1
|
Groheux D, Vaz SC, Poortmans P, Mann RM, Ulaner GA, Cook GJR, Hindié E, Pilkington Woll JP, Jacene H, Rubio IT, Vrancken Peeters MJ, Dibble EH, de Geus-Oei LF, Graff SL, Cardoso F. Role of [ 18F]FDG PET/CT in patients with invasive breast carcinoma of no special type: Literature review and comparison between guidelines. Breast 2024; 78:103806. [PMID: 39303572 PMCID: PMC11440802 DOI: 10.1016/j.breast.2024.103806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/29/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE The recently released EANM/SNMMI guideline, endorsed by several important clinical and imaging societies in the field of breast cancer (BC) care (ACR, ESSO, ESTRO, EUSOBI/ESR, EUSOMA), emphasized the role of [18F]FDG PET/CT in management of patients with no special type (NST) BC. This review identifies and summarizes similarities, discrepancies and novelties of the EANM/SNMMI guideline compared to NCCN, ESMO and ABC recommendations. METHODS The EANM/SNMMI guideline was based on a systematic literature search and the AGREE tool. The level of evidence was determined according to NICE criteria, and 85 % agreement or higher was reached regarding each statement. Comparisons with NCCN, ESMO and ABC guidelines were examined for specific clinical scenarios in patients with early stage through advanced and metastatic BC. RESULTS Regarding initial staging of patients with NST BC, [18F]FDG PET/CT is the preferred modality in the EANM-SNMMI guideline, showing superiority as a single modality to a combination of contrast-enhanced CT of thorax-abdomen-pelvis plus bone scan in head-to-head comparisons and a randomized study. Its use is recommended in patients with clinical stage IIB or higher and may be useful in certain stage IIA cases of NST BC. In NCCN, ESMO, and ABC guidelines, [18F]FDG PET/CT is instead recommended as complementary to conventional imaging to solve inconclusive findings, although ESMO and ABC also suggest [18F]FDG PET/CT can replace conventional imaging for staging patients with high-risk and metastatic NST BC. During follow up, NCCN and ESMO only recommend diagnostic imaging if there is suspicion of recurrence. Similarly, EANM-SNMMI states that [18F]FDG PET/CT is useful to detect the site and extent of recurrence only when there is clinical or laboratory suspicion of recurrence, or when conventional imaging methods are equivocal. The EANM-SNMMI guideline is the first to emphasize a role of [18F]FDG PET/CT for assessing early metabolic response to primary systemic therapy, particularly for HER2+ BC and TNBC. In the metastatic setting, EANM-SNMMI state that [18F]FDG PET/CT may help evaluate bone metastases and determine early response to treatment, in agreement with guidelines from ESMO. CONCLUSIONS The recently released EANM/SNMMI guideline reinforces the role of [18F]FDG PET/CT in the management of patients with NST BC supported by extensive evidence of its utility in several clinical scenarios.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; University Paris-Diderot, INSERM, U976, Paris, France; Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France.
| | - Sofia C Vaz
- Department of Nuclear Medicine and Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk-Antwerp, Belgium
| | - Ritse M Mann
- Department of Radiology, Radboud umc, Nijmegen, the Netherlands
| | - Gary A Ulaner
- Department of Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, United States; Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, United States
| | - Gary J R Cook
- Department of Cancer Imaging, King's College London, London, UK; King's College London and Guy's & St Thomas' PET Centre, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Elif Hindié
- Department of Nuclear Medicine, Bordeaux University Hospital, Bordeaux, France
| | | | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, and Harvard Medical School, United States
| | - Isabel T Rubio
- Department of Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Cancer Center Clinica Universidad de Navarra, Spain
| | - Marie-Jeanne Vrancken Peeters
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft, the Netherlands
| | - Stephanie L Graff
- Lifespan Cancer Institute, Providence, RI, United States; Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
2
|
Chen L, Zheng S, Chen L, Xu S, Wu K, Kong L, Xue J, Chen X, Miao W, Zhu Y. 68Ga-Labeled Fibroblast Activation Protein Inhibitor PET/CT for the Early and Late Prediction of Pathologic Response to Neoadjuvant Chemotherapy in Breast Cancer Patients: A Prospective Study. J Nucl Med 2023; 64:1899-1905. [PMID: 37918866 PMCID: PMC10690122 DOI: 10.2967/jnumed.123.266079] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
68Ga-labeled fibroblast activation protein inhibitor (68Ga-FAPI) PET/CT has demonstrated promising clinical results, with a higher SUVmax and tumor-to-background ratio (TBR) in breast cancer (BC) patients than 18F-FDG PET/CT. Here, we aimed to evaluate the suitability of 68Ga-FAPI PET/CT for the early and late prediction of the pathologic response to neoadjuvant chemotherapy (NAC) in BC. Methods: Twenty-two consecutive patients with newly diagnosed BC and an indication for NAC were prospectively included. All patients underwent standard chemotherapy and 68Ga-FAPI PET/CT at baseline, after 2 cycles of NAC (PET2), and 1 wk before surgery (PET3). SUVmax was measured in the primary tumor region and positive regional lymph nodes. The expression of fibroblast activation protein in the primary lesion was analyzed by immunohistochemistry. Results: Seven patients (31.8%) achieved a pathologic complete response (pCR), and 15 (68.2%) had residual tumors. Thirteen patients (59.1%) showed concentric withdrawal of the primary tumor, and 9 (40.9%) showed diffuse withdrawal. Between PET2 and PET3, the ΔSUVmax of the primary tumor (R 2 = 0.822; P = 0.001) and metastatic lymph nodes (R 2 = 0.645; P = 0.002) were significantly correlated. The absolute values of SUVmax and TBR at PET2 and PET3 were lower in patients with pCR than in those without pCR (P < 0.05). Moreover, a larger ΔSUVmax at any time point was strongly associated with pCR (P < 0.05). Similar downward trends in SUVmax, TBR, and ΔSUVmax were observed in the pattern of primary tumor reduction. For predicting pCR, the optimal cutoff values for ΔSUVmax after 2 chemotherapy cycles, ΔSUVmax before surgery, TBR after 2 chemotherapy cycles, and TBR before surgery of the primary tumor were 3.4 (area under the curve [AUC], 0.890), 1.1 (AUC, 0.978), -63.8% (AUC, 0.879), -90.8% (AUC, 0.978), 7.6 (AUC, 0.848), and 1.4 (AUC, 0.971), respectively. Immunohistochemistry showed that the SUVmax and TBR of 68Ga-FAPI PET/CT were positively correlated with fibroblast activation protein expression (P < 0.001 for both). Conclusion: Assessment of early changes in 68Ga-FAPI uptake during NAC by 68Ga-FAPI PET/CT can predict pCR and primary tumor concentric withdrawal in BC patients. 68Ga-FAPI PET/CT has great potential for the early and late prediction of the pathologic response to NAC in BC.
Collapse
Affiliation(s)
- Ling Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shan Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China; and
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Kunlin Wu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lingjun Kong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jiajie Xue
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China;
| | - Weibing Miao
- Department of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China;
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Groheux D, Ulaner GA, Hindie E. Breast cancer: treatment response assessment with FDG-PET/CT in the neoadjuvant and in the metastatic setting. Clin Transl Imaging 2023; 11:439-452. [DOI: 10.1007/s40336-023-00584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 01/03/2025]
|
4
|
Hayward JH, Linden OE, Lewin AA, Weinstein SP, Bachorik AE, Balija TM, Kuzmiak CM, Paulis LV, Salkowski LR, Sanford MF, Scheel JR, Sharpe RE, Small W, Ulaner GA, Slanetz PJ. ACR Appropriateness Criteria® Monitoring Response to Neoadjuvant Systemic Therapy for Breast Cancer: 2022 Update. J Am Coll Radiol 2023; 20:S125-S145. [PMID: 37236739 DOI: 10.1016/j.jacr.2023.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 05/28/2023]
Abstract
Imaging plays a vital role in managing patients undergoing neoadjuvant chemotherapy, as treatment decisions rely heavily on accurate assessment of response to therapy. This document provides evidence-based guidelines for imaging breast cancer before, during, and after initiation of neoadjuvant chemotherapy. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision process support the systematic analysis of the medical literature from peer reviewed journals. Established methodology principles such as Grading of Recommendations Assessment, Development, and Evaluation or GRADE are adapted to evaluate the evidence. The RAND/UCLA Appropriateness Method User Manual provides the methodology to determine the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where peer reviewed literature is lacking or equivocal, experts may be the primary evidentiary source available to formulate a recommendation.
Collapse
Affiliation(s)
| | - Olivia E Linden
- Research Author, University of California, San Francisco, San Francisco, California
| | - Alana A Lewin
- Panel Chair, New York University Grossman School of Medicine, New York, New York
| | - Susan P Weinstein
- Panel Vice-Chair, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Tara M Balija
- Hackensack University Medical Center, Hackensack, New Jersey; American College of Surgeons
| | - Cherie M Kuzmiak
- University of North Carolina Hospital, Chapel Hill, North Carolina
| | | | - Lonie R Salkowski
- University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin
| | | | | | | | - William Small
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, Illinois
| | - Gary A Ulaner
- Hoag Family Cancer Institute, Newport Beach, California, and University of Southern California, Los Angeles, California; Commission on Nuclear Medicine and Molecular Imaging
| | - Priscilla J Slanetz
- Specialty Chair, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
5
|
Shankar LK, Huang E, Litiere S, Hoekstra OS, Schwartz L, Collette S, Boellaard R, Bogaerts J, Seymour L, deVries EGE. Meta-Analysis of the Test-Retest Repeatability of [18F]-Fluorodeoxyglucose Standardized Uptake Values: Implications for Assessment of Tumor Response. Clin Cancer Res 2023; 29:143-153. [PMID: 36302172 DOI: 10.1158/1078-0432.ccr-21-3143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/28/2022] [Accepted: 10/24/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Currently, guidelines for PET with 18F-fluorodeoxyglucose (FDG-PET) interpretation for assessment of therapy response in oncology primarily involve visual evaluation of FDG-PET/CT scans. However, quantitative measurements of the metabolic activity in tumors may be even more useful in evaluating response to treatment. Guidelines based on such measurements, including the European Organization for Research and Treatment of Cancer Criteria and PET Response Criteria in Solid Tumors, have been proposed. However, more rigorous analysis of response criteria based on FDG-PET measurements is needed to adopt regular use in practice. EXPERIMENTAL DESIGN Well-defined boundaries of repeatability and reproducibility of quantitative measurements to discriminate noise from true signal changes are a needed initial step. An extension of the meta-analysis from de Langen and colleagues (2012) of the test-retest repeatability of quantitative FDG-PET measurements, including mean, maximum, and peak standardized uptake values (SUVmax, SUVmean, and SUVpeak, respectively), was performed. Data from 11 studies in the literature were used to estimate the relationship between the variance in test-retest measurements with uptake level and various study-level, patient-level, and lesion-level characteristics. RESULTS Test-retest repeatability of percentage fluctuations for all three types of SUV measurement (max, mean, and peak) improved with higher FDG uptake levels. Repeatability in all three SUV measurements varied for different lesion locations. Worse repeatability in SUVmean was also associated with higher tumor volumes. CONCLUSIONS On the basis of these results, recommendations regarding SUV measurements for assessing minimal detectable changes based on repeatability and reproducibility are proposed. These should be applied to differentiate between response categories for a future set of FDG-PET-based criteria that assess clinically significant changes in tumor response.
Collapse
Affiliation(s)
| | - Erich Huang
- National Cancer Institute, Bethesda, Maryland
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Otto S Hoekstra
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Larry Schwartz
- Columbia University Medical Center and New York Presbyterian Hospital, New York, New York
| | | | | | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | | |
Collapse
|
6
|
Muacevic A, Adler JR. Mammographic and Ultrasonographic Imaging Analysis for Neoadjuvant Chemotherapy Evaluation: Volume Reduction Indexes That Correlate With Pathological Complete Response. Cureus 2022; 14:e29960. [PMID: 36225243 PMCID: PMC9534532 DOI: 10.7759/cureus.29960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION We aimed to evaluate volume reduction in digital mammography (DM) and ultrasound (US) for neoadjuvant chemotherapy (NAC) evaluation, with breast cancer-specific survival and pathological complete response (pCR) associations. METHODS This is a retrospective observational cohort study analyzing recorded images in 122 selected subjects out of which 569 patients presented with advanced breast cancers. Spearman's correlation and generalized estimating equations (GEE) compared volume reduction on DM and US between pCR and non-pCR after NAC with post-surgical anatomopathology. Cox regression and Kaplan-Meier curves analyzed associations between cancer-specific survival, pCR, and volume reductions. RESULTS A total of 34.4% (N=42) obtained pCR and 65.6% (N=80) did not. Minimum percentage indexes needed to correlate with pCR over time were, at least, 28.9% for DM (p=0.006) and 10.36% for US (p=0.046), with high specificity (US=98%, DM=93%) but low sensitivity (US=7%, DM=18%). Positive predictive values were 82% (DM) and 86% (US) and negative predictive values were 37% (DM) and 36% (US). Cox regression and Kaplan-Meier curves demonstrated associations of breast cancer-specific survival with pCR (Cox regression coefficient {B}=0.209, CI 95%=0.048-0.914, p=0.038). CONCLUSIONS At least 28.9% of volume reduction on DM and 10.36% of volume reduction on US are correlated with pCR. Furthermore, pCR was associated with breast cancer-specific survival after NAC in volumetric morphological imaging analysis.
Collapse
|
7
|
Pantel AR, Viswanath V, Muzi M, Doot RK, Mankoff DA. Principles of Tracer Kinetic Analysis in Oncology, Part II: Examples and Future Directions. J Nucl Med 2022; 63:514-521. [PMID: 35361713 PMCID: PMC8973282 DOI: 10.2967/jnumed.121.263519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 02/17/2022] [Indexed: 11/29/2022] Open
Abstract
Learning Objectives: On successful completion of this activity, participants should be able to (1) describe examples of the application of PET tracer kinetic analysis to oncology; (2) list applications research and possible clinical applications in oncology where kinetic analysis is helpful; and (3) discuss future applications of kinetic modeling to cancer research and possible clinical cancer imaging practice.Financial Disclosure: This work was supported by KL2 TR001879, R01 CA211337, R01 CA113941, R33 CA225310, Komen SAC130060, R50 CA211270, and K01 DA040023. Dr. Pantel is a consultant or advisor for Progenics and Blue Earth Diagnostics and is a meeting participant or lecturer for Blue Earth Diagnostics. Dr. Mankoff is on the scientific advisory boards of GE Healthcare, Philips Healthcare, Reflexion, and ImaginAb and is the owner of Trevarx; his wife is the chief executive officer of Trevarx. The authors of this article have indicated no other relevant relationships that could be perceived as a real or apparent conflict of interest.CME Credit: SNMMI is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to sponsor continuing education for physicians. SNMMI designates each JNM continuing education article for a maximum of 2.0 AMA PRA Category 1 Credits. Physicians should claim only credit commensurate with the extent of their participation in the activity. For CE credit, SAM, and other credit types, participants can access this activity through the SNMMI website (http://www.snmmilearningcenter.org) through April 2025.Kinetic analysis of dynamic PET imaging enables the estimation of biologic processes relevant to disease. Through mathematic analysis of the interactions of a radiotracer with tissue, information can be gleaned from PET imaging beyond static uptake measures. Part I of this 2-part continuing education paper reviewed the underlying principles and methodology of kinetic modeling. In this second part, the benefits of kinetic modeling for oncologic imaging are illustrated through representative case examples that demonstrate the principles and benefits of kinetic analysis in oncology. Examples of the model types discussed in part I are reviewed here: a 1-tissue-compartment model (15O-water), an irreversible 2-tissue-compartment model (18F-FDG), and a reversible 2-tissue-compartment model (3'-deoxy-3'-18F-fluorothymidine). Kinetic approaches are contrasted with static uptake measures typically used in the clinic. Overall, this 2-part review provides the reader with background in kinetic analysis to understand related research and improve the interpretation of clinical nuclear medicine studies with a focus on oncologic imaging.
Collapse
Affiliation(s)
- Austin R Pantel
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Varsha Viswanath
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Mark Muzi
- Department of Radiology, University of Washington, Seattle, Washington
| | - Robert K Doot
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| |
Collapse
|
8
|
Ko CC, Yeh LR, Kuo YT, Chen JH. Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomark Res 2021; 9:52. [PMID: 34215324 PMCID: PMC8252278 DOI: 10.1186/s40364-021-00306-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Response Evaluation Criteria in Solid Tumors (RECIST) is the gold standard for assessment of treatment response in solid tumors. Morphologic change of tumor size evaluated by RECIST is often correlated with survival length and has been considered as a surrogate endpoint of therapeutic efficacy. However, the detection of morphologic change alone may not be sufficient for assessing response to new anti-cancer medication in all solid tumors. During the past fifteen years, several molecular-targeted therapies and immunotherapies have emerged in cancer treatment which work by disrupting signaling pathways and inhibited cell growth. Tumor necrosis or lack of tumor progression is associated with a good therapeutic response even in the absence of tumor shrinkage. Therefore, the use of unmodified RECIST criteria to estimate morphological changes of tumor alone may not be sufficient to estimate tumor response for these new anti-cancer drugs. Several studies have reported the low reliability of RECIST in evaluating treatment response in different tumors such as hepatocellular carcinoma, lung cancer, prostate cancer, brain glioma, bone metastasis, and lymphoma. There is an increased need for new medical imaging biomarkers, considering the changes in tumor viability, metabolic activity, and attenuation, which are related to early tumor response. Promising imaging techniques, beyond RECIST, include dynamic contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI), diffusion-weight imaging (DWI), magnetic resonance spectroscopy (MRS), and 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET). This review outlines the current RECIST with their limitations and the new emerging concepts of imaging biomarkers in oncology.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ren Yeh
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeon-Hor Chen
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan. .,Tu & Yuan Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, 164 Irvine Hall, Irvine, CA, 92697 - 5020, USA.
| |
Collapse
|
9
|
Skarping I, Förnvik D, Heide-Jørgensen U, Rydén L, Zackrisson S, Borgquist S. Neoadjuvant breast cancer treatment response; tumor size evaluation through different conventional imaging modalities in the NeoDense study. Acta Oncol 2020; 59:1528-1537. [PMID: 33063567 DOI: 10.1080/0284186x.2020.1830167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) is offered to an increasing number of breast cancer (BC) patients, and comprehensive monitoring of treatment response is of utmost importance. Several imaging modalities are available to follow tumor response, although likely to provide different clinical information. We aimed to examine the association between early radiological response by three conventional imaging modalities and pathological complete response (pCR). Further, we investigated the agreement between these modalities pre-, during, and post-NACT, and the accuracy of predicting pathological residual tumor burden by these imaging modalities post-NACT. MATERIAL AND METHODS This prospective Swedish cohort study included 202 BC patients assigned to NACT (2014-2019). Breast imaging with clinically used modalities: mammography, ultrasound, and tomosynthesis was performed pre-, during, and post-NACT. We investigated the agreement of tumor size by the different imaging modalities, and their accuracy of tumor size estimation. Patients with a radiological complete response or radiological partial response (≥30% decrease in tumor diameter) during NACT were classified as radiological early responders. RESULTS Patients with an early radiological response by ultrasound had 2.9 times higher chance of pCR than early radiological non-responders; the corresponding relative chance for mammography and tomosynthesis tumor size measures was 1.8 and 2.8, respectively. Post-NACT, each modality, separately, could accurately estimate tumor size (within 5 mm margin compared to pathological evaluation) in 43-46% of all tumors. The diagnostic precision in predicting pCR post-NACT was similar between the three imaging modalities; however, tomosynthesis had slightly higher specificity and positive predictive values. CONCLUSION Breast imaging modalities correctly estimated pathological tumor size in less than half of the tumors. Based on this finding, predicting residual tumor size post-NACT is challenging using conventional imaging. Patients with early radiological non-response might need improved monitoring during NACT and be considered for changed treatment plans.
Collapse
Affiliation(s)
- Ida Skarping
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Daniel Förnvik
- Department of Translational Medicine, Medical Radiation Physics, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Uffe Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Lisa Rydén
- Department of Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Sophia Zackrisson
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
10
|
Basnet B, Goyal P, Mahawar V, Bothra SJ, Agrawal C, Thapa BB, Talwar V, Jain P, Babu Koyyala VP, Goel V, Batra U, Doval DC. Role of 18F-Flurodeoxyglucose Positron-Emission Tomography/Computed Tomography in the Evaluation of Early Response to Neoadjuvant Chemotherapy in Patients with Locally Advanced Triple-Negative Breast Cancer. INDIAN JOURNAL OF NUCLEAR MEDICINE : IJNM : THE OFFICIAL JOURNAL OF THE SOCIETY OF NUCLEAR MEDICINE, INDIA 2020; 35:105-109. [PMID: 32351263 PMCID: PMC7182325 DOI: 10.4103/ijnm.ijnm_210_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 11/09/2022]
Abstract
Background: Response evaluation in locally advanced breast cancer is done through different methods ranging from clinical examination to magnetic resonance imaging, however evaluation with positron-emission tomography/computed tomography (PET/CT) in now being incorporated for the response evaluation. The aim of the present study is to correlate response to neoadjuvant chemotherapy (NACT) with PET/CT scan. Materials and Methods: The present study is a retrospective analysis of 30 locally advanced, triple-negative breast cancer patients. PET/CT scan was done pretreatment and post three and six cycles of NACT and was correlated with pathologic complete response (pCR). Responding disease was considered when there was at least a 50% reduction in the longest diameter. Results: The median pretreatment size of the breast lesion in CT scan was 3.9 ± 2.3 cm (2–12 cm) and maximum standardized uptake value (SUVmax) on PET/CT was 8.5 ± 5.5 (2.9–24). Among the responders, the median decrease in size of lesion was 3.2 ± 1.3 cm and median reduction in SUV of the tumor among was −8.1 ± 5.4 and was statistically significant when compared with nonresponders (P < 0.001). CT scan has 66% accuracy and PET has 82% accuracy at post three cycles NACT in predicting the pathological response. PET/CT had higher sensitivity and specificity when compared with CT findings alone in response evaluation. Conclusion: PET/CT scan can be considered as a sensitive tool for predicting pCRs and further larger trials are required to establish these findings.
Collapse
Affiliation(s)
- Bina Basnet
- Department of Radiology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Pankaj Goyal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Vivek Mahawar
- Department of Radiology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Sneha Jatan Bothra
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Chaturbhuj Agrawal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Bikash Bikram Thapa
- Department of Surgical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Vineet Talwar
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Parveen Jain
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | | | - Varun Goel
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Ullas Batra
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Dinesh Chandra Doval
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| |
Collapse
|
11
|
Influx rate constant of 18F-FDG increases in metastatic lymph nodes of non-small cell lung cancer patients. Eur J Nucl Med Mol Imaging 2020; 47:1198-1208. [PMID: 31974680 DOI: 10.1007/s00259-020-04682-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Primary tumor (PT) and metastatic lymph node (MLN) status have a great influence on diagnosis and treatment of lung cancer. Our main purpose was to investigate the imaging characteristics of PT or MLN by applying the 18F-FDG PET dynamic modeling approach for non-small cell lung cancer (NSCLC). METHODS Dynamic 18F-FDG PET scans were performed for 76 lung cancer patients, and 62 NSCLC cases were finally included in this study: 37 with newly diagnosed early and locally advanced lung cancer without distant metastases (group M0) and 25 metastatic lung cancer (group M1). Patlak graphic analysis (Ki calculation) based on the dynamic modeling and SUV analysis from conventional static data were performed. RESULTS For PT, both KiPT (0.050 ± 0.005 vs 0.026 ± 0.004 min-1, p < 0.001) and SUVPT (8.41 ± 0.64 vs 5.23 ± 0.73, p < 0.01) showed significant higher values in group M1 than M0. For MLN, KiMLN showed significant higher values in M1 than M0 (0.033 ± 0.005 vs 0.016 ± 0.003 min-1, p < 0.01), while no significant differences were found for SUVMLN between M0 and M1 (4.22 ± 0.49 vs 5.57 ± 0.59, p > 0.05). Both SUV PT and KiPT showed significant high values in squamous cell carcinoma than adenocarcinoma, but neither SUVPT nor KiPT showed significant differences between EGFR mutants versus wild types. The overall Spearman analysis for SUV and Ki from different groups showed variable correlation (r = 0.46-0.94). CONCLUSION The dynamic modeling for MLN (KiMLN) showed more sensitive than the static analysis (SUV) to detect metastatic lymph nodes in NSCLC, although both methods were sensitive for PT. This methodology of non-invasive imaging may become an important tool to evaluate MLN and PT status for patients who cannot undergo histological examination. CLINICAL TRIAL REGISTRATION The clinical trial registration number is NCT03679936 (http://www.clinicaltrials.gov/).
Collapse
|
12
|
Pitman KE, Alluri SR, Kristian A, Aarnes EK, Lyng H, Riss PJ, Malinen E. Influx rate of 18F-fluoroaminosuberic acid reflects cystine/glutamate antiporter expression in tumour xenografts. Eur J Nucl Med Mol Imaging 2019; 46:2190-2198. [PMID: 31264167 DOI: 10.1007/s00259-019-04375-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE 18F-fluoroaminosuberic acid (18F-FASu) is a recently developed amino acid tracer for positron emission tomography (PET) of oxidative stress that may offer improved tumour assessment over the conventional tracer 18F-fluorodeoxyglucose (18F-FDG). Our aim was to evaluate and relate dynamic 18F-FASu and 18F-FDG uptake with pharmacokinetic modelling to transporter protein expression levels in a panel of diverse tumour xenograft lines. METHODS Four different tumour xenograft lines were implanted in female athymic nude mice: MAS98.12 and HBCx3 (breast), TPMX (osteosarcoma) and A549 (lung). Dynamic PET over 60 min was performed on a small animal unit. The time-activity curves (TACs) for 18F-FASu and 18F-FDG in individual tumours were used to extract early (SUVE; 2 min p.i.) and late (SUVL; 55 min p.i.) standardised uptake values. Pharmacokinetic two-tissue compartment models were applied to the TACs to estimate rate constants K1-k4 and blood volume fraction vB. Relative levels of cystine/glutamate antiporter subunit xCT were assessed by western blotting, and expression of GLUT1 and CD31 by immunohistochemistry. RESULTS 18F-FASu showed higher SUVE, whilst 18F-FDG exhibited higher SUVL. Influx rate K1 for 18F-FASu was significantly correlated with xCT levels (p = 0.001) and was significantly higher than K1 for 18F-FDG (p < 0.001). K1 for 18F-FDG was significantly correlated with GLUT1 levels (p = 0.002). vB estimated from 18F-FASu and 18F-FDG TACs was highly consistent and significantly correlated (r = 0.85, p < 0.001). Two qualitatively different 18F-FASu uptake profiles were identified: type α with low xCT expression and low K1 (A549 and HBCx3), and type β with high xCT expression and high K1 (MAS98.12 and TPMX). CONCLUSION The influx rate of 18F-FASu reflects xCT activity in tumour xenografts. Dynamic PET with pharmacokinetic modelling is needed to fully appraise 18F-FASu distribution routes.
Collapse
Affiliation(s)
- Kathinka E Pitman
- Department of Physics, University of Oslo, P.O. Box 1048 Blindern, 0316, Oslo, Norway
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Santosh R Alluri
- Department of Chemistry, University of Oslo, P.O. Box 1048 Blindern, 0316, Oslo, Norway
| | | | | | - Heidi Lyng
- Department of Radiation Biology, Oslo University Hospital, Oslo, Norway
| | - Patrick J Riss
- Department of Chemistry, University of Oslo, P.O. Box 1048 Blindern, 0316, Oslo, Norway
| | - Eirik Malinen
- Department of Physics, University of Oslo, P.O. Box 1048 Blindern, 0316, Oslo, Norway.
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
13
|
Cochran JM, Busch DR, Leproux A, Zhang Z, O’Sullivan TD, Cerussi AE, Carpenter PM, Mehta RS, Roblyer D, Yang W, Paulsen KD, Pogue B, Jiang S, Kaufman PA, Chung SH, Schnall M, Snyder BS, Hylton N, Carp SA, Isakoff SJ, Mankoff D, Tromberg BJ, Yodh AG. Tissue oxygen saturation predicts response to breast cancer neoadjuvant chemotherapy within 10 days of treatment. JOURNAL OF BIOMEDICAL OPTICS 2018; 24:1-11. [PMID: 30338678 PMCID: PMC6194199 DOI: 10.1117/1.jbo.24.2.021202] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/30/2018] [Indexed: 05/20/2023]
Abstract
Ideally, neoadjuvant chemotherapy (NAC) assessment should predict pathologic complete response (pCR), a surrogate clinical endpoint for 5-year survival, as early as possible during typical 3- to 6-month breast cancer treatments. We introduce and demonstrate an approach for predicting pCR within 10 days of initiating NAC. The method uses a bedside diffuse optical spectroscopic imaging (DOSI) technology and logistic regression modeling. Tumor and normal tissue physiological properties were measured longitudinally throughout the course of NAC in 33 patients enrolled in the American College of Radiology Imaging Network multicenter breast cancer DOSI trial (ACRIN-6691). An image analysis scheme, employing z-score normalization to healthy tissue, produced models with robust predictions. Notably, logistic regression based on z-score normalization using only tissue oxygen saturation (StO2) measured within 10 days of the initial therapy dose was found to be a significant predictor of pCR (AUC = 0.92; 95% CI: 0.82 to 1). This observation suggests that patients who show rapid convergence of tumor tissue StO2 to surrounding tissue StO2 are more likely to achieve pCR. This early predictor of pCR occurs prior to reductions in tumor size and could enable dynamic feedback for optimization of chemotherapy strategies in breast cancer.
Collapse
Affiliation(s)
- Jeffrey M. Cochran
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
- Address all correspondence to: Jeffrey M. Cochran, E-mail:
| | - David R. Busch
- University of Texas Southwestern, Department of Anesthesiology and Pain Management, Dallas, Texas, United States
| | - Anaïs Leproux
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Zheng Zhang
- Brown University School of Public Health, Department of Biostatistics and Center for Statistical Sciences, Providence, Rhode Island, United States
| | - Thomas D. O’Sullivan
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Albert E. Cerussi
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Philip M. Carpenter
- University of Southern California, Keck School of Medicine, Department of Pathology, Los Angeles, California, United States
| | - Rita S. Mehta
- University of California Irvine, Department of Medicine, Irvine, California, United States
| | - Darren Roblyer
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Wei Yang
- University of Texas MD Anderson Cancer Center, Department of Diagnostic Radiology, Houston, Texas, United States
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States
| | - Brian Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States
| | - Shudong Jiang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States
| | - Peter A. Kaufman
- Dartmouth-Hitchcock Medical Center, Department of Hematology and Oncology, Lebanon, New Hampshire, United States
| | - So Hyun Chung
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
| | - Mitchell Schnall
- University of Pennsylvania, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - Bradley S. Snyder
- Brown University School of Public Health, Center for Statistical Sciences, Providence, Rhode Island, United States
| | - Nola Hylton
- University of California, Department of Radiology, San Francisco, California, United States
| | - Stefan A. Carp
- Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Boston, Massachusetts, United States
| | - Steven J. Isakoff
- Massachusetts General Hospital, Department of Hematology and Oncology, Boston, Massachusetts, United States
| | - David Mankoff
- University of Pennsylvania, Division of Nuclear Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - Bruce J. Tromberg
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Arjun G. Yodh
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
| |
Collapse
|
14
|
Abstract
After an overview of the principles of fludeoxyglucose-PET/computed tomography (CT) in breast cancer, its advantages and limits to evaluate treatment response are discussed. The metabolic information is helpful for early assessment of the response to neoadjuvant chemotherapy and could be used to monitor treatment, especially in aggressive breast cancer subtypes. PET/CT is also a powerful method for early assessment of the treatment response in the metastatic setting. It allows evaluation of different sites of metastases in a single examination and detection of a heterogeneous response. However, to use PET/CT to assess responses, methodology for image acquisition and analysis needs standardization.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, 1 Avenue Claude Vellefaux, Paris 75475 Cedex 10, France.
| |
Collapse
|
15
|
Slanetz PJ, Moy L, Baron P, diFlorio RM, Green ED, Heller SL, Holbrook AI, Lee SJ, Lewin AA, Lourenco AP, Niell B, Stuckey AR, Trikha S, Vincoff NS, Weinstein SP, Yepes MM, Newell MS. ACR Appropriateness Criteria ® Monitoring Response to Neoadjuvant Systemic Therapy for Breast Cancer. J Am Coll Radiol 2018; 14:S462-S475. [PMID: 29101985 DOI: 10.1016/j.jacr.2017.08.037] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 12/28/2022]
Abstract
Patients with locally advanced invasive breast cancers are often treated with neoadjuvant chemotherapy prior to definitive surgical intervention. The primary aims of this approach are to: 1) reduce tumor burden thereby permitting breast conservation rather than mastectomy; 2) promptly treat possible metastatic disease, whether or not it is detectable on preoperative staging; and 3) potentially tailor future chemotherapeutic decisions by monitoring in-vivo tumor response. Accurate radiological assessment permits optimal management and planning in this population. However, assessment of tumor size and response to treatment can vary depending on the modality used, the measurement technique (such as single longest diameter, 3-D measurements, or calculated tumor volume), and varied response of different tumor subtypes to neoadjuvant chemotherapy (such as concentric shrinkage or tumor fragmentation). As discussed in further detail, digital mammography, digital breast tomosynthesis, US and MRI represent the key modalities with potential to help guide patient management. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision include an extensive analysis of current medical literature from peer reviewed journals and the application of well-established methodologies (RAND/UCLA Appropriateness Method and Grading of Recommendations Assessment, Development, and Evaluation or GRADE) to rate the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where evidence is lacking or equivocal, expert opinion may supplement the available evidence to recommend imaging or treatment.
Collapse
Affiliation(s)
| | - Priscilla J Slanetz
- Principal Author, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| | - Linda Moy
- Panel Vice Chair, NYU Clinical Cancer Center, New York, New York
| | - Paul Baron
- Roper St. Francis Physician Partners Breast Surgery, Charleston, South Carolina; American College of Surgeons
| | | | - Edward D Green
- The University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | - Su-Ju Lee
- University of Cincinnati, Cincinnati, Ohio
| | - Alana A Lewin
- New York University School of Medicine, New York, New York
| | - Ana P Lourenco
- Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | | | - Ashley R Stuckey
- Women and Infants Hospital, Providence, Rhode Island; American Congress of Obstetricians and Gynecologists
| | | | - Nina S Vincoff
- Hofstra Northwell School of Medicine, Manhasset, New York
| | - Susan P Weinstein
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Mary S Newell
- Panel Chair, Emory University Hospital, Atlanta, Georgia
| |
Collapse
|
16
|
Sharma A, Mohan A, Bhalla AS, Sharma MC, Vishnubhatla S, Das CJ, Pandey AK, Sekhar Bal C, Patel CD, Sharma P, Agarwal KK, Kumar R. Role of Various Metabolic Parameters Derived From Baseline 18F-FDG PET/CT as Prognostic Markers in Non-Small Cell Lung Cancer Patients Undergoing Platinum-Based Chemotherapy. Clin Nucl Med 2018; 43:e8-e17. [PMID: 29112011 DOI: 10.1097/rlu.0000000000001886] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this study was to prospectively evaluate the role of various quantitative and semiquantitative metabolic parameters derived from dynamic and static baseline F-FDG PET/CT in prediction of overall survival (OS) in non-small cell lung cancer (NSCLC) patients who were planned to undergo platinum-based chemotherapy. METHODS Sixty patients (51 male and 9 female patients) with biopsy-proven NSCLC and mean age 59.55 ± 10.06 years who were planned to undergo platinum-based chemotherapy were enrolled in the study. Each patient underwent a baseline regional dynamic and a static whole-body F-FDG PET/CT after injecting 0.21 mCi/kg (5.18-7.77 MBq/kg) of F-FDG intravenously. Two dynamic PET/CT parameters, that is, net influx rate constant and glucose metabolic rate at 30 and 60 minutes, were evaluated. In addition, whole-body PET/CT parameters, that is, SUVmax, average SUV, tumor-to-background ratio, metabolic tumor volume (MTV), total lesion glycolysis (TLG) of the primary tumor, and MTV and TLG of whole-body tumor lesions, were evaluated. Best possible cutoffs for all parameters were calculated using receiver operating characteristic curve analysis. Survival analysis was performed using log-rank test, Kaplan-Meier curves, and Cox proportional hazards model to determine the prognostic markers for OS. RESULTS The median follow-up period was 4.4 months (range, 8 days to 15.9 months). In univariate analysis, the 4 static whole-body PET/CT parameters, that is, MTV, TLG, and MTV and TLG of whole-body tumor lesions, were found to be significantly associated with OS with cutoff values of 120, 800, 160, and 1350 cm and hazard ratios of 3.64 (P = 0.001), 3.35 (P = 0.002), 2.51 (P = 0.019), and 2.69 (P = 0.008), respectively. In multivariate survival analysis, MTV was found to be an independent prognostic marker for OS. CONCLUSIONS Baseline MTV and TLG evaluated from primary tumor as well as the whole-body tumor lesions are reliable prognostic markers of OS in NSCLC patients undergoing platinum-based chemotherapy. However, other baseline whole-body PET/CT parameters (SUVmax, average SUV, and tumor-to-background ratio) and dynamic PET/CT parameters (net influx rate constant, glucose metabolic rate) have no prognostic value in these patients.
Collapse
|
17
|
The use of breast imaging for predicting response to neoadjuvant lapatinib, trastuzumab and their combination in HER2-positive breast cancer: Results from Neo-ALTTO. Eur J Cancer 2017; 89:42-48. [PMID: 29227816 DOI: 10.1016/j.ejca.2017.10.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 11/22/2022]
Abstract
AIM To determine the value of mammography and breast ultrasound (US) in predicting outcomes in HER2 positive breast cancer patients (pts) within Neo-ALTTO trial. PATIENTS AND METHODS Mammography and US were required at baseline, week 6 and surgery. Two independent blinded investigators reviewed the measurements and assigned the corresponding response category. Pts showing complete or partial response according to RECIST (v1.1) were classified as responders. The association between imaging response at week 6 or prior to surgery was evaluated with respect to pathological complete response (pCR) and event-free Survival (EFS). RESULTS Of the 455 pts enrolled in the trial, 267 (61%) and 340 (77%) had evaluable mammography and US at week 6; 248 (56%) and 309 (70%) pts had evaluable mammography and US prior to surgery. At week 6, 32% and 43% of pts were classified as responders by mammography and US, respectively. pCR rates were twice as high for responders than non-responders (week 6: 46% versus 23% by US, p < 0.0001; 41% versus 24% by mammography, p = 0.007). Positive and negative predictive values of mammography and US prior to surgery were 37% and 35%, and 82% and 70%, respectively. No significant correlation was found between response by mammography and/or US at week 6/surgery and EFS. CONCLUSIONS Mammography and US were underused in Neo-ALTTO although US had the potential to assess early response whereas mammography to detect residual disease prior to surgery. Our data still emphasise the need for further imaging studies on pts treated with neoadjuvant HER2-targeted therapy.
Collapse
|
18
|
Sharma A, Mohan A, Bhalla AS, Vishnubhatla S, Pandey AK, Bal CS, Kumar R. Role of various semiquantitative parameters of 18F-FDG PET/CT studies for interim treatment response evaluation in non-small-cell lung cancer. Nucl Med Commun 2017; 38:858-867. [PMID: 28817456 DOI: 10.1097/mnm.0000000000000723] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The aim of this study was to prospectively evaluate the role of various semiquantitative parameters obtained from fluorine-18 fluorodeoxyglucose (F-FDG) PET/CT in interim treatment response assessment in biopsy-proven non-small-cell lung cancer (NSCLC) and to find the best parameter, if any. MATERIALS AND METHODS Totally, 32 patients (male/female: 25/7) with biopsy proven NSCLC and a mean age of 54.71±12.65 years were enrolled in the study. Each patient underwent whole-body F-FDG PET/CT scan after injecting 5.18-7.77 MBq/kg of F-FDG intravenously at baseline and after four cycles of chemotherapy. Five parameters - that is, target-to-background ratio (TBR), maximum standardized uptake value (SUVmax), average standardized uptake value (SUVavg), whole-body metabolic tumor volume (MTVwb), and whole-body total lesion glycolysis (TLGwb) - were evaluated for both scans along with their percentage changes ([INCREMENT]). Patients were divided into two response groups as per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria: responders and nonresponders. All parameters were compared among the two response groups using appropriate statistical methods; P value of less than 0.05 was considered significant. RESULTS All postchemotherapy parameters were found to have a significant role in the prediction of two response groups. Post-TBR had highest area under the receiver operating characteristic curve of 0.83 with a sensitivity and specificity of 75 and 82%, respectively, at a cutoff value of 4. The [INCREMENT]s, [INCREMENT]MTVwb, [INCREMENT]TLGwb, and [INCREMENT]SUVmax were significant with cutoffs of -56, -75, and -32%, respectively. [INCREMENT]MTVwb had the highest area under the receiver operating characteristic curve of 0.83 with sensitivity and specificity of 81.25%. In multivariate analysis, post-TBR and [INCREMENT]MTVwb were found to be the independent variables for prediction of interim treatment response. CONCLUSION Our study proves that a multitude of semiquantitative parameters as documented above differ significantly between two response groups in patients with advanced stage NSCLC receiving chemotherapy. Moreover, parameters in combination (ΔMTV and post-TBR) with appropriate cutoffs can predict response groups with acceptable reliability.
Collapse
Affiliation(s)
- Akshima Sharma
- Departments of aNuclear Medicine bPulmonary Medicine and Sleep Disorders cRadio Diagnosis dBiostatistics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | | | | | | | | | | | | |
Collapse
|
19
|
Gallivanone F, Panzeri MM, Canevari C, Losio C, Gianolli L, De Cobelli F, Castiglioni I. Biomarkers from in vivo molecular imaging of breast cancer: pretreatment 18F-FDG PET predicts patient prognosis, and pretreatment DWI-MR predicts response to neoadjuvant chemotherapy. MAGMA (NEW YORK, N.Y.) 2017; 30:359-373. [PMID: 28246950 PMCID: PMC5524876 DOI: 10.1007/s10334-017-0610-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Human cancers display intra-tumor phenotypic heterogeneity and recent research has focused on developing image processing methods extracting imaging descriptors to characterize this heterogeneity. This work assesses the role of pretreatment 18F-FDG PET and DWI-MR with respect to the prognosis and prediction of neoadjuvant chemotherapy (NAC) outcomes when image features are used to characterize primitive lesions from breast cancer (BC). MATERIALS AND METHODS A retrospective protocol included 38 adult women with biopsy-proven BC. Patients underwent a pre-therapy 18F-FDG PET/CT whole-body study and a pre-therapy breast multi-parametric MR study. Patients were then referred for NAC treatment and then for surgical resection, with an evaluation of the therapy response. Segmentation methods were developed in order to identify functional volumes both on 18F-FDG PET images and ADC maps. Macroscopic and histogram features were extracted from the defined functional volumes. RESULTS Our work demonstrates that macroscopic and histogram features from 18F-FDG PET are able to biologically characterize primitive BC, and define the prognosis. In addition, histogram features from ADC maps are able to predict the response to NAC. CONCLUSION Our work suggests that pre-treatment 18F-FDG PET and pre-treatment DWI-MR provide useful complementary information for biological characterization and NAC response prediction in BC.
Collapse
Affiliation(s)
- Francesca Gallivanone
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy
| | - Marta Maria Panzeri
- Department of Radiology, Centre for Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Canevari
- Department of Nuclear Medicine, Centre for Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Losio
- Department of Radiology, Centre for Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Gianolli
- Department of Nuclear Medicine, Centre for Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology, Centre for Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via Fratelli Cervi 93, Segrate, 20090, Milan, Italy.
| |
Collapse
|
20
|
Cochran JM, Chung SH, Leproux A, Baker WB, Busch DR, DeMichele AM, Tchou J, Tromberg BJ, Yodh AG. Longitudinal optical monitoring of blood flow in breast tumors during neoadjuvant chemotherapy. Phys Med Biol 2017; 62:4637-4653. [PMID: 28402286 DOI: 10.1088/1361-6560/aa6cef] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We measure tissue blood flow markers in breast tumors during neoadjuvant chemotherapy and investigate their correlation to pathologic complete response in a pilot longitudinal patient study (n = 4). Tumor blood flow is quantified optically by diffuse correlation spectroscopy (DCS), and tissue optical properties, blood oxygen saturation, and total hemoglobin concentration are derived from concurrent diffuse optical spectroscopic imaging (DOSI). The study represents the first longitudinal DCS measurement of neoadjuvant chemotherapy in humans over the entire course of treatment; it therefore offers a first correlation between DCS flow indices and pathologic complete response. The use of absolute optical properties measured by DOSI facilitates significant improvement of DCS blood flow calculation, which typically assumes optical properties based on literature values. Additionally, the combination of the DCS blood flow index and the tissue oxygen saturation from DOSI permits investigation of tissue oxygen metabolism. Pilot results from four patients suggest that lower blood flow in the lesion-bearing breast is correlated with pathologic complete response. Both absolute lesion blood flow and lesion flow relative to the contralateral breast exhibit potential for characterization of pathological response. This initial demonstration of the combined optical approach for chemotherapy monitoring provides incentive for more comprehensive studies in the future and can help power those investigations.
Collapse
Affiliation(s)
- J M Cochran
- Department of Physics and Astronomy, University of Pennsylvania, 209 S 33rd St, Philadelphia, PA 19104, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sajjadi AY, Isakoff SJ, Deng B, Singh B, Wanyo CM, Fang Q, Specht MC, Schapira L, Moy B, Bardia A, Boas DA, Carp SA. Normalization of compression-induced hemodynamics in patients responding to neoadjuvant chemotherapy monitored by dynamic tomographic optical breast imaging (DTOBI). BIOMEDICAL OPTICS EXPRESS 2017; 8:555-569. [PMID: 28270967 PMCID: PMC5330555 DOI: 10.1364/boe.8.000555] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/04/2016] [Accepted: 12/05/2016] [Indexed: 05/03/2023]
Abstract
We characterize novel breast cancer imaging biomarkers for monitoring neoadjuvant chemotherapy (NACT) and predicting outcome. Specifically, we recruited 30 patients for a pilot study in which NACT patients were imaged using dynamic tomographic optical breast imaging (DTOBI) to quantify the hemodynamic changes due to partial mammographic compression. DTOBI scans were obtained pre-treatment (referred to as day 0), as well as 7 and 30 days into therapy on female patients undergoing NACT. We present data for the 13 patients who participated in both day 0 and 7 measurements and had evaluable data, of which 7 also returned for day 30 measurements. We acquired optical images over 2 minutes following 4-8 lbs (18-36 N) of compression. The timecourses of tissue-volume averaged total hemoglobin (HbT), as well as hemoglobin oxygen saturation (SO2) in the tumor vs. surrounding tissues were compared. Outcome prediction metrics based on the differential behavior in tumor vs. normal areas for responders (>50% reduction in maximum diameter) vs. non-responders were analyzed for statistical significance. At baseline, all patients exhibit an initial decrease followed by delayed recovery in HbT, and SO2 in the tumor area, in contrast to almost immediate recovery in surrounding tissue. At day 7 and 30, this contrast is maintained in non-responders; however, in responders, the contrast in hemodynamic time-courses between tumor and normal tissue starts decreasing at day 7 and substantially disappears at day 30. At day 30 into NACT, responding tumors demonstrate "normalization" of compression induced hemodynamics vs. surrounding normal tissue whereas non-responding tumors did not. This data suggests that DTOBI imaging biomarkers, which are governed by the interplay between tissue biomechanics and oxygen metabolism, may be suitable for guiding NACT by offering early predictions of treatment outcome.
Collapse
Affiliation(s)
- Amir Y Sajjadi
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Charlestown, MA 02129, USA; These authors contributed equally to this work;
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; These authors contributed equally to this work;
| | - Bin Deng
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Bhawana Singh
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Christy M Wanyo
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Qianqian Fang
- Department of Bioengineering, Northeastern University, Boston, MA 0211, USA
| | - Michelle C Specht
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Lidia Schapira
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - David A Boas
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Stefan A Carp
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA; Department of Radiology, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
22
|
Katiyar P, Divine MR, Kohlhofer U, Quintanilla-Martinez L, Schölkopf B, Pichler BJ, Disselhorst JA. Spectral Clustering Predicts Tumor Tissue Heterogeneity Using Dynamic 18F-FDG PET: A Complement to the Standard Compartmental Modeling Approach. J Nucl Med 2016; 58:651-657. [PMID: 27811120 DOI: 10.2967/jnumed.116.181370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/19/2016] [Indexed: 12/11/2022] Open
Abstract
In this study, we described and validated an unsupervised segmentation algorithm for the assessment of tumor heterogeneity using dynamic 18F-FDG PET. The aim of our study was to objectively evaluate the proposed method and make comparisons with compartmental modeling parametric maps and SUV segmentations using simulations of clinically relevant tumor tissue types. Methods: An irreversible 2-tissue-compartmental model was implemented to simulate clinical and preclinical 18F-FDG PET time-activity curves using population-based arterial input functions (80 clinical and 12 preclinical) and the kinetic parameter values of 3 tumor tissue types. The simulated time-activity curves were corrupted with different levels of noise and used to calculate the tissue-type misclassification errors of spectral clustering (SC), parametric maps, and SUV segmentation. The utility of the inverse noise variance- and Laplacian score-derived frame weighting schemes before SC was also investigated. Finally, the SC scheme with the best results was tested on a dynamic 18F-FDG measurement of a mouse bearing subcutaneous colon cancer and validated using histology. Results: In the preclinical setup, the inverse noise variance-weighted SC exhibited the lowest misclassification errors (8.09%-28.53%) at all noise levels in contrast to the Laplacian score-weighted SC (16.12%-31.23%), unweighted SC (25.73%-40.03%), parametric maps (28.02%-61.45%), and SUV (45.49%-45.63%) segmentation. The classification efficacy of both weighted SC schemes in the clinical case was comparable to the unweighted SC. When applied to the dynamic 18F-FDG measurement of colon cancer, the proposed algorithm accurately identified densely vascularized regions from the rest of the tumor. In addition, the segmented regions and clusterwise average time-activity curves showed excellent correlation with the tumor histology. Conclusion: The promising results of SC mark its position as a robust tool for quantification of tumor heterogeneity using dynamic PET studies. Because SC tumor segmentation is based on the intrinsic structure of the underlying data, it can be easily applied to other cancer types as well.
Collapse
Affiliation(s)
- Prateek Katiyar
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Max Planck Institute for Intelligent Systems, Tuebingen, Germany; and
| | - Mathew R Divine
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University Tuebingen and Comprehensive Cancer Center, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Jonathan A Disselhorst
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
23
|
Tromberg BJ, Zhang Z, Leproux A, O'Sullivan TD, Cerussi AE, Carpenter PM, Mehta RS, Roblyer D, Yang W, Paulsen KD, Pogue BW, Jiang S, Kaufman PA, Yodh AG, Chung SH, Schnall M, Snyder BS, Hylton N, Boas DA, Carp SA, Isakoff SJ, Mankoff D. Predicting Responses to Neoadjuvant Chemotherapy in Breast Cancer: ACRIN 6691 Trial of Diffuse Optical Spectroscopic Imaging. Cancer Res 2016; 76:5933-5944. [PMID: 27527559 DOI: 10.1158/0008-5472.can-16-0346] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/03/2016] [Indexed: 02/03/2023]
Abstract
The prospective multicenter ACRIN 6691 trial was designed to evaluate whether changes from baseline to mid-therapy in a diffuse optical spectroscopic imaging (DOSI)-derived imaging endpoint, the tissue optical index (TOI), predict pathologic complete response (pCR) in women undergoing breast cancer neoadjuvant chemotherapy (NAC). DOSI instruments were constructed at the University of California, Irvine (Irvine, CA), and delivered to six institutions where 60 subjects with newly diagnosed breast tumors (at least 2 cm in the longest dimension) were enrolled over a 2-year period. Bedside DOSI images of the tissue concentrations of deoxy-hemoglobin (ctHHb), oxy-hemoglobin (ctHbO2), water (ctH2O), lipid, and TOI (ctHHb × ctH2O/lipid) were acquired on both breasts up to four times during NAC treatment: baseline, 1-week, mid-point, and completion. Of the 34 subjects (mean age 48.4 ± 10.7 years) with complete, evaluable data from both normal and tumor-containing breast, 10 (29%) achieved pCR as determined by central pathology review. The percent change in tumor-to-normal TOI ratio (%TOITN) from baseline to mid-therapy ranged from -82% to 321%, with a median of -36%. Using pCR as the reference standard and ROC curve methodology, %TOITN AUC was 0.60 (95% CI, 0.39-0.81). In the cohort of 17 patients with baseline tumor oxygen saturation (%StO2) greater than the 77% population median, %TOITN AUC improved to 0.83 (95% CI, 0.63-1.00). We conclude that the combination of baseline functional properties and dynamic optical response shows promise for clinical outcome prediction. Cancer Res; 76(20); 5933-44. ©2016 AACR.
Collapse
Affiliation(s)
- Bruce J Tromberg
- Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California.
| | - Zheng Zhang
- Department of Biostatistics and Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Anaïs Leproux
- Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
| | - Thomas D O'Sullivan
- Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
| | - Albert E Cerussi
- Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
| | | | - Rita S Mehta
- Department of Medicine, University of California Irvine, Irvine, California
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Wei Yang
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keith D Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Shudong Jiang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Peter A Kaufman
- Section of Hematology and Oncology, Dartmouth-Hitchcock Medical Center, Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Arjun G Yodh
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - So Hyun Chung
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mitchell Schnall
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bradley S Snyder
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Nola Hylton
- Department of Radiology, University of California, San Francisco, California
| | - David A Boas
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stefan A Carp
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Steven J Isakoff
- Hematology Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
24
|
Prognostic Role of Early and End-of-Neoadjuvant Treatment 18F-FDG PET/CT in Patients With Breast Cancer. Clin Nucl Med 2016; 41:e313-22. [DOI: 10.1097/rlu.0000000000001191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Goulon D, Necib H, Henaff B, Rousseau C, Carlier T, Kraeber-Bodere F. Quantitative Evaluation of Therapeutic Response by FDG-PET-CT in Metastatic Breast Cancer. Front Med (Lausanne) 2016; 3:19. [PMID: 27243012 PMCID: PMC4861036 DOI: 10.3389/fmed.2016.00019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/22/2016] [Indexed: 01/31/2023] Open
Abstract
PURPOSE To assess the therapeutic response for metastatic breast cancer with (18)F-FDG position emission tomography (PET), this retrospective study aims to compare the performance of six different metabolic metrics with PERCIST, PERCIST with optimal thresholds, and an image-based parametric approach. METHODS Thirty-six metastatic breast cancer patients underwent 128 PET scans and 123 lesions were identified. In a per-lesion and per-patient analysis, the performance of six metrics: maximum standardized uptake value (SUVmax), SUVpeak, standardized added metabolic activity (SAM), SUVmean, metabolic volume (MV), total lesion glycolysis (TLG), and a parametric approach (SULTAN) were determined and compared to the gold standard (defined by clinical assessment and biological and conventional imaging according RECIST 1.1). The evaluation was performed using PERCIST thresholds (for per-patient analysis only) and optimal thresholds (determined by the Youden criterion from the receiver operating characteristic curves). RESULTS In the per-lesion analysis, 210 pairs of lesion evolutions were studied. Using the optimal thresholds, SUVmax, SUVpeak, SUVmean, SAM, and TLG were significantly correlated with the gold standard. SUVmax, SUVpeak, and SUVmean reached the best sensitivity (91, 88, and 83%, respectively), specificity (93, 95, and 97%, respectively), and negative predictive value (NPV, 90, 88, and 83%, respectively). For the per--patient analysis, 79 pairs of PET were studied. The optimal thresholds compared to the PERCIST threshold did not improve performance for SUVmax, SUVpeak, and SUVmean. Only SUVmax, SUVpeak, SUVmean, and TLG were correlated with the gold standard. SULTAN also performed equally: 83% sensitivity, 88% specificity, and NPV 86%. CONCLUSION This study showed that SUVmax and SUVpeak were the best parameters for PET evaluation of metastatic breast cancer lesions. Parametric imaging is helpful in evaluating serial studies.
Collapse
Affiliation(s)
- Dorothée Goulon
- Service de médecine nucléaire, ICO Nantes , Saint Herblain Cedex , France
| | - Hatem Necib
- Service de radiologie, CHU Nantes , Nantes , France
| | - Brice Henaff
- Service de médecine nucléaire, CHU Nantes , Nantes , France
| | - Caroline Rousseau
- Service de médecine nucléaire, ICO Nantes, Saint Herblain Cedex, France; INSERM UM R892, Nantes, France
| | - Thomas Carlier
- Service de médecine nucléaire, CHU Nantes, Nantes, France; INSERM UM R892, Nantes, France
| | | |
Collapse
|
26
|
Jacobs MA, Wolff AC, Macura KJ, Stearns V, Ouwerkerk R, El Khouli R, Bluemke DA, Wahl R. Multiparametric and Multimodality Functional Radiological Imaging for Breast Cancer Diagnosis and Early Treatment Response Assessment. J Natl Cancer Inst Monogr 2016; 2015:40-6. [PMID: 26063885 DOI: 10.1093/jncimonographs/lgv014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the second leading cause of cancer death among US women, and the chance of a woman developing breast cancer sometime during her lifetime is one in eight. Early detection and diagnosis to allow appropriate locoregional and systemic treatment are key to improve the odds of surviving its diagnosis. Emerging data also suggest that different breast cancer subtypes (phenotypes) may respond differently to available adjuvant therapies. There is a growing understanding that not all patients benefit equally from systemic therapies, and therapeutic approaches are being increasingly personalized based on predictive biomarkers of clinical benefit. Optimal use of established and novel radiological imaging methods, such as magnetic resonance imaging and positron emission tomography, which have different biophysical mechanisms can simultaneously identify key functional parameters. These methods provide unique multiparametric radiological signatures of breast cancer, that will improve the accuracy of early diagnosis, help select appropriate therapies for early stage disease, and allow early assessment of therapeutic benefit.
Collapse
Affiliation(s)
- Michael A Jacobs
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD.
| | - Antonio C Wolff
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Katarzyna J Macura
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Vered Stearns
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Ronald Ouwerkerk
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Riham El Khouli
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - David A Bluemke
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| | - Richard Wahl
- The Russell H. Morgan Department of Radiology and Radiological Science (MAJ, KJM, RO, REK, DAB, RW), Sidney Kimmel Comprehensive Cancer Center (MAJ, ACW, KJM, VS, RW), and Department of Oncology (ACW, VS), The Johns Hopkins University School of Medicine, Baltimore, MD; National Institute of Diabetes and Digestive and Kidney Diseases (not affialted, RO) and Radiology and Imaging Sciences, National Institutes of Health Clinical Center (DAB), Bethesda, MD
| |
Collapse
|
27
|
18F-FDG PET/CT in the early prediction of pathological response in aggressive subtypes of breast cancer: review of the literature and recommendations for use in clinical trials. Eur J Nucl Med Mol Imaging 2016; 43:983-993. [DOI: 10.1007/s00259-015-3295-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/21/2015] [Indexed: 10/22/2022]
|
28
|
Kitajima K, Miyoshi Y. Present and future role of FDG-PET/CT imaging in the management of breast cancer. Jpn J Radiol 2016; 34:167-80. [DOI: 10.1007/s11604-015-0516-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/20/2015] [Indexed: 02/08/2023]
|
29
|
Utility of FDG-PET/CT in the evaluation of the response of locally advanced breast cancer to neoadjuvant chemotherapy. Int Surg 2015; 99:309-18. [PMID: 25058758 DOI: 10.9738/intsurg-d-13-00044.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) is effective in down-staging a primary tumor before surgery, and quick differentiation between responders to NAC and nonresponders is needed. We investigated the utility of [18F]fluorodeoxyglucose positron emission tomography (FDG-PET) and computed tomography (CT) in evaluating the therapeutic effectiveness of NAC. We investigated 25 patients who underwent NAC for stage II and III noninflammatory breast cancer. FDG-PET/CT was undertaken before and after NAC to determine the maximum standardized uptake value (SUVmax) reduction rate. Findings were compared with postoperative histopathologic evaluation of therapeutic response. It was not possible to accurately assess tumor response to NAC using CT. However, using the SUVmax reduction rate, we noted a significant difference (P=0.0420) between patients who were responsive and nonresponsive to NAC. The sensitivity and specificity were as high as 83.3% and 78.9%, respectively. This study demonstrated that FDG-PET/CT can differentiate responders from nonresponders. This improves patient management by avoiding unnecessary chemotherapy.
Collapse
|
30
|
Abstract
In a prior substudy of the CAN-NCIC-MA.22 clinical trial (ClinicalTrials.gov identifier NCT00066443), we observed that neoadjuvant chemotherapy reduced tumor RNA integrity in breast cancer patients, a phenomenon we term "RNA disruption." The purpose of the current study was to assess in the full patient cohort the relationship between mid-treatment tumor RNA disruption and both pCR post-treatment and, subsequently, disease-free survival (DFS) up to 108 months post-treatment. To meet these objectives, we developed the RNA disruption assay (RDA) to quantify RNA disruption and stratify it into 3 response zones of clinical importance. Zone 1 is a level of RNA disruption inadequate for pathologic complete response (pCR); Zone 2 is an intermediate level, while Zone 3 has high RNA disruption. The same RNA disruption cut points developed for pCR response were then utilized for DFS. Tumor RDA identified >fourfold more chemotherapy non-responders than did clinical response by calipers. pCR responders were clustered in RDA Zone 3, irrespective of tumor subtype. DFS was about 2-fold greater for patients with tumors in Zone 3 compared to Zone 1 patients. Kaplan-Meier survival curves corroborated these findings that high tumor RNA disruption was associated with increased DFS. DFS values for patients in zone 3 that did not achieve a pCR were similar to that of pCR recipients across tumor subtypes, including patients with hormone receptor positive tumors that seldom achieve a pCR. RDA appears superior to pCR as a chemotherapy response biomarker, supporting the prospect of its use in response-guided chemotherapy.
Collapse
|
31
|
Utility of [18F] Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography (FDG PET/CT) in the Initial Staging and Response Assessment of Locally Advanced Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Indian J Surg Oncol 2015; 6:330-6. [PMID: 27065657 DOI: 10.1007/s13193-015-0421-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 05/19/2015] [Indexed: 10/23/2022] Open
Abstract
In India up to 50 % of breast cancer patients still present as locally advanced breast cancer (LABC). The conventional methods of metastatic work up include physical examination, bone scan, chest & abdominal imaging, and biochemical tests. It is likely that the conventional staging underestimates the extent of initial spread and there is a need for more sophisticated staging procedure. The PET/CT can detect extra-axillary and occult distant metastases and also aid in predicting response to chemotherapy at an early point in time. To evaluate the utility of FDG PET/CT in initial staging and response assessment of patients with LABC receiving NACT. A prospective study of all biopsy confirmed female patients diagnosed with LABC receiving NACT from April 2013 to May 2014. The conventional work up included serum chemistry, CECT chest and abdomen and bone scan. A baseline whole body PET/CT was done in all patients. A repeat staging evaluation and a whole body PET/CT was done after 2/3rd cycle of NACT in non-responders and after 3/4 cycles in clinical responders. The histopathology report of the operative specimen was used to document the pathological response. The FDG PET/CT reported distant metastases in 11 of 38 patients, where as conventional imaging revealed metastases in only 6. Almost all the distant lesions detected by conventional imaging were detected with PET/CT, which showed additional sites of metastasis in 3 patients. In 2 patients, PET/CT detected osteolytic bone metastasis which were not detected by bone scan. In 5 patients PET CT detected N3 disease which were missed on conventional imaging. A total of 14 patients had second PET/CT done to assess the response to NACT and 11 patients underwent surgery. Two patients had complete pathological response. Of these 1 patient had complete metabolic and morphologic response and other had complete metabolic and partial morphologic response on second PET/CT scan. The 18 FDG PET/CT can detect more number of metastasis as well as additional sites of metastasis compared to conventional methods. The response assessment resulted in change of treatment regimen in 14 % of patients.
Collapse
|
32
|
Champion L, Lerebours F, Alberini JL, Fourme E, Gontier E, Bertrand F, Wartski M. 18F-FDG PET/CT to Predict Response to Neoadjuvant Chemotherapy and Prognosis in Inflammatory Breast Cancer. J Nucl Med 2015; 56:1315-21. [PMID: 26159587 DOI: 10.2967/jnumed.115.158287] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/18/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The aim of this prospective study was to assess the predictive value of (18)F-FDG PET/CT imaging for pathologic response to neoadjuvant chemotherapy (NACT) and outcome in inflammatory breast cancer (IBC) patients. METHODS Twenty-three consecutive patients (51 y ± 12.7) with newly diagnosed IBC, assessed by PET/CT at baseline (PET1), after the third course of NACT (PET2), and before surgery (PET3), were included. The patients were divided into 2 groups according to pathologic response as assessed by the Sataloff classification: pathologic complete response for complete responders (stage TA and NA or NB) and non-pathologic complete response for noncomplete responders (not stage A for tumor or not stage NA or NB for lymph nodes). In addition to maximum standardized uptake value (SUVmax) measurements, a global breast metabolic tumor volume (MTV) was delineated using a semiautomatic segmentation method. Changes in SUVmax and MTV between PET1 and PET2 (ΔSUV1-2; ΔMTV1-2) and PET1 and PET3 (ΔSUV1-3; ΔMTV1-3) were measured. RESULTS Mean SUVmax on PET1, PET2, and PET3 did not statistically differ between the 2 pathologic response groups. On receiver-operating-characteristic analysis, a 72% cutoff for ΔSUV1-3 provided the best performance to predict residual disease, with sensitivity, specificity, and accuracy of 61%, 80%, and 65%, respectively. On univariate analysis, the 72% cutoff for ΔSUV1-3 was the best predictor of distant metastasis-free survival (P = 0.05). On multivariate analysis, the 72% cutoff for ΔSUV1-3 was an independent predictor of distant metastasis-free survival (P = 0.01). CONCLUSION Our results emphasize the good predictive value of change in SUVmax between baseline and before surgery to assess pathologic response and survival in IBC patients undergoing NACT.
Collapse
Affiliation(s)
| | | | - Jean-Louis Alberini
- Service de Médecine Nucléaire, Institut Curie, Saint-Cloud, France Faculté de Médecine, Université Versailles Saint-Quentin, Saint-Quentin-en-Yvelines, France
| | | | - Eric Gontier
- Service de Médecine Nucléaire, Hôpital d'Instruction des Armées, Val de Grâce, Paris, France; and
| | | | - Myriam Wartski
- Service de Médecine Nucléaire, Institut Curie, Saint-Cloud, France
| |
Collapse
|
33
|
Dialani V, Chadashvili T, Slanetz PJ. Role of imaging in neoadjuvant therapy for breast cancer. Ann Surg Oncol 2015; 22:1416-24. [PMID: 25727555 DOI: 10.1245/s10434-015-4403-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Indexed: 12/20/2022]
Abstract
Neoadjuvant chemotherapy (NAC) involves administration of chemotherapeutic agents to patients with newly diagnosed breast cancer prior to definitive surgical treatment. Assessment of disease response to chemotherapeutic agents in vivo prior to any surgical intervention is necessary as medical oncologists are commonly tailoring or changing therapy during NAC based on response. It can also maximize the pathologic complete response (pCR) rate, resulting in more women undergoing breast conservation rather than mastectomy. Although some studies show a pCR to NAC in only 13-26 % of women, recent studies have shown higher pCR rates, especially for HER2-positive disease treated with targeted anti-HER2 therapy. Thus, accurate imaging tools for quantifying disease response are critical in the evaluation and management of patients undergoing NAC. There is currently no standard imaging method for monitoring response to therapy. Response to therapy tends to vary by tumor subtype and can be accurately assessed on imaging. We review the role of imaging before and after neoadjuvant therapy and discuss the advantages and limitations of currently available modalities, including mammography, ultrasonography, magnetic resonance imaging, and nuclear imaging.
Collapse
Affiliation(s)
- Vandana Dialani
- Division of Breast Imaging, Department of Radiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
34
|
Humbert O, Cochet A, Coudert B, Berriolo-Riedinger A, Kanoun S, Brunotte F, Fumoleau P. Role of positron emission tomography for the monitoring of response to therapy in breast cancer. Oncologist 2015; 20:94-104. [PMID: 25561512 DOI: 10.1634/theoncologist.2014-0342] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This review considers the potential utility of positron emission tomography (PET) tracers in the setting of response monitoring in breast cancer, with a special emphasis on glucose metabolic changes assessed with (18)F-fluorodeoxyglucose (FDG). In the neoadjuvant setting of breast cancer, the metabolic response can predict the final complete pathologic response after the first cycles of chemotherapy. Because tumor metabolic behavior highly depends on cancer subtype, studies are ongoing to define the optimal metabolic criteria of tumor response in each subtype. The recent multicentric randomized AVATAXHER trial has suggested, in the human epidermal growth factor 2-positive subtype, a clinical benefit of early tailoring the neoadjuvant treatment in women with poor metabolic response after the first course of treatment. In the bone-dominant metastatic setting, there is increasing clinical evidence that FDG-PET/computed tomography (CT) is the most accurate imaging modality for assessment of the tumor response to treatment when both metabolic information and morphologic information are considered. Nevertheless, there is a need to define standardized metabolic criteria of response, including the heterogeneity of response among metastases, and to evaluate the costs and health outcome of FDG-PET/CT compared with conventional imaging. New non-FDG radiotracers highlighting specific molecular hallmarks of breast cancer cells have recently emerged in preclinical and clinical studies. These biomarkers can take into account the heterogeneity of tumor biology in metastatic lesions. They may provide valuable clinical information for physicians to select and monitor the effectiveness of novel therapeutics targeting the same molecular pathways of breast tumor.
Collapse
Affiliation(s)
- Olivier Humbert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alexandre Cochet
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Bruno Coudert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alina Berriolo-Riedinger
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Salim Kanoun
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - François Brunotte
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Pierre Fumoleau
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| |
Collapse
|
35
|
Lee CI, Gold LS, Nelson HD, Chou R, Ramsey SD, Sullivan SD. Comparative effectiveness of imaging modalities to determine metastatic breast cancer treatment response. Breast 2014; 24:3-11. [PMID: 25479913 DOI: 10.1016/j.breast.2014.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/07/2014] [Accepted: 11/16/2014] [Indexed: 11/26/2022] Open
Abstract
We performed a systematic review to address the comparative effectiveness of different imaging modalities in evaluating treatment response among metastatic breast cancer patients. We searched seven multidisciplinary electronic databases for relevant publications (January 2003-December 2013) and performed dual abstraction of details and results for all clinical studies that involved stage IV breast cancer patients and evaluated imaging for detecting treatment response. Among 159 citations reviewed, 17 single-institution, non-randomized, observational studies met our inclusion criteria. Several studies demonstrate that changes in PET/CT standard uptake values are associated with changes in tumor volume as determined by bone scan, MRI, and/or CT. However, no studies evaluated comparative test performance between modalities or determined relationships between imaging findings and subsequent clinical decisions. Evidence for imaging's effectiveness in determining treatment response among metastatic breast cancer patients is limited. More rigorous research is needed to address imaging's value in this patient population.
Collapse
Affiliation(s)
- Christoph I Lee
- Department of Radiology, University of Washington School of Medicine, 825 Eastlake Avenue East, Seattle, WA 98109, USA; Department of Health Services, University of Washington School of Public Health, Box 357660, Seattle, WA 98195, USA; Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA; Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Laura S Gold
- Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Pharmaceutical Outcomes Research & Policy Program, School of Pharmacy, University of Washington, Box 257630, Seattle, WA 98195, USA.
| | - Heidi D Nelson
- Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Departments of Medical Informatics and Clinical Epidemiology and Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Providence Cancer Center, Providence Health & Services, 4805 NE Glisan Street, Portland, OR 97213, USA.
| | - Roger Chou
- Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Departments of Medical Informatics and Clinical Epidemiology and Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Scott D Ramsey
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA; Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Pharmaceutical Outcomes Research & Policy Program, School of Pharmacy, University of Washington, Box 257630, Seattle, WA 98195, USA; Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| | - Sean D Sullivan
- Department of Health Services, University of Washington School of Public Health, Box 357660, Seattle, WA 98195, USA; Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA; Pacific Northwest Evidence-Based Practice Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Pharmaceutical Outcomes Research & Policy Program, School of Pharmacy, University of Washington, Box 257630, Seattle, WA 98195, USA.
| |
Collapse
|
36
|
García Vicente AM, Cruz Mora MÁ, León Martín AA, Muñoz Sánchez MDM, Relea Calatayud F, Van Gómez López O, Espinosa Aunión R, Gonzalez Ageitos A, Soriano Castrejón Á. Glycolytic activity with 18F-FDG PET/CT predicts final neoadjuvant chemotherapy response in breast cancer. Tumour Biol 2014; 35:11613-20. [DOI: 10.1007/s13277-014-2495-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 08/13/2014] [Indexed: 11/30/2022] Open
|
37
|
Early and delayed prediction of axillary lymph node neoadjuvant response by 18F-FDG PET/CT in patients with locally advanced breast cancer. Eur J Nucl Med Mol Imaging 2014; 41:1309-18. [DOI: 10.1007/s00259-013-2657-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 11/28/2013] [Indexed: 11/26/2022]
|
38
|
Humbert O, Cochet A, Riedinger JM, Berriolo-Riedinger A, Arnould L, Coudert B, Desmoulins I, Toubeau M, Dygai-Cochet I, Guiu S, Coutant C, Fumoleau P, Brunotte F. HER2-positive breast cancer: ¹⁸F-FDG PET for early prediction of response to trastuzumab plus taxane-based neoadjuvant chemotherapy. Eur J Nucl Med Mol Imaging 2014; 41:1525-33. [PMID: 24647576 DOI: 10.1007/s00259-014-2739-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 02/17/2014] [Indexed: 02/03/2023]
Abstract
PURPOSE To investigate the value of (18)F-fluorodeoxyglucose positron emission tomography ((18)F-FDG PET/CT) to predict a pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) in women with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. MATERIAL AND METHODS Fifty-seven consecutive women with HER2-positive breast cancer, treated with trastuzumab plus taxane-based NAC, were prospectively included. Maximum Standardized Uptake Value of the primary tumor and axillary nodes were measured at baseline (PET₁.SUVmax) and after the first course of NAC (PET₂.SUVmax). Tumor metabolic volumes were assessed to determine Total Lesion Glycolysis (TLG). The tumor metabolic response (ΔSUVmax and ΔTLG) was calculated. RESULTS In univariate analysis, negative hormonal receptor status (p = 0.04), high tumor grade (p = 0.03), and low tumor PET₂.SUVmax (p = 0.001) were predictive of pCR. Tumor ΔSUVmax correlated with pCR (p = 0.03), provided that tumors with low metabolic activity at baseline were excluded. ΔTLG did not correlate with pCR. In multivariate analysis, tumor PET₂.SUVmax < 2.1 was the best independent predictive factor (Odds ratio =14.3; p = 0.004) with both negative and positive predictive values of 76 %. Although the metabolic features of the primary tumor did not depend on hormonal receptor status, both the baseline metabolism and early response of axillary nodes were higher if estrogen receptors were not expressed (p = 0.01 and p = 0.03, respectively). CONCLUSION In HER2-positive breast cancer, very low tumor residual metabolism after the first cycle of NAC (SUVmax < 2.1) was the main predictor of pCR. These results should be further explored in multicenter studies and incorporated into the design of clinical trials.
Collapse
Affiliation(s)
- Olivier Humbert
- Department of Nuclear Medicine, Centre GF Leclerc, 1 rue du Pr Marion, 21000, Dijon, France,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kurland BF, Doot RK, Linden HM, Mankoff DA, Kinahan PE. Multicenter trials using ¹⁸F-fluorodeoxyglucose (FDG) PET to predict chemotherapy response: effects of differential measurement error and bias on power calculations for unselected and enrichment designs. Clin Trials 2013; 10:886-95. [PMID: 24169628 DOI: 10.1177/1740774513506618] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Clinical validation of a predictive biomarker is especially difficult when the biomarker cannot be assessed retrospectively. A cost-effective, prospective multicenter replication study with rapid accrual is warranted prior to further validation studies such as a marker-based strategy for treatment selection. However, it is often unknown how measurement error and bias in a multicenter trial will differ from that in single-institution studies. PURPOSE Power calculations using simulated data may inform the efficient design of a multicenter study to replicate single-institution findings. This case study used serial standardized uptake value (SUV) measures from (18)F-fluorodeoxyglucose (FDG) positron emission tomography (PET) to predict early response to breast cancer neoadjuvant chemotherapy. We examined the impact of accelerating accrual through increased inclusion of secondary sites with greater levels of measurement error and bias. We also examined whether enrichment designs based on breast cancer initial uptake could increase the study power for a fixed budget (200 total scans). METHODS Reference FDG PET SUV data were selected with replacement from a single-institution trial; pathologic complete response (pCR) data were simulated using a logistic regression model predicting response by mid-therapy percent change in SUV. The impact of increased error for SUV measurements in multicenter trials was simulated by sampling from error and bias distributions: 20%-40% measurement error, 0%-40% bias, and fixed error/bias values. The proportion of patients recruited from secondary sites (with higher additional error/bias compared to primary sites) varied from 25% to 75%. RESULTS Reference power (from source data with no added error) was 0.92 for N = 100 to detect an association between percentage change in SUV and response. With moderate (20%) simulated measurement error for 3/4, 1/2, and 1/4 of measurements and 40% for the remainder, power was 0.70, 0.61, and 0.53, respectively. Reduction of study power was similar for other manifestations of measurement error (bias as a percentage of true value, absolute error, and absolute bias). Enrichment designs, which recruit additional patients by not conducting a second scan in patients with unsuitable pre-therapy uptake (low baseline SUV), did not lead to greater power for studies constrained to the same total cost. LIMITATIONS Simulation parameters could be incorrect, or not generalizable. Under a different logistic regression model relating mid-therapy percent change in SUV to pCR (with no relationship for patients with low baseline SUV, rather than the modest point estimate from reference data), the enrichment design did have somewhat greater power than the unselected design. CONCLUSION Even moderate additional measurement error substantially reduced study power under both unselected and enrichment designs.
Collapse
|
40
|
Koolen BB, Pengel KE, Wesseling J, Vogel WV, Vrancken Peeters MJTFD, Vincent AD, Gilhuijs KGA, Rodenhuis S, Rutgers EJT, Valdés Olmos RA. Sequential 18F-FDG PET/CT for early prediction of complete pathological response in breast and axilla during neoadjuvant chemotherapy. Eur J Nucl Med Mol Imaging 2013; 41:32-40. [DOI: 10.1007/s00259-013-2515-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 07/07/2013] [Indexed: 12/21/2022]
|
41
|
|
42
|
Mghanga FP, Lan X, Bakari KH, Li C, Zhang Y. Fluorine-18 fluorodeoxyglucose positron emission tomography-computed tomography in monitoring the response of breast cancer to neoadjuvant chemotherapy: a meta-analysis. Clin Breast Cancer 2013; 13:271-9. [PMID: 23714689 DOI: 10.1016/j.clbc.2013.02.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/06/2013] [Accepted: 02/04/2013] [Indexed: 10/26/2022]
Abstract
INTRODUCTION To evaluate the diagnostic performance of fluorine-18 fluorodeoxyglucose positron emission tomography (FDG-PET) in monitoring the response of breast cancers to neoadjuvant chemotherapy. METHODS Articles published in medical and oncologic journals between January 2000 and June 2012 were identified by systematic MEDLINE, Cochrane Database for Systematic Reviews, and EMBASE, and by manual searches of the references listed in original and review articles. Quality of the included studies was assessed by using the quality assessment of diagnosis accuracy studies score tool. Meta-DiSc statistical software was used to calculate the summary sensitivity and specificity, positive predictive and negative predictive values, and the summary receiver operating characteristics curve (SROC). RESULTS Fifteen studies with 745 patients were included in the study after meeting the inclusion criteria. The pooled sensitivity and specificity of FDG-PET or PET/CT were 80.5% (95% CI, 75.9%-84.5%) and 78.8% (95% CI, 74.1%-83.0%), respectively, and the positive predictive and negative predictive values were 79.8% and 79.5%, respectively. After 1 and 2 courses of chemotherapy, the pooled sensitivity and false-positive rate were 78.2% (95% CI, 73.8%-82.5%) and 11.2%, respectively; and 82.4% (95% CI, 77.4%-86.1%) and 19.3%, respectively. CONCLUSIONS Analysis of the findings suggests that FDG-PET has moderately high sensitivity and specificity in early detection of responders from nonresponders, and can be applied in the evaluation of breast cancer response to neoadjuvant chemotherapy in patients with breast cancer.
Collapse
Affiliation(s)
- Fabian Pius Mghanga
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | | | | | | | | |
Collapse
|
43
|
Hatt M, Groheux D, Martineau A, Espié M, Hindié E, Giacchetti S, de Roquancourt A, Visvikis D, Cheze-Le Rest C. Comparison between 18F-FDG PET image-derived indices for early prediction of response to neoadjuvant chemotherapy in breast cancer. J Nucl Med 2013; 54:341-9. [PMID: 23327900 DOI: 10.2967/jnumed.112.108837] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The goal of this study was to determine the best predictive factor among image-derived parameters extracted from sequential (18)F-FDG PET scans for early tumor response prediction after 2 cycles of neoadjuvant chemotherapy in breast cancer. METHODS 51 breast cancer patients were included. Responder and nonresponder status was determined by histopathologic examination according to the tumor and node Sataloff scale. PET indices (maximum and mean standardized uptake value [SUV], metabolically active tumor volume, and total lesion glycolysis [TLG]), at baseline and their variation (Δ) after 2 cycles of neoadjuvant chemotherapy were extracted from the PET images. Their predictive value was investigated using Mann-Whitney U tests and receiver-operating-characteristic analysis. Subgroup analysis was also performed by considering estrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative, triple-negative, and HER2-positive tumors separately. The impact of partial-volume correction was also investigated using an iterative deconvolution algorithm. RESULTS There were 24 pathologic nonresponders and 27 responders. None of the baseline PET parameters was correlated with response. After 2 neoadjuvant chemotherapy cycles, the reduction of each parameter was significantly associated with response, the best prediction of response being obtained with ΔTLG (96% sensitivity, 92% specificity, and 94% accuracy), which had a significantly higher area under the curve (0.91 vs. 0.82, P = 0.01) than did ΔSUVmax (63% sensitivity, 92% specificity, and 77% accuracy). Subgroup analysis confirmed a significantly higher accuracy for ΔTLG than ΔSUV for ER-positive/HER-negative but not for triple-negative and HER2-positive tumors. Partial-volume correction had no impact on the predictive value of any of the PET image-derived parameters despite significant changes in their absolute values. CONCLUSION Our results suggest that the reduction after 2 neoadjuvant chemotherapy cycles of the metabolically active volume of primary tumor measurements such as ΔTLG predicts histopathologic tumor response with higher accuracy than does ΔSUV measurements, especially for ER-positive/HER2-negative breast cancer. These results should be confirmed in a larger group of patients as they may potentially increase the clinical value and efficiency of (18)F-FDG PET for early prediction of response to neoadjuvant chemotherapy.
Collapse
|
44
|
Groheux D, Espié M, Giacchetti S, Hindié E. Performance of FDG PET/CT in the clinical management of breast cancer. Radiology 2012; 266:388-405. [PMID: 23220901 DOI: 10.1148/radiol.12110853] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this analysis, the role of metabolic imaging with fluorine 18 fluorodeoxyglucose (FDG) in breast cancer is reviewed. The analysis was limited to recent works by using state-of-the-art positron emission tomography (PET)/computed tomography (CT) technology. The strengths and limitations of FDG PET/CT are examined in various clinical settings, and the following questions are answered: Is FDG PET/CT useful to differentiate malignant from benign breast lesions? Can FDG PET/CT replace sentinel node biopsy for axillary staging? What is the role of FDG PET/CT in initial staging of inflammatory or locally advanced breast cancer? What is the role of FDG PET/CT in initial staging of clinical stage IIA and IIB and primary operable stage IIIA breast cancer? How does FDG PET/CT compare with conventional techniques in the restaging of cancer in patients who are suspected of having disease recurrence? What is the role of FDG PET/CT in the assessment of early response to neoadjuvant therapy and of response to therapy for metastatic disease? Some recommendations for clinical practice are given.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine and Department of Medical Oncology, Breast Diseases Unit, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475 Paris Cedex 10, France.
| | | | | | | |
Collapse
|
45
|
Im HJ, Kim YK, Kim YI, Lee JJ, Lee WW, Kim SE. Usefulness of Combined Metabolic-Volumetric Indices of (18)F-FDG PET/CT for the Early Prediction of Neoadjuvant Chemotherapy Outcomes in Breast Cancer. Nucl Med Mol Imaging 2012; 47:36-43. [PMID: 24895506 DOI: 10.1007/s13139-012-0181-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 10/08/2012] [Accepted: 10/09/2012] [Indexed: 11/25/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the usefulness of metabolic-volumetric indices of (18)F- fluorodeoxy-D-glucose ((18)F-FDG) positron emission tomography/computed tomography (PET/CT) for the evaluation of neoadjuvant chemotherapy outcomes in breast cancer. METHODS Twenty-four patients with locally advanced breast cancer were enrolled in the study. They underwent baseline (18)F-FDG PET/CT scan and received four or six cycles of neoadjuvant chemotherapy, interim (18)F-FDG PET/CT was done after second cycle of chemotherapy. Maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) of the primary lesions were calculated. Reduction rates of these parameters were obtained between baseline and interim (18)F-FDG PET/CT. Chemotherapy outcomes were assessed using tumor size reduction rate and histological grading system (Miller and Payne system). Reduction rates of SUVmax, MTV, and TLG correlated with chemotherapy outcomes. RESULTS MTV and TLG reduction rates showed significant correlation with tumor size reduction rate (R = 0.68, P = 0.0004; R = 0.62, P = 0.002, respectively). However, SUVmax reduction rate showed no significant correlation. MTV and TLG reduction rates were significantly higher in responders than nonresponders, as determined by Miller and Payne system (P < 0.0007, P < 0.002). However, SUVmax reduction rate showed no significant difference. On ROC analysis, the area under the MTV and TLG curves was 0.886, and that of SUVmax was 0.743. Sensitivity, specificity, positive predictive value, and negative predictive value to predict histopathologic response were the same for MTV and TLG, and the values were 100 %, 85.7 %, 83.3 %, and 100 %, respectively (at the reduction rate of 93.2 % for MTV, and 95.8 % for TLG). CONCLUSION Changes of metabolic-volumetric indices successfully reflected the neoadjuvant chemotherapy outcomes. MTV and TLG could be robust indices in discriminating pathologic responder as SUVmax, after neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Hyung-Jun Im
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea ; Department of Molecular Medicine and Biopharmaceutical Sciences, WCU Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea ; Department of Nuclear Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Borame-gil 41, Dongjak-gu, Seoul, 156-707 Korea
| | - Yong-Il Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea ; Department of Molecular Medicine and Biopharmaceutical Sciences, WCU Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Jong Jin Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Won Woo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea ; Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggyi Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea ; Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggyi Korea
| |
Collapse
|
46
|
|
47
|
Buchbender C, Kuemmel S, Hoffmann O, Stahl AR, Kimmig R, Otterbach F, Ladd S, Koeninger A, Forsting M, Bockisch A, Antoch G, Heusner TA. FDG-PET/CT for the early prediction of histopathological complete response to neoadjuvant chemotherapy in breast cancer patients: initial results. Acta Radiol 2012; 53:628-36. [PMID: 22761341 DOI: 10.1258/ar.2012.110699] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Up to about one-quarter of patients treated with neoadjuvant chemotherapy do not adequately respond to the given treatment. By a differentiation between responders and non-responders ineffective toxic therapies can be prevented. PURPOSE To retrospectively test if FDG-PET/CT is able to early differentiate between breast cancer lesions with pathological complete response (pCR) and lesions without pathological complete response (npCR) after two cycles of neoadjuvant chemotherapy (NACT). MATERIAL AND METHODS In this retrospective study 26 breast cancer patients (mean age, 46.9 years ± 9.9 years) underwent a pre-therapeutic FDG-PET/CT scan and a subsequent FDG-PET/CT after the second cycle of NACT. Histopathology of resected specimen served as the reference standard. Maximum standardized uptake values (SUVmax) of cancer lesions before and after the second cycle of NACT were measured. Two evaluation algorithms were used: (a) pCR: Sinn Score 3 and 4, npCR: Sinn Score 0-2; (b) pCR: Sinn Score 4, npCR: Sinn Score 0-3. The absolute and relative decline of the SUVmax (ΔSUVmax, ΔSUVmax(%))was calculated. Differences of the SUVmax as well as of the SUVmax decline between pCR lesions and npCR lesions were tested for statistical significance P < 0.05. To identify the optimal cut-off value of ΔSUVmax(%) to differentiate between pCR lesions and npCR lesions a receiver-operating curve (ROC) analysis was performed. RESULTS Using evaluation algorithm A the ΔSUVmax was 13.5 (pCR group) and 3.9 (npCR group) (P = 0.006); the ΔSUVmax(%) was 79% and 47%, respectively (P = 0.001). On ROC analysis an optimal cut-off ΔSUVmax(%) of 66% was found. Using evaluation algorithm B the ΔSUVmax was 17.5 (pCR group) and 4.9 (npCR group) (P = 0.013); the ΔSUVmax(%) was 89% and 51%, respectively (P = 0.003). On ROC analysis an optimal cut-off ΔSUVmax(%) of 88% was found. CONCLUSION FDG-PET/CT may be able to early differentiate between pCR and npCR of primary breast cancer lesions after two cycles of NACT.
Collapse
Affiliation(s)
- Christian Buchbender
- Univ Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, Dusseldorf
- Univ Duisburg-Essen, Medical Faculty, Department of Diagnostic and Interventional Radiology and Neuroradiology, Essen
| | - Sherko Kuemmel
- Univ Duisburg-Essen, Medical Faculty, Department of Gynecology and Obstetrics, Essen
| | - Oliver Hoffmann
- Univ Duisburg-Essen, Medical Faculty, Department of Gynecology and Obstetrics, Essen
| | - Alexander R Stahl
- Univ Duisburg-Essen, Medical Faculty, Department of Nuclear Medicine, Essen
| | - Rainer Kimmig
- Univ Duisburg-Essen, Medical Faculty, Department of Gynecology and Obstetrics, Essen
| | - Friedrich Otterbach
- Univ Duisburg-Essen, Medical Faculty, Institute of Pathology and Neuropathology, Essen, Germany
| | - Susanne Ladd
- Univ Duisburg-Essen, Medical Faculty, Department of Diagnostic and Interventional Radiology and Neuroradiology, Essen
| | - Angela Koeninger
- Univ Duisburg-Essen, Medical Faculty, Department of Gynecology and Obstetrics, Essen
| | - Michael Forsting
- Univ Duisburg-Essen, Medical Faculty, Department of Diagnostic and Interventional Radiology and Neuroradiology, Essen
| | - Andreas Bockisch
- Univ Duisburg-Essen, Medical Faculty, Department of Nuclear Medicine, Essen
| | - Gerald Antoch
- Univ Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, Dusseldorf
| | - Till A Heusner
- Univ Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, Dusseldorf
- Univ Duisburg-Essen, Medical Faculty, Department of Diagnostic and Interventional Radiology and Neuroradiology, Essen
| |
Collapse
|
48
|
Kang H, Lee HY, Lee KS, Kim JH. Imaging-based tumor treatment response evaluation: review of conventional, new, and emerging concepts. Korean J Radiol 2012; 13:371-90. [PMID: 22778559 PMCID: PMC3384819 DOI: 10.3348/kjr.2012.13.4.371] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/14/2012] [Indexed: 01/07/2023] Open
Abstract
Tumor response may be assessed readily by the use of Response Evaluation Criteria in Solid Tumor version 1.1. However, the criteria mainly depend on tumor size changes. These criteria do not reflect other morphologic (tumor necrosis, hemorrhage, and cavitation), functional, or metabolic changes that may occur with targeted chemotherapy or even with conventional chemotherapy. The state-of-the-art multidetector CT is still playing an important role, by showing high-quality, high-resolution images that are appropriate enough to measure tumor size and its changes. Additional imaging biomarker devices such as dual energy CT, positron emission tomography, MRI including diffusion-weighted MRI shall be more frequently used for tumor response evaluation, because they provide detailed anatomic, and functional or metabolic change information during tumor treatment, particularly during targeted chemotherapy. This review elucidates morphologic and functional or metabolic approaches, and new concepts in the evaluation of tumor response in the era of personalized medicine (targeted chemotherapy).
Collapse
Affiliation(s)
- Hee Kang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | | | | | | |
Collapse
|
49
|
Cochet A, Generali D, Fox SB, Ferrozzi F, Hicks RJ. Positron emission tomography and neoadjuvant therapy of breast cancer. J Natl Cancer Inst Monogr 2012; 2011:111-5. [PMID: 22043055 DOI: 10.1093/jncimonographs/lgr035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The increasing use of neoadjuvant therapy for breast cancer has led to the development of early surrogate markers of response. Positron emission tomography (PET) allows noninvasive study of fundamental biologic processes in the tumor; furthermore, PET provides various markers to assess tumor response early in the course of therapy. Numerous studies have shown that changes in tumor glucose metabolism during therapy are significantly correlated with final response and patient outcome. Moreover, new PET tracers that are currently being developed or under evaluation, providing specific information on tumor characteristics or receptor expression, will assist the development of new targeted anticancer agents.
Collapse
Affiliation(s)
- Alexandre Cochet
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria 3002, Australia.
| | | | | | | | | |
Collapse
|
50
|
Design considerations for using PET as a response measure in single site and multicenter clinical trials. Acad Radiol 2012; 19:184-90. [PMID: 22104290 DOI: 10.1016/j.acra.2011.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Revised: 09/14/2011] [Accepted: 10/04/2011] [Indexed: 01/08/2023]
Abstract
RATIONALE AND OBJECTIVES Positron emission tomography (PET) is used to evaluate response to therapy with increasing interest in having PET provide endpoints for clinical trials. Here we demonstrate impacts of PET measurement error and choice of quantification method on clinical trial design. MATERIALS AND METHODS Sample size was calculated for two-arm randomized trials with percent change in (18)F-fluorodeoxyglucose (FDG) PET uptake as an efficacy endpoint. Two methods of uptake quantification were considered: standardized uptake values (SUVs) and kinetic measures from dynamic imaging. Calculations assumed a 20 percentage point difference in treatment groups' average percent change, and yielded 80% power at α = 0.05. The range of precision (10%-40%) in PET uptake measures was based on review of the literature. The range of SUV sensitivities (50%-100%) relative to kinetic analyses was based on a study of 75 locally advanced breast cancer patients. RESULTS Sample sizes increased from 8 to 126 as PET precision worsened from 10% to 40% at full measurement sensitivity to true change. In a subgroup with low initial FDG uptake, a sample size of 126 was required under 20% standard deviation using clinical SUVs. More sophisticated imaging quantification could reduce this sample size to 32. CONCLUSIONS The dependence of sample size on measurement precision and the sensitivity of imaging measures to true change should be considered in single site and multicenter PET trials to avoid underpowered studies with inconclusive results. Sophisticated PET imaging methods that are more sensitive to changes in uptake may be advantageous in early studies with limited patient numbers.
Collapse
|