accuracy of 97%, respectively, in predicting the final treatment response. Seven patients were predicted to be non-responders (observational arm (n=2), experimental arm (n=5)). Three of 5 non-responders in the experimental arm had chemotherapy regimens adapted with an early initiation of taxane therapy or chemotherapy intensification, or early surgery and ended up as responders on final evaluation. Conclusion The study demonstrates the feasibility of QUS-radiomics adapted guided NAC for patients with breast cancer. The ability of a QUS-based model in the early prediction of treatment response was prospectively validated in the current study. Clinical trial registration clinicaltrials.gov, ID NCT04050228.
Collapse
Affiliation(s)
- Archya Dasgupta
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Sonal Gandhi
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rossana C. Pezo
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrea Eisen
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ellen Warner
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frances C. Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, ON, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | | | - Maureen Trudeau
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gregory J. Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Osapoetra LO, Dasgupta A, DiCenzo D, Fatima K, Quiaoit K, Saifuddin M, Karam I, Poon I, Husain Z, Tran WT, Sannachi L, Czarnota GJ. Quantitative US Delta Radiomics to Predict Radiation Response in Individuals with Head and Neck Squamous Cell Carcinoma. Radiol Imaging Cancer 2024; 6:e230029. [PMID: 38391311 PMCID: PMC10988345 DOI: 10.1148/rycan.230029] [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: 03/21/2023] [Revised: 11/24/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024]
Abstract
Purpose To investigate the role of quantitative US (QUS) radiomics data obtained after the 1st week of radiation therapy (RT) in predicting treatment response in individuals with head and neck squamous cell carcinoma (HNSCC). Materials and Methods This prospective study included 55 participants (21 with complete response [median age, 65 years {IQR: 47-80 years}, 20 male, one female; and 34 with incomplete response [median age, 59 years {IQR: 39-79 years}, 33 male, one female) with bulky node-positive HNSCC treated with curative-intent RT from January 2015 to October 2019. All participants received 70 Gy of radiation in 33-35 fractions over 6-7 weeks. US radiofrequency data from metastatic lymph nodes were acquired prior to and after 1 week of RT. QUS analysis resulted in five spectral maps from which mean values were extracted. We applied a gray-level co-occurrence matrix technique for textural analysis, leading to 20 QUS texture and 80 texture-derivative parameters. The response 3 months after RT was used as the end point. Model building and evaluation utilized nested leave-one-out cross-validation. Results Five delta (Δ) parameters had statistically significant differences (P < .05). The support vector machines classifier achieved a sensitivity of 71% (15 of 21), a specificity of 76% (26 of 34), a balanced accuracy of 74%, and an area under the receiver operating characteristic curve of 0.77 on the test set. For all the classifiers, the performance improved after the 1st week of treatment. Conclusion A QUS Δ-radiomics model using data obtained after the 1st week of RT from individuals with HNSCC predicted response 3 months after treatment completion with reasonable accuracy. Keywords: Computer-Aided Diagnosis (CAD), Ultrasound, Radiation Therapy/Oncology, Head/Neck, Radiomics, Quantitative US, Radiotherapy, Head and Neck Squamous Cell Carcinoma, Machine Learning Clinicaltrials.gov registration no. NCT03908684 Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
| | | | - Daniel DiCenzo
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Kashuf Fatima
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Karina Quiaoit
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Murtuza Saifuddin
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Irene Karam
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Ian Poon
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Zain Husain
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - William T. Tran
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Lakshmanan Sannachi
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| | - Gregory J. Czarnota
- From the Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P.,
Z.H., W.T.T., G.J.C.), Medical Oncology (W.T.T.), and Medicine (W.T.T.),
Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON, Canada M4N
3M5; Departments of Radiation Oncology (L.O.O., A.D., I.K., I.P., Z.H., W.T.T.,
G.J.C.) and Medical Biophysics (G.J.C.), University of Toronto, Toronto, Canada;
and Departments of Physical Sciences (L.O.O., A.D., D.D., K.F., K.Q., M.S.,
L.S., G.J.C.) and Evaluative Clinical Sciences (W.T.T.), Sunnybrook Research
Institute, Toronto, Canada
| |
Collapse
|
6
|
Mireștean CC, Iancu RI, Iancu DPT. Image Guided Radiotherapy (IGRT) and Delta (Δ) Radiomics-An Urgent Alliance for the Front Line of the War against Head and Neck Cancers. Diagnostics (Basel) 2023; 13:2045. [PMID: 37370940 DOI: 10.3390/diagnostics13122045] [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: 03/15/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The identification of a biomarker that is response predictive could offer a solution for the stratification of the treatment of head and neck cancers (HNC) in the context of high recurrence rates, especially those associated with loco-regional failure. Delta (Δ) radiomics, a concept based on the variation of parameters extracted from medical imaging using artificial intelligence (AI) algorithms, demonstrates its potential as a predictive biomarker of treatment response in HNC. The concept of image-guided radiotherapy (IGRT), including computer tomography simulation (CT) and position control imaging with cone-beam-computed tomography (CBCT), now offers new perspectives for radiomics applied in radiotherapy. The use of Δ features of texture, shape, and size, both from the primary tumor and from the tumor-involved lymph nodes, demonstrates the best predictive accuracy. If, in the case of treatment response, promising Δ radiomics results could be obtained, even after 24 h from the start of treatment, for radiation-induced xerostomia, the evaluation of Δ radiomics in the middle of treatment could be recommended. The fused models (clinical and Δ radiomics) seem to offer benefits, both in comparison to the clinical model and to the radiomic model. The selection of patients who benefit from induction chemotherapy is underestimated in Δ radiomic studies and may be an unexplored territory with major potential. The advantage offered by "in house" simulation CT and CBCT favors the rapid implementation of Δ radiomics studies in radiotherapy departments. Positron emission tomography (PET)-CT Δ radiomics could guide the new concepts of dose escalation on radio-resistant sub-volumes based on radiobiological criteria, but also guide the "next level" of HNC adaptive radiotherapy (ART).
Collapse
Affiliation(s)
- Camil Ciprian Mireștean
- Department of Oncology and Radiotherapy, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Surgery, Railways Clinical Hospital Iasi, 700506 Iași, Romania
| | - Roxana Irina Iancu
- Oral Pathology Department, "Gr. T. Popa" Faculty of Dental Medicine, University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Clinical Laboratory, "St. Spiridon" Emergency Universitary Hospital, 700111 Iași, Romania
| | - Dragoș Petru Teodor Iancu
- Oncology and Radiotherapy Department, Faculty of Medicine, "Gr. T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Radiation Oncology, Regional Institute of Oncology, 700483 Iași, Romania
| |
Collapse
|
7
|
Chiesa-Estomba CM, Mayo-Yanez M, Guntinas-Lichius O, Vander-Poorten V, Takes RP, de Bree R, Halmos GB, Saba NF, Nuyts S, Ferlito A. Radiomics in Hypopharyngeal Cancer Management: A State-of-the-Art Review. Biomedicines 2023; 11:805. [PMID: 36979783 PMCID: PMC10045560 DOI: 10.3390/biomedicines11030805] [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: 01/29/2023] [Revised: 02/25/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
(1) Background: Hypopharyngeal squamous cell carcinomas usually present with locally advanced disease and a correspondingly poor prognosis. Currently, efforts are being made to improve tumor characterization and provide insightful information for outcome prediction. Radiomics is an emerging area of study that involves the conversion of medical images into mineable data; these data are then used to extract quantitative features based on shape, intensity, texture, and other parameters; (2) Methods: A systematic review of the peer-reviewed literature was conducted; (3) Results: A total of 437 manuscripts were identified. Fifteen manuscripts met the inclusion criteria. The main targets described were the evaluation of textural features to determine tumor-programmed death-ligand 1 expression; a surrogate for microvessel density and heterogeneity of perfusion; patient stratification into groups at high and low risk of progression; prediction of early recurrence, 1-year locoregional failure and survival outcome, including progression-free survival and overall survival, in patients with locally advanced HPSCC; thyroid cartilage invasion, early disease progression, recurrence, induction chemotherapy response, treatment response, and prognosis; and (4) Conclusions: our findings suggest that radiomics represents a potentially useful tool in the diagnostic workup as well as during the treatment and follow-up of patients with HPSCC. Large prospective studies are essential to validate this technology in these patients.
Collapse
Affiliation(s)
- Carlos M. Chiesa-Estomba
- Otorhinolaryngology-Head & Neck Surgery Department, Hospital Universitario Donostia, Biodonostia Research Institute, Faculty of Medicine, Deusto University, 20014 San Sebastian, Spain
| | - Miguel Mayo-Yanez
- Otorhinolaryngology-Head and Neck Surgery Department, Complexo Hospitalario Universitario A Coruña (CHUAC), 15006 A Coruña, Spain
| | | | - Vincent Vander-Poorten
- Section Head and Neck Oncology, Department of Oncology, KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Robert P. Takes
- Department of Otolaryngology/Head and Neck Surgery, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Gyorgy B. Halmos
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, The Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Sandra Nuyts
- Department of Radiation Oncology, University Hospitals Leuven, KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Alfio Ferlito
- Coordinator of the International Head and Neck Scientific Group, 35125 Padua, Italy
| |
Collapse
|
8
|
Bang C, Bernard G, Le WT, Lalonde A, Kadoury S, Bahig H. Artificial intelligence to predict outcomes of head and neck radiotherapy. Clin Transl Radiat Oncol 2023; 39:100590. [PMID: 36935854 PMCID: PMC10014342 DOI: 10.1016/j.ctro.2023.100590] [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: 01/13/2023] [Revised: 01/28/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
Head and neck radiotherapy induces important toxicity, and its efficacy and tolerance vary widely across patients. Advancements in radiotherapy delivery techniques, along with the increased quality and frequency of image guidance, offer a unique opportunity to individualize radiotherapy based on imaging biomarkers, with the aim of improving radiation efficacy while reducing its toxicity. Various artificial intelligence models integrating clinical data and radiomics have shown encouraging results for toxicity and cancer control outcomes prediction in head and neck cancer radiotherapy. Clinical implementation of these models could lead to individualized risk-based therapeutic decision making, but the reliability of the current studies is limited. Understanding, validating and expanding these models to larger multi-institutional data sets and testing them in the context of clinical trials is needed to ensure safe clinical implementation. This review summarizes the current state of the art of machine learning models for prediction of head and neck cancer radiotherapy outcomes.
Collapse
Key Words
- ADASYN, adaptive synthetic sampling
- AI, artificial intelligence
- ANN, artificial neural network
- AUC, Area Under the ROC Curve
- Artificial intelligence
- BMI, body mass index
- C-Index, concordance index
- CART, Classification and Regression Tree
- CBCT, cone-beam computed tomography
- CIFE, conditional informax feature extraction
- CNN, convolutional neural network
- CRT, chemoradiation
- CT, computed tomography
- Cancer outcomes
- DL, deep learning
- DM, distant metastasis
- DSC, Dice Similarity Coefficient
- DSS, clinical decision support systems
- DT, Decision Tree
- DVH, Dose-volume histogram
- GANs, Generative Adversarial Networks
- GB, Gradient boosting
- GPU, graphical process units
- HNC, head and neck cancer
- HPV, human papillomavirus
- HR, hazard ratio
- Head and neck cancer
- IAMB, incremental association Markov blanket
- IBDM, image based data mining
- IBMs, image biomarkers
- IMRT, intensity-modulated RT
- KNN, k nearest neighbor
- LLR, Local linear forest
- LR, logistic regression
- LRR, loco-regional recurrence
- MIFS, mutual information based feature selection
- ML, machine learning
- MRI, Magnetic resonance imaging
- MRMR, Minimum redundancy feature selection
- Machine learning
- N-MLTR, Neural Multi-Task Logistic Regression
- NPC, nasopharynx
- NTCP, Normal Tissue Complication Probability
- OPC, oropharyngeal cancer
- ORN, osteoradionecrosis
- OS, overall survival
- PCA, Principal component analysis
- PET, Positron emission tomography
- PG, parotid glands
- PLR, Positive likelihood ratio
- PM, pharyngeal mucosa
- PTV, Planning target volumes
- PreSANet, deep preprocessor module and self-attention
- Predictive modeling
- QUANTEC, Quantitative Analyses of Normal Tissue Effects in the Clinic
- RF, random forest
- RFC, random forest classifier
- RFS, recurrence free survival
- RLR, Rigid logistic regression
- RRF, Regularized random forest
- RSF, random survival forest
- RT, radiotherapy
- RTLI, radiation-induced temporal lobe injury
- Radiomic
- SDM, shared decision making
- SMG, submandibular glands
- SMOTE, synthetic minority over-sampling technique
- STIC, sticky saliva
- SVC, support vector classifier
- SVM, support vector machine
- XGBoost, extreme gradient boosting
Collapse
Affiliation(s)
- Chulmin Bang
- Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Galaad Bernard
- Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - William T. Le
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Polytechnique Montréal, Montreal, QC, Canada
| | - Arthur Lalonde
- Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Université de Montréal, Montreal, QC, Canada
| | - Samuel Kadoury
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Polytechnique Montréal, Montreal, QC, Canada
| | - Houda Bahig
- Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
9
|
Caprini E, D'Agnese G, Brennan PA, Rahimi S. Human papilomaviru-related oropharyngeal squamous cell carcinoma and radiomics: A new era? J Oral Pathol Med 2023; 52:300-304. [PMID: 36847112 DOI: 10.1111/jop.13419] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/21/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND The increase of the incidence of human papillomavirus dependent oropharyngeal squamous cell carcinoma is alarming, although we have greatly progressed in the classification and staging of this disease. We now know that human papillomavirus related oropharyngeal squamous cell carcinoma is a sub-type of head and neck squamous cell carcinoma with favourable prognosis and good response to therapy that needs a proper system of classification and staging. Thus, in routine practice it is essential to test patients for the presence of human papillomavirus. The most popular technique to assess human papillomavirus status is immunohistochemistry on biopsy samples with p16, which is an excellent surrogate for high-risk human papillomavirus infection. Another highly sensitive and specific tissue-based technique for the detection of human papillomavirus is RNAscope In situ hybridization that has a prohibitive cost, limiting its use in routine practice. Radiomics is an artificial intelligence based non-invasive method of computational analysis of computed tomography, magnetic resonance imaging, positron emission tomography, and ultrasound images. METHODS In this review, we summarise the last findings of radiomics applied to human papillomavirus associated oropharyngeal squamous cell carcinoma. RESULTS A growing body of evidence suggest that radiomics is able to characterise and detect early relapse after treatment, and enable development of tailored therapy of human papillomavirus positive oropharyngeal squamous cell carcinoma.
Collapse
Affiliation(s)
- Elisabetta Caprini
- Anatomia Patologica, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Giampaolo D'Agnese
- Information Technology, Istituto Dermopatico dell'immacolata IDI-IRCCS, Rome, Italy
| | - Peter A Brennan
- Department of Oral and Maxillofacial Surgery, Queen Alexandra Hospital, Portsmouth, UK
| | - Siavash Rahimi
- Anatomia Patologica, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| |
Collapse
|
10
|
Current Role of Delta Radiomics in Head and Neck Oncology. Int J Mol Sci 2023; 24:ijms24032214. [PMID: 36768535 PMCID: PMC9916410 DOI: 10.3390/ijms24032214] [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: 11/27/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The latest developments in the management of head and neck cancer show an increasing trend in the implementation of novel approaches using artificial intelligence for better patient stratification and treatment-related risk evaluation. Radiomics, or the extraction of data from various imaging modalities, is a tool often used to evaluate specific features related to the tumour or normal tissue that are not identifiable by the naked eye and which can add value to existing clinical data. Furthermore, the assessment of feature variations from one time point to another based on subsequent images, known as delta radiomics, was shown to have even higher value for treatment-outcome prediction or patient stratification into risk categories. The information gathered from delta radiomics can, further, be used for decision making regarding treatment adaptation or other interventions found to be beneficial to the patient. The aim of this work is to collate the existing studies on delta radiomics in head and neck cancer and evaluate its role in tumour response and normal-tissue toxicity predictions alike. Moreover, this work also highlights the role of holomics, which brings under the same umbrella clinical and radiomic features, for a more complex patient characterization and treatment optimisation.
Collapse
|
11
|
Implementation of Non-Invasive Quantitative Ultrasound in Clinical Cancer Imaging. Cancers (Basel) 2022; 14:cancers14246217. [PMID: 36551702 PMCID: PMC9776858 DOI: 10.3390/cancers14246217] [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: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Quantitative ultrasound (QUS) is a non-invasive novel technique that allows treatment response monitoring. Studies have shown that QUS backscatter variables strongly correlate with changes observed microscopically. Increases in cell death result in significant alterations in ultrasound backscatter parameters. In particular, the parameters related to scatterer size and scatterer concentration tend to increase in relation to cell death. The use of QUS in monitoring tumor response has been discussed in several preclinical and clinical studies. Most of the preclinical studies have utilized QUS for evaluating cell death response by differentiating between viable cells and dead cells. In addition, clinical studies have incorporated QUS mostly for tissue characterization, including classifying benign versus malignant breast lesions, as well as responder versus non-responder patients. In this review, we highlight some of the important findings of previous preclinical and clinical studies and expand the applicability and therapeutic benefits of QUS in clinical settings. We summarized some recent clinical research advances in ultrasound-based radiomics analysis for monitoring and predicting treatment response and characterizing benign and malignant breast lesions. We also discuss current challenges, limitations, and future prospects of QUS-radiomics.
Collapse
|
12
|
MR-Guided Adaptive Radiotherapy for OAR Sparing in Head and Neck Cancers. Cancers (Basel) 2022; 14:cancers14081909. [PMID: 35454816 PMCID: PMC9028510 DOI: 10.3390/cancers14081909] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Normal tissue toxicities in head and neck cancer persist as a cause of decreased quality of life and are associated with poorer treatment outcomes. The aim of this article is to review organ at risk (OAR) sparing approaches available in MR-guided adaptive radiotherapy and present future developments which hope to improve treatment outcomes. Increasing the spatial conformity of dose distributions in radiotherapy is an important first step in reducing normal tissue toxicities, and MR-guided treatment devices presents a new opportunity to use biological information to drive treatment decisions on a personalized basis. Abstract MR-linac devices offer the potential for advancements in radiotherapy (RT) treatment of head and neck cancer (HNC) by using daily MR imaging performed at the time and setup of treatment delivery. This article aims to present a review of current adaptive RT (ART) methods on MR-Linac devices directed towards the sparing of organs at risk (OAR) and a view of future adaptive techniques seeking to improve the therapeutic ratio. This ratio expresses the relationship between the probability of tumor control and the probability of normal tissue damage and is thus an important conceptual metric of success in the sparing of OARs. Increasing spatial conformity of dose distributions to target volume and OARs is an initial step in achieving therapeutic improvements, followed by the use of imaging and clinical biomarkers to inform the clinical decision-making process in an ART paradigm. Pre-clinical and clinical findings support the incorporation of biomarkers into ART protocols and investment into further research to explore imaging biomarkers by taking advantage of the daily MR imaging workflow. A coherent understanding of this road map for RT in HNC is critical for directing future research efforts related to sparing OARs using image-guided radiotherapy (IGRT).
Collapse
|
13
|
Bhardwaj D, Dasgupta A, DiCenzo D, Brade S, Fatima K, Quiaoit K, Trudeau M, Gandhi S, Eisen A, Wright F, Look-Hong N, Curpen B, Sannachi L, Czarnota GJ. Early Changes in Quantitative Ultrasound Imaging Parameters during Neoadjuvant Chemotherapy to Predict Recurrence in Patients with Locally Advanced Breast Cancer. Cancers (Basel) 2022; 14:cancers14051247. [PMID: 35267555 PMCID: PMC8909335 DOI: 10.3390/cancers14051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study was conducted to explore the use of quantitative ultrasound (QUS) in predicting recurrence for patients with locally advanced breast cancer (LABC) early during neoadjuvant chemotherapy (NAC). METHODS Eighty-three patients with LABC were scanned with 7 MHz ultrasound before starting NAC (week 0) and during treatment (week 4). Spectral parametric maps were generated corresponding to tumor volume. Twenty-four textural features (QUS-Tex1) were determined from parametric maps acquired using grey-level co-occurrence matrices (GLCM) for each patient, which were further processed to generate 64 texture derivatives (QUS-Tex1-Tex2), leading to a total of 95 features from each time point. Analysis was carried out on week 4 data and compared to baseline (week 0) data. ∆Week 4 data was obtained from the difference in QUS parameters, texture features (QUS-Tex1), and texture derivatives (QUS-Tex1-Tex2) of week 4 data and week 0 data. Patients were divided into two groups: recurrence and non-recurrence. Machine learning algorithms using k-nearest neighbor (k-NN) and support vector machines (SVMs) were used to generate radiomic models. Internal validation was undertaken using leave-one patient out cross-validation method. RESULTS With a median follow up of 69 months (range 7-118 months), 28 patients had disease recurrence. The k-NN classifier was the best performing algorithm at week 4 with sensitivity, specificity, accuracy, and area under curve (AUC) of 87%, 75%, 81%, and 0.83, respectively. The inclusion of texture derivatives (QUS-Tex1-Tex2) in week 4 QUS data analysis led to the improvement of the classifier performances. The AUC increased from 0.70 (0.59 to 0.79, 95% confidence interval) without texture derivatives to 0.83 (0.73 to 0.92) with texture derivatives. The most relevant features separating the two groups were higher-order texture derivatives obtained from scatterer diameter and acoustic concentration-related parametric images. CONCLUSIONS This is the first study highlighting the utility of QUS radiomics in the prediction of recurrence during the treatment of LABC. It reflects that the ongoing treatment-related changes can predict clinical outcomes with higher accuracy as compared to pretreatment features alone.
Collapse
Affiliation(s)
- Divya Bhardwaj
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Archya Dasgupta
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Stephen Brade
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Kashuf Fatima
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Karina Quiaoit
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Maureen Trudeau
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Sonal Gandhi
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Andrea Eisen
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Frances Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Medical Imaging, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Lakshmanan Sannachi
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +416-480-6128
| |
Collapse
|
14
|
Zhang X, Zhang Y, Zhang G, Qiu X, Tan W, Yin X, Liao L. Deep Learning With Radiomics for Disease Diagnosis and Treatment: Challenges and Potential. Front Oncol 2022; 12:773840. [PMID: 35251962 PMCID: PMC8891653 DOI: 10.3389/fonc.2022.773840] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
The high-throughput extraction of quantitative imaging features from medical images for the purpose of radiomic analysis, i.e., radiomics in a broad sense, is a rapidly developing and emerging research field that has been attracting increasing interest, particularly in multimodality and multi-omics studies. In this context, the quantitative analysis of multidimensional data plays an essential role in assessing the spatio-temporal characteristics of different tissues and organs and their microenvironment. Herein, recent developments in this method, including manually defined features, data acquisition and preprocessing, lesion segmentation, feature extraction, feature selection and dimension reduction, statistical analysis, and model construction, are reviewed. In addition, deep learning-based techniques for automatic segmentation and radiomic analysis are being analyzed to address limitations such as rigorous workflow, manual/semi-automatic lesion annotation, and inadequate feature criteria, and multicenter validation. Furthermore, a summary of the current state-of-the-art applications of this technology in disease diagnosis, treatment response, and prognosis prediction from the perspective of radiology images, multimodality images, histopathology images, and three-dimensional dose distribution data, particularly in oncology, is presented. The potential and value of radiomics in diagnostic and therapeutic strategies are also further analyzed, and for the first time, the advances and challenges associated with dosiomics in radiotherapy are summarized, highlighting the latest progress in radiomics. Finally, a robust framework for radiomic analysis is presented and challenges and recommendations for future development are discussed, including but not limited to the factors that affect model stability (medical big data and multitype data and expert knowledge in medical), limitations of data-driven processes (reproducibility and interpretability of studies, different treatment alternatives for various institutions, and prospective researches and clinical trials), and thoughts on future directions (the capability to achieve clinical applications and open platform for radiomics analysis).
Collapse
Affiliation(s)
- Xingping Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Yanchun Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Guijuan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, China
| | - Xiaoxia Yin
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| |
Collapse
|
15
|
Application of Machine Learning Methods to Improve the Performance of Ultrasound in Head and Neck Oncology: A Literature Review. Cancers (Basel) 2022; 14:cancers14030665. [PMID: 35158932 PMCID: PMC8833587 DOI: 10.3390/cancers14030665] [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: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Ultrasound (US) is a non-invasive imaging method that is routinely utilized in head and neck cancer patients to assess the anatomic extent of tumors, nodal and non-nodal neck masses and for imaging the salivary glands. In this review, we summarize the present evidence on whether the application of machine learning (ML) methods can potentially improve the performance of US in head and neck cancer patients. We found that published clinical literature on ML methods applied to US datasets was limited but showed evidence of improved diagnostic and prognostic performance. However, a majority of these studies were based on retrospective evaluation and conducted at a single center with a limited number of datasets. The conduct of multi-center studies could help better validate the performance of ML-based US radiomics and facilitate the integration of these approaches into routine clinical practice. Abstract Radiomics is a rapidly growing area of research within radiology that involves the extraction and modeling of high-dimensional quantitative imaging features using machine learning/artificial intelligence (ML/AI) methods. In this review, we describe the published clinical evidence on the application of ML methods to improve the performance of ultrasound (US) in head and neck oncology. A systematic search of electronic databases (MEDLINE, PubMed, clinicaltrials.gov) was conducted according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Of 15,080 initial articles identified, 34 studies were selected for in-depth analysis. Twenty-five out of 34 studies (74%) focused on the diagnostic application of US radiomics while 6 (18%) studies focused on response assessment and 3 (8%) studies utilized US radiomics for modeling normal tissue toxicity. Support vector machine (SVM) was the most commonly employed ML method (47%) followed by multivariate logistic regression (24%) and k-nearest neighbor analysis (21%). Only 11/34 (~32%) of the studies included an independent validation set. A majority of studies were retrospective in nature (76%) and based on single-center evaluation (85%) with variable numbers of patients (12–1609) and imaging datasets (32–1624). Despite these limitations, the application of ML methods resulted in improved diagnostic and prognostic performance of US highlighting the potential clinical utility of this approach.
Collapse
|
16
|
Nardone V, Reginelli A, Grassi R, Boldrini L, Vacca G, D'Ippolito E, Annunziata S, Farchione A, Belfiore MP, Desideri I, Cappabianca S. Delta radiomics: a systematic review. Radiol Med 2021; 126:1571-1583. [PMID: 34865190 DOI: 10.1007/s11547-021-01436-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Radiomics can provide quantitative features from medical imaging that can be correlated with various biological features and clinical endpoints. Delta radiomics, on the other hand, consists in the analysis of feature variation at different acquisition time points, usually before and after therapy. The aim of this study was to provide a systematic review of the different delta radiomics approaches. METHODS Eligible articles were searched in Embase, PubMed, and ScienceDirect using a search string that included free text and/or Medical Subject Headings (MeSH) with three key search terms: "radiomics", "texture", and "delta". Studies were analysed using QUADAS-2 and the RQS tool. RESULTS Forty-eight studies were finally included. The studies were divided into preclinical/methodological (five studies, 10.4%); rectal cancer (six studies, 12.5%); lung cancer (twelve studies, 25%); sarcoma (five studies, 10.4%); prostate cancer (three studies, 6.3%), head and neck cancer (six studies, 12.5%); gastrointestinal malignancies excluding rectum (seven studies, 14.6%), and other disease sites (four studies, 8.3%). The median RQS of all studies was 25% (mean 21% ± 12%), with 13 studies (30.2%) achieving a quality score < 10% and 22 studies (51.2%) < 25%. CONCLUSIONS Delta radiomics shows potential benefit for several clinical endpoints in oncology (differential diagnosis, prognosis and prediction of treatment response, and evaluation of side effects). Nevertheless, the studies included in this systematic review suffer from the bias of overall low quality, so that the conclusions are currently heterogeneous, not robust, and not replicable. Further research with prospective and multicentre studies is needed for the clinical validation of delta radiomics approaches.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy.
| | - Roberta Grassi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Luca Boldrini
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giovanna Vacca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Emma D'Ippolito
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Salvatore Annunziata
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Alessandra Farchione
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia - Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Maria Paola Belfiore
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
17
|
Guiot J, Vaidyanathan A, Deprez L, Zerka F, Danthine D, Frix AN, Lambin P, Bottari F, Tsoutzidis N, Miraglio B, Walsh S, Vos W, Hustinx R, Ferreira M, Lovinfosse P, Leijenaar RTH. A review in radiomics: Making personalized medicine a reality via routine imaging. Med Res Rev 2021; 42:426-440. [PMID: 34309893 DOI: 10.1002/med.21846] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
Radiomics is the quantitative analysis of standard-of-care medical imaging; the information obtained can be applied within clinical decision support systems to create diagnostic, prognostic, and/or predictive models. Radiomics analysis can be performed by extracting hand-crafted radiomics features or via deep learning algorithms. Radiomics has evolved tremendously in the last decade, becoming a bridge between imaging and precision medicine. Radiomics exploits sophisticated image analysis tools coupled with statistical elaboration to extract the wealth of information hidden inside medical images, such as computed tomography (CT), magnetic resonance (MR), and/or Positron emission tomography (PET) scans, routinely performed in the everyday clinical practice. Many efforts have been devoted in recent years to the standardization and validation of radiomics approaches, to demonstrate their usefulness and robustness beyond any reasonable doubts. However, the booming of publications and commercial applications of radiomics approaches warrant caution and proper understanding of all the factors involved to avoid "scientific pollution" and overly enthusiastic claims by researchers and clinicians alike. For these reasons the present review aims to be a guidebook of sorts, describing the process of radiomics, its pitfalls, challenges, and opportunities, along with its ability to improve clinical decision-making, from oncology and respiratory medicine to pharmacological and genotyping studies.
Collapse
Affiliation(s)
- Julien Guiot
- Department of Pneumology, University Hospital of Liège, Liège, Belgium
| | - Akshayaa Vaidyanathan
- Radiomics (Oncoradiomics SA), Liège, Belgium.,The D-Lab, Department of Precision Medicine, Department of Nuclear Medicine, GROW-School for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Louis Deprez
- Department of Radiology, University Hospital of Liège, Liège, Belgium
| | - Fadila Zerka
- Radiomics (Oncoradiomics SA), Liège, Belgium.,The D-Lab, Department of Precision Medicine, Department of Nuclear Medicine, GROW-School for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Denis Danthine
- Department of Radiology, University Hospital of Liège, Liège, Belgium
| | - Anne-Noelle Frix
- Department of Pneumology, University Hospital of Liège, Liège, Belgium
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, Department of Nuclear Medicine, GROW-School for Oncology, Maastricht University, Maastricht, The Netherlands
| | | | | | | | - Sean Walsh
- Radiomics (Oncoradiomics SA), Liège, Belgium
| | - Wim Vos
- Radiomics (Oncoradiomics SA), Liège, Belgium
| | - Roland Hustinx
- Department of Nuclear Medicine and Oncological Imaging, University Hospital of Liege, Liege, Belgium.,GIGA-CRC in vivo imaging, University of Liège, Liège, Belgium
| | - Marta Ferreira
- GIGA-CRC in vivo imaging, University of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Department of Nuclear Medicine and Oncological Imaging, University Hospital of Liege, Liege, Belgium
| | | |
Collapse
|