1
|
Sharma D, Sannachi L, Osapoetra LO, Cartar H, Cui W, Giles A, Czarnota GJ. Noninvasive Evaluation of Breast Tumor Response to Combined Ultrasound-Stimulated Microbubbles and Hyperthermia Therapy Using Quantitative Ultrasound-Based Texture Analysis Method. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:137-150. [PMID: 37873733 DOI: 10.1002/jum.16347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/19/2023] [Accepted: 09/23/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVES Quantitative ultrasound (QUS) is a noninvasive imaging technique that can be used for assessing response to anticancer treatment. In the present study, tumor cell death response to the ultrasound-stimulated microbubbles (USMB) and hyperthermia (HT) treatment was monitored in vivo using QUS. METHODS Human breast cancer cell lines (MDA-MB-231) were grown in mice and were treated with HT (10, 30, 50, and 60 minutes) alone, or in combination with USMB. Treatment effects were examined using QUS with a center frequency of 25 MHz (bandwidth range: 16 to 32 MHz). Backscattered radiofrequency (RF) data were acquired from tumors subjected to treatment. Ultrasound parameters such as average acoustic concentration (AAC) and average scatterer diameter (ASD), were estimated 24 hours prior and posttreatment. Additionally, texture features: contrast (CON), correlation (COR), energy (ENE), and homogeneity (HOM) were extracted from QUS parametric maps. All estimated parameters were compared with histopathological findings. RESULTS The findings of our study demonstrated a significant increase in QUS parameters in both treatment conditions: HT alone (starting from 30 minutes of heat exposure) and combined treatment of HT plus USMB finally reaching a maximum at 50 minutes of heat exposure. Increase in AAC for 50 minutes HT alone and USMB +50 minutes was found to be 5.19 ± 0.417% and 5.91 ± 1.11%, respectively, compared to the control group with AAC value of 1.00 ± 0.44%. Furthermore, between the treatment groups, ΔASD-ENE values for USMB +30 minutes HT significantly reduced, depicting 0.00062 ± 0.00096% compared to 30 minutes HT only group, showing 0.0058 ± 0.0013%. Further, results obtained from the histological analysis indicated greater cell death and reduced nucleus size in both HT alone and HT combined with USMB. CONCLUSION The texture-based QUS parameters indicated a correlation with microstructural changes obtained from histological data. This work demonstrated the use of QUS to detect HT treatment effects in breast cancer tumors in vivo.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Lakshmanan Sannachi
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Laurentius Oscar Osapoetra
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Holliday Cartar
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Wentao Cui
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Anoja Giles
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Sannachi L, Osapoetra LO, DiCenzo D, Halstead S, Wright F, Look-Hong N, Slodkowska E, Gandhi S, Curpen B, Kolios MC, Oelze M, Czarnota GJ. A priori prediction of breast cancer response to neoadjuvant chemotherapy using quantitative ultrasound, texture derivative and molecular subtype. Sci Rep 2023; 13:22687. [PMID: 38114526 PMCID: PMC10730572 DOI: 10.1038/s41598-023-49478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023] Open
Abstract
The purpose of this study was to investigate the performances of the tumor response prediction prior to neoadjuvant chemotherapy based on quantitative ultrasound, tumour core-margin, texture derivative analyses, and molecular parameters in a large cohort of patients (n = 208) with locally advanced and earlier-stage breast cancer and combined them to best determine tumour responses with machine learning approach. Two multi-features response prediction algorithms using a k-nearest neighbour and support vector machine were developed with leave-one-out and hold-out cross-validation methods to evaluate the performance of the response prediction models. In a leave-one-out approach, the quantitative ultrasound-texture analysis based model attained good classification performance with 80% of accuracy and AUC of 0.83. Including molecular subtype in the model improved the performance to 83% of accuracy and 0.87 of AUC. Due to limited number of samples in the training process, a model developed with a hold-out approach exhibited a slightly higher bias error in classification performance. The most relevant features selected in predicting the response groups are core-to-margin, texture-derivative, and molecular subtype. These results imply that that baseline tumour-margin, texture derivative analysis methods combined with molecular subtype can potentially be used for the prediction of ultimate treatment response in patients prior to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Lakshmanan Sannachi
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075, Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Laurentius O Osapoetra
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075, Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Daniel DiCenzo
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075, Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Schontal Halstead
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075, Bayview Avenue, Toronto, ON, M4N 3M5, Canada
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Frances Wright
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Nicole Look-Hong
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Elzbieta Slodkowska
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sonal Gandhi
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Michael Oelze
- Department of Electrical and Computer Engineering, Univerity of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Gregory J Czarnota
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075, Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Sukhadia SS, Muller KE, Workman AA, Nagaraj SH. Machine Learning-Based Prediction of Distant Recurrence in Invasive Breast Carcinoma Using Clinicopathological Data: A Cross-Institutional Study. Cancers (Basel) 2023; 15:3960. [PMID: 37568776 PMCID: PMC10416932 DOI: 10.3390/cancers15153960] [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: 06/09/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer is the most common type of cancer worldwide. Alarmingly, approximately 30% of breast cancer cases result in disease recurrence at distant organs after treatment. Distant recurrence is more common in some subtypes such as invasive breast carcinoma (IBC). While clinicians have utilized several clinicopathological measurements to predict distant recurrences in IBC, no studies have predicted distant recurrences by combining clinicopathological evaluations of IBC tumors pre- and post-therapy with machine learning (ML) models. The goal of our study was to determine whether classification-based ML techniques could predict distant recurrences in IBC patients using key clinicopathological measurements, including pathological staging of the tumor and surrounding lymph nodes assessed both pre- and post-neoadjuvant therapy, response to therapy via standard-of-care imaging, and binary status of adjuvant therapy administered to patients. We trained and tested four clinicopathological ML models using a dataset (144 and 17 patients for training and testing, respectively) from Duke University and validated the best-performing model using an external dataset (8 patients) from Dartmouth Hitchcock Medical Center. The random forest model performed better than the C-support vector classifier, multilayer perceptron, and logistic regression models, yielding AUC values of 1.0 in the testing set and 0.75 in the validation set (p < 0.002) across both institutions, thereby demonstrating the cross-institutional portability and validity of ML models in the field of clinical research in cancer. The top-ranking clinicopathological measurement impacting the prediction of distant recurrences in IBC were identified to be tumor response to neoadjuvant therapy as evaluated via SOC imaging and pathology, which included tumor as well as node staging.
Collapse
Affiliation(s)
- Shrey S. Sukhadia
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH 03766, USA; (K.E.M.); (A.A.W.)
| | - Kristen E. Muller
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH 03766, USA; (K.E.M.); (A.A.W.)
| | - Adrienne A. Workman
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH 03766, USA; (K.E.M.); (A.A.W.)
| | - Shivashankar H. Nagaraj
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
4
|
Kheirkhah N, Kornecki A, Czarnota GJ, Samani A, Sadeghi-Naini A. Enhanced full-inversion-based ultrasound elastography for evaluating tumor response to neoadjuvant chemotherapy in patients with locally advanced breast cancer. Phys Med 2023; 112:102619. [PMID: 37343438 DOI: 10.1016/j.ejmp.2023.102619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/15/2023] [Accepted: 06/05/2023] [Indexed: 06/23/2023] Open
Abstract
PURPOSE An enhanced ultrasound elastography technique is proposed for early assessment of locally advanced breast cancer (LABC) response to neoadjuvant chemotherapy (NAC). METHODS The proposed elastography technique inputs ultrasound radiofrequency data obtained through tissue quasi-static stimulation and adapts a strain refinement algorithm formulated based on fundamental principles of continuum mechanics, coupled with an iterative inverse finite element method to reconstruct the breast Young's modulus (E) images. The technique was explored for therapy response assessment using data acquired from 25 LABC patients before and at weeks 1, 2, and 4 after the NAC initiation (100 scans). The E ratio of tumor to the surrounding tissue was calculated at different scans and compared to the baseline for each patient. Patients' response to NAC was determined many months later using standard clinical and histopathological criteria. RESULTS Reconstructed E ratio changes obtained as early as one week after the NAC onset demonstrate very good separation between the two cohorts of responders and non-responders to NAC. Statistically significant differences were observed in the E ratio changes between the two patient cohorts at weeks 1 to 4 after treatment (p-value < 0.001; statistical power greater than 97%). A significant difference in axial strain ratio changes was observed only at week 4 (p-value = 0.01; statistical power = 76%). No significant difference was observed in tumor size changes at weeks 1, 2 or 4. CONCLUSION The proposed elastography technique demonstrates a high potential for chemotherapy response monitoring in LABC patients and superior performance compared to strain imaging.
Collapse
Affiliation(s)
- Niusha Kheirkhah
- School of Biomedical Engineering, Western University, London, ON, Canada
| | - Anat Kornecki
- Department of Medical Imaging, Western University, London, ON, Canada
| | - Gregory J Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Abbas Samani
- School of Biomedical Engineering, Western University, London, ON, Canada; Departments of Medical Biophysics, Western University, London, ON, Canada; Department of Electrical and Computer Engineering, Western University, London, ON, Canada; Imaging Research, Robarts Research Institute, Western University, London, ON, Canada
| | - Ali Sadeghi-Naini
- School of Biomedical Engineering, Western University, London, ON, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Electrical Engineering and Computer Science, York University, Toronto, ON, Canada.
| |
Collapse
|
5
|
Tai H, Margolis R, Li J, Hoyt K. H-Scan Ultrasound Monitoring of Breast Cancer Response to Chemotherapy and Validation With Diffusion-Weighted Magnetic Resonance Imaging. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:1297-1306. [PMID: 36468546 DOI: 10.1002/jum.16143] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 05/18/2023]
Abstract
OBJECTIVES H-scan ultrasound (US) imaging is a novel tissue characterization technique to detect apoptosis-induced changes in cancer cells after the initiation of effective drug treatment. The objective of the proposed research was to assess the sensitivity of 3-dimensional (3D) H-scan US technique for monitoring the response of breast cancer-bearing animals to neoadjuvant chemotherapy and correlate results to diffusion-weighted magnetic resonance imaging (DW-MRI) measurements of programmed cancer cell death. METHODS Experimental studies used female mice (N = 18) implanted with human breast cancer cells. Animals underwent H-scan US and DW-MRI imaging on days 0, 1, 3, 7, and 10. After imaging at day 0, breast tumor-bearing nude mice were treated biweekly with an apoptosis-inducing drug. Texture analysis of H-scan US images explored spatial relationships between local US scattering. At day 10, H-scan measurements were compared with DW-MRI-derived apparent diffusion coefficient (ADC) values and histological findings. RESULTS H-scan US imaging of low and high dose cisplatin-treated cancer-bearing animals revealed changes in image intensity suggesting a progressive decrease in aggregate US scatterer size that was not observed in control animals. Longitudinal trends discovered in H-scan US result matched with texture analysis and DW-MRI (P < .01). Further, analysis of the H-scan US image intensity and corresponding DW-MRI-derived ADC values revealed a strong linear correlation (R2 = .93, P < .001). These changes were due to cancer cell apoptotic activity and consider as early detectable biomarker during treatment. CONCLUSIONS The 3D H-scan technique holds promise for assisting clinicians in monitoring the early response of breast cancer tumor to neoadjuvant chemotherapy and adding value to traditional diagnostic ultrasound examinations.
Collapse
Affiliation(s)
- Haowei Tai
- Department of Electrical and Computer Engineering, University of Texas at Dallas, Richardson, Texas, USA
| | - Ryan Margolis
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA
| | - Junjie Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
6
|
Khairalseed M, Hoyt K. High-Resolution Ultrasound Characterization of Local Scattering in Cancer Tissue. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:951-960. [PMID: 36681609 PMCID: PMC9974749 DOI: 10.1016/j.ultrasmedbio.2022.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Ultrasound (US) has afforded an approach to tissue characterization for more than a decade. The challenge is to reveal hidden patterns in the US data that describe tissue function and pathology that cannot be seen in conventional US images. Our group has developed a high-resolution analysis technique for tissue characterization termed H-scan US, an imaging method used to interpret the relative size of acoustic scatterers. In the present study, the objective was to compare local H-scan US image intensity with registered histologic measurements made directly at the cellular level. Human breast cancer cells (MDA-MB 231, American Type Culture Collection, Manassas, VA, USA) were orthotopically implanted into female mice (N = 5). Tumors were allowed to grow for approximately 4 wk before the study started. In vivo imaging of tumor tissue was performed using a US system (Vantage 256, Verasonics Inc., Kirkland, WA, USA) equipped with a broadband capacitive micromachined ultrasonic linear array transducer (Kolo Medical, San Jose, CA, USA). A 15-MHz center frequency was used for plane wave imaging with five angles for spatial compounding. H-scan US image reconstruction involved use of parallel convolution filters to measure the relative strength of backscattered US signals. Color codes were applied to filter outputs to form the final H-scan US image display. For histologic processing, US imaging cross-sections were carefully marked on the tumor surface, and tumors were excised and sliced along the same plane. By use of optical microscopy, whole tumor tissue sections were scanned and digitized after nuclear staining. US images were interpolated to have the same number of pixels as the histology images and then spatially aligned. Each nucleus from the histologic sections was automatically segmented using custom MATLAB software (The MathWorks Inc., Natick, MA, USA). Nuclear size and spacing from the histology images were then compared with local H-scan US image features. Overall, local H-scan US image intensity exhibited a significant correlation with both cancer cell nuclear size (R2 > 0.27, p < 0.001) and the inverse relationship with nuclear spacing (R2 > 0.17, p < 0.001).
Collapse
Affiliation(s)
- Mawia Khairalseed
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas, USA.
| |
Collapse
|
7
|
Hoerig C, Wallace K, Wu M, Mamou J. Classification of Metastatic Lymph Nodes In Vivo Using Quantitative Ultrasound at Clinical Frequencies. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:787-801. [PMID: 36470739 DOI: 10.1016/j.ultrasmedbio.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/28/2022] [Accepted: 10/30/2022] [Indexed: 06/17/2023]
Abstract
Quantitative ultrasound (QUS) methods characterizing the backscattered echo signal have been of use in assessing tissue microstructure. High-frequency (30 MHz) QUS methods have been successful in detecting metastases in surgically excised lymph nodes (LNs), but limited evidence exists regarding the efficacy of QUS for evaluating LNs in vivo at clinical frequencies (2-10 MHz). In this study, a clinical scanner and 10-MHz linear probe were used to collect radiofrequency (RF) echo data of LNs in vivo from 19 cancer patients. QUS methods were applied to estimate parameters derived from the backscatter coefficient (BSC) and statistics of the envelope-detected RF signal. QUS parameters were used to train classifiers based on linear discriminant analysis (LDA) and support vector machines (SVMs). Two BSC-based parameters, scatterer diameter and acoustic concentration, were the most effective for accurately detecting metastatic LNs, with both LDA and SVMs achieving areas under the receiver operating characteristic (AUROC) curve ≥0.94. A strategy of classifying LNs based on the echo frame with the highest cancer probability improved performance to 88% specificity at 100% sensitivity (AUROC = 0.99). These results provide encouraging evidence that QUS applied at clinical frequencies may be effective at accurately identifying metastatic LNs in vivo, helping in diagnosis while reducing unnecessary biopsies and surgical treatments.
Collapse
Affiliation(s)
- Cameron Hoerig
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA.
| | | | - Maoxin Wu
- Department of Pathology, Stony Brook University, Stony Brook, New York, USA
| | - Jonathan Mamou
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
8
|
Sebastian JA, Strohm EM, Baranger J, Villemain O, Kolios MC, Simmons CA. Assessing engineered tissues and biomaterials using ultrasound imaging: In vitro and in vivo applications. Biomaterials 2023; 296:122054. [PMID: 36842239 DOI: 10.1016/j.biomaterials.2023.122054] [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: 07/12/2022] [Revised: 01/24/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023]
Abstract
Quantitative assessment of the structural, functional, and mechanical properties of engineered tissues and biomaterials is fundamental to their development for regenerative medicine applications. Ultrasound (US) imaging is a non-invasive, non-destructive, and cost-effective technique capable of longitudinal and quantitative monitoring of tissue structure and function across centimeter to sub-micron length scales. Here we present the fundamentals of US to contextualize its application for the assessment of biomaterials and engineered tissues, both in vivo and in vitro. We review key studies that demonstrate the versatility and broad capabilities of US for clinical and pre-clinical biomaterials research. Finally, we highlight emerging techniques that further extend the applications of US, including for ultrafast imaging of biomaterials and engineered tissues in vivo and functional monitoring of stem cells, organoids, and organ-on-a-chip systems in vitro.
Collapse
Affiliation(s)
- Joseph A Sebastian
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, Canada.
| | - Eric M Strohm
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
| | - Jérôme Baranger
- Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Olivier Villemain
- Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute of Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Canada; Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada.
| |
Collapse
|
9
|
Rafat M, Kaffas AE, Swarnakar A, Shostak A, Graves EE. Technical note: Noninvasive monitoring of normal tissue radiation damage using spectral quantitative ultrasound spectroscopy. Med Phys 2023; 50:1251-1256. [PMID: 36564922 PMCID: PMC9940792 DOI: 10.1002/mp.16184] [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: 07/03/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND While radiation therapy (RT) is a critical component of breast cancer therapy and is known to decrease overall local recurrence rates, recent studies have shown that normal tissue radiation damage may increase recurrence risk. Fibrosis is a well-known consequence of RT, but the specific sequence of molecular and mechanical changes induced by RT remains poorly understood. PURPOSE To improve cancer therapy outcomes, there is a need to understand the role of the irradiated tissue microenvironment in tumor recurrence. This study seeks to evaluate the use of spectral quantitative ultrasound (spectral QUS) for real time determination of the normal tissue characteristic radiation response and to correlate these results to molecular features in irradiated tissues. METHODS Murine mammary fat pads (MFPs) were irradiated to 20 Gy, and spectral QUS was used to analyze tissue physical properties pre-irradiation as well as at 1, 5, and 10 days post-irradiation. Tissues were processed for scanning electron microscopy imaging as well as histological and immunohistochemical staining to evaluate morphology and structure. RESULTS Tissue morphological and structural changes were observed non-invasively following radiation using mid-band fit (MBF), spectral slope (SS), and spectral intercept (SI) measurements obtained from spectral QUS. Statistically significant shifts in MBF and SI indicate structural tissue changes in real time, which matched histological observations. Radiation damage was indicated by increased adipose tissue density and extracellular matrix (ECM) deposition. CONCLUSIONS Our findings demonstrate the potential of using spectral QUS to noninvasively evaluate normal tissue changes resulting from radiation damage. This supports further pre-clinical studies to determine how the tissue microenvironment and physical properties change in response to therapy, which may be important for improving treatment strategies.
Collapse
Affiliation(s)
- Marjan Rafat
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212 USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ahmed El Kaffas
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Ankush Swarnakar
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Anastasia Shostak
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Edward E. Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
10
|
Feleppa EJ. Quantitative Ultrasound: An Emerging Technology for Detecting, Diagnosing, Imaging, Evaluating, and Monitoring Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1403:3-17. [PMID: 37495911 DOI: 10.1007/978-3-031-21987-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Ultrasound has been a popular clinical imaging modality for decades. It is a well-established means of displaying the macroscopic anatomy of soft-tissue structures. While conventional ultrasound methods, i.e., B-mode and Doppler methods, are well proven and continue to advance technically in many ways, e.g., by extending into higher frequencies and taking advantage of harmonic phenomena in tissues, fundamentally new so-called quantitative ultrasound (QUS) technologies also are emerging and offer exciting promise for making significant improvements in clinical imaging and characterization of disease. These emerging quantitative methods include spectrum analysis, image statistics, elasticity imaging, contrast-agent methods, and flow-detection and -measurement techniques. Each provides independent information. When used alone, each can provide clinically valuable imaging capabilities; when combined with each other, their capabilities may be more powerful in many applications. Furthermore, all can be used fused with other imaging modalities, such as computed tomography (CT), magnetic-resonance (MR), positron-emission-tomography (PET), or single-photon emission computerized tomography (SPECT) imaging, to offer possibly even greater improvements in detecting, diagnosing, imaging, evaluating, and monitoring disease. This chapter focuses on QUS methods that are based on spectrum analysis and image statistics.
Collapse
Affiliation(s)
- Ernest J Feleppa
- Department of Radiology, Massachusetts General Hospital, Center for Ultrasound Research and Translation (CURT), Boston, MA, USA
- Riverside Research, New York, NY, USA
| |
Collapse
|
11
|
Rheinheimer S, Christopoulos P, Erdmann S, Saupe J, Golpon H, Vogel-Claussen J, Dinkel J, Thomas M, Heussel CP, Kauczor HU, Heussel G. Dynamic contrast enhanced MRI of pulmonary adenocarcinomas for early risk stratification: higher contrast uptake associated with response and better prognosis. BMC Med Imaging 2022; 22:215. [PMID: 36471318 PMCID: PMC9724354 DOI: 10.1186/s12880-022-00943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To explore the prognostic value of serial dynamic contrast-enhanced (DCE) MRI in patients with advanced pulmonary adenocarcinoma undergoing first-line therapy with either tyrosine-kinase inhibitors (TKI) or platinum-based chemotherapy (PBC). METHODS Patients underwent baseline (day 0, n = 98), and post-therapeutic DCE MRI (PBC: day + 1, n = 52); TKI: day + 7, n = 46) at 1.5T. Perfusion curves were acquired at 10, 40, and 70 s after contrast application and analysed semiquantitatively. Treatment response was evaluated at 6 weeks by CT (RECIST 1.1); progression-free survival (PFS) and overall survival were analysed with respect to clinical and perfusion parameters. Relative uptake was defined as signal difference between contrast and non-contrast images, divided by the non-contrast signal. Predictors of survival were selected using Cox regression analysis. Median follow-up was 825 days. RESULTS In pre-therapeutic and early post-therapeutic MRI, treatment responders (n = 27) showed significantly higher relative contrast uptake within the tumor at 70 s after application as compared to non-responders (n = 71, p ≤ 0.02), response defined as PR by RECIST 1.1 at 6 weeks. There was no significant change of perfusion at early MRI after treatment. In multivariate regression analysis of selected parameters, the strongest association with PFS were relative uptake at 40 s in the early post-treatment MRI and pre-treatment clinical data (presence of liver metastases, ECOG performance status). CONCLUSION Higher contrast uptake within the tumor at pre-treatment and early post-treatment MRI was associated with treatment response and better prognosis. DCE MRI of pulmonary adenocarcinoma may provide important prognostic information.
Collapse
Affiliation(s)
- Stephan Rheinheimer
- grid.7700.00000 0001 2190 4373Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,Radiology, Asklepios Hospital Munich, Robert-Koch-Allee 2, 82131 Gauting, Germany
| | - Petros Christopoulos
- grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Thoracic Oncology, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Stella Erdmann
- Medical Biometry, Institute of Medical Biometry, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Julia Saupe
- grid.7700.00000 0001 2190 4373Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Heiko Golpon
- grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.10423.340000 0000 9529 9877Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Jens Vogel-Claussen
- grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,Diagnostic and Interventional Radiology and Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,grid.10423.340000 0000 9529 9877Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Julien Dinkel
- grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,Radiology, Asklepios Hospital Munich, Robert-Koch-Allee 2, 82131 Gauting, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Michael Thomas
- grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Thoracic Oncology, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Claus Peter Heussel
- grid.7700.00000 0001 2190 4373Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Hans-Ulrich Kauczor
- grid.5253.10000 0001 0328 4908Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| | - Gudula Heussel
- grid.7700.00000 0001 2190 4373Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Thoracic Oncology, Thoraxklinik at University of Heidelberg, Röntgenstrasse 1, 69126 Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
12
|
Cui Y, Yin FF. Impact of image quality on radiomics applications. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac7fd7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Radiomics features extracted from medical images have been widely reported to be useful in the patient specific outcome modeling for variety of assessment and prediction purposes. Successful application of radiomics features as imaging biomarkers, however, is dependent on the robustness of the approach to the variation in each step of the modeling workflow. Variation in the input image quality is one of the main sources that impacts the reproducibility of radiomics analysis when a model is applied to broader range of medical imaging data. The quality of medical image is generally affected by both the scanner related factors such as image acquisition/reconstruction settings and the patient related factors such as patient motion. This article aimed to review the published literatures in this field that reported the impact of various imaging factors on the radiomics features through the change in image quality. The literatures were categorized by different imaging modalities and also tabulated based on the imaging parameters and the class of radiomics features included in the study. Strategies for image quality standardization were discussed based on the relevant literatures and recommendations for reducing the impact of image quality variation on the radiomics in multi-institutional clinical trial were summarized at the end of this article.
Collapse
|
13
|
Tai H, Song J, Li J, Reddy S, Khairalseed M, Hoyt K. Three-Dimensional H-Scan Ultrasound Imaging of Early Breast Cancer Response to Neoadjuvant Therapy in a Murine Model. Invest Radiol 2022; 57:222-232. [PMID: 34652291 PMCID: PMC8916970 DOI: 10.1097/rli.0000000000000831] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Three-dimensional (3D) H-scan is a new ultrasound (US) technique that images the relative size of acoustic scatterers. The goal of this research was to evaluate use of 3D H-scan US imaging for monitoring early breast cancer response to neoadjuvant therapy using a preclinical murine model of breast cancer. MATERIALS AND METHODS Preclinical studies were conducted using luciferase-positive breast cancer-bearing mice (n = 40). Anesthetized animals underwent US imaging at baseline before administration with an apoptosis-inducing drug or a saline control. Image data were acquired using a US scanner equipped with a volumetric transducer following either a shorter- or longer-term protocol. The later included bioluminescent imaging to quantify tumor cell viability. At termination, tumors were excised for ex vivo analysis. RESULTS In vivo results showed that 3D H-scan US imaging is considerably more sensitive to tumor changes after apoptosis-inducing drug therapy as compared with traditional B-scan US. Although there was no difference at baseline (P > 0.99), H-scan US results from treated tumors exhibited progressive decreases in image intensity (up to 62.2% by day 3) that had a significant linear correlation with cancer cell nuclear size (R2 > 0.51, P < 0.001). Results were validated by histological data and a secondary longitudinal study with survival as the primary end point. DISCUSSION Experimental results demonstrate that noninvasive 3D H-scan US imaging can detect an early breast tumor response to apoptosis-inducing drug therapy. Local in vivo H-scan US image intensity correlated with cancer cell nuclear size, which is one of the first observable changes of a cancer cell undergoing apoptosis and confirmed using histological techniques. Early imaging results seem to provide prognostic insight on longer-term tumor response. Overall, 3D H-scan US imaging is a promising technique that visualizes the entire tumor and detects breast cancer response at an early stage of therapy.
Collapse
Affiliation(s)
- Haowei Tai
- Department of Electrical and Computer Engineering, University of Texas at Dallas, Richardson, Texas
| | - Jane Song
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Junjie Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Shreya Reddy
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Mawia Khairalseed
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
14
|
Breast Tumor Classification Using Intratumoral Quantitative Ultrasound Descriptors. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1633858. [PMID: 35295204 PMCID: PMC8920646 DOI: 10.1155/2022/1633858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer is a global epidemic, responsible for one of the highest mortality rates among women. Ultrasound imaging is becoming a popular tool for breast cancer screening, and quantitative ultrasound (QUS) techniques are being increasingly applied by researchers in an attempt to characterize breast tissue. Several different quantitative descriptors for breast cancer have been explored by researchers. This study proposes a breast tumor classification system using the three major types of intratumoral QUS descriptors which can be extracted from ultrasound radiofrequency (RF) data: spectral features, envelope statistics features, and texture features. A total of 16 features were extracted from ultrasound RF data across two different datasets, of which one is balanced and the other is severely imbalanced. The balanced dataset contains RF data of 100 patients with breast tumors, of which 48 are benign and 52 are malignant. The imbalanced dataset contains RF data of 130 patients with breast tumors, of which 104 are benign and 26 are malignant. Holdout validation was used to split the balanced dataset into 60% training and 40% testing sets. Feature selection was applied on the training set to identify the most relevant subset for the classification of benign and malignant breast tumors, and the performance of the features was evaluated on the test set. A maximum classification accuracy of 95% and an area under the receiver operating characteristic curve (AUC) of 0.968 was obtained on the test set. The performance of the identified relevant features was further validated on the imbalanced dataset, where a hybrid resampling strategy was firstly utilized to create an optimal balance between benign and malignant samples. A maximum classification accuracy of 93.01%, sensitivity of 94.62%, specificity of 91.4%, and AUC of 0.966 were obtained. The results indicate that the identified features are able to distinguish between benign and malignant breast lesions very effectively, and the combination of the features identified in this research has the potential to be a significant tool in the noninvasive rapid and accurate diagnosis of breast cancer.
Collapse
|
15
|
Deep learning of quantitative ultrasound multi-parametric images at pre-treatment to predict breast cancer response to chemotherapy. Sci Rep 2022; 12:2244. [PMID: 35145158 PMCID: PMC8831592 DOI: 10.1038/s41598-022-06100-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, a novel deep learning-based methodology was investigated to predict breast cancer response to neo-adjuvant chemotherapy (NAC) using the quantitative ultrasound (QUS) multi-parametric imaging at pre-treatment. QUS multi-parametric images of breast tumors were generated using the data acquired from 181 patients diagnosed with locally advanced breast cancer and planned for NAC followed by surgery. The ground truth response to NAC was identified for each patient after the surgery using the standard clinical and pathological criteria. Two deep convolutional neural network (DCNN) architectures including the residual network and residual attention network (RAN) were explored for extracting optimal feature maps from the parametric images, with a fully connected network for response prediction. In different experiments, the features maps were derived from the tumor core only, as well as the core and its margin. Evaluation results on an independent test set demonstrate that the developed model with the RAN architecture to extract feature maps from the expanded parametric images of the tumor core and margin had the best performance in response prediction with an accuracy of 88% and an area under the receiver operating characteristic curve of 0.86. Ten-year survival analyses indicate statistically significant differences between the survival of the responders and non-responders identified based on the model prediction at pre-treatment and the standard criteria at post-treatment. The results of this study demonstrate the promising capability of DCNNs with attention mechanisms in predicting breast cancer response to NAC prior to the start of treatment using QUS multi-parametric images.
Collapse
|
16
|
Can we use radiomics in ultrasound imaging? Impact of preprocessing on feature repeatability. Diagn Interv Imaging 2021; 102:659-667. [PMID: 34690106 DOI: 10.1016/j.diii.2021.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE The purpose of this study was to assess the inter-slice radiomic feature repeatability in ultrasound imaging and the impact of preprocessing using intensity standardization and grey-level discretization to help improve radiomics reproducibility. MATERIALS AND METHODS This single-center study enrolled consecutive patients with an orbital lesion who underwent ultrasound examination of the orbit from December 2015 to July 2019. Two images per lesion were randomly assigned to two subsets. Radiomic features were extracted and inter-slice repeatability was assessed using the intraclass correlation coefficient (ICC) between the subsets. The impact of preprocessing on feature repeatability was assessed using image intensity standardization with or without outliers removal on whole images, bounding boxes or regions of interest (ROI), and fixed bin size or fixed bin number grey-level discretization. Number of inter-slice repeatable features (ICC ≥0.7) between methods was compared. RESULTS Eighty-eight patients (37 men, 51 women) with a mean age of 51.5 ± 17 (SD) years (range: 20-88 years) were enrolled. Without preprocessing, 29/101 features (28.7%) were repeatable between slices. The greatest number of repeatable features (41/101) was obtained using intensity standardization with outliers removal on the ROI and fixed bin size discretization. Standardization performed better with outliers removal than without (P < 0.001), and on ROIs than on native images (P < 0.001). Fixed bin size discretization performed better than fixed bin number (P = 0.008). CONCLUSION Radiomic features extracted from ultrasound images are impacted by the slice and preprocessing. The use of intensity standardization with outliers removal applied to the ROI and a fixed bin size grey-level discretization may improve feature repeatability.
Collapse
|
17
|
Venkatasamy A, Guerin E, Blanchet A, Orvain C, Devignot V, Jung M, Jung AC, Chenard MP, Romain B, Gaiddon C, Mellitzer G. Ultrasound and Transcriptomics Identify a Differential Impact of Cisplatin and Histone Deacetylation on Tumor Structure and Microenvironment in a Patient-Derived In Vivo Model of Gastric Cancer. Pharmaceutics 2021; 13:1485. [PMID: 34575561 PMCID: PMC8467189 DOI: 10.3390/pharmaceutics13091485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/06/2023] Open
Abstract
The reasons behind the poor efficacy of transition metal-based chemotherapies (e.g., cisplatin) or targeted therapies (e.g., histone deacetylase inhibitors, HDACi) on gastric cancer (GC) remain elusive and recent studies suggested that the tumor microenvironment could contribute to the resistance. Hence, our objective was to gain information on the impact of cisplatin and the pan-HDACi SAHA (suberanilohydroxamic acid) on the tumor substructure and microenvironment of GC, by establishing patient-derived xenografts of GC and a combination of ultrasound, immunohistochemistry, and transcriptomics to analyze. The tumors responded partially to SAHA and cisplatin. An ultrasound gave more accurate tumor measures than a caliper. Importantly, an ultrasound allowed a noninvasive real-time access to the tumor substructure, showing differences between cisplatin and SAHA. These differences were confirmed by immunohistochemistry and transcriptomic analyses of the tumor microenvironment, identifying specific cell type signatures and transcription factor activation. For instance, cisplatin induced an "epithelial cell like" signature while SAHA favored a "mesenchymal cell like" one. Altogether, an ultrasound allowed a precise follow-up of the tumor progression while enabling a noninvasive real-time access to the tumor substructure. Combined with transcriptomics, our results underline the different intra-tumoral structural changes caused by both drugs that impact differently on the tumor microenvironment.
Collapse
Affiliation(s)
- Aina Venkatasamy
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
- IHU-Strasbourg (Institut Hospitalo-Universitaire), 67091 Strasbourg, France
| | - Eric Guerin
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| | - Anais Blanchet
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| | - Christophe Orvain
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| | - Véronique Devignot
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| | | | - Alain C. Jung
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
- Laboratoire de Biologie Tumorale, ICANS, 67200 Strasbourg, France
| | - Marie-Pierre Chenard
- Pathology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
| | - Benoit Romain
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
- Digestive Surgery Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | - Christian Gaiddon
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| | - Georg Mellitzer
- Streinth Lab (Stress Response and Innovative Therapies), Strasbourg University, Inserm UMR_S 1113 IRFAC (Interface Recherche Fondamental et Appliquée à la Cancérologie), 67200 Strasbourg, France; (A.V.); (E.G.); (A.B.); (C.O.); (V.D.); (A.C.J.); (B.R.)
| |
Collapse
|
18
|
Characterizing intra-tumor regions on quantitative ultrasound parametric images to predict breast cancer response to chemotherapy at pre-treatment. Sci Rep 2021; 11:14865. [PMID: 34290259 PMCID: PMC8295369 DOI: 10.1038/s41598-021-94004-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023] Open
Abstract
The efficacy of quantitative ultrasound (QUS) multi-parametric imaging in conjunction with unsupervised classification algorithms was investigated for the first time in characterizing intra-tumor regions to predict breast tumor response to chemotherapy before the start of treatment. QUS multi-parametric images of breast tumors were generated using the ultrasound radiofrequency data acquired from 181 patients diagnosed with locally advanced breast cancer and planned for neo-adjuvant chemotherapy followed by surgery. A hidden Markov random field (HMRF) expectation maximization (EM) algorithm was applied to identify distinct intra-tumor regions on QUS multi-parametric images. Several features were extracted from the segmented intra-tumor regions and tumor margin on different parametric images. A multi-step feature selection procedure was applied to construct a QUS biomarker consisting of four features for response prediction. Evaluation results on an independent test set indicated that the developed biomarker coupled with a decision tree model with adaptive boosting (AdaBoost) as the classifier could predict the treatment response of patient at pre-treatment with an accuracy of 85.4% and an area under the receiver operating characteristic (ROC) curve (AUC) of 0.89. In comparison, the biomarkers consisted of the features derived from the entire tumor core (without consideration of the intra-tumor regions), and the entire tumor core and the tumor margin could predict the treatment response of patients with an accuracy of 74.5% and 76.4%, and an AUC of 0.79 and 0.76, respectively. Standard clinical features could predict the therapy response with an accuracy of 69.1% and an AUC of 0.6. Long-term survival analyses indicated that the patients predicted by the developed model as responders had a significantly better survival compared to the non-responders. Similar findings were observed for the two response cohorts identified at post-treatment based on standard clinical and pathological criteria. The results obtained in this study demonstrated the potential of QUS multi-parametric imaging integrated with unsupervised learning methods in identifying distinct intra-tumor regions in breast cancer to characterize its responsiveness to chemotherapy prior to the start of treatment.
Collapse
|
19
|
Baek J, Parker KJ. H-scan trajectories indicate the progression of specific diseases. Med Phys 2021; 48:5047-5058. [PMID: 34287952 DOI: 10.1002/mp.15108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/18/2023] Open
Abstract
PURPOSE The ability of ultrasound to assess pathology is increasing with the development of quantitative parameters. Among these are a set of parameters derived from the recent H-scan analysis of subresolvable scattering. The emergence of these quantitative measures of tissue/ultrasound interactions now enables a study of the unique trajectories of multiparametric features in multidimensional space, representing the progression of specific diseases over time. We develop the mathematical and visual tools that are effective for classifying, quantifying, and visualizing the steady progression of several diseases from independent studies, all within a uniform framework. METHODS After applying the H-scan analysis of ultrasound echoes, we trained a support vector machine (SVM) to classify the unique trajectories of progressive liver disease from fibrosis, steatosis, and pancreatic ductal adenocarcinoma (PDAC) metastasis. Our approaches include the development of trajectory maps and disease-specific color imaging stains. RESULTS The multidimensional SVM image classification reached 100% accuracy across the three different studies. CONCLUSION H-scan trajectories can be useful to track the progression of multiple classes of diseases, improving diagnosis, staging, and assessing the response to therapy.
Collapse
Affiliation(s)
- Jihye Baek
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, New York, USA
| | - Kevin J Parker
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, New York, USA
| |
Collapse
|
20
|
Moghadas-Dastjerdi H, Rahman SETH, Sannachi L, Wright FC, Gandhi S, Trudeau ME, Sadeghi-Naini A, Czarnota GJ. Prediction of chemotherapy response in breast cancer patients at pre-treatment using second derivative texture of CT images and machine learning. Transl Oncol 2021; 14:101183. [PMID: 34293685 PMCID: PMC8319580 DOI: 10.1016/j.tranon.2021.101183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023] Open
Abstract
Textural and second derivative textural features of CT images can be used in conjunction with machine learning models to predict breast cancer response to chemotherapy prior to the start of treatment. The proposed predictive model separates the patients at pre-treatment into two cohorts (responders/non-responders) with significantly different survival. The proposed methodology is a step forward towards the precision oncology paradigm for breast cancer patients.
Although neoadjuvant chemotherapy (NAC) is a crucial component of treatment for locally advanced breast cancer (LABC), only about 70% of patients respond to it. Effective adjustment of NAC for individual patients can significantly improve survival rates of those resistant to standard regimens. Thus, the early prediction of NAC outcome is of great importance in facilitating a personalized paradigm for breast cancer therapeutics. In this study, quantitative computed tomography (qCT) parametric imaging in conjunction with machine learning techniques were investigated to predict LABC tumor response to NAC. Textural and second derivative textural (SDT) features of CT images of 72 patients diagnosed with LABC were analysed before the initiation of NAC to quantify intra-tumor heterogeneity. These quantitative features were processed through a correlation-based feature reduction followed by a sequential feature selection with a bootstrap 0.632+ area under the receiver operating characteristic (ROC) curve (AUC0.632+) criterion. The best feature subset consisted of a combination of one textural and three SDT features. Using these features, an AdaBoost decision tree could predict the patient response with a cross-validated AUC0.632+ accuracy, sensitivity and specificity of 0.88, 85%, 88% and 75%, respectively. This study demonstrates, for the first time, that a combination of textural and SDT features of CT images can be used to predict breast cancer response NAC prior to the start of treatment which can potentially facilitate early therapy adjustments.
Collapse
Affiliation(s)
- Hadi Moghadas-Dastjerdi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Shan-E-Tallat Hira Rahman
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Lakshmanan Sannachi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Frances C Wright
- Surgical Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, and Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Sonal Gandhi
- Division of Medical Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, and Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Maureen E Trudeau
- Division of Medical Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, and Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ali Sadeghi-Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada.
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Byra M, Dobruch-Sobczak K, Klimonda Z, Piotrzkowska-Wroblewska H, Litniewski J. Early Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer Sonography Using Siamese Convolutional Neural Networks. IEEE J Biomed Health Inform 2021; 25:797-805. [PMID: 32749986 DOI: 10.1109/jbhi.2020.3008040] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Early prediction of response to neoadjuvant chemotherapy (NAC) in breast cancer is crucial for guiding therapy decisions. In this work, we propose a deep learning based approach for the early NAC response prediction in ultrasound (US) imaging. We used transfer learning with deep convolutional neural networks (CNNs) to develop the response prediction models. The usefulness of two transfer learning techniques was examined. First, a CNN pre-trained on the ImageNet dataset was utilized. Second, we applied double transfer learning, the CNN pre-trained on the ImageNet dataset was additionally fine-tuned with breast mass US images to differentiate malignant and benign lesions. Two prediction tasks were investigated. First, a L1 regularized logistic regression prediction model was developed based on generic neural features extracted from US images collected before the chemotherapy (a priori prediction). Second, Siamese CNNs were used to quantify differences between US images collected before the treatment and after the first and second course of NAC. The proposed methods were evaluated using US data collected from 39 tumors. The better performing deep learning models achieved areas under the receiver operating characteristic curve of 0.797 and 0.847 in the case of the a priori prediction and the Siamese model, respectively. The proposed approach was compared with a method based on handcrafted morphological features. Our study presents the feasibility of using transfer learning with CNNs for the NAC response prediction in US imaging.
Collapse
|
22
|
Wiskin J, Malik B, Borup D, Pirshafiey N, Klock J. Full wave 3D inverse scattering transmission ultrasound tomography in the presence of high contrast. Sci Rep 2020; 10:20166. [PMID: 33214569 PMCID: PMC7677558 DOI: 10.1038/s41598-020-76754-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/02/2020] [Indexed: 12/29/2022] Open
Abstract
We present here a quantitative ultrasound tomographic method yielding a sub-mm resolution, quantitative 3D representation of tissue characteristics in the presence of high contrast media. This result is a generalization of previous work where high impedance contrast was not present and may provide a clinically and laboratory relevant, relatively inexpensive, high resolution imaging method for imaging in the presence of bone. This allows tumor, muscle, tendon, ligament or cartilage disease monitoring for therapy and general laboratory or clinical settings. The method has proven useful in breast imaging and is generalized here to high-resolution quantitative imaging in the presence of bone. The laboratory data are acquired in ~ 12 min and the reconstruction in ~ 24 min-approximately 200 times faster than previously reported simulations in the literature. Such fast reconstructions with real data require careful calibration, adequate data redundancy from a 2D array of 2048 elements and a paraxial approximation. The imaging results show that tissue surrounding the high impedance region is artifact free and has correct speed of sound at sub-mm resolution.
Collapse
|
23
|
Jalalifar A, Soliman H, Ruschin M, Sahgal A, Sadeghi-Naini A. A Brain Tumor Segmentation Framework Based on Outlier Detection Using One-Class Support Vector Machine. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:1067-1070. [PMID: 33018170 DOI: 10.1109/embc44109.2020.9176263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Accurate segmentation of brain tumors is a challenging task and also a crucial step in diagnosis and treatment planning for cancer patients. Magnetic resonance imaging (MRI) is the standard imaging modality for detection, characterization, treatment planning and outcome evaluation of brain tumors. MRI scans are usually acquired at multiple sessions before and after the treatment. An automatic segmentation framework is highly desirable to segment brain tumors in MR images as it streamlines the image-guided radiation therapy workflow considerably. Automatic segmentation of brain tumors also facilitates an incremental development of data-driven systems for therapy outcome prediction based on radiomics analysis. In this study, an outlier-detection-based segmentation framework is proposed to delineate brain tumors in magnetic resonance (MR) images automatically. The proposed method considers the tumor and edema pixels in an MR image as outliers compared to the pixels associated with the healthy tissue. The framework generates two outlier masks using independent one-class support vector machines that operate on post-contrast T1-weighted (T1w) and T2-weighted-fluid-attenuation-inversion-recovery (T2-FLAIR) images. The outlier masks are subsequently refined and fused using a number of morphological and logical operators to estimate a tumor mask for each image slice. The framework was constructed and evaluated using the MRI data acquired from 35 and 5 patients with brain metastasis, respectively. The obtained results demonstrated an average Dice similarity coefficient and Hausdorff distance of 0.84 ± 0.06 and 1.85 ± 0.48 mm, respectively, between the manual (ground truth) and automatic tumor contours, on the independent test set.
Collapse
|
24
|
Quantitative ultrasound delta-radiomics during radiotherapy for monitoring treatment responses in head and neck malignancies. Future Sci OA 2020; 6:FSO624. [PMID: 33235811 PMCID: PMC7668124 DOI: 10.2144/fsoa-2020-0073] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: We investigated quantitative ultrasound (QUS) in patients with node-positive head and neck malignancies for monitoring responses to radical radiotherapy (RT). Materials & methods: QUS spectral and texture parameters were acquired from metastatic lymph nodes 24 h, 1 and 4 weeks after starting RT. K-nearest neighbor and naive-Bayes machine-learning classifiers were used to build prediction models for each time point. Response was detected after 3 months of RT, and patients were classified into complete and partial responders. Results: Single-feature naive-Bayes classification performed best with a prediction accuracy of 80, 86 and 85% at 24 h, week 1 and 4, respectively. Conclusion: QUS-radiomics can predict RT response at 3 months as early as 24 h with reasonable accuracy, which further improves into 1 week of treatment. Patients with head and neck cancer are often treated with radiation, which usually spans over 6–7 weeks. The response is usually measured 3 months after treatment completion. In this study, we had performed ultrasound scans from the patient’s neck node during radiation treatment (after 24 h, 1 and 4 weeks). Artificial intelligence was used to interpret the ultrasound imaging and predict the response to radiation at the end of 3 months. The scans obtained after the first week were able to predict the treatment response with reasonable accuracy (86%).
Collapse
|
25
|
Quiaoit K, DiCenzo D, Fatima K, Bhardwaj D, Sannachi L, Gangeh M, Sadeghi-Naini A, Dasgupta A, Kolios MC, Trudeau M, Gandhi S, Eisen A, Wright F, Look-Hong N, Sahgal A, Stanisz G, Brezden C, Dinniwell R, Tran WT, Yang W, Curpen B, Czarnota GJ. Quantitative ultrasound radiomics for therapy response monitoring in patients with locally advanced breast cancer: Multi-institutional study results. PLoS One 2020; 15:e0236182. [PMID: 32716959 PMCID: PMC7384762 DOI: 10.1371/journal.pone.0236182] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) is the standard of care for patients with locally advanced breast cancer (LABC). The study was conducted to investigate the utility of quantitative ultrasound (QUS) carried out during NAC to predict the final tumour response in a multi-institutional setting. METHODS Fifty-nine patients with LABC were enrolled from three institutions in North America (Sunnybrook Health Sciences Centre (Toronto, Canada), MD Anderson Cancer Centre (Texas, USA), and Princess Margaret Cancer Centre (Toronto, Canada)). QUS data were collected before starting NAC and subsequently at weeks 1 and 4 during chemotherapy. Spectral tumour parametric maps were generated, and textural features determined using grey-level co-occurrence matrices. Patients were divided into two groups based on their pathological outcomes following surgery: responders and non-responders. Machine learning algorithms using Fisher's linear discriminant (FLD), K-nearest neighbour (K-NN), and support vector machine (SVM-RBF) were used to generate response classification models. RESULTS Thirty-six patients were classified as responders and twenty-three as non-responders. Among all the models, SVM-RBF had the highest accuracy of 81% at both weeks 1 and week 4 with area under curve (AUC) values of 0.87 each. The inclusion of week 1 and 4 features led to an improvement of the classifier models, with the accuracy and AUC from baseline features only being 76% and 0.68, respectively. CONCLUSION QUS data obtained during NAC reflect the ongoing treatment-related changes during chemotherapy and can lead to better classifier performances in predicting the ultimate pathologic response to treatment compared to baseline features alone.
Collapse
Affiliation(s)
- Karina Quiaoit
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Daniel DiCenzo
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Kashuf Fatima
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Divya Bhardwaj
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Lakshmanan Sannachi
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Mehrdad Gangeh
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, Canada
| | - Archya Dasgupta
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | | | - Maureen Trudeau
- Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Sonal Gandhi
- Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Andrea Eisen
- Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Frances Wright
- Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Nicole Look-Hong
- Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Greg Stanisz
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Christine Brezden
- Department of Medical Oncology, Saint Michael's Hospital, University of Toronto, Toronto, Canada
| | - Robert Dinniwell
- Department of Radiation Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
- Department of Radiation Oncology, London Health Sciences Centre, London, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - William T. Tran
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Wei Yang
- Department of Diagnostic Radiology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Gregory J. Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, Canada
- Department of Physics, Ryerson University, Toronto, Canada
- * E-mail:
| |
Collapse
|
26
|
Zhang Q, Lou Y, Bai XL, Liang TB. Intratumoral heterogeneity of hepatocellular carcinoma: From single-cell to population-based studies. World J Gastroenterol 2020; 26:3720-3736. [PMID: 32774053 PMCID: PMC7383842 DOI: 10.3748/wjg.v26.i26.3720] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/02/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by high heterogeneity in both intratumoral and interpatient manners. While interpatient heterogeneity is related to personalized therapy, intratumoral heterogeneity (ITH) largely influences the efficacy of therapies in individuals. ITH contributes to tumor growth, metastasis, recurrence, and drug resistance and consequently limits the prognosis of patients with HCC. There is an urgent need to understand the causes, characteristics, and consequences of tumor heterogeneity in HCC for the purposes of guiding clinical practice and improving survival. Here, we summarize the studies and technologies that describe ITH in HCC to gain insight into the origin and evolutionary process of heterogeneity. In parallel, evidence is collected to delineate the dynamic relationship between ITH and the tumor ecosystem. We suggest that conducting comprehensive studies of ITH using single-cell approaches in temporal and spatial dimensions, combined with population-based clinical trials, will help to clarify the clinical implications of ITH, develop novel intervention strategies, and improve patient prognosis.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang Province, China
| | - Yu Lou
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang Province, China
| | - Ting-Bo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
27
|
Moghadas-Dastjerdi H, Sha-E-Tallat HR, Sannachi L, Sadeghi-Naini A, Czarnota GJ. A priori prediction of tumour response to neoadjuvant chemotherapy in breast cancer patients using quantitative CT and machine learning. Sci Rep 2020; 10:10936. [PMID: 32616912 PMCID: PMC7331583 DOI: 10.1038/s41598-020-67823-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Response to Neoadjuvant chemotherapy (NAC) has demonstrated a high correlation to survival in locally advanced breast cancer (LABC) patients. An early prediction of responsiveness to NAC could facilitate treatment adjustments on an individual patient basis that would be expected to improve treatment outcomes and patient survival. This study investigated, for the first time, the efficacy of quantitative computed tomography (qCT) parametric imaging to characterize intra-tumour heterogeneity and its application in predicting tumour response to NAC in LABC patients. Textural analyses were performed on CT images acquired from 72 patients before the start of chemotherapy to determine quantitative features of intra-tumour heterogeneity. The best feature subset for response prediction was selected through a sequential feature selection with bootstrap 0.632 + area under the receiver operating characteristic (ROC) curve (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\mathrm{A}\mathrm{U}\mathrm{C}}_{0.632+}$$\end{document}AUC0.632+) as a performance criterion. Several classifiers were evaluated for response prediction using the selected feature subset. Amongst the applied classifiers an Adaboost decision tree provided the best results with cross-validated \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\mathrm{A}\mathrm{U}\mathrm{C}}_{0.632+}$$\end{document}AUC0.632+, accuracy, sensitivity and specificity of 0.89, 84%, 80% and 88%, respectively. The promising results obtained in this study demonstrate the potential of the proposed biomarkers to be used as predictors of LABC tumour response to NAC prior to the start of treatment.
Collapse
Affiliation(s)
- Hadi Moghadas-Dastjerdi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Hira Rahman Sha-E-Tallat
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Lakshmanan Sannachi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Ali Sadeghi-Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Nasr R, Falou O, Shahin A, Hysi E, Wirtzfeld LA, Berndl ESL, Kolios MC. Mean Scatterer Spacing Estimation Using Cepstrum-Based Continuous Wavelet Transform. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:1118-1126. [PMID: 31905136 DOI: 10.1109/tuffc.2020.2963955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The goal of this study was to develop an ultrasound (US) scatterer spacing estimation method using an enhanced cepstral analysis based on continuous wavelet transforms (CWTs). Simulations of backscattering media containing periodic and quasi-periodic scatterers were carried out to test the developed algorithm. Experimental data from HT-29 pellets and in vivo PC3 tumors were then used to estimate the mean scatterer spacing. For simulated media containing quasi-periodic scatterers at 1-mm and 100- [Formula: see text] spacing with 5% positional variation, the developed algorithm yielded a spacing estimation error of ~1% for 25- and 55-MHz US pulses. The mean scatterer spacing of HT-29 cell pellets (31.97 [Formula: see text]) was within 3% of the spacing obtained from histology and agreed with the predicted spacing from simulations based on the same pellets for both frequencies. The agreement extended to in vivo PC3 tumors estimation of the spacing with a variance of 1.68% between the spacing derived from the tumor histology and the application of the CWT to the experimental results. The developed technique outperformed the traditional cepstral methods as it can detect nonprominent peaks from quasi-random scatterer configurations. This work can be potentially used to detect morphological tissue changes during normal development or disease treatment.
Collapse
|
29
|
Sannachi L, Gangeh M, Naini AS, Bhargava P, Jain A, Tran WT, Czarnota GJ. Quantitative Ultrasound Monitoring of Breast Tumour Response to Neoadjuvant Chemotherapy: Comparison of Results Among Clinical Scanners. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1142-1157. [PMID: 32111456 DOI: 10.1016/j.ultrasmedbio.2020.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Abstract
Quantitative ultrasound (QUS) techniques have been demonstrated to detect cell death in vitro and in vivo. Recently, multi-feature classification models have been incorporated into QUS texture-feature analysis methods to increase further the sensitivity and specificity of detecting treatment response in locally advanced breast cancer patients. To effectively incorporate these analytic methods into clinical applications, QUS and texture-feature estimations should be independent of data acquisition systems. The study here investigated the consistencies of QUS and texture-feature estimation techniques relative to several factors. These included the ultrasound system properties, the effects of tissue heterogeneity and the effects of these factors on the monitoring of response to neoadjuvant chemotherapy. Specifically, tumour-response-detection performance based on QUS and texture parameters using two clinical ultrasound systems was compared. Observed variations in data between the systems were small and the results exhibited good agreement in tumour response predictions obtained from both ultrasound systems. The results obtained in this study suggest that tissue heterogeneity was a dominant feature in the parameters measured with the two different ultrasound systems; whereas differences in ultrasound system beam properties only exhibited a minor impact on texture features. The McNemar statistical test performed on tumour response prediction results from the two systems did not reveal significant differences. Overall, the results in this study demonstrate the potential to achieve reliable and consistent QUS and texture-based analyses across different ultrasound imaging platforms.
Collapse
Affiliation(s)
- Lakshmanan Sannachi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mehrdad Gangeh
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ali-Sadeghi Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Priya Bhargava
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Aparna Jain
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - William Tyler Tran
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Gregory Jan Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
30
|
Karami E, Ruschin M, Soliman H, Sahgal A, Stanisz GJ, Sadeghi-Naini A. An MR Radiomics Framework for Predicting the Outcome of Stereotactic Radiation Therapy in Brain Metastasis. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:1022-1025. [PMID: 31946067 DOI: 10.1109/embc.2019.8856558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Despite recent advances in cancer treatment, patients with brain metastasis still suffer from poor overall survival (OS) after standard treatment. Predicting the treatment outcome before or early after the treatment can potentially assist the physicians in improving the therapy outcome by adjusting a standard treatment on an individual patient basis. In this study, a data-driven computational framework was proposed and investigated to predict the local control/failure (LC/LF) outcome in patients with brain metastasis treated with hypo-fractionated stereotactic radiation therapy (SRT). The framework extracted several geometrical and textural features from the magnetic resonance (MR) images of the tumour and edema regions acquired for 38 patients. Subsequent to a multi-step feature reduction/selection, a quantitative MR biomarker consisting of two features was constructed. A support vector machine classifier was used for outcome prediction using the constructed MR biomarker. The bootstrap .632+ and leave-one-patient-out cross-validation methods were used to assess the model's performance. The results indicated that the outcome of LF after SRT could be predicted with an area under the curve of 0.80 and a cross-validated accuracy of 82%. The results obtained implied a good potential of the proposed framework for local outcome prediction in patients with brain metastasis treated with SRT and encourage further investigations on a larger cohort of patients.
Collapse
|
31
|
Nguyen TN, Tam AJ, Do MN, Oelze ML. Estimation of Backscatter Coefficients Using an In Situ Calibration Source. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:308-317. [PMID: 31567079 PMCID: PMC7075368 DOI: 10.1109/tuffc.2019.2944305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The objective of this article is to demonstrate the feasibility of estimating the backscatter coefficient (BSC) using an in situ calibration source. Traditional methods of estimating the BSC in vivo using a reference phantom technique do not account for transmission losses due to intervening layers between the ultrasonic source and the tissue region to be interrogated, leading to increases in bias and variance of BSC-based estimates. To account for transmission losses, an in situ calibration approach is proposed. The in situ calibration technique employs a titanium sphere that is well-characterized ultrasonically, biocompatible, and embedded inside the sample. A set of experiments was conducted to evaluate the embedded titanium spheres as in situ calibration targets for BSC estimation. The first experiment quantified the backscattered signal strength from titanium spheres of three sizes: 0.5, 1, and 2 mm in diameter. The second set of experiments assessed the repeatability of BSC estimates from the titanium spheres and compared these BSCs to theory. The third set of experiments quantified the ability of the titanium bead to provide an in situ reference spectrum in the presence of a lossy layer on top of the sample. The final set of experiments quantified the ability of the bead to provide a calibration spectrum over multiple depths in the sample. All experiments were conducted using an L9-4/38 linear array connected to a SonixOne system. The strongest signal was observed from the 2-mm titanium bead with the signal-to-noise ratio (SNR) of 11.6 dB with respect to the background speckle. Using an analysis bandwidth of 2.5-5.5 MHz, the mean differences between the experimentally derived BSCs and BSCs derived from the Faran theory were 0.54 and 0.76 dB using the array and a single-element transducer, respectively. The BSCs estimated using the in situ calibration approach without the layer and with the layer and using the reference phantom approach with the layer were compared to the reference phantom approach without the layer present. The mean differences in BSCs were 0.15, 0.73, and -9.69 dB, respectively. The mean differences of the BSCs calculated from data blocks located at depths that were either 30 pulse lengths above or below the actual bead depth compared to the BSC calculated at bead depth were -1.55 and -1.48 dB, respectively. The results indicate that an in situ calibration target can account for overlaying tissue losses, thereby improving the robustness of BSC-based estimates.
Collapse
|
32
|
Karami E, Jalalifar A, Ruschin M, Soliman H, Sahgal A, Stanisz GJ, Sadeghi-Naini A. An Automatic Framework for Segmentation of Brain Tumours at Follow-up Scans after Radiation Therapy .. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:463-466. [PMID: 31945938 DOI: 10.1109/embc.2019.8856858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Brain metastasis is the most common intracranial malignancy with a poor overall survival (OS) after treatment. The standard stereotactic radiation therapy (SRT) planning procedure for brain metastasis requires delineating the tumour volume on magnetic resonance (MR) images. MR images are also acquired at multiple follow-up scans after SRT to monitor the treatment outcome through measuring changes in the physical dimensions of the tumour. Such measurements require manual segmentation of the tumour volume on multiple slices of several follow-up images which is tedious and impedes the SRT evaluation work flow considerably. In this study, an automatic framework was proposed to segment the tumour volume on longitudinal MR images acquired at standard follow-up scans after SRT. The multi-step segmentation framework was based on region growing and morphological snakes models that applied the standard SRT planning tumour contour as a basis to approximate the tumour shape and location at each follow-up scan for an accurate automatic segmentation of tumour volume. The framework was evaluated using the MR imaging data acquired from five patients prior to and at three follow-up scans after SRT. The preliminary results indicated that the Dice similarity coefficient between the ground truth tumour masks and their automatically segmented counterparts ranged between 0.84 and 0.90, while the average Dice coefficient for all the follow-up scans was 0.88. The results obtained implied a good potential of the proposed framework for being incorporated into the SRT treatment planning and evaluation systems as well as outcome prediction models.
Collapse
|
33
|
Karami E, Soliman H, Ruschin M, Sahgal A, Myrehaug S, Tseng CL, Czarnota GJ, Jabehdar-Maralani P, Chugh B, Lau A, Stanisz GJ, Sadeghi-Naini A. Quantitative MRI Biomarkers of Stereotactic Radiotherapy Outcome in Brain Metastasis. Sci Rep 2019; 9:19830. [PMID: 31882597 PMCID: PMC6934477 DOI: 10.1038/s41598-019-56185-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/08/2019] [Indexed: 02/08/2023] Open
Abstract
About 20-40% of cancer patients develop brain metastases, causing significant morbidity and mortality. Stereotactic radiation treatment is an established option that delivers high dose radiation to the target while sparing the surrounding normal tissue. However, up to 20% of metastatic brain tumours progress despite stereotactic treatment, and it can take months before it is evident on follow-up imaging. An early predictor of radiation therapy outcome in terms of tumour local failure (LF) is crucial, and can facilitate treatment adjustments or allow for early salvage treatment. In this study, an MR-based radiomics framework was proposed to derive and investigate quantitative MRI (qMRI) biomarkers for the outcome of LF in brain metastasis patients treated with hypo-fractionated stereotactic radiation therapy (SRT). The qMRI biomarkers were constructed through a multi-step feature extraction/reduction/selection framework using the conventional MR imaging data acquired from 100 patients (133 lesions), and were applied in conjunction with machine learning techniques for outcome prediction and risk assessment. The results indicated that the majority of the features in the optimal qMRI biomarkers characterize the heterogeneity in the surrounding regions of tumour including edema and tumour/lesion margins. The optimal qMRI biomarker consisted of five features that predict the outcome of LF with an area under the curve (AUC) of 0.79, and a cross-validated sensitivity and specificity of 81% and 79%, respectively. The Kaplan-Meier analyses showed a statistically significant difference in local control (p-value < 0.0001) and overall survival (p = 0.01). Findings from this study are a step towards using qMRI for early prediction of local failure in brain metastasis patients treated with SRT. This may facilitate early adjustments in treatment, such as surgical resection or salvage radiation, that can potentially improve treatment outcomes. Investigations on larger cohorts of patients are, however, required for further validation of the technique.
Collapse
Affiliation(s)
- Elham Karami
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hany Soliman
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Mark Ruschin
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | | | - Brige Chugh
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Angus Lau
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - Ali Sadeghi-Naini
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| |
Collapse
|
34
|
Predictive quantitative ultrasound radiomic markers associated with treatment response in head and neck cancer. Future Sci OA 2019; 6:FSO433. [PMID: 31915534 PMCID: PMC6920736 DOI: 10.2144/fsoa-2019-0048] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: We aimed to identify quantitative ultrasound (QUS)-radiomic markers to predict radiotherapy response in metastatic lymph nodes of head and neck cancer. Materials & methods: Node-positive head and neck cancer patients underwent pretreatment QUS imaging of their metastatic lymph nodes. Imaging features were extracted using the QUS spectral form, and second-order texture parameters. Machine-learning classifiers were used for predictive modeling, which included a logistic regression, naive Bayes, and k-nearest neighbor classifiers. Results: There was a statistically significant difference in the pretreatment QUS-radiomic parameters between radiological complete responders versus partial responders (p < 0.05). The univariable model that demonstrated the greatest classification accuracy included: spectral intercept (SI)-contrast (area under the curve = 0.741). Multivariable models were also computed and showed that the SI-contrast + SI-homogeneity demonstrated an area under the curve = 0.870. The three-feature model demonstrated that the spectral slope-correlation + SI-contrast + SI-homogeneity-predicted response with accuracy of 87.5%. Conclusion: Multivariable QUS-radiomic features of metastatic lymph nodes can predict treatment response a priori. In this study, quantitative ultrasound (QUS) and machine-learning classification was used to predict treatment outcomes in head and neck cancer patients. Metastatic lymph nodes in the neck were scanned using conventional frequency ultrasound (US). Quantitative data were collected from the US-radiofrequency signal a priori. Machine-learning classification models were computed using QUS features; these included the linear fit parameters of the power spectrum, and second-order texture parameters of the QUS parametric images. Treatment outcomes were measured based on radiological response. Patients were classified into binary groups: radiologic complete response (CR) or radiological partial response (PR), which was assessed 3 months following treatment. Initial results demonstrate high accuracy (%Acc = 87.5%) for predicting radiological response. The results of this study suggest that QUS can be used to predict head and neck cancer response to radiotherapy a priori.
Collapse
|
35
|
Ritter A, Hirschfeld M, Berner K, Rücker G, Jäger M, Weiss D, Medl M, Nöthling C, Gassner S, Asberger J, Erbes T. Circulating non‑coding RNA‑biomarker potential in neoadjuvant chemotherapy of triple negative breast cancer? Int J Oncol 2019; 56:47-68. [PMID: 31789396 PMCID: PMC6910196 DOI: 10.3892/ijo.2019.4920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022] Open
Abstract
Due to the positive association between neoadjuvant chemotherapy (NACT) and the promising early response rates of patients with triple negative breast cancer (TNBC), including probabilities of pathological complete response, NACT is increasingly used in TNBC management. Liquid biopsy-based biomarkers with the power to diagnose the early response to NACT may support established monitoring tools, which are to a certain extent imprecise and costly. Simple serum- or urine-based analyses of non-coding RNA (ncRNA) expression may allow for fast, minimally-invasive testing and timely adjustment of the therapy regimen. The present study investigated breast cancer-related ncRNAs [microRNA (miR)-7, -9, -15a, -17, -18a, -19b, -21, -30b, -222 and -320c, PIWI-interacting RNA-36743 and GlyCCC2] in triple positive BT-474 cells and three TNBC cell lines (BT-20, HS-578T and MDA-MB-231) treated with various chemotherapeutic agents using reverse transcription-quantitative PCR. Intracellular and secreted microvesicular ncRNA expression levels were analysed using a multivariable statistical regression analysis. Chemotherapy-driven effects were investigated by analysing cell cycle determinants at the mRNA and protein levels. Serum and urine specimens from 8 patients with TNBC were compared with 10 healthy females using two-sample t-tests. Samples from the patients with TNBC were compared at two time points. Chemotherapeutic treatments induced distinct changes in ncRNA expression in TNBC cell lines and the BT-474 cell line in intra- and extracellular compartments. Serum and urine-based ncRNA expression analysis was able to discriminate between patients with TNBC and controls. Time point comparisons in the urine samples of patients with TNBC revealed a general rise in the level of ncRNA. Serum data suggested a potential association between piR-36743, miR-17, -19b and -30b expression levels and an NACT-driven complete clinical response. The present study highlighted the potential of ncRNAs as liquid biopsy-based biomarkers in TNBC chemotherapy treatment. The ncRNAs tested in the present study have been previously investigated for their involvement in BC or TNBC chemotherapy responses; however, these previous studies were restricted to patient tissue or in vitro models. The data from the present study offer novel insight into ncRNA expression in liquid samples from patients with TNBC, and the study serves as an initial step in the evaluation of ncRNAs as diagnostic biomarkers in the monitoring of TNBC therapy.
Collapse
Affiliation(s)
- Andrea Ritter
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Marc Hirschfeld
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Kai Berner
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Gerta Rücker
- Institute of Medical Biometry and Statistics, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79104 Freiburg, Germany
| | - Markus Jäger
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Daniela Weiss
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Markus Medl
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Claudia Nöthling
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Sandra Gassner
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Jasmin Asberger
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| | - Thalia Erbes
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical Center‑University of Freiburg, D‑79106 Freiburg, Germany
| |
Collapse
|
36
|
Dobruch-Sobczak K, Piotrzkowska-Wróblewska H, Klimoda Z, Secomski W, Karwat P, Markiewicz-Grodzicka E, Kolasińska-Ćwikła A, Roszkowska-Purska K, Litniewski J. Monitoring the response to neoadjuvant chemotherapy in patients with breast cancer using ultrasound scattering coefficient: A preliminary report. J Ultrason 2019; 19:89-97. [PMID: 31355579 PMCID: PMC6750328 DOI: 10.15557/jou.2019.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/10/2019] [Indexed: 11/22/2022] Open
Abstract
Objective: Neoadjuvant chemotherapy was initially used in locally advanced breast cancer, and currently it is recommended for patients with Stage 3 and with early-stage disease with human epidermal growth factor receptors positive or triple-negative breast cancer. Ultrasound imaging in combination with a quantitative ultrasound method is a novel diagnostic approach. Aim of study: The aim of this study was to analyze the variability of the integrated backscatter coefficient, and to evaluate their use to predict the effectiveness of treatment and compare to ultrasound examination results. Material and method: Ten patients (mean age 52.9) with 13 breast tumors (mean dimension 41 mm) were selected for neoadjuvant chemotherapy. Ultrasound was performed before the treatment and one week after each course of neoadjuvant chemotherapy. The dimensions were assessed adopting the RECIST criteria. Tissue responses were classified as pathological response into the following categories: not responded to the treatment (G1, cell reduction by ≤9%) and responded to the treatment partially: G2, G3, G4, cell reduction by 10-29% (G2), 30-90% (G3), >90% (G4), respectively, and completely. Results: In B-mode examination partial response was observed in 9/13 cases (completely, G1, G3, G4), and stable disease was demonstrated in 3/13 cases (completely, G1, G4). Complete response was found in 1/13 cases. As for backscatter coefficient, 10/13 tumors (completely, and G2, G3, and G4) were characterized by an increased mean value of 153%. Three tumors 3/13 (G1) displayed a decreased mean value of 31%. Conclusion: The variability of backscatter coefficient, could be associated with alterations in the structure of the tumor tissue during neoadjuvant chemotherapy. There were unequivocal differences between responded and non-responded patients. The backscatter coefficient analysis correlated better with the results of histopathological verification than with the B-mode RECIST criteria. Objective: Neoadjuvant chemotherapy was initially used in locally advanced breast cancer, and currently it is recommended for patients with Stage 3 and with early-stage disease with human epidermal growth factor receptors positive or triple-negative breast cancer. Ultrasound imaging in combination with a quantitative ultrasound method is a novel diagnostic approach. Aim of study: The aim of this study was to analyze the variability of the integrated backscatter coefficient, and to evaluate their use to predict the effectiveness of treatment and compare to ultrasound examination results. Material and method: Ten patients (mean age 52.9) with 13 breast tumors (mean dimension 41 mm) were selected for neoadjuvant chemotherapy. Ultrasound was performed before the treatment and one week after each course of neoadjuvant chemotherapy. The dimensions were assessed adopting the RECIST criteria. Tissue responses were classified as pathological response into the following categories: not responded to the treatment (G1, cell reduction by ≤9%) and responded to the treatment partially: G2, G3, G4, cell reduction by 10–29% (G2), 30–90% (G3), >90% (G4), respectively, and completely. Results: In B-mode examination partial response was observed in 9/13 cases (completely, G1, G3, G4), and stable disease was demonstrated in 3/13 cases (completely, G1, G4). Complete response was found in 1/13 cases. As for backscatter coefficient, 10/13 tumors (completely, and G2, G3, and G4) were characterized by an increased mean value of 153%. Three tumors 3/13 (G1) displayed a decreased mean value of 31%. Conclusion: The variability of backscatter coefficient, could be associated with alterations in the structure of the tumor tissue during neoadjuvant chemotherapy. There were unequivocal differences between responded and non-responded patients. The backscatter coefficient analysis correlated better with the results of histopathological verification than with the B-mode RECIST criteria.
Collapse
Affiliation(s)
- Katarzyna Dobruch-Sobczak
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland ; Radiology Department , M. Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Hanna Piotrzkowska-Wróblewska
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland
| | - Ziemowit Klimoda
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland
| | - Wojciech Secomski
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland
| | - Piotr Karwat
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland
| | - Ewa Markiewicz-Grodzicka
- Chemiotherapy Department , M. Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Agnieszka Kolasińska-Ćwikła
- Chemiotherapy Department , M. Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Katarzyna Roszkowska-Purska
- Zakład Patomorfologii , M. Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Jerzy Litniewski
- Ultrasound Department , Institute of Fundamental Technological Research , Polish Academy of Sciences , Warsaw , Poland
| |
Collapse
|
37
|
Hysi E, Fadhel MN, Moore MJ, Zalev J, Strohm EM, Kolios MC. Insights into photoacoustic speckle and applications in tumor characterization. PHOTOACOUSTICS 2019; 14:37-48. [PMID: 31080733 PMCID: PMC6505056 DOI: 10.1016/j.pacs.2019.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 05/20/2023]
Abstract
In ultrasound imaging, fully-developed speckle arises from the spatiotemporal superposition of pressure waves backscattered by randomly distributed scatterers. Speckle appearance is affected by the imaging system characteristics (lateral and axial resolution) and the random-like nature of the underlying tissue structure. In this work, we examine speckle formation in acoustic-resolution photoacoustic (PA) imaging using simulations and experiments. Numerical and physical phantoms were constructed to demonstrate that PA speckle carries information related to unresolved absorber structure in a manner similar to ultrasound speckle and unresolved scattering structures. A fractal-based model of the tumor vasculature was used to study PA speckle from unresolved cylindrical vessels. We show that speckle characteristics and the frequency content of PA signals can be used to monitor changes in average vessel size, linked to tumor growth. Experimental validation on murine tumors demonstrates that PA speckle can be utilized to characterize the unresolved vasculature in acoustic-resolution photoacoustic imaging.
Collapse
Affiliation(s)
- Eno Hysi
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology, Li Ka Shing Knowledge Institute, Keenan Research Centre, St. Michael’s Hospital, Toronto, ON, Canada
| | - Muhannad N. Fadhel
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology, Li Ka Shing Knowledge Institute, Keenan Research Centre, St. Michael’s Hospital, Toronto, ON, Canada
| | - Michael J. Moore
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology, Li Ka Shing Knowledge Institute, Keenan Research Centre, St. Michael’s Hospital, Toronto, ON, Canada
| | - Jason Zalev
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology, Li Ka Shing Knowledge Institute, Keenan Research Centre, St. Michael’s Hospital, Toronto, ON, Canada
| | - Eric M. Strohm
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Michael C. Kolios
- Department of Physics, Ryerson University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology, Li Ka Shing Knowledge Institute, Keenan Research Centre, St. Michael’s Hospital, Toronto, ON, Canada
- Corresponding author.
| |
Collapse
|
38
|
Piotrzkowska-Wróblewska H, Dobruch-Sobczak K, Klimonda Z, Karwat P, Roszkowska-Purska K, Gumowska M, Litniewski J. Monitoring breast cancer response to neoadjuvant chemotherapy with ultrasound signal statistics and integrated backscatter. PLoS One 2019; 14:e0213749. [PMID: 30870478 PMCID: PMC6417657 DOI: 10.1371/journal.pone.0213749] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Background Neoadjuvant chemotherapy (NAC) is used in patients with breast cancer to reduce tumor focus, metastatic risk, and patient mortality. Monitoring NAC effects is necessary to capture resistant patients and stop or change treatment. The existing methods for evaluating NAC results have some limitations. The aim of this study was to assess the tumor response at an early stage, after the first doses of the NAC, based on the variability of the backscattered ultrasound energy, and backscatter statistics. The backscatter statistics has not previously been used to monitor NAC effects. Methods The B-mode ultrasound images and raw radio frequency data from breast tumors were obtained using an ultrasound scanner before chemotherapy and 1 week after each NAC cycle. The study included twenty-four malignant breast cancers diagnosed in sixteen patients and qualified for neoadjuvant treatment before surgery. The shape parameter of the homodyned K distribution and integrated backscatter, along with the tumor size in the longest dimension, were determined based on ultrasound data and used as markers for NAC response. Cancer tumors were assigned to responding and non-responding groups, according to histopathological evaluation, which was a reference in assessing the utility of markers. Statistical analysis was performed to rate the ability of markers to predict the final NAC response based on data obtained after subsequent therapeutic doses. Results Statistically significant differences (p<0.05) between groups were obtained after 2, 3, 4, and 5 doses of NAC for quantitative ultrasound markers and after 5 doses for the assessment based on maximum tumor dimension. Statistical analysis showed that, after the second and third NAC courses the classification based on integrated backscatter marker was characterized by an AUC of 0.69 and 0.82, respectively. The introduction of the second quantitative marker describing the statistical properties of scattering increased the corresponding AUC values to 0.82 and 0.91. Conclusions Quantitative ultrasound information can characterize the tumor's pathological response better and at an earlier stage of therapy than the assessment of the reduction of its dimensions. The introduction of statistical parameters of ultrasonic backscatter to monitor the effects of chemotherapy can increase the effectiveness of monitoring and contribute to a better personalization of NAC therapy.
Collapse
Affiliation(s)
| | - Katarzyna Dobruch-Sobczak
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Radiology Department, Cancer Center and Institute of Oncology, M. Skłodowska-Curie Memorial, Warsaw, Poland
| | - Ziemowit Klimonda
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Karwat
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Roszkowska-Purska
- Department of Pathology, Cancer Center and Institute of Oncology, M. Skłodowska-Curie Memorial, Warsaw, Poland
| | - Magdalena Gumowska
- Radiology Department, Cancer Center and Institute of Oncology, M. Skłodowska-Curie Memorial, Warsaw, Poland
| | - Jerzy Litniewski
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- * E-mail:
| |
Collapse
|