1
|
Mariano L, Nicosia L, Latronico A, Bozzini AC, Dominelli V, Pupo D, Pesapane F, Pizzamiglio M, Cassano E. The role and potential of digital breast tomosynthesis in neoadjuvant systemic therapy evaluation for optimising breast cancer management: a pictorial essay. Br J Radiol 2025; 98:485-495. [PMID: 39724185 PMCID: PMC11919077 DOI: 10.1093/bjr/tqae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/27/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
Neoadjuvant therapy (NT) has become the gold standard for treating locally advanced breast cancer (BC). The assessment of pathological response (pR) post-NT plays a crucial role in predicting long-term survival, with contrast-enhanced MRI currently recognised as the preferred imaging modality for its evaluation. Traditional imaging techniques, such as digital mammography (DM) and ultrasonography (US), encounter difficulties in post-NT assessments due to breast density, lesion changes, fibrosis, and molecular patterns. Digital breast tomosynthesis (DBT) offers solutions to prevalent challenges in DM, such as tissue overlap, and facilitates a comprehensive assessment of lesion morphology, dimensions, and margins. Studies suggest that DBT correlates more accurately with pathology than DM and US, showcasing its potential advantages. This pictorial essay demonstrates the potential of DBT as a complementary tool to DM for assessing pR after NT, including instances of true- and false-positive assessments correlated with histopathological findings. In conclusion, DBT emerges as a valuable adjunct to DM, effectively addressing its limitations in post-NT assessment. The technology's potential to diminish tissue overlap, improve discrimination, and provide multi-dimensional perspectives demonstrates promising results, indicating its utility in scenarios where MRI is contraindicated or inaccessible.
Collapse
Affiliation(s)
- Luciano Mariano
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Luca Nicosia
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Antuono Latronico
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Anna Carla Bozzini
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Valeria Dominelli
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Davide Pupo
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Filippo Pesapane
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Maria Pizzamiglio
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| | - Enrico Cassano
- Division of Breast Radiology, Department of Medical Imaging and Radiation Sciences, IEO European Institute of Oncology, IRCCS, 20141, Via Ripamonti 435, Milano, Italy
| |
Collapse
|
2
|
Catanuto G, Gentile D, Martorana F, Tomatis M, Ponti A, Marotti L, Aristei C, Cardoso MJ, Cheung KL, Curigliano G, De Vries J, Karakatsanis A, Santini D, Sardanelli F, Van Dam P, Rubio IT. Clinico-pathological features predicting indication to mastectomy in breast cancer patients achieving complete response after neoadjuvant therapy: A retrospective analysis of the EUSOMA database. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109643. [PMID: 40009908 DOI: 10.1016/j.ejso.2025.109643] [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: 12/02/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
AIMS We investigated factors related to the type of surgery, i.e. mastectomy versus breast conserving surgery (BCS), in breast cancer (BC) patients with complete pathologic response in the breast (ypT0) after neoadjuvant therapy (NAT). METHODS A retrospective analysis from the EUSOMA database was performed using data from 55 certified centers across 14 European countries, including ypT0 BC patients (i.e., neither invasive nor in situ residuals), treated between 2017 and 2022. Variables analyzed included year of surgery, age, number and distribution of tumor focality, extent, clinical and pathological stages, and biologic subtype. Logistic regression was used to identify predictors of surgical choice. The Kaplan-Meier method was used for comparison of local recurrence-free survival (LRFS) between surgical groups. RESULTS Of 1416 BC patients included, 67.5 % underwent BCS and 32.5 % mastectomy. At multivariable analysis, factors increasing the likelihood of mastectomy included: more recent year of surgery [odds ratio (OR) 2.61, 95 % confidence interval (95%CI): 1.51-4.51,p = 0.001], younger age (OR: 0.96, 95%CI: 0.95-0.97,p < 0.001), multifocality (OR: 2.20, 95%CI: 1.61-3.00,p < 0.001) and multicentricity (OR: 12.66, 95%CI: 6.82-23.49,p < 0.001), advanced clinical tumor stage (OR: 14.54, 95%CI: 5.80-36.47,p < 0.001), and baseline axillary nodal involvement (OR: 1.56, 95%CI: 1.12-2.17,p = 0.009). Comparison between groups did not show a significant difference in LRFS (p = 0.389). CONCLUSION Many BC patients undergo mastectomy despite achieving complete response of primary tumor after NAT. Patients-related and tumor-related features, as well as having surgery in more recent years, seems to influence this choice. Our findings suggest the need for an optimized decision-making to spare unnecessary mastectomies.
Collapse
Affiliation(s)
| | - Damiano Gentile
- Breast Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federica Martorana
- Humanitas-Istituto Clinico Catanese Misterbianco, Catania, Italy; Department of Clinical and Experimental Medicine, University of Catania, Italy.
| | - Mariano Tomatis
- European Society of Breast Cancer Specialists (EUSOMA), Florence, Italy
| | - Antonio Ponti
- European Society of Breast Cancer Specialists (EUSOMA), Florence, Italy; CPO Piemonte, Turin, Italy
| | - Lorenza Marotti
- European Society of Breast Cancer Specialists (EUSOMA), Florence, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia, Italy; Perugia General Hospital Sant'Andrea delle Fratte Perugia, Italy
| | - Maria Joao Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Portugal; Lisbon University Faculty of Medicine, Lisbon, Portugal
| | - Kwok Leung Cheung
- Nottingham Breast Cancer Research Centre, University of Nottingham, United Kingdom; School of Medicine, University of Nottingham, United Kingdom
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Italy
| | | | - Andreas Karakatsanis
- Department for Surgical Sciences, Uppsala University, Uppsala, Sweden; Section for Breast Surgery, Department of Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Donatella Santini
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | | | - Peter Van Dam
- Multidisciplinary Oncologic Center, Antwerp University Hospital, Edegem, Belgium
| | - Isabel T Rubio
- Breast Surgical Oncology, Clinica Universidad de Navarra, Cancer Center Universidad de Navarra, Madrid, Spain
| |
Collapse
|
3
|
Chia JLL, He GS, Ngiam KY, Hartman M, Ng QX, Goh SSN. Harnessing Artificial Intelligence to Enhance Global Breast Cancer Care: A Scoping Review of Applications, Outcomes, and Challenges. Cancers (Basel) 2025; 17:197. [PMID: 39857979 PMCID: PMC11764353 DOI: 10.3390/cancers17020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND In recent years, Artificial Intelligence (AI) has shown transformative potential in advancing breast cancer care globally. This scoping review seeks to provide a comprehensive overview of AI applications in breast cancer care, examining how they could reshape diagnosis, treatment, and management on a worldwide scale and discussing both the benefits and challenges associated with their adoption. METHODS In accordance with PRISMA-ScR and ensuing guidelines on scoping reviews, PubMed, Web of Science, Cochrane Library, and Embase were systematically searched from inception to end of May 2024. Keywords included "Artificial Intelligence" and "Breast Cancer". Original studies were included based on their focus on AI applications in breast cancer care and narrative synthesis was employed for data extraction and interpretation, with the findings organized into coherent themes. RESULTS Finally, 84 articles were included. The majority were conducted in developed countries (n = 54). The majority of publications were in the last 10 years (n = 83). The six main themes for AI applications were AI for breast cancer screening (n = 32), AI for image detection of nodal status (n = 7), AI-assisted histopathology (n = 8), AI in assessing post-neoadjuvant chemotherapy (NACT) response (n = 23), AI in breast cancer margin assessment (n = 5), and AI as a clinical decision support tool (n = 9). AI has been used as clinical decision support tools to augment treatment decisions for breast cancer and in multidisciplinary tumor board settings. Overall, AI applications demonstrated improved accuracy and efficiency; however, most articles did not report patient-centric clinical outcomes. CONCLUSIONS AI applications in breast cancer care show promise in enhancing diagnostic accuracy and treatment planning. However, persistent challenges in AI adoption, such as data quality, algorithm transparency, and resource disparities, must be addressed to advance the field.
Collapse
Affiliation(s)
- Jolene Li Ling Chia
- NUS Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr. S117597, Singapore 119077, Singapore (G.S.H.)
| | - George Shiyao He
- NUS Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr. S117597, Singapore 119077, Singapore (G.S.H.)
| | - Kee Yuen Ngiam
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| | - Mikael Hartman
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| | - Qin Xiang Ng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore 169857, Singapore
| | - Serene Si Ning Goh
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| |
Collapse
|
4
|
AlBuainain RY, Bunajem FY, Abdulla HA. Assessment of Tumor Response to Neoadjuvant Chemotherapy in Breast Cancer Using MRI and 18F-FDG PET/CT. Eur J Breast Health 2025; 21:46-51. [PMID: 39744907 PMCID: PMC11706120 DOI: 10.4274/ejbh.galenos.2024.2024-8-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025]
Abstract
Objective Neoadjuvant chemotherapy (NACT) has been the primary treatment method for patients with local advanced breast cancer. A pathological complete response (pCR) to therapy correlates with better overall disease prognosis. Magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT) have been widely used to monitor the response to NACT in breast cancer. The aim of this study was to assess tumor response to NACT by MRI and PET/CT, to determine which imaging modality is more accurate in detecting tumor response post NACT in breast cancer. Materials and Methods A retrospective review of our database revealed 34 women with breast cancer that had MRI and PET/CT performed prior to and after NACT, followed by definitive surgery. For response assessment, we calculated the difference in maximum diameter of the tumor in MRI and difference in standard uptake values in PET/CT. The correspondence rate between the imaging modalities and pCR were calculated. For the prediction of pCR, the sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and accuracy where analyzed. Results The assessment of tumor response to NACT showed 11 cases with pCR (32%), 15 pathological partial response (44%) and eight pathological no response (24%). The correspondence rate between MRI and pathological response was 50% (17/34), compared to 65% (22/34) for PET/CT. For prediction of pCR, MRI showed higher specificity compared to PET/CT (78.2% vs. 73.9%, p = 0.024), while the accuracy of PET/CT was significantly higher (79.4% vs. 70.5%, p = 0.004). PET/CT also had a higher NPV compared to MRI (94.4% vs. 78.2%, p = 0.002). There were no differences in terms of sensitivity and PPV between MRI and PET/CT. Conclusion Compared to MRI, PET/CT was more likely to correlate with the pathological response after NACT. For the prediction of pCR, PET/CT proved to be a more accurate imaging modality to monitor response after NACT than MRI.
Collapse
Affiliation(s)
- Reem Yusuf AlBuainain
- Department of Surgery, Salmaniya Medical Complex, Government Hospitals, Manama, Bahrain
| | - Fatema Yusuf Bunajem
- Department of Radiology, Salmaniya Medical Complex, Government Hospitals, Manama, Bahrain
| | - Hussain Adnan Abdulla
- Department of Surgery, Salmaniya Medical Complex, Government Hospitals, Manama, Bahrain
| |
Collapse
|
5
|
Álvarez-Benito M. Imaging evaluation of neoadjuvant breast cancer treatment: where do we stand? Eur Radiol 2024; 34:6271-6272. [PMID: 38753195 PMCID: PMC11399156 DOI: 10.1007/s00330-024-10799-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/10/2024] [Accepted: 04/25/2024] [Indexed: 09/15/2024]
Affiliation(s)
- Marina Álvarez-Benito
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC) Córdoba, Córdoba, Spain.
- Breast Cancer Unit, Department of Diagnostic Radiology, Reina Sofía University Hospital, Córdoba, Spain.
- University of Córdoba, Córdoba, Spain.
| |
Collapse
|
6
|
Vidali S, Irmici G, Depretto C, Bellini C, Pugliese F, Incardona LA, Di Naro F, De Benedetto D, Di Filippo G, Ferraro F, De Berardinis C, Miele V, Scaperrotta G, Nori Cucchiari J. Performance of Contrast-Enhanced Mammography (CEM) for Monitoring Neoadjuvant Chemotherapy Response among Different Breast Cancer Subtypes. Cancers (Basel) 2024; 16:2694. [PMID: 39123423 PMCID: PMC11311316 DOI: 10.3390/cancers16152694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Neoadjuvant chemotherapy (NAT) plays a crucial role in breast cancer (BC) treatment, both in advanced BC and in early-stage BC, with different rates of pathological complete response (pCR) among the different BC molecular subtypes. Imaging monitoring is mandatory to evaluate the NAT efficacy. This study evaluates the diagnostic performance of Contrast-Enhanced Mammography (CEM) in BC patients undergoing NAT. This retrospective two-center study included 174 patients. The breast lesions were classified based on the molecular subtypes in hormone receptor (HR+)/HER2-, HER2+, and triple-negative breast cancer (TNBC). The histopathological analysis performed following surgery was used as a reference standard for the pCR. Sensitivity, specificity, PPV, and NPV were measured overall and for the different subtypes. We enrolled 174 patients, 79/174 (46%) HR+/HER2-, 59/174 (33.9%) HER2+, and 35/174 (20.1%) TNBC; the pCR was found in 64/174 (36.8%), of which 57.1% were TNBCs. In the total population, the CEM sensitivity and specificity were 66.2% and 75.2%, with a PPV of 61.4% and an NPV of 78.8%. The highest specificity (80.9%) and NPV (91.7%) were found in HR+/HER2-, while the highest sensitivity (70%) and PPV appeared (73.7%) in TNBC. The results indicate that CEM is a valid tool to assess the pCR, with different performances among the subtypes of BC.
Collapse
Affiliation(s)
- Sofia Vidali
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Giovanni Irmici
- Breast Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Catherine Depretto
- Breast Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Chiara Bellini
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| | - Francesca Pugliese
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| | - Ludovica Anna Incardona
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| | - Federica Di Naro
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| | - Diego De Benedetto
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| | - Giacomo Di Filippo
- UOC Endocrinochirurgia, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| | - Fabiola Ferraro
- Department of Biomedicine Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90133 Palermo, Italy
| | - Claudia De Berardinis
- Breast Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy
| | | | - Jacopo Nori Cucchiari
- Breast Imaging Unit, Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (C.B.); (L.A.I.); (D.D.B.)
| |
Collapse
|
7
|
Lo Gullo R, Marcus E, Huayanay J, Eskreis-Winkler S, Thakur S, Teuwen J, Pinker K. Artificial Intelligence-Enhanced Breast MRI: Applications in Breast Cancer Primary Treatment Response Assessment and Prediction. Invest Radiol 2024; 59:230-242. [PMID: 37493391 PMCID: PMC10818006 DOI: 10.1097/rli.0000000000001010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
ABSTRACT Primary systemic therapy (PST) is the treatment of choice in patients with locally advanced breast cancer and is nowadays also often used in patients with early-stage breast cancer. Although imaging remains pivotal to assess response to PST accurately, the use of imaging to predict response to PST has the potential to not only better prognostication but also allow the de-escalation or omission of potentially toxic treatment with undesirable adverse effects, the accelerated implementation of new targeted therapies, and the mitigation of surgical delays in selected patients. In response to the limited ability of radiologists to predict response to PST via qualitative, subjective assessments of tumors on magnetic resonance imaging (MRI), artificial intelligence-enhanced MRI with classical machine learning, and in more recent times, deep learning, have been used with promising results to predict response, both before the start of PST and in the early stages of treatment. This review provides an overview of the current applications of artificial intelligence to MRI in assessing and predicting response to PST, and discusses the challenges and limitations of their clinical implementation.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Eric Marcus
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jorge Huayanay
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Radiology, National Institute of Neoplastic Diseases, Lima, Peru
| | - Sarah Eskreis-Winkler
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Sunitha Thakur
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonas Teuwen
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| |
Collapse
|
8
|
Pfob A, Cai L, Schneeweiss A, Rauch G, Thomas B, Schaefgen B, Kuemmel S, Reimer T, Hahn M, Thill M, Blohmer JU, Hackmann J, Malter W, Bekes I, Friedrichs K, Wojcinski S, Joos S, Paepke S, Degenhardt T, Rom J, Rody A, van Mackelenbergh M, Banys-Paluchowski M, Große R, Reinisch M, Karsten MM, Sidey-Gibbons C, Wallwiener M, Golatta M, Heil J. Minimally Invasive Breast Biopsy After Neoadjuvant Systemic Treatment to Identify Breast Cancer Patients with Residual Disease for Extended Neoadjuvant Treatment: A New Concept. Ann Surg Oncol 2024; 31:957-965. [PMID: 37947974 PMCID: PMC10761434 DOI: 10.1245/s10434-023-14551-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Breast cancer patients with residual disease after neoadjuvant systemic treatment (NAST) have a worse prognosis compared with those achieving a pathologic complete response (pCR). Earlier identification of these patients might allow timely, extended neoadjuvant treatment strategies. We explored the feasibility of a vacuum-assisted biopsy (VAB) after NAST to identify patients with residual disease (ypT+ or ypN+) prior to surgery. METHODS We used data from a multicenter trial, collected at 21 study sites (NCT02948764). The trial included women with cT1-3, cN0/+ breast cancer undergoing routine post-neoadjuvant imaging (ultrasound, MRI, mammography) and VAB prior to surgery. We compared the findings of VAB and routine imaging with the histopathologic evaluation of the surgical specimen. RESULTS Of 398 patients, 34 patients with missing ypN status and 127 patients with luminal tumors were excluded. Among the remaining 237 patients, tumor cells in the VAB indicated a surgical non-pCR in all patients (73/73, positive predictive value [PPV] 100%), whereas PPV of routine imaging after NAST was 56.0% (75/134). Sensitivity of the VAB was 72.3% (73/101), and 74.3% for sensitivity of imaging (75/101). CONCLUSION Residual cancer found in a VAB specimen after NAST always corresponds to non-pCR. Residual cancer assumed on routine imaging after NAST corresponds to actual residual cancer in about half of patients. Response assessment by VAB is not safe for the exclusion of residual cancer. Response assessment by biopsies after NAST may allow studying the new concept of extended neoadjuvant treatment for patients with residual disease in future trials.
Collapse
Affiliation(s)
- André Pfob
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany.
| | - Lie Cai
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Thomas
- Coordination Centre for Clinical Trials (KKS), University Heidelberg, Heidelberg, Germany
| | - Benedikt Schaefgen
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Sherko Kuemmel
- Breast Unit, Kliniken Essen-Mitte, Essen, Germany
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Toralf Reimer
- Department of Gynecology/Breast Unit, University Hospital Rostock, Rostock, Germany
| | - Markus Hahn
- Department of Gynecology/Breast Unit, University Hospital Tuebingen, Tübingen, Germany
| | - Marc Thill
- Department of Gynecology and Gynecological Oncology/Breast Unit, Agaplesion Markus Hospital Frankfurt, Frankfurt, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - John Hackmann
- Department of Gynecology/Breast Unit, Marienhospital, Witten, Germany
| | - Wolfram Malter
- Department of Gynecology and Obstetrics, Medical Faculty, Breast Cancer Center, University of Cologne, Cologne, Germany
| | - Inga Bekes
- Department of Gynecology/Breast Unit, University Hospital Ulm, Ulm, Germany
| | - Kay Friedrichs
- Department of Gynecology/Breast Unit, Jerusalem Hospital Hamburg, Hamburg, Germany
| | - Sebastian Wojcinski
- Department of Gynecology and Obstetrics, Breast Cancer Center, Klinikum Bielefeld Mitte GmbH, Bielefeld, Germany
| | - Sylvie Joos
- Radiologische Allianz Hamburg, Hamburg, Germany
| | - Stefan Paepke
- Frauenklinik, Interdisziplinäres Brustzentrum des Klinikums rechts der Isar der Technischen Universität München, Munich, Germany
| | - Tom Degenhardt
- Department of Gynecology/Breast Unit, University Hospital Munich, Munich, Germany
| | - Joachim Rom
- Department of Gynecology/Breast Unit, Klinikum Frankfurt-Höchst, Frankfurt, Germany
| | - Achim Rody
- Department of Gynecology/Breast Unit, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | | | - Regina Große
- Department of Gynecology/Breast Unit, University Hospital Halle, Halle, Germany
| | | | - Maria Margarete Karsten
- Department of Gynecology with Breast Center, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Chris Sidey-Gibbons
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Markus Wallwiener
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Michael Golatta
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Breast Unit, Klinikum Sankt Elisabeth, Heidelberg, Germany
| | - Joerg Heil
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Breast Unit, Klinikum Sankt Elisabeth, Heidelberg, Germany
| |
Collapse
|
9
|
Guo X, Zhang J, Gong X, Wang J, Dai H, Jiao D, Ling R, Zhao Y, Yang H, Liu Y, Liu K, Zhang J, Mao D, He J, Yu Z, Liu Y, Fu P, Wang J, Jiang H, Zhao Z, Tian X, Cao Z, Wu K, Song A, Jin F, Fan Z, Liu Z. Axillary lymph node dissection in triple-negative or HER2-positive breast cancer patients with clinical N2 achieving pathological complete response after neoadjuvant therapy: Is it necessary? Breast 2024; 73:103671. [PMID: 38277714 PMCID: PMC10832498 DOI: 10.1016/j.breast.2024.103671] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/26/2023] [Accepted: 01/05/2024] [Indexed: 01/28/2024] Open
Abstract
AIM This study aims to identify suitable candidates for axillary sentinel lymph node biopsy (SLNB) or targeted axillary dissection (TAD) among clinical N2 (cN2) triple-negative (TN) or HER2 positive (HER2+)breast cancer patients following neoadjuvant therapy(NAT). BACKGROUND Despite the substantial axillary burden in cN2 breast cancer patients, high pathological response rates can be achieved with NAT in TN or HER2+ subtypes, thus enabling potential downstaging of axillary surgery. METHODS A retrospective analysis was conducted on data from the CSBrS-012 study, screening 709 patients with initial cN2, either HER2+ or TN subtype, from January 1, 2010 to December 31, 2020. The correlation between axillary pathologic complete response (pCR) (yPN0) and breast pCR was examined. RESULTS Among the 177 cN2 patients who achieved breast pCR through NAT, 138 (78.0 %) also achieved axillary pCR. However, in the 532 initial clinical N2 patients who did not achieve breast pCR, residual axillary lymph node metastasis persisted in 77.4 % (412/532) of cases. The relative risk of residual axillary lymph node metastasis in patients who did not achieve breast pCR was 12.4 (8.1-19.1), compared to those who did achieve breast pCR, P < 0.001. CONCLUSION For cN2 TN or HER2+ breast cancer patients who achieve breast pCR following NAT, consideration could be given to downstaging and performing an axillary SLNB or TAD.
Collapse
Affiliation(s)
- Xuhui Guo
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Jiao Zhang
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Xilong Gong
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Jia Wang
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Hao Dai
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Dechuang Jiao
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China
| | - Rui Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi Zhao
- Surgical Oncology Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110022, China
| | - Hongjian Yang
- Department of Breast Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang Province, 310022, China
| | - Yunjiang Liu
- Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 052360, China
| | - Ke Liu
- Fourth Department of Breast Surgery, Jilin Cancer Hospital. Changchun, Jilin Province, 130012, China
| | - Jianguo Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Dahua Mao
- Department of Breast Surgery, Affiliated Wudang Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550009, China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250033, China
| | - Yinhua Liu
- Breast Disease Center, Peking University First Hospital, Beijing, 100034, China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Jiandong Wang
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100852, China
| | - Hongchuan Jiang
- Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zuowei Zhao
- Department of Breast Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China
| | - Xingsong Tian
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, 250021, China
| | - Zhongwei Cao
- Department of Thyroid, Breast, Hernia Surgery, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia Autonomous Region, 010017, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200433, China
| | - Ailin Song
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu Province, 730000, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110002, China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, 130021, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Dongming Road, Zhengzhou, Henan Province, 450008, China.
| |
Collapse
|
10
|
Sui L, Yan Y, Jiang T, Ou D, Chen C, Lai M, Ni C, Zhu X, Wang L, Yang C, Li W, Yao J, Xu D. Ultrasound and clinicopathological characteristics-based model for prediction of pathologic response to neoadjuvant chemotherapy in HER2-positive breast cancer: a case-control study. Breast Cancer Res Treat 2023; 202:45-55. [PMID: 37639063 PMCID: PMC10504141 DOI: 10.1007/s10549-023-07057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND The objective of this study was to develop a model combining ultrasound (US) and clinicopathological characteristics to predict the pathologic response to neoadjuvant chemotherapy (NACT) in human epidermal growth factor receptor 2 (HER2)-positive breast cancer. MATERIALS AND METHODS This is a retrospective study that included 248 patients with HER2-positive breast cancer who underwent NACT from March 2018 to March 2022. US and clinicopathological characteristics were collected from all patients in this study, and characteristics obtained using univariate analysis at p < 0.1 were subjected to multivariate analysis and then the conventional US and clinicopathological characteristics independently associated with pathologic complete response (pCR) from the analysis were used to develop US models, clinicopathological models, and their combined models by the area under the receiver operating characteristic (ROC) curve (AUC), accuracy, sensitivity, and specificity to assess their predictive efficacy. RESULTS The combined model had an AUC of 0.808, a sensitivity of 88.72%, a specificity of 60.87%, and an accuracy of 75.81% in predicting pCR of HER2-positive breast cancer after NACT, which was significantly better than the clinicopathological model (AUC = 0.656) and the US model (AUC = 0.769). In addition, six characteristics were screened as independent predictors, namely the Clinical T stage, Clinical N stage, PR status, posterior acoustic, margin, and calcification. CONCLUSION The conventional US combined with clinicopathological characteristics to construct a combined model has a good diagnostic effect in predicting pCR in HER2-positive breast cancer and is expected to be a useful tool to assist clinicians in effectively determining the efficacy of NACT in HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Lin Sui
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Yuqi Yan
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Tian Jiang
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Di Ou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Chen Chen
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
- Graduate School, Wannan Medical College, Wuhu, China
| | - Min Lai
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Ni
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Xi Zhu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Liping Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Chen Yang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Wei Li
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Jincao Yao
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, China
| | - Dong Xu
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| |
Collapse
|
11
|
Chen Y, Qi Y, Wang K. Neoadjuvant chemotherapy for breast cancer: an evaluation of its efficacy and research progress. Front Oncol 2023; 13:1169010. [PMID: 37854685 PMCID: PMC10579937 DOI: 10.3389/fonc.2023.1169010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) for breast cancer is widely used in the clinical setting to improve the chance of surgery, breast conservation and quality of life for patients with advanced breast cancer. A more accurate efficacy evaluation system is important for the decision of surgery timing and chemotherapy regimen implementation. However, current methods, encompassing imaging techniques such as ultrasound and MRI, along with non-imaging approaches like pathological evaluations, often fall short in accurately depicting the therapeutic effects of NAC. Imaging techniques are subjective and only reflect macroscopic morphological changes, while pathological evaluation is the gold standard for efficacy assessment but has the disadvantage of delayed results. In an effort to identify assessment methods that align more closely with real-world clinical demands, this paper provides an in-depth exploration of the principles and clinical applications of various assessment approaches in the neoadjuvant chemotherapy process.
Collapse
Affiliation(s)
- Yushi Chen
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan, China
| | - Yu Qi
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, Basic Medical School, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Abstract
Breast cancer (BC) remains one of the leading causes of death among women. The management and outcome in BC are strongly influenced by a multidisciplinary approach, which includes available treatment options and different imaging modalities for accurate response assessment. Among breast imaging modalities, MR imaging is the modality of choice in evaluating response to neoadjuvant therapy, whereas F-18 Fluorodeoxyglucose positron emission tomography, conventional computed tomography (CT), and bone scan play a vital role in assessing response to therapy in metastatic BC. There is an unmet need for a standardized patient-centric approach to use different imaging methods for response assessment.
Collapse
Affiliation(s)
- Saima Muzahir
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, 1364 Clifton Road, Atlanta GA 30322, USA; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, Room E152, 1364 Clifton Road, Atlanta, GA 30322, USA.
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, USA; Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - David M Schuster
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, Room E152, 1364 Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Caracciolo M, Castello A, Urso L, Borgia F, Marzola MC, Uccelli L, Cittanti C, Bartolomei M, Castellani M, Lopci E. Comparison of MRI vs. [ 18F]FDG PET/CT for Treatment Response Evaluation of Primary Breast Cancer after Neoadjuvant Chemotherapy: Literature Review and Future Perspectives. J Clin Med 2023; 12:5355. [PMID: 37629397 PMCID: PMC10455346 DOI: 10.3390/jcm12165355] [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: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
The purpose of this systematic review was to investigate the diagnostic accuracy of [18F]FDG PET/CT and breast MRI for primary breast cancer (BC) response assessment after neoadjuvant chemotherapy (NAC) and to evaluate future perspectives in this setting. We performed a critical review using three bibliographic databases (i.e., PubMed, Scopus, and Web of Science) for articles published up to the 6 June 2023, starting from 2012. The Quality Assessment of Diagnosis Accuracy Study (QUADAS-2) tool was adopted to evaluate the risk of bias. A total of 76 studies were identified and screened, while 14 articles were included in our systematic review after a full-text assessment. The total number of patients included was 842. Eight out of fourteen studies (57.1%) were prospective, while all except one study were conducted in a single center. In the majority of the included studies (71.4%), 3.0 Tesla (T) MRI scans were adopted. Three out of fourteen studies (21.4%) used both 1.5 and 3.0 T MRI and only two used 1.5 T. [18F]FDG was the radiotracer used in every study included. All patients accepted surgical treatment after NAC and each study used pathological complete response (pCR) as the reference standard. Some of the studies have demonstrated the superiority of [18F]FDG PET/CT, while others proved that MRI was superior to PET/CT. Recent studies indicate that PET/CT has a better specificity, while MRI has a superior sensitivity for assessing pCR in BC patients after NAC. The complementary value of the combined use of these modalities represents probably the most important tool to improve diagnostic performance in this setting. Overall, larger prospective studies, possibly randomized, are needed, hopefully evaluating PET/MR and allowing for new tools, such as radiomic parameters, to find a proper place in the setting of BC patients undergoing NAC.
Collapse
Affiliation(s)
- Matteo Caracciolo
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Luca Urso
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy
| | - Francesca Borgia
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, 44124 Ferrara, Italy
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Maria Cristina Marzola
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy
| | - Licia Uccelli
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, 44124 Ferrara, Italy
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Corrado Cittanti
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, 44124 Ferrara, Italy
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, 44124 Ferrara, Italy
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS—Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| |
Collapse
|
14
|
Lopez BP, Kappadath SC. Monte Carlo-derived 99m Tc uptake quantification with commercial planar MBI: Tumor and breast activity concentrations. Med Phys 2023; 50:4388-4398. [PMID: 36625713 PMCID: PMC10331527 DOI: 10.1002/mp.16213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/23/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Current molecular breast imaging (MBI) images are limited to qualitative evaluation, not absolute measurement, of 99m Tc uptake in benign and malignant breast tissues. PURPOSE This work assesses the accuracy of previously-published and newly-proposed tumor and normal breast tissue 99m Tc uptake MBI measurements using simulations of a commercial dual-headed planar MBI system under typical clinical and acquisition protocols. METHODS Quantification techniques were tested in over 4000 simulated acquisitions of spherical and ellipsoid tumors with clinically relevant uptake conditions using a validated Monte Carlo application of the GE Discovery NM750b system. The evaluated techniques consisted of four tumor total activity methodologies (two single-detector-based and two geometric-mean-based), two tumor MBI volume methodologies (diameter-based and ROI-based), and two normal tissue activity concentration methodologies (single-detector-based and geometric-mean-based). The most accurate of these techniques were then used to estimate tumor activity concentrations and tumor to normal tissue relative activity concentrations (RC). RESULTS Single-detector techniques for tumor total activity quantification achieved mean (standard deviation) relative errors of 0.2% (4.3%) and 1.6% (4.4%) when using the near and far detector images, respectively and were more accurate and precise than the measured 8.1% (5.8%) errors of a previously published geometric-mean technique. Using these activity estimates and the true tumor volumes resulted in tumor activity concentration and RC errors within 10% of simulated values. The precision of tumor activity concentration and RC when using only MBI measurements were largely driven by the errors in estimating tumor MBI volume using planar images (± 30% inter-quartile range). CONCLUSIONS Planar MBI images were shown to accurately and reliably be used to estimate tumor total activities and normal tissue activity concentrations in this simulation study. However, volumetric tumor uptake measurements (i.e., absolute and relative concentrations) are limited by inaccuracies in MBI volume estimation using two-dimensional images, highlighting the need for either tomographic MBI acquisitions or anatomical volume estimates for accurate three-dimensional tumor uptake estimates.
Collapse
Affiliation(s)
- Benjamin P Lopez
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - S Cheenu Kappadath
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
15
|
Tang WJ, Chen SY, Hu WK, Li XL, Zheng BJ, Wang ZS, Ding HJ, Chen LX, Zhang QQ, Yu XM, Sui Y, Wei XH, Guo Y. Abbreviated Versus Full-Protocol MRI for Breast Cancer Neoadjuvant Chemotherapy Response Assessment: Diagnostic Performance by General and Breast Radiologists. AJR Am J Roentgenol 2023; 220:817-825. [PMID: 36752371 DOI: 10.2214/ajr.22.28686] [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] [Indexed: 02/09/2023]
Abstract
BACKGROUND. Abbreviated protocols could allow wider adoption of MRI in patients undergoing breast cancer neoadjuvant chemotherapy (NAC). However, abbreviated MRI has been explored primarily in screening settings. OBJECTIVE. The purpose of this article was to compare diagnostic performance of abbreviated MRI and full-protocol MRI for evaluation of breast cancer NAC response, stratifying by radiologists' breast imaging expertise. METHODS. This retrospective study included 203 patients with breast cancer (mean age, 52.1 ± 11.2 [SD] years) from two hospitals who underwent MRI before NAC initiation and after NAC completion before surgical resection from March 2017 to April 2021. Abbreviated MRI was extracted from full-protocol MRI and included the axial T2-weighted sequence and precontrast and single early postcontrast T1-weighted sequences. Three general radiologists and three breast radiologists independently interpreted abbreviated and full-protocol MRI in separate sessions, identifying enhancing lesions to indicate residual tumor and measuring lesion size. The reference standard was presence and size of residual tumor on pathologic assessment of post-NAC surgical specimens. RESULTS. A total of 50 of 203 patients had pathologic complete response (pCR). Intraobserver and interobserver agreement for abbreviated and full-protocol MRI for general and breast radiologists ranged from substantial to nearly perfect (κ = 0.70-0.81). Abbreviated MRI compared with full-protocol MRI showed no significant difference for general radiologists in sensitivity (54.7% vs 57.3%, p > .99), specificity (92.8% vs 95.6%, p = .29), or accuracy (83.4% vs 86.2%, p = .30), nor for breast radiologists in sensitivity (60.0% vs 61.3%, p > .99), specificity (94.6% vs 97.4%, p = .22), or accuracy (86.0% vs 88.5%, p = .30). Sensitivity, specificity, and accuracy were not significantly different between protocols for any reader individually (p > .05). Mean difference in residual tumor size on MRI relative to pathology for abbreviated protocol ranged for general radiologists from -0.19 to 0.03 mm and for breast radiologists from -0.15 to -0.05 mm, and for full protocol ranged for general radiologists from 0.57 to 0.65 mm and for breast radiologists from 0.66 to 0.79 mm. CONCLUSION. Abbreviated compared with full-protocol MRI showed similar intraobserver and interobserver agreement and no significant difference in diagnostic performance. Full-protocol MRI but not abbreviated MRI slightly overestimated pathologic tumor sizes. CLINICAL IMPACT. Abbreviated protocols may facilitate use of MRI for post-NAC response assessment by general and breast radiologists.
Collapse
Affiliation(s)
- Wen-Jie Tang
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Si-Yi Chen
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Wen-Ke Hu
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Xue-Li Li
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Bing-Jie Zheng
- Department of Radiology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen-Sui Wang
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Han-Jun Ding
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Lei-Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Qiong-Qiong Zhang
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Xiao-Meng Yu
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Yi Sui
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Xin-Hua Wei
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| | - Yuan Guo
- Department of Radiology, Guangzhou First People's Hospital, No 1 Panfu Rd, Guangzhou, 510180, China
| |
Collapse
|
16
|
Wang X, Hua H, Han J, Zhong X, Liu J, Chen J. Evaluation of Multiparametric MRI Radiomics-Based Nomogram in Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer: A Two-Center study. Clin Breast Cancer 2023:S1526-8209(23)00134-9. [PMID: 37321954 DOI: 10.1016/j.clbc.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 06/17/2023]
Abstract
INTRODUCTION This study evaluated the performance of primary foci of breast cancer on multiparametric magnetic resonance imaging (MRI) contributing to establish and validate radiomics-based nomograms for predicting the different pathological outcome of breast cancer patients after neoadjuvant chemotherapy (NAC). MATERIALS AND METHODS Retrospectively collected 387 patients with locally advanced breast cancer, all treated with NAC and received breast dynamic contrast-enhanced MRI (DCE-MRI) before NAC. Radiomics signatures were extracted from region of interest (ROI) on multiparametric MRI to build rad score. Clinical-pathologic data and radiological features established the clinical model. The comprehensive model featured rad-score, predictive clinical-pathologic data and radiological features, which was ultimately displayed as a nomogram. Patients were grouped in 2 different ways in accordance with the Miller-Payne (MP) grading of surgical specimens. The first grouping method: 181 patients with pathological reaction grades Ⅳ∼Ⅴ were included in the significant remission group, while 206 patients with pathological reaction grades Ⅰ∼Ⅲ were included in the nonsignificant remission group. The second grouping method: 117 patients with pathological complete response (pCR) were assigned to the pCR group, and 270 patients who failed to meet pCR were assigned to in the non-pCR group. Two combined nomograms are created from 2 grouped data for predicting different pathological responses to NAC. The area under the curves (AUC) of the receiver operating characteristic curves (ROC) were used to evaluate the performance of each model. While decision curve analysis (DCA) and calibration curves were used for estimating the clinical application value of the nomogram. RESULTS Two combined nomograms embodying rad score and clinical-pathologic data outperformed, showing good calibrations for predicting response to NAC. The combined nomogram predicting pCR showed the best performance with the AUC values of 0.97, 0.90 and 0.86 in the training, testing, and external validation cohorts respectively. The AUC values of another combined nomogram predicting significant remission: 0.98, 0.88 0.80 in the training, testing and external validation cohorts. DCA showed the comprehensive model nomogram obtained the most clinical benefit. CONCLUSIONS The combined nomogram could preoperatively predict significant remission or even pCR to NAC in breast cancer based on multiparametric MRI and clinical-pathologic data.
Collapse
Affiliation(s)
- Xiaolin Wang
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Hua
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqi Han
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Zhong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Liu
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Chen
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
17
|
Pfob A, Heil J. Artificial intelligence to de-escalate loco-regional breast cancer treatment. Breast 2023; 68:201-204. [PMID: 36842193 PMCID: PMC9988657 DOI: 10.1016/j.breast.2023.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
In this review, we evaluate the potential and recent advancements in using artificial intelligence techniques to de-escalate loco-regional breast cancer therapy, with a special focus on surgical treatment after neoadjuvant systemic treatment (NAST). The increasing use and efficacy of NAST make the optimal loco-regional management of patients with pathologic complete response (pCR) a clinically relevant knowledge gap. It is hypothesized that patients with pCR do not benefit from therapeutic surgery because all tumor has already been eradicated by NAST. It is unclear, however, how residual cancer after NAST can be reliably excluded prior to surgery to identify patients eligible for omitting breast cancer surgery. Evidence from clinical trials evaluating the potential of imaging and minimally-invasive biopsies to exclude residual cancer suggests that there is a high risk of missing residual cancer. More recently, AI-based algorithms have shown promising results to reliably exclude residual cancer after NAST. This example illustrates the great potential of AI-based algorithms to further de-escalate and individualize loco-regional breast cancer treatment.
Collapse
Affiliation(s)
- André Pfob
- Department of Obstetrics & Gynecology, Heidelberg University Hospital, Germany; National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Joerg Heil
- Department of Obstetrics & Gynecology, Heidelberg University Hospital, Germany; Breast Centre Heidelberg, Klinik St. Elisabeth, Heidelberg, Germany
| |
Collapse
|
18
|
Han X, Li H, Dong SS, Zhou SY, Wang CH, Guo L, Yang J, Zhang GL. Application of triple evaluation method in predicting the efficacy of neoadjuvant therapy for breast cancer. World J Surg Oncol 2023; 21:116. [PMID: 36978164 PMCID: PMC10052864 DOI: 10.1186/s12957-023-02998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVE To analyze the factors related to the efficacy of neoadjuvant therapy for breast cancer and find appropriate evaluation methods for evaluating the efficacy of neoadjuvant therapy METHODS: A total of 143 patients with breast cancer treated by neoadjuvant chemotherapy at Baotou Cancer Hospital were retrospectively analyzed. The chemotherapy regimen was mainly paclitaxel combined with carboplatin for 1 week, docetaxel combined with carboplatin for 3 weeks, and was replaced with epirubicin combined with cyclophosphamide after evaluation of disease progression. All HER2-positive patients were treated with simultaneous targeted therapy, including trastuzumab single-target therapy and trastuzumab combined with pertuzumab double-target therapy. Combined with physical examination, color Doppler ultrasound, and magnetic resonance imaging (MRI), a systematic evaluation system was initially established-the "triple evaluation method." A baseline evaluation was conducted before treatment. The efficacy was evaluated by physical examination and color Doppler every cycle, and the efficacy was evaluated by physical examination, color Doppler, and MRI every two cycles. RESULTS The increase in ultrasonic blood flow after treatment could affect the efficacy of monitoring. The presence of two preoperative time-signal intensity curves is a therapeutically effective protective factor for inflow. The triple evaluation determined by physical examination, color Doppler ultrasound, and MRI in determining clinical efficacy is consistent with the effectiveness of the pathological gold standard. CONCLUSION The therapeutic effect of neoadjuvant therapy can be better evaluated by combining clinical physical examination, color ultrasound, and nuclear magnetic resonance evaluation. The three methods complement each other to avoid the insufficient evaluation of a single method, which is convenient for most prefecty-level hospitals. Additionally, this method is simple, feasible, and suitable for promotion.
Collapse
Affiliation(s)
- Xu Han
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Hui Li
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Sha-Sha Dong
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Shui-Ying Zhou
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Cai-Hong Wang
- Department of Operating Room, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - Lin Guo
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Jie Yang
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Gang-Ling Zhang
- Department of Breast Surgery, Baotou Cancer Hospital, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China.
| |
Collapse
|
19
|
Portnow LH, Kochkodan-Self JM, Maduram A, Barrios M, Onken AM, Hong X, Mittendorf EA, Giess CS, Chikarmane SA. Multimodality Imaging Review of HER2-positive Breast Cancer and Response to Neoadjuvant Chemotherapy. Radiographics 2023; 43:e220103. [PMID: 36633970 DOI: 10.1148/rg.220103] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2/neu or ErbB2)-positive breast cancers comprise 15%-20% of all breast cancers. The most common manifestation of HER2-positive breast cancer at mammography or US is an irregular mass with spiculated margins that often contains calcifications; at MRI, HER2-positive breast cancer may appear as a mass or as nonmass enhancement. HER2-positive breast cancers are often of intermediate to high nuclear grade at histopathologic analysis, with increased risk of local recurrence and metastases and poorer overall prognosis. However, treatment with targeted monoclonal antibody therapies such as trastuzumab and pertuzumab provides better local-regional control and leads to improved survival outcome. With neoadjuvant treatments, including monoclonal antibodies, taxanes, and anthracyclines, women are now potentially able to undergo breast conservation therapy and sentinel lymph node biopsy versus mastectomy and axillary lymph node dissection. Thus, the radiologist's role in assessing the extent of local-regional disease and response to neoadjuvant treatment at imaging is important to inform surgical planning and adjuvant treatment. However, assessment of treatment response remains difficult, with the potential for different imaging modalities to result in underestimation or overestimation of disease to varying degrees when compared with surgical pathologic analysis. In particular, the presence of calcifications at mammography is especially difficult to correlate with the results of pathologic analysis after chemotherapy. Breast MRI findings remain the best predictor of pathologic response. The authors review the initial manifestations of HER2-positive tumors, the varied responses to neoadjuvant chemotherapy, and the challenges in assessing residual cancer burden through a multimodality imaging review with pathologic correlation. © RSNA, 2023 Quiz questions for this article are available through the Online Learning Center.
Collapse
Affiliation(s)
- Leah H Portnow
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Jeanne M Kochkodan-Self
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Amy Maduram
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Mirelys Barrios
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Allison M Onken
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Xuefei Hong
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Elizabeth A Mittendorf
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Catherine S Giess
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Sona A Chikarmane
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| |
Collapse
|
20
|
Pfob A, Dubsky P. The underused potential of breast conserving therapy after neoadjuvant system treatment - Causes and solutions. Breast 2023; 67:110-115. [PMID: 36669994 PMCID: PMC9982288 DOI: 10.1016/j.breast.2023.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/08/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Breast conserving therapy (BCT), consisting of breast conserving surgery and subsequent radiotherapy, is an equivalent option to mastectomy for women with early breast cancer. Although BCT after neoadjuvant systemic treatment (NAST) has been routinely recommend by international guidelines since many years, the rate of BCT worldwide varies largely and its potential is still underused. While the rate of BCT in western countries has increased over the past decades to currently about 70%, the rate of BCT is as low as 10% in other countries. In this review, we will evaluate the underused potential of breast conservation after NAST, identify causes, and discuss possible solutions. We identified clinical and non-clinical causes for the underuse of BCT after NAST including uncertainties within the community regarding oncologic outcomes, the correct tumor localization after NAST, the management of multifocal and multicentric tumors, margin assessment, disparities of socio-economic aspects on a patient and national level, and psychological biases affecting the shared decision-making process between patients and clinicians. Possible solutions to mitigate the underuse of BCT after NAST include interdisciplinary teams that keep the whole patient pathway in mind, optimized treatment counseling and shared decision-making, and targeted financial support to alleviate disparities.
Collapse
Affiliation(s)
- André Pfob
- Department of Obstetrics & Gynecology, Heidelberg University Hospital, Germany; National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Peter Dubsky
- Breast Centre, Hirslanden Klinik St. Anna, Luzern, Switzerland,Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, Austria
| |
Collapse
|
21
|
Zheng D, He X, Jing J. Overview of Artificial Intelligence in Breast Cancer Medical Imaging. J Clin Med 2023; 12:419. [PMID: 36675348 PMCID: PMC9864608 DOI: 10.3390/jcm12020419] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
The heavy global burden and mortality of breast cancer emphasize the importance of early diagnosis and treatment. Imaging detection is one of the main tools used in clinical practice for screening, diagnosis, and treatment efficacy evaluation, and can visualize changes in tumor size and texture before and after treatment. The overwhelming number of images, which lead to a heavy workload for radiologists and a sluggish reporting period, suggests the need for computer-aid detection techniques and platform. In addition, complex and changeable image features, heterogeneous quality of images, and inconsistent interpretation by different radiologists and medical institutions constitute the primary difficulties in breast cancer screening and imaging diagnosis. The advancement of imaging-based artificial intelligence (AI)-assisted tumor diagnosis is an ideal strategy for improving imaging diagnosis efficient and accuracy. By learning from image data input and constructing algorithm models, AI is able to recognize, segment, and diagnose tumor lesion automatically, showing promising application prospects. Furthermore, the rapid advancement of "omics" promotes a deeper and more comprehensive recognition of the nature of cancer. The fascinating relationship between tumor image and molecular characteristics has attracted attention to the radiomic and radiogenomics, which allow us to perform analysis and detection on the molecular level with no need for invasive operations. In this review, we integrate the current developments in AI-assisted imaging diagnosis and discuss the advances of AI-based breast cancer precise diagnosis from a clinical point of view. Although AI-assisted imaging breast cancer screening and detection is an emerging field and draws much attention, the clinical application of AI in tumor lesion recognition, segmentation, and diagnosis is still limited to research or in limited patients' cohort. Randomized clinical trials based on large and high-quality cohort are lacking. This review aims to describe the progress of the imaging-based AI application in breast cancer screening and diagnosis for clinicians.
Collapse
Affiliation(s)
| | | | - Jing Jing
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
22
|
Montagna G. Estimating the Benefit of Preoperative Systemic Therapy to Reduce the Extent of Breast Cancer Surgery: Current Standard and Future Directions. Cancer Treat Res 2023; 188:149-174. [PMID: 38175345 DOI: 10.1007/978-3-031-33602-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Once reserved for locally advanced tumors which were deemed inoperable at presentation, preoperative systemic therapy (PST) is nowadays increasingly used to treat early breast cancer. PST allows for in vivo assessment of tumor response, for tailoring of adjuvant systemic therapy and for de-escalation of breast and the axillary surgery. Increased rates of pathological complete response together with more accurate response assessment and surgical planning have led to a significant reduction in surgical morbidity. While surgical assessment remains the standard of care, ongoing studies are evaluating whether surgery can be omitted in patients who achieve a complete pathological response. In this chapter, I will review the impact of PST on surgical de-escalation and the data supporting the safety of this approach.
Collapse
Affiliation(s)
- Giacomo Montagna
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 300 East 66Th Street, New York, NY, 10065, USA.
| |
Collapse
|
23
|
Pavlov MV, Bavrina AP, Plekhanov VI, Golubyatnikov GY, Orlova AG, Subochev PV, Davydova DA, Turchin IV, Maslennikova AV. Changes in the tumor oxygenation but not in the tumor volume and tumor vascularization reflect early response of breast cancer to neoadjuvant chemotherapy. Breast Cancer Res 2023; 25:12. [PMID: 36717842 PMCID: PMC9887770 DOI: 10.1186/s13058-023-01607-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Breast cancer neoadjuvant chemotherapy (NACT) allows for assessing tumor sensitivity to systemic treatment, planning adjuvant treatment and follow-up. However, a sufficiently large number of patients fail to achieve the desired level of pathological tumor response while optimal early response assessment methods have not been established now. In our study, we simultaneously assessed the early chemotherapy-induced changes in the tumor volume by ultrasound (US), the tumor oxygenation by diffuse optical spectroscopy imaging (DOSI), and the state of the tumor vascular bed by Doppler US to elaborate the predictive criteria of breast tumor response to treatment. METHODS A total of 133 patients with a confirmed diagnosis of invasive breast cancer stage II to III admitted to NACT following definitive breast surgery were enrolled, of those 103 were included in the final analysis. Tumor oxygenation by DOSI, tumor volume by US, and tumor vascularization by Doppler US were determined before the first and second cycle of NACT. After NACT completion, patients underwent surgery followed by pathological examination and assessment of the pathological tumor response. On the basis of these, data regression predictive models were created. RESULTS We observed changes in all three parameters 3 weeks after the start of the treatment. However, a high predictive potential for early assessment of tumor sensitivity to NACT demonstrated only the level of oxygenation, ΔStO2, (ρ = 0.802, p ≤ 0.01). The regression model predicts the tumor response with a high probability of a correct conclusion (89.3%). The "Tumor volume" model and the "Vascularization index" model did not accurately predict the absence of a pathological tumor response to treatment (60.9% and 58.7%, respectively), while predicting a positive response to treatment was relatively better (78.9% and 75.4%, respectively). CONCLUSIONS Diffuse optical spectroscopy imaging appeared to be a robust tool for early predicting breast cancer response to chemotherapy. It may help identify patients who need additional molecular genetic study of the tumor in order to find the source of resistance to treatment, as well as to correct the treatment regimen.
Collapse
Affiliation(s)
- Mikhail V. Pavlov
- Nizhny Novgorod Regional Clinical Oncology Dispensary, Delovaya St., 11/1, Nizhny Novgorod, Russia 603126
| | - Anna P. Bavrina
- grid.416347.30000 0004 0386 1631Privolzhsky Research Medical University, Minina Square, 10/1, Nizhny Novgorod, Russia 603950
| | - Vladimir I. Plekhanov
- grid.410472.40000 0004 0638 0147Institute of Applied Physics RAS, Ul’yanov Street, 46, Nizhny Novgorod, Russia 603950
| | - German Yu. Golubyatnikov
- grid.410472.40000 0004 0638 0147Institute of Applied Physics RAS, Ul’yanov Street, 46, Nizhny Novgorod, Russia 603950
| | - Anna G. Orlova
- grid.410472.40000 0004 0638 0147Institute of Applied Physics RAS, Ul’yanov Street, 46, Nizhny Novgorod, Russia 603950
| | - Pavel V. Subochev
- grid.410472.40000 0004 0638 0147Institute of Applied Physics RAS, Ul’yanov Street, 46, Nizhny Novgorod, Russia 603950
| | - Diana A. Davydova
- Nizhny Novgorod Regional Clinical Oncology Dispensary, Delovaya St., 11/1, Nizhny Novgorod, Russia 603126
| | - Ilya V. Turchin
- grid.410472.40000 0004 0638 0147Institute of Applied Physics RAS, Ul’yanov Street, 46, Nizhny Novgorod, Russia 603950
| | - Anna V. Maslennikova
- grid.416347.30000 0004 0386 1631Privolzhsky Research Medical University, Minina Square, 10/1, Nizhny Novgorod, Russia 603950 ,grid.28171.3d0000 0001 0344 908XNational Research Lobachevsky State University of Nizhny Novgorod, Gagarin Ave., 23, Nizhny Novgorod, Russia 603022
| |
Collapse
|
24
|
Sabatino V, Pignata A, Valentini M, Fantò C, Leonardi I, Campora M. Assessment and Response to Neoadjuvant Treatments in Breast Cancer: Current Practice, Response Monitoring, Future Approaches and Perspectives. Cancer Treat Res 2023; 188:105-147. [PMID: 38175344 DOI: 10.1007/978-3-031-33602-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Neoadjuvant treatments (NAT) for breast cancer (BC) consist in the administration of chemotherapy-more rarely endocrine therapy-before surgery. Firstly, it was introduced 50 years ago to downsize locally advanced (inoperable) BCs. NAT are now widespread and so effective to be used also at the early stage of the disease. NAT are heterogeneous in terms of therapeutic patterns, class of used drugs, dosage, and duration. The poly-chemotherapy regimen and administration schedule are established by a multi-disciplinary team, according to the stage of disease, the tumor subtype and the age, the physical status, and the drug sensitivity of BC patients. Consequently, an accurate monitoring of treatment response can provide significant clinical advantages, such as the treatment de-escalation in case of early recognition of complete response or, on the contrary, the switch to an alternative treatment path in case of early detection of resistance to the ongoing therapy. Future is going toward increasingly personalized therapies and the prediction of individual response to treatment is the key to practice customized care pathways, preserving oncological safety and effectiveness. To gain such goal, the development of an accurate monitoring system, reproducible and reliable alone or as part of more complex diagnostic algorithms, will be promising.
Collapse
Affiliation(s)
- Vincenzo Sabatino
- Breast Imaging Department, Santa Chiara Hospital, APSS, Trento, Italy.
| | - Alma Pignata
- Breast Center, Spedali Civili Hospital, ASST, Brescia, Italy
| | - Marvi Valentini
- Breast Imaging Department, Santa Chiara Hospital, APSS, Trento, Italy
| | - Carmen Fantò
- Breast Imaging Department, Santa Chiara Hospital, APSS, Trento, Italy
| | - Irene Leonardi
- Breast Imaging Department, Santa Chiara Hospital, APSS, Trento, Italy
| | - Michela Campora
- Pathology Department, Santa Chiara Hospital, APSS, Trento, Italy
| |
Collapse
|
25
|
Sobhi A, Talaat hamed S, Hussein ES, Lasheen S, Hussein M, Ebrahim Y. Predicting pathological response of locally advanced breast cancer to neoadjuvant chemotherapy: comparing the performance of whole body 18F-FDG PETCT versus DCE-MRI of the breast. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2022. [DOI: 10.1186/s43055-022-00743-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
With the expansion of the use of the neoadjuvant chemotherapy(NAC) in locally advanced breast cancer (LABC), both dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET CT) are promising methods for assessment of the tumor response during chemotherapy. We aimed to evaluate the diagnostic accuracy of DCE-MRI of breast &18 F-FDG PETCT regarding the assessment of early response to neoadjuvant chemotherapy (NAC) in locally advanced breast cancer patients (LABC) and pathologic complete response (pCR) prediction.
Results
A total of forty LABC patients who had NAC were included in the study. Before and during NAC, PET/CT and DCE-MRI were used. Various morphological and functional criteria were compared and linked with post-operative pathology for both. The MRI sensitivity and specificity in assessing NAC response in conjunction with pathological data were 100% (p = 0.001) and 12.5% (p = 0.18) respectively. The equivalent readings for PET/CT were 94.1% (p = 0.001) and 25% (p = 0.18), respectively, although the estimated total accuracy for both MRI and PETCT was the same measuring 94.1% (p = 0.001) and 25% (p = 0.18) (72%). PETCT had a higher overall accuracy than MRI in assessing the response of axillary lymph nodes (ALN) to NAC (64% and 56%, respectively). Longest diameter of lesion, ADC value, and maximal enhancement in baseline MRI, SUVmax and SUV mean in baseline PETCT were all significant predictors of rCR.
Conclusion
During NAC in the primary breast mass and ALN, DCE-MRI demonstrated a better sensitivity in predicting pCR in LABC patients. Although both MRI and PETCT were equally accurate in detecting pCR of LABC patients to NAC, PETCT was more accurate in detecting pathological response of ALN to NAC.
Collapse
|
26
|
Phadke S. Optimization of Neoadjuvant Therapy for Early-Stage Triple-Negative and HER2 + Breast Cancer. Curr Oncol Rep 2022; 24:1779-1789. [PMID: 36181611 DOI: 10.1007/s11912-022-01331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Neoadjuvant, or pre-operative, therapy for the treatment of early-stage breast cancer has several potential benefits, especially for patients with triple-negative or HER2 + subtypes. This review provides an overview of optimal practices for utilizing neoadjuvant therapy, guidelines for decision-making, and ongoing clinical trials that are expected to help refine therapy choices. RECENT FINDINGS For triple-negative disease, the addition of the checkpoint inhibitor pembrolizumab to chemotherapy has shown remarkable efficacy, increasing response rates and survival. In the HER2 + setting, we are now able to safely avoid use of anthracyclines in most patients and refine adjuvant treatment choices based on response to neoadjuvant therapy. Results from recent clinical studies highlight advancements in systemic therapy and mark steps toward precision medicine, although reliable biomarkers of therapy response are still needed.
Collapse
Affiliation(s)
- Sneha Phadke
- Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
27
|
Zhang MQ, Du Y, Zha HL, Liu XP, Cai MJ, Chen ZH, Chen R, Wang J, Wang SJ, Zhang JL, Li CY. Construction and validation of a personalized nomogram of ultrasound for pretreatment prediction of breast cancer patients sensitive to neoadjuvant chemotherapy. Br J Radiol 2022; 95:20220626. [PMID: 36378247 PMCID: PMC9733610 DOI: 10.1259/bjr.20220626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/26/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To construct a combined radiomics model based on pre-treatment ultrasound for predicting of advanced breast cancers sensitive to neoadjuvant chemotherapy (NAC). METHODS A total of 288 eligible breast cancer patients who underwent NAC before surgery were enrolled in the retrospective study cohort. Radiomics features reflecting the phenotype of the pre-NAC tumors were extracted. With features selected using the least absolute shrinkage and selection operator (LASSO) regression, radiomics signature (Rad-score) was established based on the pre-NAC ultrasound. Then, radiomics nomogram of ultrasound (RU) was established on the basis of the best radiomic signature incorporating independent clinical features. The performance of RU was evaluated in terms of calibration curve, area under the curve (AUC), and decision curve analysis (DCA). RESULTS Nine features were selected to construct the radiomics signature in the training cohort. Combined with independent clinical characteristics, the performance of RU for identifying Grade 4-5 patients was significantly superior than the clinical model and Rad-score alone (p < 0.05, as per the Delong test), which achieved an AUC of 0.863 (95% CI, 0.814-0.963) in the training group and 0.854 (95% CI, 0.776-0.931) in the validation group. DCA showed that this model satisfactory clinical utility, suggesting its robustness as a response predictor. CONCLUSION This study demonstrated that RU has a potential role in predicting drug-sensitive breast cancers. ADVANCES IN KNOWLEDGE Aiming at early detection of Grade 4-5 breast cancer patients, the radiomics nomogram based on ultrasound has been approved as a promising indicator with high clinical utility. It is the first application of ultrasound-based radiomics nomogram to distinguish drug-sensitive breast cancers.
Collapse
Affiliation(s)
- Man-Qi Zhang
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Du
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Ling Zha
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Pei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng-Jun Cai
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Hui Chen
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Chen
- Department of Breast surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jue Wang
- Department of Breast surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shou-Ju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu-Lou Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Ying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Madani M, Behzadi MM, Nabavi S. The Role of Deep Learning in Advancing Breast Cancer Detection Using Different Imaging Modalities: A Systematic Review. Cancers (Basel) 2022; 14:5334. [PMID: 36358753 PMCID: PMC9655692 DOI: 10.3390/cancers14215334] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 12/02/2022] Open
Abstract
Breast cancer is among the most common and fatal diseases for women, and no permanent treatment has been discovered. Thus, early detection is a crucial step to control and cure breast cancer that can save the lives of millions of women. For example, in 2020, more than 65% of breast cancer patients were diagnosed in an early stage of cancer, from which all survived. Although early detection is the most effective approach for cancer treatment, breast cancer screening conducted by radiologists is very expensive and time-consuming. More importantly, conventional methods of analyzing breast cancer images suffer from high false-detection rates. Different breast cancer imaging modalities are used to extract and analyze the key features affecting the diagnosis and treatment of breast cancer. These imaging modalities can be divided into subgroups such as mammograms, ultrasound, magnetic resonance imaging, histopathological images, or any combination of them. Radiologists or pathologists analyze images produced by these methods manually, which leads to an increase in the risk of wrong decisions for cancer detection. Thus, the utilization of new automatic methods to analyze all kinds of breast screening images to assist radiologists to interpret images is required. Recently, artificial intelligence (AI) has been widely utilized to automatically improve the early detection and treatment of different types of cancer, specifically breast cancer, thereby enhancing the survival chance of patients. Advances in AI algorithms, such as deep learning, and the availability of datasets obtained from various imaging modalities have opened an opportunity to surpass the limitations of current breast cancer analysis methods. In this article, we first review breast cancer imaging modalities, and their strengths and limitations. Then, we explore and summarize the most recent studies that employed AI in breast cancer detection using various breast imaging modalities. In addition, we report available datasets on the breast-cancer imaging modalities which are important in developing AI-based algorithms and training deep learning models. In conclusion, this review paper tries to provide a comprehensive resource to help researchers working in breast cancer imaging analysis.
Collapse
Affiliation(s)
- Mohammad Madani
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Mohammad Mahdi Behzadi
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Sheida Nabavi
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
29
|
Yi X, Huang D, Li Z, Wang X, Yang T, Zhao M, Wu J, Zhong T. The role and application of small extracellular vesicles in breast cancer. Front Oncol 2022; 12:980404. [PMID: 36185265 PMCID: PMC9515427 DOI: 10.3389/fonc.2022.980404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy and the leading cause of cancer-related deaths in women worldwide. Currently, patients’ survival remains a challenge in BC due to the lack of effective targeted therapies and the difficult condition of patients with higher aggressiveness, metastasis and drug resistance. Small extracellular vesicles (sEVs), which are nanoscale vesicles with lipid bilayer envelopes released by various cell types in physiological and pathological conditions, play an important role in biological information transfer between cells. There is growing evidence that BC cell-derived sEVs may contribute to the establishment of a favorable microenvironment that supports cancer cells proliferation, invasion and metastasis. Moreover, sEVs provide a versatile platform not only for the diagnosis but also as a delivery vehicle for drugs. This review provides an overview of current new developments regarding the involvement of sEVs in BC pathogenesis, including tumor proliferation, invasion, metastasis, immune evasion, and drug resistance. In addition, sEVs act as messenger carriers carrying a variety of biomolecules such as proteins, nucleic acids, lipids and metabolites, making them as potential liquid biopsy biomarkers for BC diagnosis and prognosis. We also described the clinical applications of BC derived sEVs associated MiRs in the diagnosis and treatment of BC along with ongoing clinical trials which will assist future scientific endeavors in a more organized direction.
Collapse
Affiliation(s)
- Xiaomei Yi
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tong Yang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Minghong Zhao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jiyang Wu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Tianyu Zhong,
| |
Collapse
|
30
|
Wang H, Li Y, Qi Y, Zhao E, Kong X, Yang C, Yang Q, Zhang C, Liu Y, Song Z. Pegylated Liposomal Doxorubicin, Docetaxel, and Trastuzumab as Neoadjuvant Treatment for HER2-Positive Breast Cancer Patients: A Phase II and Biomarker Study. Front Oncol 2022; 12:909426. [PMID: 35875123 PMCID: PMC9304895 DOI: 10.3389/fonc.2022.909426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
Background Combined neoadjuvant chemotherapy with trastuzumab and pertuzumab is the standard regimen for human epidermal growth receptor 2 (HER2)-positive breast cancer (BC). However, pertuzumab is not available because it is not on the market or covered by medicare in some regions or poor economy. Anthracyclines and taxanes are cornerstones in BC chemotherapy, and their combination contributes to satisfactory efficiency in neoadjuvant settings. Nonetheless, concomitant administration of trastuzumab and an anthracycline is generally avoided clinically due to cardiotoxicity. Pegylated liposomal doxorubicin (PLD) is less cardiotoxic compared with traditional anthracyclines. Here, we conducted this prospective study to evaluate the efficacy, safety, and potential biomarkers for PLD plus trastuzumab and docetaxel as neoadjuvant treatment in HER2-positive BC. Patients and Methods Patients with stage II or III HER2-positive BC were recruited in this multicenter, open-label, single-arm, phase II study. Eligible patients were given 6 cycles of PLD plus docetaxel and trastuzumab. Primary endpoint was total pathological complete response (tpCR, ypT0/is ypN0). Secondary endpoints were breast pathological complete response (bpCR, ypT0/is), objective response rate (ORR), operation rate, breast-conserving surgery rate, and safety. Metadherin (MTDH), glutaminyl-peptide cyclotransferase (QPCT), topoisomerase II alpha (TOP2A), programmed death ligand 1 (PD-L1), and tumor-infiltrating lymphocytes (TILs) were evaluated in BC tissues pre-neoadjuvant for potential biomarkers. Results Between March 2019 and February 2021, 54 patients were enrolled, 50 were included in the analysis, and 35 (70.0%) completed 6 cycles of neoadjuvant treatment. Forty-nine (98.0%) patients underwent surgery with a breast-conserving rate of 44.0%. The tpCR rate, bpCR rate, and ORR were 48.0% (95% CI, 33.7%–62.6%), 60.0% (95% CI, 45.2%–73.6%), and 84.0% (95% CI, 70.9%–92.8%), respectively. tpCR was associated with MTDH (p = 0.002) and QPCT (p = 0.036) expression but not with TOP2A (p = 0.75), PD-L1 (p = 0.155), or TILs (p = 0.76). Patients with HR-negative status were more likely to achieve bpCR compared with those with HR-positive status (76.2% vs. 48.3%, p = 0.047). Grade ≥3 adverse events occurred in 38.0% of patients. Left ventricular ejection fraction decline by ≥10% was reported in 18.0% of patients, and no patient experienced congestive heart failure. Conclusions PLD plus docetaxel and trastuzumab might be a potential neoadjuvant regimen for HER2-positive BC with a high tpCR rate and manageable tolerability. MTDH and QPCT are potential predictive markers for tpCR.
Collapse
Affiliation(s)
- Haoqi Wang
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Yuntao Li
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Yixin Qi
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Erbao Zhao
- Department of Breast Center, Shanxi Cancer Hospital, Taiyuan, China
| | - Xiangshun Kong
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, China
| | - Chao Yang
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Qiqi Yang
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Chengyuan Zhang
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
| | - Yueping Liu
- Pathology Department, Fourth Hospital of Hebei Medical University, Hebei Province Key Laboratory of Breast Cancer Molecular Medicine, Shijiazhuang, China
- *Correspondence: Zhenchuan Song, ; Yueping Liu,
| | - Zhenchuan Song
- Breast Center, Fourth Hospital of Hebei Medical University, Key Laboratory for Breast Cancer Molecular Medicine of Hebei Province, Shijiazhuang, China
- *Correspondence: Zhenchuan Song, ; Yueping Liu,
| |
Collapse
|
31
|
Pfob A, Sidey-Gibbons C, Rauch G, Thomas B, Schaefgen B, Kuemmel S, Reimer T, Hahn M, Thill M, Blohmer JU, Hackmann J, Malter W, Bekes I, Friedrichs K, Wojcinski S, Joos S, Paepke S, Degenhardt T, Rom J, Rody A, van Mackelenbergh M, Banys-Paluchowski M, Große R, Reinisch M, Karsten M, Golatta M, Heil J. Intelligent Vacuum-Assisted Biopsy to Identify Breast Cancer Patients With Pathologic Complete Response (ypT0 and ypN0) After Neoadjuvant Systemic Treatment for Omission of Breast and Axillary Surgery. J Clin Oncol 2022; 40:1903-1915. [PMID: 35108029 DOI: 10.1200/jco.21.02439] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/24/2021] [Accepted: 01/05/2022] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Neoadjuvant systemic treatment (NST) elicits a pathologic complete response in 40%-70% of women with breast cancer. These patients may not need surgery as all local tumor has already been eradicated by NST. However, nonsurgical approaches, including imaging or vacuum-assisted biopsy (VAB), were not able to accurately identify patients without residual cancer in the breast or axilla. We evaluated the feasibility of a machine learning algorithm (intelligent VAB) to identify exceptional responders to NST. METHODS We trained, tested, and validated a machine learning algorithm using patient, imaging, tumor, and VAB variables to detect residual cancer after NST (ypT+ or in situ or ypN+) before surgery. We used data from 318 women with cT1-3, cN0 or +, human epidermal growth factor receptor 2-positive, triple-negative, or high-proliferative Luminal B-like breast cancer who underwent VAB before surgery (ClinicalTrials.gov identifier: NCT02948764, RESPONDER trial). We used 10-fold cross-validation to train and test the algorithm, which was then externally validated using data of an independent trial (ClinicalTrials.gov identifier: NCT02575612). We compared findings with the histopathologic evaluation of the surgical specimen. We considered false-negative rate (FNR) and specificity to be the main outcomes. RESULTS In the development set (n = 318) and external validation set (n = 45), the intelligent VAB showed an FNR of 0.0%-5.2%, a specificity of 37.5%-40.0%, and an area under the receiver operating characteristic curve of 0.91-0.92 to detect residual cancer (ypT+ or in situ or ypN+) after NST. Spiegelhalter's Z confirmed a well-calibrated model (z score -0.746, P = .228). FNR of the intelligent VAB was lower compared with imaging after NST, VAB alone, or combinations of both. CONCLUSION An intelligent VAB algorithm can reliably exclude residual cancer after NST. The omission of breast and axillary surgery for these exceptional responders may be evaluated in future trials.
Collapse
Affiliation(s)
- André Pfob
- University Breast Unit, Department of Obstetrics & Gynecology, Heidelberg University Hospital, Heidelberg, Germany
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chris Sidey-Gibbons
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Thomas
- Coordination Centre for Clinical Trials (KKS), University Heidelberg, Heidelberg, Germany
| | - Benedikt Schaefgen
- University Breast Unit, Department of Obstetrics & Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Toralf Reimer
- Department of Gynecology/Breast Unit, University Hospital Rostock, Rostock, Germany
| | - Markus Hahn
- Department of Gynecology/Breast Unit, University Hospital Tuebingen, Tuebingen, Germany
| | - Marc Thill
- Department of Gynecology and Gynecological Oncology/Breast Unit, Agaplesion Markus Hospital Frankfurt, Frankfurt, Germany
| | - Jens-Uwe Blohmer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Gynecology with Breast Center, Berlin, Germany
| | - John Hackmann
- Department of Gynecology/Breast Unit, Marienhospital, Witten, Germany
| | - Wolfram Malter
- Department of Gynecology and Obstetrics, Breast Cancer Center, Medical Faculty, University of Cologne, Cologne, Germany
| | - Inga Bekes
- Department of Gynecology/Breast Unit, University Hospital Ulm, Ulm, Germany
| | - Kay Friedrichs
- Department of Gynecology/Breast Unit, Jerusalem Hospital Hamburg, Hamburg, Germany
| | - Sebastian Wojcinski
- Department of Gynecology and Obstetrics, Breast Cancer Center, Klinikum Bielefeld Mitte GmbH, Bielefeld, Germany
| | - Sylvie Joos
- Radiologische Allianz Hamburg, Hamburg, Germany
| | - Stefan Paepke
- Department of Gynecology/Breast Unit, Hospital rechts der Isar, Munich, Germany
| | - Tom Degenhardt
- Department of Gynecology/Breast Unit, University Hospital Munich, Munich, Germany
| | - Joachim Rom
- Department of Gynecology/Breast Unit, Klinikum Frankfurt-Höchst, Frankfurt, Germany
| | - Achim Rody
- Department of Gynecology/Breast Unit, University Hospital Schleswig-Holstein, Luebeck, Germany
| | | | - Maggie Banys-Paluchowski
- Department of Gynecology/Breast Unit, University Hospital Schleswig-Holstein, Luebeck, Germany
- Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Regina Große
- Department of Gynecology/Breast Unit, University Hospital Halle, Halle, Germany
| | | | - Maria Karsten
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Gynecology with Breast Center, Berlin, Germany
| | - Michael Golatta
- University Breast Unit, Department of Obstetrics & Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Heil
- University Breast Unit, Department of Obstetrics & Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
32
|
Management of the axilla in T1-2N1 breast cancer. NPJ Breast Cancer 2022; 8:69. [PMID: 35637226 PMCID: PMC9151923 DOI: 10.1038/s41523-022-00432-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022] Open
|
33
|
Kong X, Zhang Q, Wu X, Zou T, Duan J, Song S, Nie J, Tao C, Tang M, Wang M, Zou J, Xie Y, Li Z, Li Z. Advances in Imaging in Evaluating the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer. Front Oncol 2022; 12:816297. [PMID: 35669440 PMCID: PMC9163342 DOI: 10.3389/fonc.2022.816297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) is increasingly widely used in breast cancer treatment, and accurate evaluation of its response provides essential information for treatment and prognosis. Thus, the imaging tools used to quantify the disease response are critical in evaluating and managing patients treated with NAC. We discussed the recent progress, advantages, and disadvantages of common imaging methods in assessing the efficacy of NAC for breast cancer.
Collapse
Affiliation(s)
- Xianshu Kong
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Qian Zhang
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Xuemei Wu
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Tianning Zou
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jiajun Duan
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Shujie Song
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jianyun Nie
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Chu Tao
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Mi Tang
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Maohua Wang
- First Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jieya Zou
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yu Xie
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhen Li
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| |
Collapse
|
34
|
Hatazawa J. The Clinical Value of Breast Specific Gamma Imaging and Positron Imaging: An Update. Semin Nucl Med 2022; 52:619-627. [PMID: 35346487 DOI: 10.1053/j.semnuclmed.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 01/15/2023]
Abstract
In the management of patients with breast cancer (BC), a mammography contributed to screen an early-stage patient, to plan a therapy strategy, to evaluate a therapy outcome, to detect a recurrence, and to reduce a mortality. Currently, various imaging modalities, such as CT, MR, Ultrasound (US), SPECT/CT, PET/CT, PET/MR have been utilized for the management of BC patients. In order to overcome a limited spatial resolution and sensitivity of whole-body systems in nuclear medicine imaging, dedicated breast imaging modalities were developed. One is a gamma imaging system with single/dual head scintillation detectors or semiconductor detectors associated with light compression device for breast parenchyma. Radiopharmaceutical for the gamma imaging is 99mTc-sestamibi. Another is a positron imaging system with opposite-type panel detectors and ring-shaped type detectors. Radiopharmaceutical for positron imaging is 18F-fluorodeoxyglucose. The breast-specific gamma and positron imaging systems were utilized mainly to detect small lesions less than 1 cm in diameter especially in patients with dense breast, to evaluate an effect of preoperative neo-adjuvant therapy, to plan surgical procedures (conservative-surgery vs mastectomy), and to detect a recurrence. By combining higher sensitivity and spatial resolution scanners with new radiopharmaceuticals, an information on molecular-level pathology of BC is increasingly available in an individual patient. This article reviewed clinical impact and future perspective of this field.
Collapse
Affiliation(s)
- Jun Hatazawa
- Department of Quantum Cancer Therapy, Research Center for Nuclear Physics, Osaka University, Osaka, Japan; Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan; Institute for Radiation Sciences, Osaka University, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
35
|
Bitencourt A, Iima M, Langs G, Pinker K. Editorial: Impact of Breast MRI on Breast Cancer Treatment and Prognosis. Front Oncol 2022; 12:825101. [PMID: 35359360 PMCID: PMC8963269 DOI: 10.3389/fonc.2022.825101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Almir Bitencourt
- Imaging Department, A.C.Camargo Cancer Center, São Paulo, Brazil
- Breast Imaging Section, DASA, São Paulo, Brazil
| | - Mami Iima
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Georg Langs
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
36
|
Heil J, Pfob A, Sinn HP, Rauch G, Bach P, Thomas B, Schaefgen B, Kuemmel S, Reimer T, Hahn M, Thill M, Blohmer JU, Hackmann J, Malter W, Bekes I, Friedrichs K, Wojcinski S, Joos S, Paepke S, Ditsch N, Rody A, Große R, van Mackelenbergh M, Reinisch M, Karsten M, Golatta M. Diagnosing Pathologic Complete Response in the Breast After Neoadjuvant Systemic Treatment of Breast Cancer Patients by Minimal Invasive Biopsy: Oral Presentation at the San Antonio Breast Cancer Symposium on Friday, December 13, 2019, Program Number GS5-03. Ann Surg 2022; 275:576-581. [PMID: 32657944 DOI: 10.1097/sla.0000000000004246] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We evaluated the ability of minimally invasive, image-guided vacuum-assisted biopsy (VAB) to reliably diagnose a pathologic complete response in the breast (pCR-B). SUMMARY BACKGROUND DATA Neoadjuvant systemic treatment (NST) elicits a pathologic complete response in up to 80% of women with breast cancer. In such cases, breast surgery, the gold standard for confirming pCR-B, may be considered overtreatment. METHODS This multicenter, prospective trial enrolled 452 women presenting with initial stage 1-3 breast cancer of all biological subtypes. Fifty-four women dropped out; 398 were included in the full analysis. All participants had an imaging-confirmed partial or complete response to NST and underwent study-specific image-guided VAB before guideline-adherent breast surgery. The primary endpoint was the false-negative rate (FNR) of VAB-confirmed pCR-B. RESULTS Image-guided VAB alone did not detect surgically confirmed residual tumor in 37 of 208 women [FNR, 17.8%; 95% confidence interval (CI), 12.8-23.7%]. Of these 37 women, 12 (32.4%) had residual DCIS only, 20 (54.1%) had minimal residual tumor (<5 mm), and 19 of 25 (76.0%) exhibited invasive cancer cellularity of ≤10%. In 19 of the 37 cases (51.4%), the false-negative result was potentially avoidable. Exploratory analysis showed that performing VAB with the largest needle by volume (7-gauge) resulted in no false-negative results and that combining imaging and image-guided VAB into a single diagnostic test lowered the FNR to 6.2% (95% CI, 3.4%-10.5%). CONCLUSIONS Image-guided VAB missed residual disease more often than expected. Refinements in procedure and patient selection seem possible and necessary before omitting breast surgery.
Collapse
Affiliation(s)
- Joerg Heil
- Department of Gynecology/Breast Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - André Pfob
- Department of Gynecology/Breast Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Peter Sinn
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Paul Bach
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Bettina Thomas
- Coordination Centre for Clinical Trials (KKS), University Heidelberg, Heidelberg, Germany
| | - Benedikt Schaefgen
- Department of Gynecology/Breast Unit, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Toralf Reimer
- Department of Gynecology/Breast Unit, University Hospital Rostock, Rostock, Germany
| | - Markus Hahn
- Department of Gynecology/Breast Unit, University Hospital Tuebingen, Tuebingen, Germany
| | - Marc Thill
- Department of Gynecology and Gynecological Oncology/Breast Unit, Agaplesion Markus Hospital Frankfurt, Frankfurt, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology/Breast Unit, University Hospital Berlin, Berlin, Germany
| | - John Hackmann
- Department of Gynecology/Breast Unit, Marienhospital, Witten, Germany
| | - Wolfram Malter
- Department of Gynecology/Breast Unit, University Hospital of Cologne, Köln, Germany
| | - Inga Bekes
- Department of Gynecology/Breast Unit, University Hospital Ulm, Ulm, Germany
| | - Kay Friedrichs
- Department of Gynecology/Breast Unit, Jerusalem Hospital Hamburg, Hamburg, Germany
| | - Sebastian Wojcinski
- Department of Gynecology/Breast Unit, Franziskus Hospital Bielefeld, Bielefeld, Germany
| | - Sylvie Joos
- Department of Radiology, Visiorad, Pinneberg, Germany
| | - Stefan Paepke
- Department of Gynecology/Breast Unit, Hospital rechts der Isar, Munich, Germany
| | - Nina Ditsch
- Department of Gynecology/Breast Unit, University Hospital Munich, Munich, Germany
- Department of Gynecology/Breast Unit, University Hospital Augsburg, Augsburg, Germany
| | - Achim Rody
- Department of Gynecology/Breast Unit, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Regina Große
- Department of Gynecology/Breast Unit, University Hospital Halle, Halle, Germany
| | | | | | - Maria Karsten
- Department of Gynecology/Breast Unit, University Hospital Berlin, Berlin, Germany
| | - Michael Golatta
- Department of Gynecology/Breast Unit, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
37
|
Pfob A, Heil J. Breast and axillary surgery after neoadjuvant systemic treatment - A review of clinical routine recommendations and the latest clinical research. Breast 2022; 62 Suppl 1:S7-S11. [PMID: 35135710 PMCID: PMC9097799 DOI: 10.1016/j.breast.2022.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/27/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Breast and axillary surgery after neoadjuvant systemic treatment for women with breast cancer has undergone multiple paradigm changes within the past years. In this review, we provide a state-of-the-art overview of breast and axillary surgery after neoadjuvant systemic treatment from both, a clinical routine perspective and a clinical research perspective. For axillary disease, axillary lymph node dissection, sentinel lymph node biopsy, or targeted axillary dissection are nowadays recommended depending on the lymph node status before and after neoadjuvant systemic treatment. For the primary tumor in the breast, breast conserving surgery remains the standard of care. The clinical management of exceptional responders to neoadjuvant systemic treatment is a pressing knowledge gap due to the increasing number of patients who achieve a pathologic complete response to neoadjuvant systemic treatment and for whom surgery may have no therapeutic benefit. Current clinical research evaluates whether less invasive procedures can exclude residual cancer after neoadjuvant systemic treatment as reliably as surgery to possibly omit surgery for those patients in the future. Breast and axillary surgery after neoadjuvant systemic treatment has evolved. Choice of axillary surgery depends on lymph node status before and after treatment. Optimal management of exceptional responders to neoadjuvant treatment is unclear. Clinical research aims to reliably exclude residual cancer without surgery. For exceptional responders, breast cancer surgery may be omitted in the future.
Collapse
Affiliation(s)
- André Pfob
- University Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Heil
- University Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
38
|
Lee SC, Tchelepi H, Khadem N, Desai B, Yamashita M, Hovanessian-Larsen L. Imaging of Benign and Malignant Breast Lesions Using Contrast-Enhanced Ultrasound: A Pictorial Essay. Ultrasound Q 2022; 38:2-12. [PMID: 35239626 DOI: 10.1097/ruq.0000000000000574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Contrast-enhanced ultrasound is a promising noninvasive imaging technique for evaluating benign and malignant breast lesions, as contrast provides information about perfusion and microvasculature. Contrast-enhanced ultrasound is currently off-label use in the breast in the United States, but its clinical and investigational use in breast imaging is gaining popularity. It is important for radiologists to be familiar with the imaging appearances of benign and malignant breast masses using contrast-enhanced ultrasound. This pictorial essay illustrates enhancement patterns of various breast masses from our own experience. Pathologies include subtypes of invasive breast cancer, fibroadenomas, papillary lesions, fibrocystic change, and inflammatory processes. Contrast-enhanced ultrasound pitfalls and limitations are discussed.
Collapse
Affiliation(s)
- Sandy C Lee
- Department of Radiology, Keck School of Medicine, University of Southern California
| | - Hisham Tchelepi
- Department of Radiology, Keck School of Medicine, University of Southern California
| | - Nasim Khadem
- Department of Radiology, VA Long Beach Medical Center, Long Beach, CA
| | - Bhushan Desai
- Department of Radiology, Keck School of Medicine, University of Southern California
| | - Mary Yamashita
- Department of Radiology, Keck School of Medicine, University of Southern California
| | | |
Collapse
|
39
|
Rubio IT, Sobrido C. Neoadjuvant approach in patients with early breast cancer: patient assessment, staging, and planning. Breast 2022; 62 Suppl 1:S17-S24. [PMID: 34996668 PMCID: PMC9097809 DOI: 10.1016/j.breast.2021.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/30/2021] [Indexed: 11/30/2022] Open
Abstract
Neoadjuvant treatment (NAT) has become an option in early stage (stage I-II) breast cancer (EBC). New advances in systemic and targeted therapies have increased rates of pathologic complete response increasing the number of patients undergoing NAT. Clear benefits of NAT are downstaging the tumor and the axillary nodes to de-escalate surgery and to evaluate response to treatment. Selection of patients for NAT in EBC rely in several factors that are related to patient characteristics (i.e, age and comorbidities), to tumor histology, to stage at diagnosis and to the potential changes in surgical or adjuvant treatments when NAT is administered. Imaging and histologic confirmation is performed to assess extent of disease y to confirm diagnosis. Besides mammogram and ultrasound, functional breast imaging MRI has been incorporated to better predict treatment response and residual disease. Contrast enhanced mammogram (CEM), shear wave elastography (SWE), or Dynamic Optical Breast Imaging (DOBI) are emerging techniques under investigation for assessment of response to neoadjuvant therapy as well as for predicting response. Surgical plan should be delineated after NAT taking into account baseline characteristics, tumor response and patient desire. In the COVID era, we have witnessed also the increasing use of NAT in patients who may be directed to surgery, unable to have it performed as surgery has been reserved for emergency cases only.
Collapse
|
40
|
Increasing Imaging Value to Breast Cancer Care Through Prognostic Modeling of Multiparametric MRI Features in Patients Undergoing Neoadjuvant Chemotherapy. Acad Radiol 2022; 29 Suppl 1:S164-S165. [PMID: 35033453 DOI: 10.1016/j.acra.2021.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022]
|
41
|
Diagnostic performance of breast imaging with ultrasonography, magnetic resonance and mammography in the assessment of residual tumor after neoadjuvant chemotherapy in breast cancer patients. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1034379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
42
|
Deep learning radiomics of ultrasonography can predict response to neoadjuvant chemotherapy in breast cancer at an early stage of treatment: a prospective study. Eur Radiol 2021; 32:2099-2109. [PMID: 34654965 DOI: 10.1007/s00330-021-08293-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Breast cancer (BC) is the most common cancer in women worldwide, and neoadjuvant chemotherapy (NAC) is considered the standard of treatment for most patients with BC. However, response rates to NAC vary among patients, which leads to delays in appropriate treatment and affects the prognosis for patients who ineffectively respond to NAC. This study aimed to investigate the feasibility of deep learning radiomics (DLR) in the prediction of NAC response at an early stage. METHODS In total, 168 patients with clinicopathologically confirmed BC were enrolled in this prospective study, from March 2016 to December 2020. All patients completed NAC treatment and underwent ultrasonography (US) at three time points (before NAC, after the second course, and after the fourth course). We developed two DLR models, DLR-2 and DLR-4, for predicting responses after the second and fourth courses of NAC. Furthermore, a novel deep learning radiomics pipeline (DLRP) was proposed for stepwise prediction of response at different time points of NAC administration. RESULTS In the validation cohort, DLR-2 achieved an AUC of 0.812 (95% CI: 0.770-0.851) with an NPV of 83.3% (95% CI: 76.5-89.6). DLR-4 achieved an AUC of 0.937 (95% CI: 0.913-0.955) with a specificity of 90.5% (95% CI: 86.3-94.2). Moreover, 19 of 21 non-response patients were successfully identified by DLRP, suggesting that they could benefit from treatment strategy adjustment at an early stage of NAC. CONCLUSIONS The proposed DLRP strategy holds promise for effectively predicting NAC response at its early stage for BC patients. KEY POINTS • We proposed two novel deep learning radiomics (DLR) models to predict response to neoadjuvant chemotherapy (NAC) in breast cancer (BC) patients based on US images at different NAC time points. • Combining two DLR models, a deep learning radiomics pipeline (DLRP) was proposed for stepwise prediction of response to NAC. • The DLRP may provide BC patients and physicians with an effective and feasible tool to predict response to NAC at an early stage and to determine further personalized treatment options.
Collapse
|
43
|
Koelbel V, Pfob A, Schaefgen B, Sinn P, Feisst M, Golatta M, Gomez C, Stieber A, Bach P, Rauch G, Heil J. Vacuum-Assisted Breast Biopsy After Neoadjuvant Systemic Treatment for Reliable Exclusion of Residual Cancer in Breast Cancer Patients. Ann Surg Oncol 2021; 29:1076-1084. [PMID: 34581923 PMCID: PMC8724060 DOI: 10.1245/s10434-021-10847-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/05/2021] [Indexed: 11/18/2022]
Abstract
Background About 40 % of women with breast cancer achieve a pathologic complete response in the breast after neoadjuvant systemic treatment (NST). To identify these women, vacuum-assisted biopsy (VAB) was evaluated to facilitate risk-adaptive surgery. In confirmatory trials, the rates of missed residual cancer [false-negative rates (FNRs)] were unacceptably high (> 10%). This analysis aimed to improve the ability of VAB to exclude residual cancer in the breast reliably by identifying key characteristics of false-negative cases. Methods Uni- and multivariable logistic regressions were performed using data of a prospective multicenter trial (n = 398) to identify patient and VAB characteristics associated with false-negative cases (no residual cancer in the VAB but in the surgical specimen). Based on these findings FNR was exploratively re-calculated. Results In the multivariable analysis, a false-negative VAB result was significantly associated with accompanying ductal carcinoma in situ (DCIS) in the initial diagnostic biopsy [odds ratio (OR), 3.94; p < 0.001], multicentric disease on imaging before NST (OR, 2.74; p = 0.066), and age (OR, 1.03; p = 0.034). Exclusion of women with DCIS or multicentric disease (n = 114) and classication of VABs that did not remove the clip marker as uncertain representative VABs decreased the FNR to 2.9% (3/104). Conclusion For patients without accompanying DCIS or multicentric disease, performing a distinct representative VAB (i.e., removing a well-placed clip marker) after NST suggests that VAB might reliably exclude residual cancer in the breast without surgery. This evidence will inform the design of future trials evaluating risk-adaptive surgery for exceptional responders to NST.
Collapse
Affiliation(s)
- Vivian Koelbel
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - André Pfob
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Schaefgen
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Sinn
- Department of Pathology, Heidelberg University, Heidelberg, Germany
| | - Manuel Feisst
- Institute of Medical Biometry and Informatics (IMBI), Heidelberg University, Heidelberg, Germany
| | - Michael Golatta
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christina Gomez
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anne Stieber
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Paul Bach
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Joerg Heil
- Breast Unit, Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
44
|
Hunt KN. Molecular Breast Imaging: A Scientific Review. JOURNAL OF BREAST IMAGING 2021; 3:416-426. [PMID: 38424795 DOI: 10.1093/jbi/wbab039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Indexed: 03/02/2024]
Abstract
Molecular breast imaging (MBI) is a nuclear medicine technique that has evolved considerably over the past two decades. Technical advances have allowed reductions in administered doses to the point that they are now acceptable for screening. The most common radiotracer used in MBI, 99mTc-sestamibi, has a long history of safe use. Biopsy capability has become available in recent years, with early clinical experience demonstrating technically successful biopsies of MBI-detected lesions. MBI has been shown to be an effective supplemental screening tool in women with dense breasts and is also utilized for breast cancer staging, assessment of response to neoadjuvant chemotherapy, problem solving, and as an alternative to breast MRI in women who have a contraindication to MRI. The degree of background parenchymal uptake on MBI shows promise as a tool for breast cancer risk stratification. Radiologist interpretation is guided by a validated MBI lexicon that mirrors the BI-RADS lexicon. With short interpretation times, a fast learning curve for radiologists, and a substantially lower cost than breast MRI, MBI provides many benefits in the practices in which it is utilized. This review will discuss the current state of MBI technology, clinical applications of MBI, MBI interpretation, radiation dose associated with MBI, and the future of MBI.
Collapse
Affiliation(s)
- Katie N Hunt
- Mayo Clinic, Department of Radiology, Rochester, MN, USA
| |
Collapse
|
45
|
Romeo V, Accardo G, Perillo T, Basso L, Garbino N, Nicolai E, Maurea S, Salvatore M. Assessment and Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer: A Comparison of Imaging Modalities and Future Perspectives. Cancers (Basel) 2021; 13:3521. [PMID: 34298733 PMCID: PMC8303777 DOI: 10.3390/cancers13143521] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) is becoming the standard of care for locally advanced breast cancer, aiming to reduce tumor size before surgery. Unfortunately, less than 30% of patients generally achieve a pathological complete response and approximately 5% of patients show disease progression while receiving NAC. Accurate assessment of the response to NAC is crucial for subsequent surgical planning. Furthermore, early prediction of tumor response could avoid patients being overtreated with useless chemotherapy sections, which are not free from side effects and psychological implications. In this review, we first analyze and compare the accuracy of conventional and advanced imaging techniques as well as discuss the application of artificial intelligence tools in the assessment of tumor response after NAC. Thereafter, the role of advanced imaging techniques, such as MRI, nuclear medicine, and new hybrid PET/MRI imaging in the prediction of the response to NAC is described in the second part of the review. Finally, future perspectives in NAC response prediction, represented by AI applications, are discussed.
Collapse
Affiliation(s)
- Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (T.P.); (S.M.)
| | - Giuseppe Accardo
- Department of Breast Surgery, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, 85028 Potenza, Italy;
| | - Teresa Perillo
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (T.P.); (S.M.)
| | - Luca Basso
- IRCCS SDN, 80143 Naples, Italy; (L.B.); (N.G.); (E.N.); (M.S.)
| | - Nunzia Garbino
- IRCCS SDN, 80143 Naples, Italy; (L.B.); (N.G.); (E.N.); (M.S.)
| | | | - Simone Maurea
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (T.P.); (S.M.)
| | - Marco Salvatore
- IRCCS SDN, 80143 Naples, Italy; (L.B.); (N.G.); (E.N.); (M.S.)
| |
Collapse
|
46
|
Hatzipanagiotou ME, Huber D, Gerthofer V, Hetterich M, Ripoll BR, Ortmann O, Seitz S. Feasibility of ABUS as an Alternative to Handheld Ultrasound for Response Control in Neoadjuvant Breast Cancer Treatment. Clin Breast Cancer 2021; 22:e142-e146. [PMID: 34219020 DOI: 10.1016/j.clbc.2021.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 11/03/2022]
Abstract
INTRODUCTION The Invenia Automated Breast Ultrasound Screening (ABUS) is indicated as an adjunct to mammography for breast cancer screening in asymptomatic women with high-density breast tissue. ABUS provides time-efficient evaluation of the 3-dimensional recordings within 3 to 6 minutes. The role and advantages of ABUS in everyday clinical practice, especially in routine examination during neoadjuvant chemotherapy (NACT), is not clear. The aim of this monocentric, noninterventional retrospective study is to evaluate the use of ABUS in patients who are under NACT treatment for response control. METHODS Regular sonographic response check with handheld ultrasound (HHUS) examination and with ABUS were conducted in 83 women who underwent NACT. The response controls were conducted every 3 to 6 weeks during NACT. The handheld sonography was performed with GE Voluson S8. Handheld sonographic measurements and ABUS measurements were compared with the final pathologic tumor size. RESULTS There was no statistical difference between the measurements with HHUS examination or ABUS compared with final pathologic tumor size (P = .47). The average difference from ABUS measured tumor size to final pathologic tumor size was 9.8 mm. The average difference from handheld measured tumor size to final pathologic tumor size was 9/3 mm. Both the specificity of ABUS and HHUS examination in predicting pathologic complete remission was 100%. CONCLUSION ABUS seems to be a suitable method to conduct response control in neoadjuvant breast cancer treatment. ABUS may facilitate preoperative planning and offers remarkable time saving for physicians compared with HHUS examination and thus should be considered for clinical practice.
Collapse
Affiliation(s)
| | - Deborah Huber
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| | - Valeria Gerthofer
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| | - Madeleine Hetterich
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| | - Blanca Roca Ripoll
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| | - Olaf Ortmann
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| | - Stephan Seitz
- University Medical Centre Regensburg, Department of Gynecology and Obstetrics, 93053 Regensburg, Germany
| |
Collapse
|
47
|
Prognostic value of response evaluation based on breast MRI after neoadjuvant treatment: a retrospective cohort study. Eur Radiol 2021; 31:9520-9528. [PMID: 34036420 DOI: 10.1007/s00330-021-08042-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/18/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate the impact of response evaluation after neoadjuvant chemotherapy (NAC) in breast cancer patients, assessed by both magnetic resonance imaging (MRI) and pathology, on disease-free survival (DFS). METHODS This single-center, retrospective cohort study included consecutive breast cancer patients who underwent NAC and preoperative breast MRI. Resolution of invasive carcinoma in the breast and axilla was defined as complete pathological response (pCR). Radiological complete response (rCR) was defined as the absence of abnormal enhancement in the tumor site. Kaplan-Meier estimator was used to estimate the disease-free survival on 60 months. Cox regression analysis was used to estimate hazard ratio (HR) values. RESULTS In total, 317 patients were included with a mean age of 47.3 years and a mean tumor size of 39.8 mm. The most common immunophenotype was luminal (44.9%), followed by triple-negative (26.8%). Overall, 126 patients (39.7%) had an rCR, while 119 (37.5%) had pCR; the radiological and pathological responses agreed in 252 cases (79.5%). During follow-up, patients who had rCR and pCR had a better DFS curve compared to patients with non-rCR and non-pCR, while those who had rCR or pCR presented an intermediate curve (Log-rank p = 0.003). Multivariate analysis showed a higher risk of recurrence in patients with non-rCR and non-pCR (HR: 5,626; p = 0.020) and those who had a complete response on MRI or pathology only (HR: 4,369; p = 0.067), when compared to patients with rCR and pCR. CONCLUSIONS The association of MRI and pathological responses after NAC might better stratify the risk of recurrence and prognosis in breast cancer patients. KEY POINTS • Association of response evaluation after neoadjuvant chemotherapy by pathology and MRI allows better stratification of prognosis. • Complete response to neoadjuvant chemotherapy on pathology and MRI was related to better disease-free survival. • Complete response on MRI or pathology only had a greater risk of recurrence.
Collapse
|
48
|
Azhar Y, Agustina H, Hernowo BS. Primary Systemic Therapy for HER2/Neu-Positive Operable Breast Cancer Increases the Number of Breast-Conserving Surgery and Disease-Free Survival: Retrospective Cohort Analysis at Single Institution. ASIAN JOURNAL OF ONCOLOGY 2021. [DOI: 10.1055/s-0041-1729481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Abstract
Objective The aim of this study was to evaluate the efficacy and cardiotoxicity profile, and to reduce the extend of breast cancer surgery in primary systemic therapy (PST) HER2/neu–positive operable breast cancer patients.
Materials and Methods A total of 152 patients diagnosed from 2010 to 2015 were included in the study. The PST consisted of a sequential regimen of taxanes and anthracyclines plus trastuzumab. The clinical and pathological responses and the type of breast cancer surgery were evaluated and correlated with clinical and biological factors. The cardiotoxicity profile and long-term benefits were analyzed.
Results The median patient age was 47 (37–67) years, with T2 and T3 67 (44.1%) and 85 (55.9%), respectively. Axillary lymph node breast cancer at diagnosis N0 was 104 (68.4%) and N1 and N2 were 28.9% and 2.6%, respectively. A total of 95.7% of patients had nonspecific type of breast cancer, 67% of tumors were hormonal receptor–negative, 75.5% were grade III, 100% Ki67 > 20%, and 90% of tumors were confirmed to be HER2/neu–positive through immunohistochemistry. Following PST, pathological complete response (pCR) rate was achieved in 44.7% evaluable patients. The pCR rate was higher in HR-negative (93.1% vs. 6.9%) cancer and in grade III (86.2%) than in grade I and II (13.8%) cancer; only 75.5% of complete response (CR) on ultrasound and magnetic resonance imaging were also CR on pathology results. Breast conserving surgery was performed in 41.4%. Regarding type of chemotherapy, there were no significant differences between chemotherapy with anthracycline backbone or taxanes to achieved pathological complete response. Despite that, we were unable to demonstrate an association between pCR and better DFS with p = 0.096; HR 5.7 95.0% CI (0.73–45.52). Patients who are hormonal receptor positive tend to have lower disease-free survival (DFS) than those who are hormonal receptor negative; HR = 6.34, 95.0% CI (1.54–26.00) and p = 0.010. Five years DFS was higher for those who achieved pCR compare with those who did not. Even in this research we failed to show it is statistically significant.
Conclusion A sequential regimen of taxanes and anthracyclines plus trastuzumab was effective with high pCR rates and increases the possibility to do breast conservation surgery and had tolerable cardiotoxicity profile.
Collapse
Affiliation(s)
| | | | - Bethy S Hernowo
- Department of Pathology Anatomy, Hasan Sadikin General Hospital/Faculty of Medicine Universitas Padjadjaran, Bandung West Java, Indonesia
| |
Collapse
|
49
|
Nakashima K, Uematsu T, Harada TL, Takahashi K, Nishimura S, Tadokoro Y, Hayashi T, Watanabe J, Sugino T, Notsu A. Can breast MRI and adjunctive Doppler ultrasound improve the accuracy of predicting pathological complete response after neoadjuvant chemotherapy? Breast Cancer 2021; 28:1120-1130. [PMID: 33837896 DOI: 10.1007/s12282-021-01249-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 04/06/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND To examine the accuracy of MRI and Doppler ultrasound (US) for detecting residual tumor after neoadjuvant chemotherapy (NAC) for breast cancer and evaluate whether adjunctive Doppler US improves the MRI accuracy. METHODS We reviewed 276 invasive breast cancer cases treated with NAC. Tumors were classified into four subtypes based on estrogen receptor and HER2 status. Response to NAC was evaluated using contrast-enhanced MRI and Doppler US. Residual Doppler flow was assumed to indicate a residual tumor. MRI and Doppler findings were compared with the histopathological findings of resected specimens. Pathological complete response (pCR) was defined as neither in situ nor invasive cancer left. RESULTS Of the 276 tumors, imaging complete responses were observed using MRI and Doppler US in 62 (22%) and 111 (40%), respectively, whereas pCR was achieved in 44 (16%). MRI and Doppler US predicted residual tumor with 88% and 69% sensitivity, 80% and 91% specificity, 87% and 73% accuracy, 96% and 98% PPV, and 56% and 36% NPV, respectively. The accuracies of MRI and Doppler US were significantly higher for HER2-negative than HER2-positive tumors (p < 0.001 and p = 0.043, respectively). Seven (26%) of 27 false-negative cases identified by MRI were correctly diagnosed as positives with adjunctive Doppler US. CONCLUSIONS Although MRI accurately detected residual tumor with 87% accuracy, this was still not sufficient to meet clinical demands and differed with tumor subtype. Adjunctive Doppler US in cases that appear to show a complete response on MRI might reduce chances of false negatives and increase the NPV of MRI for predicting residual tumor.
Collapse
Affiliation(s)
- Kazuaki Nakashima
- Division of Breast Imaging and Breast Interventional Radiology, Shizuoka Cancer Center Hospital, Nagaizumi, Shizuoka, 411-8777, Japan.
| | - Takayoshi Uematsu
- Division of Breast Imaging and Breast Interventional Radiology, Shizuoka Cancer Center Hospital, Nagaizumi, Shizuoka, 411-8777, Japan
| | - Taiyo L Harada
- Division of Breast Imaging and Breast Interventional Radiology, Shizuoka Cancer Center Hospital, Nagaizumi, Shizuoka, 411-8777, Japan
| | - Kaoru Takahashi
- Division of Breast Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | | | - Yukiko Tadokoro
- Division of Breast Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Tomomi Hayashi
- Division of Breast Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Junichiro Watanabe
- Division of Breast Oncology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Akifumi Notsu
- Clinical Research Promotion Unit, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| |
Collapse
|
50
|
Huang Y, Le J, Miao A, Zhi W, Wang F, Chen Y, Zhou S, Chang C. Prediction of treatment responses to neoadjuvant chemotherapy in breast cancer using contrast-enhanced ultrasound. Gland Surg 2021; 10:1280-1290. [PMID: 33968680 DOI: 10.21037/gs-20-836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Elucidation the efficacy of neoadjuvant chemotherapy (NAC) in breast cancer is important for informing therapeutic decisions. This study aimed at evaluating the potential value of contrast-enhanced ultrasound (CEUS) parameters in predicting breast cancer responses to NAC. Methods We performed CEUS examinations before and after two cycles of NAC. Quantitative CEUS parameters [maximum intensity (IMAX), rise time (RT), time to peak (TTP), and mean transit time (mTT)], tumor diameter, and their changes were measured and compared to histopathological responses, according to the Miller-Payne Grading (MPG) system (score 1, 2, or 3: minor response; score 4 or 5: good response). Prediction models for good response were developed by multiple logistic regression analysis and internally validated through bootstrap analysis. The receiver operating characteristic (ROC) curve was used to evaluate the performance of prediction models. Results A total of 143 patients were enrolled in this study among whom 98 (68.5%) achieved a good response and while 45 (31.5%) exhibited a minor response. Several imaging variables including diameter, IMAX, changes in diameter (Δdiameter), IMAX (ΔIMAX) and TTP (ΔTTP) were found to be significantly associated with good therapeutic responses (P<0.05). The areas under the curve (AUC) increased from 0.748 to 0.841 in the multivariate model that combined CEUS parameters and molecular subtypes with a sensitivity and specificity of 0.786, 0.745, respectively. Tumor molecular subtype was the primary predictor of primary endpoint. Conclusions CEUS is a potential tool for predicting responses to NAC in locally advanced breast cancer patients. Compared to the other molecular subtypes, triple negative and HER2+/ER- subtypes are more likely to exhibit a good response to NAC.
Collapse
Affiliation(s)
- Yunxia Huang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Le
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Aiyu Miao
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxiang Zhi
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fen Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaling Chen
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shichong Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai Chang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|