1
|
Papakonstantinou A, Foukakis T. Neoadjuvant treatment of HER2-positive breast cancer: Has the era of antibody-drug conjugates arrived? Ann Oncol 2025:S0923-7534(25)00133-4. [PMID: 40194621 DOI: 10.1016/j.annonc.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Affiliation(s)
- A Papakonstantinou
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - T Foukakis
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Hennessy MA, Cimino-Mathews A, Carter JM, Kachergus JM, Ma Y, Leal JP, Solnes LB, Denbow R, Abramson VG, Carey LA, Rimawi M, Specht J, Storniolo AM, Valero V, Vaklavas C, Winer EP, Krop IE, Wolff AC, Wahl RL, Perez EA, Huang CY, Stearns V, Thompson EA, Connolly RM. Multiplex Spatial Proteomic Analysis of HER2-Positive Breast Tumors Reveals Unique Molecular and Immunologic Features Associated With Treatment Response. JCO Precis Oncol 2025; 9:e2400546. [PMID: 40179327 PMCID: PMC11968088 DOI: 10.1200/po-24-00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/17/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE Predictive biomarkers to better tailor therapy for patients with early-stage human epidermal growth factor 2 (HER2)-positive breast cancer are a priority. We hypothesized that HER2 and immune-based biomarkers would be predictive of pathologic complete response (pCR) to preoperative trastuzumab/pertuzumab (HP). MATERIALS AND METHODS Patients with stage II/III, estrogen receptor (ER)-negative, HER2-positive breast cancer received neoadjuvant HP in the TBCRC026 clinical trial. The pCR after receiving HP alone was 22% (18/83). Tumor biopsies were performed at baseline. Secondary correlative objectives were to determine the relationship between HER2-based biomarkers and immune processes with pCR. NanoString code sets BC360 and IO360 were used to compare differential gene expression in baseline tumors that underwent pCR versus no pCR. NanoString GeoMx digital spatial profiling was used to assess immune protein abundance in intraepithelial and stromal segments. Stromal tumor-infiltrating lymphocytes and Ki67 were evaluated by hematoxylin and eosin and immunohistochemistry, respectively. RESULTS Intraepithelial HER2 protein abundance was significantly associated with pCR (P = .001). Low HER2 abundance tumors were primarily basal-like, and essentially all (19/20) failed to achieve pCR. High HER2 abundance tumors that achieved pCR (14/51) exhibited a high degree of immune cell activity, whereas high HER2 abundance tumors that failed to achieve pCR tumors (37/51) were enriched for M-phase processes and epidermal growth factor receptor signaling. Baseline Ki67 was significantly higher in nonresponders (P = .04). CONCLUSION ER-negative, HER2-positive breast cancer has unique molecular and immunologic features that may predict pCR after neoadjuvant HP. Validation of these potential biomarkers and composite biomarker analyses may guide design of future clinical trials.
Collapse
Affiliation(s)
| | - Ashley Cimino-Mathews
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Yaohua Ma
- Department of Quantitative Health Science, Mayo Clinic Florida, Jacksonville, FL
| | - Jeffrey P. Leal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Lilja B. Solnes
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Rita Denbow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | - Anna Maria Storniolo
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN
| | - Vicente Valero
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Antonio C. Wolff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | - Edith A. Perez
- Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, FL
| | - Chiung-Yu Huang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
- University of California, San Francisco, CA
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | | | - Roisin M. Connolly
- Cancer Research @UCC, University College Cork, Cork, Ireland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
3
|
Hennessy MA, Cimino-Mathews A, Carter JM, Kachergus JM, Ma Y, Leal JP, Solnes LB, Abramson VG, Carey LA, Rimawi M, Specht J, Storniolo AM, Vaklavas C, Krop I, Winer E, Denbow R, Valero V, Wolff AC, Wahl RL, Huang CY, Stearns V, Thompson EA, Connolly RM. A composite 18F-FDG PET/CT and HER2 tissue-based biomarker to predict response to neoadjuvant pertuzumab and trastuzumab in HER2-positive breast cancer (TBCRC026). Breast 2025; 81:104432. [PMID: 40049115 PMCID: PMC11928837 DOI: 10.1016/j.breast.2025.104432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/10/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Early metabolic change on PET/CT was predictive of response to neoadjuvant trastuzumab/pertuzumab (HP) in TBCRC026. We hypothesized that a composite biomarker incorporating PET/CT and HER2 tissue-based biomarkers could improve biomarker performance. METHODS 83 patients with estrogen receptor-negative/HER2-positive breast cancer received neoadjuvant HP alone [pathologic complete response (pCR) 22 %]. PET/CT was performed at baseline and 15 days post initiation of therapy (C1D15). Promising imaging biomarkers included ≥40 % SULmax decline between baseline and C1D15, and C1D15 SULmax ≤3. Baseline tissue-based biomarkers included HER2-enriched intrinsic subtype (72 %, 46/64; NanoString), tumor HER2 protein abundance (median log2 13.5, range log2 7.1-15.9; NanoString DSP), and HER2 3+ (83 %, 64/77; immunohistochemistry). Logistic regressions were fitted to predict pCR with HER2/PET-CT biomarkers. The C statistic assessed overall prediction power. The optimal composite score cut-off was determined by maximizing Youden's index. RESULTS Factors most predictive for pCR in single predictor models included C1D15 SULmax (OR 0.43; p = 0.007, c = 0.77), % reduction in SULmax (OR 1.03, p = 0.006, c = 0.72) and tumor HER2 protein abundance (OR 1.75; p = 0.01, c = 0.76). The composite of C1D15 SULmax and % reduction in SULmax and their interaction term, had improved probability (c = 0.89 from c = 0.78), with high sensitivity (100 %) and negative predictive value (100 %). The addition of tumor HER2 protein did not further improve prediction power (c = 0.90). CONCLUSION The HER2/PET-CT biomarker had high prediction power for pCR, however was not superior to the prediction power of PET/CT alone. Non-invasive PET/CT biomarkers may facilitate a response-guided approach to neoadjuvant therapy, allowing intensification and de-intensification of treatment, pending further evaluation.
Collapse
Affiliation(s)
| | - Ashley Cimino-Mathews
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Yaohua Ma
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, FL, USA
| | - Jeffrey P Leal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lilja B Solnes
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Lisa A Carey
- University of North Carolina, Chapel Hill, NC, USA
| | | | | | - Anna Maria Storniolo
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
| | | | - Ian Krop
- Yale Cancer Center, New Haven, CT, USA
| | | | - Rita Denbow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Antonio C Wolff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Chiung-Yu Huang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA; University of California, San Francisco, CA, USA
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Roisin M Connolly
- Cancer Research @UCC, University College Cork, Ireland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Liu X, Eriksson Bergman L, Boman C, Foukakis T, Matikas A. Long-term outcome for neoadjuvant versus adjuvant chemotherapy in early breast cancer and the prognostic impact of nodal therapy response: A population-based study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109587. [PMID: 39794172 DOI: 10.1016/j.ejso.2025.109587] [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: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
INTRODUCTION Although neoadjuvant systemic treatment for non-metastatic breast cancer has gained ground during the past decade, there is no compelling evidence that it improves overall survival compared to primary tumor resection and adjuvant treatment. At the same time, the approach to responders to neoadjuvant treatment in the axilla is evolving. MATERIALS AND METHODS This is a retrospective analysis of a prospectively collected population-based registry. Patients that received neoadjuvant (n = 2126) or adjuvant chemotherapy (n = 4754) for non-metastatic breast cancer during 2007-2020 in the Stockholm-Gotland region, which comprises 25 % of the entire Swedish population, were included. Overall survival of patients treated preoperatively and postoperatively was compared using inverse probability treatment weighting and landmark analysis. The prognostic impact of change between prechemotherapy clinical to postchemotherapy pathologic nodal stage (cN/pN) in women receiving neoadjuvant treatment was investigated. RESULTS Median follow-up was 4.93 years. There was no difference in adjusted overall survival between adjuvant (reference) and neoadjuvant treatment in the entire population (HR = 1.38, 95 % CI 0.98-1.93, p = 0.062) or in breast cancer subtypes. Patients converting from positive clinical to negative pathologic nodal stage (cN+/pN0) had improved outcomes compared to cN0/pN0 or patients with pN0 following primary surgery. These patients had a particular disease trajectory, with early peak in risk of death followed by quick and sustained decrease. CONCLUSION There was no difference in survival of patients treated with neoadjuvant versus adjuvant systemic therapy for non-metastatic breast cancer. Patients with cN+/pN0 have excellent prognosis and represent potential candidates for de-escalation of local and systemic treatment.
Collapse
Affiliation(s)
- Xingrong Liu
- Karolinska Institutet, Oncology/Pathology Department, Stockholm, Sweden
| | - Louise Eriksson Bergman
- Karolinska Institutet, Oncology/Pathology Department, Stockholm, Sweden; Department of Surgery and Oncology, Capio Sankt Göran Hospital, Stockholm, Sweden
| | - Caroline Boman
- Karolinska Institutet, Oncology/Pathology Department, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Theodoros Foukakis
- Karolinska Institutet, Oncology/Pathology Department, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Alexios Matikas
- Karolinska Institutet, Oncology/Pathology Department, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center, Stockholm, Sweden.
| |
Collapse
|
5
|
Liu S, Yu M, Mou E, Wang M, Liu S, Xia L, Li H, Tang H, Feng Y, Yu X, Mi K, Wang H. The optimal neoadjuvant treatment strategy for HR+/HER2 + breast cancer: a network meta-analysis. Sci Rep 2025; 15:713. [PMID: 39753653 PMCID: PMC11699132 DOI: 10.1038/s41598-024-84039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
The efficacy of neoadjuvant therapy varies significantly with hormone receptor (HR) status for patients with human epidermal growth factor receptor 2 (HER2) positive breast cancer (BC). Despite extensive research on HER2 + BC, the optimal neoadjuvant strategy for HR+/HER2 + BC remains inconclusive. This study aimed to identify the optimal neoadjuvant regimen for HR+/HER2 + BC treatment. We conducted a systematic search for trials comparing neoadjuvant regimens for HR+/HER2 + BC and a network meta-analysis. Odds ratios for pathological complete response (pCR) and hazard ratios for event-free survival (EFS) were calculated. Treatment regimens were ranked using the surface under the cumulative ranking curve. 20 trials with 2809 patients were included. In pCR analysis, three neoadjuvant regimens sequentially ranked at the top, namely those comprising T-DM1, pertuzumab with trastuzumab, and tyrosine kinase inhibitor with trastuzumab, demonstrating significantly higher pCR rates than monotherapies. In EFS analysis, pertuzumab with trastuzumab ranked the first while T-DM1 containing regimen ranked the last. Anthracycline-free regimens showed a marginally higher pCR rate than anthracycline-containing regimens, while carboplatin-containing regimens demonstrated a numerically higher pCR rate than carboplatin-free regimens. Significant heterogeneity was observed in endocrine therapy analysis, which may be caused by different strategies for incorporating endocrine therapy. In conclusion, trastuzumab plus pertuzumab stands out as the optimal neoadjuvant HER2-targeting regimen for HR+/HER2 + BC Furthermore, anthracycline-free carboplatin-containing chemotherapy emerges as a promising combination treatment. Further investigation is required to clarify the role of endocrine therapy in HR+/HER2 + BC to guide its clinical application.
Collapse
Affiliation(s)
- Shiwei Liu
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Miao Yu
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Exian Mou
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Meihua Wang
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuanghua Liu
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Xia
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Li
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Tang
- Shanghai Roche Pharmaceuticals Ltd, Shanghai, China
| | - Yajing Feng
- Shanghai Roche Pharmaceuticals Ltd, Shanghai, China
| | - Xin Yu
- Shanghai Roche Pharmaceuticals Ltd, Shanghai, China
| | - Kun Mi
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Hao Wang
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
6
|
Vidal JM, Tsiknakis N, Staaf J, Bosch A, Ehinger A, Nimeus E, Salgado R, Bai Y, Rimm DL, Hartman J, Acs B. The analytical and clinical validity of AI algorithms to score TILs in TNBC: can we use different machine learning models interchangeably? EClinicalMedicine 2024; 78:102928. [PMID: 39634035 PMCID: PMC11615110 DOI: 10.1016/j.eclinm.2024.102928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
Background Pathologist-read tumor-infiltrating lymphocytes (TILs) have showcased their predictive and prognostic potential for early and metastatic triple-negative breast cancer (TNBC) but it is still subject to variability. Artificial intelligence (AI) is a promising approach toward eliminating variability and objectively automating TILs assessment. However, demonstrating robust analytical and prognostic validity is the key challenge currently preventing their integration into clinical workflows. Methods We evaluated the impact of ten AI models on TILs scoring, emphasizing their distinctions in TILs analytical and prognostic validity. Several AI-based TILs scoring models (seven developed and three previously validated AI models) were tested in a retrospective analytical cohort and in an independent prospective cohort to compare prognostic validation against invasive disease-free survival endpoint with 4 years median follow-up. The development and analytical validity set consisted of diagnostic tissue slides of 79 women with surgically resected primary invasive TNBC tumors diagnosed between 2012 and 2016 from the Yale School of Medicine. An independent set comprising of 215 TNBC patients from Sweden diagnosed between 2010 and 2015, was used for testing prognostic validity. Findings A significant difference in analytical validity (Spearman's r = 0.63-0.73, p < 0.001) is highlighted across AI methodologies and training strategies. Interestingly, the prognostic performance of digital TILs is demonstrated for eight out of ten AI models, even less extensively trained ones, with similar and overlapping hazard ratios (HR) in the external validation cohort (Cox regression analysis based on IDFS-endpoint, HR = 0.40-0.47; p < 0.004). Interpretation The demonstrated prognostic validity for most of the AI TIL models can be attributed to the intrinsic robustness of host anti-tumor immunity (measured by TILs) as a biomarker. However, the discrepancies between AI models should not be overlooked; rather, we believe that there is a critical need for an accessible, large, multi-centric dataset that will serve as a benchmark ensuring the comparability and reliability of different AI tools in clinical implementation. Funding Nikos Tsiknakis is supported by the Swedish Research Council (Grant Number 2021-03061, Theodoros Foukakis). Balazs Acs is supported by The Swedish Society for Medical Research (Svenska Sällskapet för Medicinsk Forskning) postdoctoral grant. Roberto Salgado is supported by a grant from Breast Cancer Research Foundation (BCRF).
Collapse
Affiliation(s)
- Joan Martínez Vidal
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Nikos Tsiknakis
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Ana Bosch
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Region Skåne, Lund, Sweden
| | - Anna Ehinger
- Department of Genetics, Pathology and Molecular Diagnostics, Laboratory Medicine, Region Skåne, Lund, Sweden
| | - Emma Nimeus
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Yalai Bai
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - David L. Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Balazs Acs
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
7
|
Yang Y, Zheng Y, Sun X, Zhao A, Wu Y. Antibody drug conjugate, a level-up version of monoclonal antibody? Int J Surg 2024; 110:5944-5948. [PMID: 38833359 PMCID: PMC11392205 DOI: 10.1097/js9.0000000000001748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 05/19/2024] [Indexed: 06/06/2024]
Affiliation(s)
- Yuqi Yang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center,West China Hospital, Sichuan University
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yue Zheng
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center,West China Hospital, Sichuan University
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University
| | - Xu Sun
- Department of Hematology West China Hospital, Sichuan University
| | - Ailin Zhao
- Department of Hematology West China Hospital, Sichuan University
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center,West China Hospital, Sichuan University
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University
| |
Collapse
|
8
|
Yang Y, Wang J, Ren Q, Yu R, Yuan Z, Jiang Q, Guan S, Tang X, Duan T, Meng X. Multimodal data integration using machine learning to predict the risk of clear cell renal cancer metastasis: a retrospective multicentre study. Abdom Radiol (NY) 2024; 49:2311-2324. [PMID: 38879708 DOI: 10.1007/s00261-024-04418-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE To develop and validate a predictive combined model for metastasis in patients with clear cell renal cell carcinoma (ccRCC) by integrating multimodal data. MATERIALS AND METHODS In this retrospective study, the clinical and imaging data (CT and ultrasound) of patients with ccRCC confirmed by pathology from three tertiary hospitals in different regions were collected from January 2013 to January 2023. We developed three models, including a clinical model, a radiomics model, and a combined model. The performance of the model was determined based on its discriminative power and clinical utility. The evaluation indicators included area under the receiver operating characteristic curve (AUC) value, accuracy, sensitivity, specificity, negative predictive value, positive predictive value and decision curve analysis (DCA) curve. RESULTS A total of 251 patients were evaluated. Patients (n = 166) from Shandong University Qilu Hospital (Jinan) were divided into the training cohort, of which 50 patients developed metastases; patients (n = 37) from Shandong University Qilu Hospital (Qingdao) were used as internal testing, of which 15 patients developed metastases; patients (n = 48) from Changzhou Second People's Hospital were used as external testing, of which 13 patients developed metastases. In the training set, the combined model showed the highest performance (AUC, 0.924) in predicting lymph node metastasis (LNM), while the clinical and radiomics models both had AUCs of 0.845 and 0.870, respectively. In the internal testing, the combined model had the highest performance (AUC, 0.877) for predicting LNM, while the AUCs of the clinical and radiomics models were 0.726 and 0.836, respectively. In the external testing, the combined model had the highest performance (AUC, 0.849) for predicting LNM, while the AUCs of the clinical and radiomics models were 0.708 and 0.804, respectively. The DCA curve showed that the combined model had a significant prediction probability in predicting the risk of LNM in ccRCC patients compared with the clinical model or the radiomics model. CONCLUSION The combined model was superior to the clinical and radiomics models in predicting LNM in ccRCC patients.
Collapse
Affiliation(s)
- YouChang Yang
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - JiaJia Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - QingGuo Ren
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Rong Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - ZiYi Yuan
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - QingJun Jiang
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Shuai Guan
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - XiaoQiang Tang
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - TongTong Duan
- Department of Ultrasound, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - XiangShui Meng
- Department of Radiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China.
| |
Collapse
|
9
|
Helgadottir H, Matikas A, Fernebro J, Frödin JE, Ekman S, Rodriguez-Wallberg KA. Fertility and reproductive concerns related to the new generation of cancer drugs and the clinical implication for young individuals undergoing treatments for solid tumors. Eur J Cancer 2024; 202:114010. [PMID: 38520926 DOI: 10.1016/j.ejca.2024.114010] [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: 01/19/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
The treatment landscape of solid tumors has changed markedly in the last years. Molecularly targeted treatments and immunotherapies have been implemented and have, in many cancers, lowered the risk of relapse and prolonged survival. Patients with tumors harboring specific targetable molecular alterations or mutations are often of a younger age, and hence future fertility and family building can be important concerns in this group. However, there are great uncertainties regarding the effect of the new drugs on reproductive functions, including fertility, pregnancy and lactation and how young patients with cancers, both women and men should be advised. The goal with this review is to gather the current knowledge regarding oncofertility and the different novel therapies, including immune checkpoint inhibitors, antibody-drug conjugates, small molecules and monoclonal antibody targeted therapies. The specific circumstances and reproductive concerns in different patient groups where novel treatments have been broadly introduced are also discussed, including those with melanoma, lung, breast, colorectal and gynecological cancers. It is clear, that more awareness is needed regarding potential drug toxicity on reproductive tissues, and it is of essence that individuals are informed based on current expertise and on available fertility preservation methods.
Collapse
Affiliation(s)
- Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Skin Cancer Centrum, Theme Cancer, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| | - Alexios Matikas
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gynecological Cancer, Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Jan-Erik Frödin
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gastrointestinal Oncology, Department of Upper abdomen, Karolinska University Hospital, Sweden
| | - Simon Ekman
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Pérez-García JM, Cortés J, Ruiz-Borrego M, Colleoni M, Stradella A, Bermejo B, Dalenc F, Escrivá-de-Romaní S, Calvo Martínez L, Ribelles N, Marmé F, Cortés A, Albacar C, Gebhart G, Prat A, Kerrou K, Schmid P, Braga S, Di Cosimo S, Gion M, Antonarelli G, Popa C, Szostak E, Alcalá-López D, Gener P, Rodríguez-Morató J, Mina L, Sampayo-Cordero M, Llombart-Cussac A. 3-year invasive disease-free survival with chemotherapy de-escalation using an 18F-FDG-PET-based, pathological complete response-adapted strategy in HER2-positive early breast cancer (PHERGain): a randomised, open-label, phase 2 trial. Lancet 2024; 403:1649-1659. [PMID: 38582092 DOI: 10.1016/s0140-6736(24)00054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND PHERGain was designed to assess the feasibility, safety, and efficacy of a chemotherapy-free treatment based on a dual human epidermal growth factor receptor 2 (HER2) blockade with trastuzumab and pertuzumab in patients with HER2-positive early breast cancer (EBC). It used an 18fluorine-fluorodeoxyglucose-PET-based, pathological complete response (pCR)-adapted strategy. METHODS PHERGain was a randomised, open-label, phase 2 trial that took place in 45 hospitals in seven European countries. It randomly allocated patients in a 1:4 ratio with centrally confirmed, HER2-positive, stage I-IIIA invasive, operable breast cancer with at least one PET-evaluable lesion to either group A, where patients received docetaxel (75 mg/m2, intravenous), carboplatin (area under the curve 6 mg/mL per min, intravenous), trastuzumab (600 mg fixed dose, subcutaneous), and pertuzumab (840 mg loading dose followed by 420 mg maintenance doses, intravenous; TCHP), or group B, where patients received trastuzumab and pertuzumab with or without endocrine therapy, every 3 weeks. Random allocation was stratified by hormone receptor status. Centrally reviewed PET was conducted at baseline and after two treatment cycles. Patients in group B were treated according to on-treatment PET results. Patients in group B who were PET-responders continued with trastuzumab and pertuzumab with or without endocrine therapy for six cycles, while PET-non-responders were switched to receive six cycles of TCHP. After surgery, patients in group B who were PET-responders who did not achieve a pCR received six cycles of TCHP, and all patients completed up to 18 cycles of trastuzumab and pertuzumab. The primary endpoints were pCR in patients who were group B PET-responders after two treatment cycles (the results for which have been reported previously) and 3-year invasive disease-free survival (iDFS) in patients in group B. The study is registered with ClinicalTrials.gov (NCT03161353) and is ongoing. FINDINGS Between June 26, 2017, and April 24, 2019, a total of 356 patients were randomly allocated (71 patients in group A and 285 patients in group B), and 63 (89%) and 267 (94%) patients proceeded to surgery in groups A and B, respectively. At this second analysis (data cutoff: Nov 4, 2022), the median duration of follow-up was 43·3 months (range 0·0-63·0). In group B, the 3-year iDFS rate was 94·8% (95% CI 91·4-97·1; p=0·001), meeting the primary endpoint. No new safety signals were identified. Treatment-related adverse events and serious adverse events (SAEs) were numerically higher in patients allocated to group A than to group B (grade ≥3 62% vs 33%; SAEs 28% vs 14%). Group B PET-responders with pCR presented the lowest incidence of treatment-related grade 3 or higher adverse events (1%) without any SAEs. INTERPRETATION Among HER2-positive EBC patients, a PET-based, pCR-adapted strategy was associated with an excellent 3-year iDFS. This strategy identified about a third of patients who had HER2-positive EBC who could safely omit chemotherapy. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- José Manuel Pérez-García
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona 08022, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona 08022, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | | | - Agostina Stradella
- Medical Oncology Department, Institut Català D'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Begoña Bermejo
- Medical Oncology, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, Spain; Medicine Department, Universidad de Valencia, Spain; Oncology Biomedical Research National Network (CIBERONC-ISCIII), Madrid, Spain
| | - Florence Dalenc
- Oncopole Claudius Regaud- IUCT, Inserm, Department of Medical Oncology, Toulouse, France
| | - Santiago Escrivá-de-Romaní
- Medical Oncology Department, Breast Cancer Group, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Nuria Ribelles
- UGC Oncología Intercentros, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Málaga, Spain
| | - Frederik Marmé
- University Hospital Mannheim; Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Cinta Albacar
- Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Geraldine Gebhart
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies Group, IDIBAPS, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Khaldoun Kerrou
- APHP, Tenon Hospital IUC-UPMC, Nuclear Medicine and PET Center Department, Sorbonne University, Paris, France; INSERM U938 (Cancer Biology and Therapeutics), Paris, France
| | - Peter Schmid
- Barts Experimental Cancer Medicine Centre, Barts Cancer Institute, Queen Mary University of London, UK; Barts Hospital NHS Trust, London, UK
| | - Sofia Braga
- Unidade de Mama, Instituto CUF de Oncologia, Lisbon, Portugal; NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Gion
- University Hospital Ramón y Cajal, Madrid, Spain
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Crina Popa
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Emilia Szostak
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | | | - Petra Gener
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | | | - Leonardo Mina
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | | | - Antonio Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Hospital Arnau de Vilanova, Universidad Católica de Valencia, Valencia, Spain.
| |
Collapse
|
11
|
Zhu Y, Zerdes I, Matikas A, Cruz IR, Bergqvist M, Elinder E, Bosch A, Lindman H, Einbeigi Z, Andersson A, Carlsson L, Dreifaldt AC, Isaksson-Friman E, Hellstrom M, Johansson H, Wang K, Bergh JCS, Hatschek T, Foukakis T. The role of serum thymidine kinase 1 activity in neoadjuvant-treated HER2-positive breast cancer: biomarker analysis from the Swedish phase II randomized PREDIX HER2 trial. Breast Cancer Res Treat 2024; 204:299-308. [PMID: 38175448 PMCID: PMC10948570 DOI: 10.1007/s10549-023-07200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Thymidine kinase 1 (TK1) plays a pivotal role in DNA synthesis and cellular proliferation. TK1 has been studied as a prognostic marker and as an early indicator of treatment response in human epidermal growth factor 2 (HER2)-negative early and metastatic breast cancer (BC). However, the prognostic and predictive value of serial TK1 activity in HER2-positive BC remains unknown. METHODS In the PREDIX HER2 trial, 197 HER2-positive BC patients were randomized to neoadjuvant trastuzumab, pertuzumab, and docetaxel (DPH) or trastuzumab emtansine (T-DM1), followed by surgery and adjuvant epirubicin and cyclophosphamide. Serum samples were prospectively collected from all participants at multiple timepoints: at baseline, after cycle 1, 2, 4, and 6, at end of adjuvant therapy, annually for a total period of 5 years and/or at the time of recurrence. The associations of sTK1 activity with baseline characteristics, pathologic complete response (pCR), event-free survival (EFS), and disease-free survival (DFS) were evaluated. RESULTS No association was detected between baseline sTK1 levels and all the baseline clinicopathologic characteristics. An increase of TK1 activity from baseline to cycle 2 was seen in all cases. sTK1 level at baseline, after 2 and 4 cycles was not associated with pCR status. After a median follow-up of 58 months, 23 patients had EFS events. There was no significant effect between baseline or cycle 2 sTK1 activity and time to event. A non-significant trend was noted among patents with residual disease (non-pCR) and high sTK1 activity at the end of treatment visit, indicating a potentially worse long-term prognosis. CONCLUSION sTK1 activity increased following neoadjuvant therapy for HER2-positive BC but was not associated with patient outcomes or treatment benefit. However, the post-surgery prognostic value in patients that have not attained pCR warrants further investigation. TRIAL REGISTRATION ClinicalTrials.gov, NCT02568839. Registered on 6 October 2015.
Collapse
Affiliation(s)
- Yajing Zhu
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden.
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Ivette Raices Cruz
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Ana Bosch
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Henrik Lindman
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Southern Älvsborg Hospital, Borås, Sweden
| | | | - Lena Carlsson
- Department of Oncology, Sundsvall Hospital, Sundsvall, Sweden
| | | | | | - Mats Hellstrom
- Centre for Clinical Cancer Studies, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Hemming Johansson
- Centre for Clinical Cancer Studies, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kang Wang
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
| | - Jonas C S Bergh
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Hatschek
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska Vägen A2:07, Solna, 171 64, Stockholm, Sweden
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Loibl S, André F, Bachelot T, Barrios CH, Bergh J, Burstein HJ, Cardoso MJ, Carey LA, Dawood S, Del Mastro L, Denkert C, Fallenberg EM, Francis PA, Gamal-Eldin H, Gelmon K, Geyer CE, Gnant M, Guarneri V, Gupta S, Kim SB, Krug D, Martin M, Meattini I, Morrow M, Janni W, Paluch-Shimon S, Partridge A, Poortmans P, Pusztai L, Regan MM, Sparano J, Spanic T, Swain S, Tjulandin S, Toi M, Trapani D, Tutt A, Xu B, Curigliano G, Harbeck N. Early breast cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2024; 35:159-182. [PMID: 38101773 DOI: 10.1016/j.annonc.2023.11.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 147.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Affiliation(s)
- S Loibl
- GBG Forschungs GmbH, Neu-Isenburg; Centre for Haematology and Oncology, Bethanien, Frankfurt, Germany
| | - F André
- Breast Cancer Unit, Medical Oncology Department, Gustave Roussy, Cancer Campus, Villejuif
| | - T Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - C H Barrios
- Oncology Department, Latin American Cooperative Oncology Group and Oncoclínicas, Porto Alegre, Brazil
| | - J Bergh
- Department of Oncology-Pathology, Bioclinicum, Karolinska Institutet and Breast Cancer Centre, Karolinska Comprehensive Cancer Centre and University Hospital, Stockholm, Sweden
| | - H J Burstein
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M J Cardoso
- Breast Unit, Champalimaud Foundation, Champalimaud Cancer Centre, Lisbon; Faculty of Medicine, Lisbon University, Lisbon, Portugal
| | - L A Carey
- Division of Medical Oncology, The University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, USA
| | - S Dawood
- Department of Oncology, Mediclinic City Hospital, Dubai, UAE
| | - L Del Mastro
- Medical Oncology Clinic, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Internal Medicine and Medical Specialities, School of Medicine, University of Genoa, Genoa, Italy
| | - C Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Giessen and Marburg, Marburg
| | - E M Fallenberg
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - P A Francis
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - H Gamal-Eldin
- Department of Surgical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - K Gelmon
- Department of Medical Oncology, British Columbia Cancer, Vancouver, Canada
| | - C E Geyer
- Department of Internal Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA
| | - M Gnant
- Comprehensive Cancer Centre, Medical University of Vienna, Vienna, Austria
| | - V Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova; Oncology 2 Unit, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - S Gupta
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - S B Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - D Krug
- Department of Radiation Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - M Martin
- Hospital General Universitario Gregorio Maranon, Universidad Complutense, GEICAM, Madrid, Spain
| | - I Meattini
- Department of Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence; Department of Experimental and Clinical Biomedical Sciences 'M. Serio', University of Florence, Florence, Italy
| | - M Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - W Janni
- Department of Obstetrics and Gynaecology, University of Ulm, Ulm, Germany
| | - S Paluch-Shimon
- Sharett Institute of Oncology Department, Hadassah University Hospital & Faculty of Medicine Hebrew University, Jerusalem, Israel
| | - A Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - P Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - L Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven
| | - M M Regan
- Division of Biostatistics, Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - J Sparano
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - S Swain
- Medicine Department, Georgetown University Medical Centre and MedStar Health, Washington, USA
| | - S Tjulandin
- N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - M Toi
- Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Bunkyo-ku, Japan
| | - D Trapani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - A Tutt
- Breast Cancer Research Division, The Institute of Cancer Research, London; Comprehensive Cancer Centre, Division of Cancer Studies, Kings College London, London, UK
| | - B Xu
- Department of Medical Oncology, National Cancer Centre/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - G Curigliano
- Early Drug Development for Innovative Therapies Division, Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - N Harbeck
- Breast Centre, Department of Obstetrics & Gynaecology and Comprehensive Cancer Centre Munich, LMU University Hospital, Munich, Germany
| |
Collapse
|
13
|
Filis P, Zerdes I, Soumala T, Matikas A, Foukakis T. The ever-expanding landscape of antibody-drug conjugates (ADCs) in solid tumors: A systematic review. Crit Rev Oncol Hematol 2023; 192:104189. [PMID: 37866413 DOI: 10.1016/j.critrevonc.2023.104189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The advent of targeted therapies signaled novel avenues for more optimal oncological outcomes. Antibody-drug conjugates (ADCs) have risen as a cornerstone of the ever-expanding targeted therapy era. The purpose of this systematic review is to delineate the rapidly evolving clinical landscape of ADCs for solid tumors. METHODS A literature search was performed in Medline, Embase and Cochrane databases for phase II and III clinical trials. Outcomes of interest were the objective response rate, overall survival, progression-free survival and adverse events. RESULTS A total of 92 clinical trials (76 phase II and 16 phase III) evaluated the efficacy and safety of ADCs for a plethora of solid tumors. Out of the 30 investigated ADCs, 8 have received approval by regulatory organizations for solid tumors. Currently, 52 phase III clinical trials for ADCs are ongoing. CONCLUSION ADCs have shown promising results for several solid tumors and various cancer settings.
Collapse
Affiliation(s)
- Panagiotis Filis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece.
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodora Soumala
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Hennessy MA, Leal JP, Huang CY, Solnes LB, Denbow R, Abramson VG, Carey LA, Liu MC, Rimawi M, Specht J, Storniolo AM, Valero V, Vaklavas C, Winer EP, Krop IE, Wolff AC, Cimino-Mathews A, Wahl RL, Stearns V, Connolly RM. Correlation of SUV on Early Interim PET with Recurrence-Free Survival and Overall Survival in Primary Operable HER2-Positive Breast Cancer (the TBCRC026 Trial). J Nucl Med 2023; 64:1690-1696. [PMID: 37652539 DOI: 10.2967/jnumed.123.265853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/06/2023] [Indexed: 09/02/2023] Open
Abstract
Predictive biomarkers of response to human epidermal growth factor receptor 2 (HER2)-directed therapy are essential to inform treatment decisions. The TBCRC026 trial reported that early declines in tumor SUVs corrected for lean body mass (SULmax) on 18F-FDG PET/CT predicted a pathologic complete response (pCR) to HER2 therapy with neoadjuvant trastuzumab and pertuzumab (HP) without chemotherapy in estrogen receptor (ER)-negative, HER2-positive breast cancer. We hypothesized that 18F-FDG PET/CT SULmax parameters would predict recurrence-free survival (RFS) and overall survival (OS). Methods: Patients with stage II/III ER-negative, HER2-positive breast cancer received neoadjuvant HP (n = 88). pCR after HP alone was 22% (18/83), additional nonstudy neoadjuvant therapy was administered in 28% (25/88), and the majority received adjuvant therapy per physician discretion. 18F-FDG PET/CT was performed at baseline and at cycle 1, day 15 (C1D15). RFS and OS were summarized using the Kaplan-Meier method and compared between subgroups using logrank tests. Associations between 18F-FDG PET/CT (≥40% decline in SULmax between baseline and C1D15, or C1D15 SULmax ≤ 3) and pCR were evaluated using Cox regressions, where likelihood ratio CIs were reported because of the small numbers of events. Results: Median follow-up was 53.7 mo (83/88 evaluable), with 6 deaths and 14 RFS events. Estimated RFS and OS at 3 y was 84% (95% CI, 76%-92%) and 92% (95% CI, 87%-98%), respectively. A C1D15 SULmax of 3 or less was associated with improved RFS (hazard ratio [HR], 0.36; 95% CI, 0.11-1.05; P = 0.06) and OS (HR, 0.14; 95% CI, 0.01-0.85; P = 0.03), the latter statistically significant. The association of an SULmax decline of at least 40% (achieved in 59%) with RFS and OS did not reach statistical significance. pCR was associated with improved RFS (HR, 0.25; 95% CI, 0.01-1.24; P = 0.10) but did not reach statistical significance. Conclusion: For the first time, we report a potential association between a C1D15 SULmax of 3 or less on 18F-FDG PET/CT and RFS and OS outcomes in patients with ER-negative, HER2-positive breast cancer receiving neoadjuvant HP alone. If confirmed in future studies, this imaging-based biomarker may facilitate early individualization of therapy.
Collapse
Affiliation(s)
| | - Jeffrey P Leal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| | - Chiung-Yu Huang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
- University of California, San Francisco, California
| | - Lilja B Solnes
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| | - Rita Denbow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| | | | - Lisa A Carey
- University of North Carolina, Chapel Hill, North Carolina
| | | | | | | | - Anna Maria Storniolo
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana
| | | | | | | | - Ian E Krop
- Yale Cancer Center, New Haven, Connecticut; and
| | - Antonio C Wolff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| | | | | | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| | - Roisin M Connolly
- Cancer Research @UCC, Cork, Ireland;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland
| |
Collapse
|
15
|
Gebhart G. 18F-FDG PET "Metabolic Response" to Neoadjuvant Systemic Therapy for Breast Cancer: Quo Vadis? J Nucl Med 2023; 64:1697-1698. [PMID: 37918851 DOI: 10.2967/jnumed.123.266303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Indexed: 11/04/2023] Open
Affiliation(s)
- Geraldine Gebhart
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
16
|
Wittwer NL, Brown MP, Liapis V, Staudacher AH. Antibody drug conjugates: hitting the mark in pancreatic cancer? J Exp Clin Cancer Res 2023; 42:280. [PMID: 37880707 PMCID: PMC10598980 DOI: 10.1186/s13046-023-02868-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Pancreatic cancer is one of the most common causes of cancer-related death, and the 5-year survival rate has only improved marginally over the last decade. Late detection of the disease means that in most cases the disease has advanced locally and/or metastasized, and curative surgery is not possible. Chemotherapy is still the first-line treatment however, this has only had a modest impact in improving survival, with associated toxicities. Therefore, there is an urgent need for targeted approaches to better treat pancreatic cancer, while minimizing treatment-induced side-effects. Antibody drug conjugates (ADCs) are one treatment option that could fill this gap. Here, a monoclonal antibody is used to deliver extremely potent drugs directly to the tumor site to improve on-target killing while reducing off-target toxicity. In this paper, we review the current literature for ADC targets that have been examined in vivo for treating pancreatic cancer, summarize current and on-going clinical trials using ADCs to treat pancreatic cancer and discuss potential strategies to improve their therapeutic window.
Collapse
Affiliation(s)
- Nicole L Wittwer
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia.
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| |
Collapse
|
17
|
Villacampa G, Matikas A, Oliveira M, Prat A, Pascual T, Papakonstantinou A. Landscape of neoadjuvant therapy in HER2-positive breast cancer: a systematic review and network meta-analysis. Eur J Cancer 2023:S0959-8049(23)00188-0. [PMID: 37142539 DOI: 10.1016/j.ejca.2023.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND The recommended preoperative approach for HER2-positive breast cancer is unclear. We aimed to investigate the following: i) what is the optimal neoadjuvant regimen and ii) whether anthracyclines could be excluded. METHODS A systematic literature search in Medline, Embase and Web of Science databases was performed. Studies had to satisfy the following criteria: i) randomised controlled trials (RCTs), ii) enroled patients treated preoperatively for HER2-positive BC (breast cancer), iii) at least one treatment group received an anti-HER2 agent, iv) available information of any efficacy end-point and v) published in English. A network meta-analysis with a frequentist framework using random-effects model was used to pool direct and indirect evidence. Pathologic complete response (pCR), event-free survival (EFS) and overall survival (OS) were the efficacy end-points of interest, and selected safety end-points were also analysed. RESULTS A total of 11,049 patients with HER2-positive BC (46 RCTs) were included in the network meta-analysis, and 32 different regimens were evaluated. Dual anti-HER2-therapy, with pertuzumab or tyrosine kinase inhibitors, combined with chemotherapy was significantly superior to trastuzumab and chemotherapy in terms of pCR, EFS and OS. However, a higher risk of cardiotoxicity was observed with dual anti-HER2-therapy. Anthracycline-based chemotherapy was not associated with better efficacy outcomes in comparison with non-anthracycline-based chemotherapy. In anthracycline-free regimens, the addition of carboplatin presented numerically better efficacy outcomes. CONCLUSION Dual HER2 blockade with chemotherapy is the recommended choice as neoadjuvant therapy for HER2-positive breast cancer, preferably by omitting anthracyclines in favour of carboplatin.
Collapse
Affiliation(s)
- Guillermo Villacampa
- SOLTI Breast Cancer Research Group, Spain; The Institute of Cancer Research, London, United Kingdom; Oncology Data Science, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Alexios Matikas
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Breast Cancer, Endocrine Tumours and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Mafalda Oliveira
- SOLTI Breast Cancer Research Group, Spain; Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Aleix Prat
- SOLTI Breast Cancer Research Group, Spain; Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain
| | - Tomás Pascual
- SOLTI Breast Cancer Research Group, Spain; Translational Genomics and Targeted Therapies in Solid Tumours, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Spain
| | - Andri Papakonstantinou
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Breast Cancer, Endocrine Tumours and Sarcoma, Karolinska University Hospital, Stockholm, Sweden; Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
18
|
Huang Y, Zhu T, Zhang X, Li W, Zheng X, Cheng M, Ji F, Zhang L, Yang C, Wu Z, Ye G, Lin Y, Wang K. Longitudinal MRI-based fusion novel model predicts pathological complete response in breast cancer treated with neoadjuvant chemotherapy: a multicenter, retrospective study. EClinicalMedicine 2023; 58:101899. [PMID: 37007742 PMCID: PMC10050775 DOI: 10.1016/j.eclinm.2023.101899] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 04/04/2023] Open
Abstract
Background Accurate identification of pCR to neoadjuvant chemotherapy (NAC) is essential for determining appropriate surgery strategy and guiding resection extent in breast cancer. However, a non-invasive tool to predict pCR accurately is lacking. Our study aims to develop ensemble learning models using longitudinal multiparametric MRI to predict pCR in breast cancer. Methods From July 2015 to December 2021, we collected pre-NAC and post-NAC multiparametric MRI sequences per patient. We then extracted 14,676 radiomics and 4096 deep learning features and calculated additional delta-value features. In the primary cohort (n = 409), the inter-class correlation coefficient test, U-test, Boruta and the least absolute shrinkage and selection operator regression were used to select the most significant features for each subtype of breast cancer. Five machine learning classifiers were then developed to predict pCR accurately for each subtype. The ensemble learning strategy was used to integrate the single-modality models. The diagnostic performances of models were evaluated in the three external cohorts (n = 343, 170 and 340, respectively). Findings A total of 1262 patients with breast cancer from four centers were enrolled in this study, and pCR rates were 10.6% (52/491), 54.3% (323/595) and 37.5% (66/176) in HR+/HER2-, HER2+ and TNBC subtype, respectively. Finally, 20, 15 and 13 features were selected to construct the machine learning models in HR+/HER2-, HER2+ and TNBC subtypes, respectively. The multi-Layer Perception (MLP) yields the best diagnostic performances in all subtypes. For the three subtypes, the stacking model integrating pre-, post- and delta-models yielded the highest AUCs of 0.959, 0.974 and 0.958 in the primary cohort, and AUCs of 0.882-0.908, 0.896-0.929 and 0.837-0.901 in the external validation cohorts, respectively. The stacking model had accuracies of 85.0%-88.9%, sensitivities of 80.0%-86.3%, and specificities of 87.4%-91.5% in the external validation cohorts. Interpretation Our study established a novel tool to predict the responses of breast cancer to NAC and achieve excellent performance. The models could help to determine post-NAC surgery strategy for breast cancer. Funding This study is supported by grants from the National Natural Science Foundation of China (82171898, 82103093), the Deng Feng project of high-level hospital construction (DFJHBF202109), the Guangdong Basic and Applied Basic Research Foundation (grant number, 2020A1515010346, 2022A1515012277), the Science and Technology Planning Project of Guangzhou City (202002030236), the Beijing Medical Award Foundation (YXJL-2020-0941-0758), and the Beijing Science and Technology Innovation Medical Development Foundation (KC2022-ZZ-0091-5). Funding sources were not involved in the study design, data collection, analysis and interpretation, writing of the report, or decision to submit the article for publication.
Collapse
Affiliation(s)
- YuHong Huang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - XiaoLing Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Li
- Department of Breast Cancer, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - XingXing Zheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - MinYi Cheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - Fei Ji
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - LiuLu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - CiQiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - ZhiYong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
- Corresponding author. Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
| | - GuoLin Ye
- Department of Breast Cancer, The First People's Hospital of Foshan, Foshan, Guangdong, China
- Corresponding author. Department of Breast Cancer, The First People's Hospital of Foshan, Foshan, 528000, China.
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Corresponding author. Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
- Corresponding author. Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| |
Collapse
|