1
|
Raghavendra AS, Bassett R, Damodaran S, Barcenas CH, Mouabbi JA, Layman R, Tripathy D. Clinical Characteristics and Survival Outcomes of Metastatic Invasive Lobular and Ductal Carcinoma. JAMA Netw Open 2025; 8:e251888. [PMID: 40293750 PMCID: PMC12038507 DOI: 10.1001/jamanetworkopen.2025.1888] [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: 10/05/2024] [Accepted: 01/24/2025] [Indexed: 04/30/2025] Open
Abstract
Importance Comparing the clinical and molecular features of metastatic invasive lobular carcinoma (mILC) and metastatic invasive ductal carcinoma (mIDC) is essential to enhance understanding of breast cancer biology and improve personalized treatment approaches. Objective To compare mILC and mIDC in terms of survival outcomes and to investigate the association of clinicopathologic characteristics with those outcomes. Design, Setting, and Participants This cohort study included adult patients at the University of Texas MD Anderson Cancer Center with their first metastatic diagnosis occurring between January 1997 and December 2020. Patient records were obtained from an institutional database. The study follow-up concluded in July 2023, and data were analyzed from July to December 2024. Exposure Diagnosis of mIDC and mILC. Main Outcomes and Measures Progression-free survival (PFS), overall survival (OS), and disease-free interval (DFI) were estimated using the Kaplan-Meier method. Survival distributions were compared using the log-rank test. Multivariable Cox proportional hazards regression was used to assess the association of metastasis onset, estrogen receptor (ER) expression level, and tumor grade with OS and PFS. Results The analysis included a total of 9714 patients (9628 women [99%]), 8535 with mIDC and 1179 with mILC. The median age at metastasis was 53.3 years (range, 17.6-62.0 years). Generally, patients with mILC were older and had lower nuclear grade tumors and fewer metastasis sites than patients with mIDC. Patients with mILC had longer PFS (median, 0.65 years; 95% CI, 0.58-0.74) than patients with mIDC (median, 0.46 years; 95% CI, 0.45-0.48) (hazard ratio [HR], 0.78; 95% CI, 0.73-0.84; P < .001). For OS, patients with mILC had longer OS (median, 3.06 years; 95% CI, 2.87-3.29) than patients with mIDC (median, 2.60 years; 95% CI, 2.52-2.67 years) (HR, 0.91; 95% CI, 0.84-0.98; P = .01). Overall, patients with mILC had longer DFI than patients with mIDC (HR, 0.69; 95% CI, 0.64-0.75; P < .001). At initial diagnosis, patients with mILC were less likely to present with visceral metastasis (522 patients [44.3%]) than patients with mIDC (4909 patients [57.5%]). A higher proportion of patients with mILC (931 patients [79.0%]) than patients with mIDC (5431 patients [63.6%]) had bone-only metastasis. Conclusions and Relevance In this cohort study, patients with mILC had longer OS and PFS than those with mIDC. Metastasis onset, ER positivity, and tumor grade were associated with survival outcomes, and distinct metastatic patterns of mIDC and mILC were also associated with survival for mIDC and mILC, which may help guide more personalized treatment strategies for each subtype.
Collapse
Affiliation(s)
- Akshara S. Raghavendra
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Senthil Damodaran
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Carlos H. Barcenas
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jason A. Mouabbi
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Rachel Layman
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Debu Tripathy
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
2
|
Bullock E, Brunton VG. E-Cadherin-Mediated Cell-Cell Adhesion and Invasive Lobular Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:259-275. [PMID: 39821030 DOI: 10.1007/978-3-031-70875-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
E-cadherin is a transmembrane protein and central component of adherens junctions (AJs). The extracellular domain of E-cadherin forms homotypic interactions with E-cadherin on adjacent cells, facilitating the formation of cell-cell adhesions, known as AJs, between neighbouring cells. The intracellular domain of E-cadherin interacts with α-, β- and p120-catenins, linking the AJs to the actin cytoskeleton. Functional AJs maintain epithelial tissue identity and integrity. Transcriptional downregulation of E-cadherin is the first step in epithelial-to-mesenchymal transition (EMT), a process essential in development and tissue repair, which, in breast cancer, can contribute to tumour progression and metastasis. In addition, loss-of-function mutations in E-cadherin are a defining feature of invasive lobular breast cancer (also known as invasive lobular carcinoma (ILC)), the second most common histological subtype of breast cancer. ILC displays a discohesive, single-file invasive growth pattern due to the loss of functional AJs. Despite being so prevalent, until recently there has been limited ILC-focused research and historically ILC patients have often been excluded from clinical trials. Despite displaying a number of good prognostic indicators, such as low grade and high rates of estrogen receptor positivity, ILC patients tend to have similar or poorer outcomes relative to the most common subtype of breast cancer, invasive ductal carcinoma (IDC). In ILC, E-cadherin loss promotes hyperactivation of growth factor receptors, in particular insulin-like growth factor 1 receptor, anoikis resistance and synthetic lethality with ROS1 inhibition. These features introduce clinical vulnerabilities that could potentially be exploited to improve outcomes for ILC patients, for whom there are currently limited tailored treatments available.
Collapse
Affiliation(s)
- Esme Bullock
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK
| | - Valerie G Brunton
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Zhang W, Huang Y, Zhou Y, Xue J, Gao S, Kang L, Shi J, Zhou T, Duan Y, Guo S, Li Q. Delving into female breast cancer: Distinct disease-specific survival outcomes between invasive lobular and ductal carcinomas revealed by propensity score matching. PLoS One 2024; 19:e0300116. [PMID: 39715246 DOI: 10.1371/journal.pone.0300116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 02/22/2024] [Indexed: 12/25/2024] Open
Abstract
PURPOSE The difference in prognosis between invasive lobular carcinoma (ILC) and invasive ductal carcinoma (IDC) is still controversial in the academic community. Resolving this controversy can help to more accurately determine patients' prognosis, provide further personalized treatment, alleviate unnecessary psychological burden for some patients, and provide direction for further fundamental research. PATIENTS AND METHODS A retrospective cohort study was conducted using the SEER Research Plus Data 8 Registries, Nov 2021 sub (1978-2019), including female breast cancer patients diagnosed with ILC or IDC between 2010 and 2015. Univariate and multivariate Cox regression analyses were performed, and key covariates affecting prognosis were selected. Propensity score matching (PSM) was employed to match patients, and balance tests were conducted to evaluate covariate distribution. Disease-specific survival (DSS) differences between the matched IDC and ILC groups were compared. RESULTS Following PSM, the covariate differences between the IDC and ILC groups were significantly reduced. The survival analysis revealed a significantly better prognosis for the IDC group than the ILC group (Log-rank test p < 0.001), with a 28.0% increased risk observed in the ILC group. CONCLUSION This study provides evidence supporting the existence of significant differences in prognosis between IDC and ILC patients after rigorous matching. The IDC group displayed a significantly better prognosis than the ILC group. Notably, these findings have implications for personalized treatment in clinical practice and contribute to the ongoing academic debate on survival differences between IDC and ILC. However, further research is needed to investigate the biological mechanisms, gene expression, and signaling pathway disparities between IDC and ILC, aiming to provide more targeted guidance for clinical decision-making.
Collapse
Affiliation(s)
- Wu Zhang
- Graduate School, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yuquan Huang
- Guilin Medical University, Guilin, Guangxi, China
| | - Ye Zhou
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Jiaojiao Xue
- College of Postgraduate, Hebei Medical University, Shijiazhuang, China
| | - Shan Gao
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, China
| | - Lin Kang
- Department of Pathology, Hebei General Hospital, Shijiazhuang, China
| | - Jian Shi
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Zhou
- Department of Breast Cancer Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yalong Duan
- School of Graduate Studies, Hebei North University, Zhangjiakou, China
| | - Sihan Guo
- Department of Computer Science, Durham University, Durham, United Kingdom
| | - Qingxia Li
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
4
|
van Ommen-Nijhof A, Steenbruggen TG, Wiersma TG, Balduzzi S, Daletzakis A, Holtkamp MJ, Delfos M, Schot M, Beelen K, Siemerink EJM, Heijns J, Mandjes IA, Wesseling J, Rosenberg EH, Vrancken Peeters MJT, Linn SC, Sonke GS. Intensified alkylating chemotherapy for patients with oligometastatic breast cancer harboring homologous recombination deficiency: Primary outcomes from the randomized phase III OLIGO study. Eur J Cancer 2024; 213:115083. [PMID: 39489924 DOI: 10.1016/j.ejca.2024.115083] [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: 07/05/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Oligometastatic breast cancer (OMBC) is a clinical entity with a prospect of long-term survival, but uncertainty remains on its optimal treatment. We studied whether intensified alkylating chemotherapy (IACT) improves long-term outcome compared to conventional-dose chemotherapy (CDCT) as part of a multimodality approach for patients with OMBC harboring homologous recombination deficiency (HRD). PATIENTS AND METHODS Eligible patients had HER2-negative OMBC, harboring HRD, with ≤ 3 distant metastases, pathologic proof of distant disease and a favorable response to three cycles CDCT. Participants were randomized 1:1 to continue with either CDCT or IACT. IACT consisted of one mobilization course followed by two cycles of mini-CTC (carboplatin, thiotepa and cyclophosphamide) supported by peripheral blood progenitor cell reinfusion. Primary outcome was event-free survival (EFS). Secondary endpoints included overall survival (OS), quality of life and safety. RESULTS Seventy-five patients were randomized to either IACT (n = 36) or CDCT (n = 39). Twenty-three (31 %) patients had hormone receptor-positive disease and 52 (69 %) had triple-negative disease. Median EFS in the IACT-group was 28 months (95 % confidence interval [CI] 21-not reached [NR]) versus 25 months (95 %CI 14-NR) in the CDCT-group (hazard ratio [HR] for recurrence or death 0.78, 95 %CI 0.42-1.42). Median OS was 67 months (95 %CI 37-NR) in the IACT-group and 36 (95 %CI 26-NR) in the CDCT-group (HR 0.74, 95 %CI 0.37-1.48). CONCLUSIONS The entire study population experienced long-term survival, with median OS well over five years. IACT compared to CDCT did not improve outcome in patients with OMBC harboring study-defined HRD. The optimal therapy for patients with OMBC requires further study. TRIAL REGISTRATION ClinicalTrials.gov: NCT01646034.
Collapse
Affiliation(s)
| | - T G Steenbruggen
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands; Department of internal medicine, St Antonius hospital, Nieuwegein, the Netherlands
| | - T G Wiersma
- Department of radiotherapy, NKI-AVL, Amsterdam, the Netherlands
| | - S Balduzzi
- Department of biometrics, NKI-AVL, Amsterdam, the Netherlands
| | - A Daletzakis
- Department of biometrics, NKI-AVL, Amsterdam, the Netherlands
| | - M J Holtkamp
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands
| | - M Delfos
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands
| | - M Schot
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands
| | - K Beelen
- Department of internal medicine, Rijnstate, Arnhem, the Netherlands
| | - E J M Siemerink
- Department of internal medicine, Ziekenhuisgroep Twente, Almelo, Hengelo, the Netherlands
| | - J Heijns
- Department of internal medicine, Amphia, Breda, the Netherlands
| | - I A Mandjes
- Department of biometrics, NKI-AVL, Amsterdam, the Netherlands
| | - J Wesseling
- Department of pathology, NKI-AVL, Amsterdam, the Netherlands; Department of pathology, LUMC, Leiden, the Netherlands
| | - E H Rosenberg
- Department of pathology, NKI-AVL, Amsterdam, the Netherlands
| | - M J T Vrancken Peeters
- Department of surgical oncology, NKI-AVL, Amsterdam, the Netherlands; Department of surgical oncology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - S C Linn
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands; Department of pathology, Utrecht University, Utrecht, the Netherlands
| | - G S Sonke
- Department of medical oncology, NKI-AVL, Amsterdam, the Netherlands; Department of medical oncology, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Gilardi L, Airò Farulla LS, Bonatto E, Ceci F. Novel PET tracers in breast cancer for treatment optimization: clinical utility and future perspectives. Curr Opin Oncol 2024; 36:514-520. [PMID: 39011728 DOI: 10.1097/cco.0000000000001057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW There is a critical need for timely and accurate decisions in breast cancer management. This narrative review aims to clarify the potential role of novel PET tracers in optimizing treatment strategies for different breast cancer subtypes. RECENT FINDINGS 2-deoxy-2-[18F]-fluoro-D-glucose PET/computed tomography (FDG PET/CT) has a pivotal role in response assessment in metastatic breast cancer, despite its limitations in certain histological and molecular subtypes. Further PET radiopharmaceuticals have been proposed to address these clinical needing. SUMMARY FES PET/CT demonstrates its usefulness in assessing ER expression and predicting response to therapy in luminal breast cancer, with implications for treatment optimization and monitoring. In HER2-positive and HER2-low breast cancer, HER2-targeted PET tracers show potential in assessing HER2 status, while their accuracy in predicting response to targeted therapies is still debated. PARP-targeted PET imaging holds potential for selecting patients for PARP inhibitors treatments, particularly in triple-negative breast cancer (TNBC), where imaging tools are crucial due to the absence of specific targets. Immunotherapy and antibody-drug conjugates (ADCs) are emerging treatment options for TNBC, and PET imaging targeting immune checkpoints could aid in treatment selection and response monitoring. The dynamic role of PET/CT imaging in tailoring breast cancer treatments requires further multidisciplinary research to validate the clinical utility of targeted tracers.
Collapse
Affiliation(s)
- Laura Gilardi
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
| | - Lighea Simona Airò Farulla
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Bonatto
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Agostinetto E, Bruzzone M, Hamy AS, Kim HJ, Chiodi C, Bernstein-Molho R, Linn S, Pogoda K, Carrasco E, Derouane F, Bajpai J, Nader-Marta G, Lopetegui-Lia N, Partridge AH, Cortesi L, Rousset-Jablonski C, Giugliano F, Renaud T, Ferrari A, Paluch-Shimon S, Fruscio R, Cui W, Wong SM, Vernieri C, Ruddy KJ, Dieci MV, Matikas A, Rozenblit M, Aguilar Y Mendez D, De Marchis L, Borea R, Puglisi F, Pistelli M, Kufel-Grabowska J, Di Rocco R, Mariamidze E, Atzori F, Kourie HR, Popovic L, de Azambuja E, Blondeaux E, Lambertini M. Characteristics and clinical outcomes of breast cancer in young BRCA carriers according to tumor histology. ESMO Open 2024; 9:103714. [PMID: 39288653 PMCID: PMC11421331 DOI: 10.1016/j.esmoop.2024.103714] [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: 03/22/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Young women with breast cancer (BC) have an increased chance of carrying germline BRCA pathogenic variants (PVs). Limited data exist on the prognostic impact of tumor histology (i.e. ductal versus lobular) in hereditary breast cancer. METHODS This multicenter retrospective cohort study included women aged ≤40 years with early-stage breast cancer diagnosed between January 2000 and December 2020 and known to carry germline PVs in BRCA1/2. Histology was locally assessed in each center. The Kaplan-Meier method and Cox regression analysis were used to assess disease-free survival and overall survival. RESULTS Of 4628 patients included from 78 centers worldwide, 3969 (86%) had pure ductal, 135 (3%) pure lobular, and 524 (11%) other histologies. Compared with ductal tumors, lobular tumors were more often grade 1/2 (57.7% versus 22.1%), stage III (29.6% versus 18.5%), and luminal A-like (42.2% versus 12.2%). Lobular tumors were more often associated with BRCA2 PVs (71.1% BRCA2), while ductal tumors were more often associated with BRCA1 PVs (65.7% BRCA1). Patients with lobular tumors more often had mastectomy (68.9% versus 58.3%), and less often received chemotherapy (83.7% versus 92.9%). With a median follow-up of 7.8 years, no significant differences were observed in disease-free survival (adjusted hazard ratio 1.01, 95% confidence interval 0.74-1.37) or overall survival (hazard ratio 0.96, 95% confidence interval 0.62-1.50) between patients with ductal versus lobular tumors. No significant survival differences were observed according to specific BRCA gene, breast cancer subtype, or body mass index. CONCLUSIONS In this large global cohort of young BRCA carriers with breast cancer, the incidence of pure lobular histology was low and associated with higher disease stage at diagnosis, luminal-like disease and BRCA2 PVs. Histology did not appear to impact prognosis.
Collapse
Affiliation(s)
- E Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium.
| | - M Bruzzone
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - A-S Hamy
- Department of Medical Oncology, Universite Paris Cité, Institut Curie, Paris, France
| | - H J Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - C Chiodi
- Cancer Survivorship Program - Molecular Predictors and New Targets in Oncology, INSERM Unit 981, Gustave Roussy, Villejuif, France
| | - R Bernstein-Molho
- Susanne Levy Gertner Oncogenetics Unit, The Danek Gertner Institute of Human Genetics, Chaim Sheba Medical Center Affiliated to Tel Aviv University, Tel Hashomer, Israel
| | - S Linn
- Department of Molecular Pathology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - K Pogoda
- Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - E Carrasco
- Hereditary Cancer Genetics Unit, Medical Oncology Department, Vall d´Hebron University Hospital, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - F Derouane
- Department of General Medical Oncology and Multidisciplinary Breast Center, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - J Bajpai
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - G Nader-Marta
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - N Lopetegui-Lia
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland
| | - A H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - L Cortesi
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - C Rousset-Jablonski
- Department of Surgery, Leon Berard Cancer Center, Lyon; Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron; Unité INSERM U1290 RESHAPE, Lyon
| | - F Giugliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - T Renaud
- Cancer Genetics Unit, Bergonie Institute, Bordeaux, France
| | - A Ferrari
- Hereditary Breast and Ovarian Cancer (HBOC) Unit and General Surgery 3 - Senology, Surgical Department, Fondazione IRCCS Policlinico San Matteo, Pavia; University of Pavia, Pavia, Italy
| | - S Paluch-Shimon
- Breast Oncology Unit, Sharett Institute of Oncology, Hadassah University Hospital, Jerusalem; Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - R Fruscio
- UO Gynecology, Fondazione IRCCS San Gerardo, Monza; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - W Cui
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria; Department of Medical Oncology, Peter MacCallum Cancer Center, Melbourne, Australia
| | - S M Wong
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal; McGill University Medical School, Montreal, Canada
| | - C Vernieri
- Medical Oncology Department, Breast Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan; Oncology and Hematology-Oncology Department, University of Milan, Milan, Italy
| | - K J Ruddy
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, USA
| | - M V Dieci
- Department of Surgical, Oncological and Gastroenterological Sciences, Università di Padova, Padua; Oncologia 2, Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - A Matikas
- Department of Oncology/Pathology, Karolinska Institute, Stockholm; Breast Center, Karolinska University Hospital, Stockholm, Sweden
| | - M Rozenblit
- Yale University, Medical Oncology, New Haven, USA
| | - D Aguilar Y Mendez
- Tecnologico de Monterrey, Breast Cancer Center, Hospital Zambrano Hellion - TecSalud, Monterrey, Mexico
| | - L De Marchis
- Division of Medical Oncology, Department of Radiological, Oncological and Pathological Sciences, 'La Sapienza' University of Rome, Rome; Oncology Unit, Umberto 1 University Hospital, Rome
| | - R Borea
- Medical Oncology Department, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genoa
| | - F Puglisi
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano; Department of Medicine, University of Udine, Udine
| | - M Pistelli
- Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | | | - R Di Rocco
- Department of Medical Oncology, Azienda USL Toscana Sud Est - Misericordia Hospital, Grosseto, Italy
| | - E Mariamidze
- Medical Oncology Department, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Oncology and Hematology, Todua Clinic, Tbilisi, Georgia
| | - F Atzori
- Azienda Ospedaliero Universitaria di Cagliari, Cagliari, Italy
| | - H R Kourie
- Saint Joseph University of Beirut, Beirut, Lebanon
| | - L Popovic
- Oncology Institute of Vojvodina - Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - E de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - E Blondeaux
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - M Lambertini
- Medical Oncology Department, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genoa
| |
Collapse
|
7
|
Ndlovu H, Lawal IO, Mokoala KMG, Sathekge MM. Imaging Molecular Targets and Metabolic Pathways in Breast Cancer for Improved Clinical Management: Current Practice and Future Perspectives. Int J Mol Sci 2024; 25:1575. [PMID: 38338854 PMCID: PMC10855575 DOI: 10.3390/ijms25031575] [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: 12/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Breast cancer is the most frequently diagnosed cancer and leading cause of cancer-related deaths worldwide. Timely decision-making that enables implementation of the most appropriate therapy or therapies is essential for achieving the best clinical outcomes in breast cancer. While clinicopathologic characteristics and immunohistochemistry have traditionally been used in decision-making, these clinical and laboratory parameters may be difficult to ascertain or be equivocal due to tumor heterogeneity. Tumor heterogeneity is described as a phenomenon characterized by spatial or temporal phenotypic variations in tumor characteristics. Spatial variations occur within tumor lesions or between lesions at a single time point while temporal variations are seen as tumor lesions evolve with time. Due to limitations associated with immunohistochemistry (which requires invasive biopsies), whole-body molecular imaging tools such as standard-of-care [18F]FDG and [18F]FES PET/CT are indispensable in addressing this conundrum. Despite their proven utility, these standard-of-care imaging methods are often unable to image a myriad of other molecular pathways associated with breast cancer. This has stimulated interest in the development of novel radiopharmaceuticals targeting other molecular pathways and processes. In this review, we discuss validated and potential roles of these standard-of-care and novel molecular approaches. These approaches' relationships with patient clinicopathologic and immunohistochemical characteristics as well as their influence on patient management will be discussed in greater detail. This paper will also introduce and discuss the potential utility of novel PARP inhibitor-based radiopharmaceuticals as non-invasive biomarkers of PARP expression/upregulation.
Collapse
Affiliation(s)
- Honest Ndlovu
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| | - Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Kgomotso M. G. Mokoala
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| | - Mike M. Sathekge
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| |
Collapse
|
8
|
Li C, Yuan Q, Deng T, Xu G, Hou J, Zheng L, Wu G. Prognosis difference between HER2-low and HER2-zero breast cancer patients: a systematic review and meta-analysis. Breast Cancer 2023; 30:965-975. [PMID: 37470943 DOI: 10.1007/s12282-023-01487-w] [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: 04/10/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND HER2-low breast cancer (BC) is proposed to be a special population of patients with an immunohistochemistry (IHC) score of 1 + or 2 + and non-amplified in situ hybridization (ISH) results. The role and prognostic impact of HER2-low BC is still controversial. This meta-analysis aims to explore the prognostic difference between of HER2-low and HER2-zero characteristic in BC patients. METHODS A meta-analysis was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and eligible studies were search in PubMed, Web of Science and EMBASE databases. Quality assessment of included studies were performed by Quality in Prognostic Studies (QUIPS) tool. Hazard ratios (HRs) and corresponding 95% confidence interval (CI) for overall survival (OS) and disease-free survival (DFS) were pooled in a meta-analysis. Furthermore, subgroup analysis, sensitivity analysis, and analysis for publication bias were conducted. RESULTS Eighteen studies comprising a total of 93,317 patients were included for meta-analysis. BC patients with HER2-low characteristic have longer OS (HRs 0.87, 95% CI 0.81-0.93, p < 0.0001) and DFS (HRs 0.82, 95% CI 0.73-0.93, p = 0.001) compared to those with HER2-zero characteristic. Subgroup analysis indicate that the source of heterogeneity may come from the hormone receptor (HR) status group. Although, the publication bias was detected, sensitivity analysis and the trim-and-fill method analysis demonstrated the stability and reliability of the results. CONCLUSION HER2-low BC patients have longer OS and DFS compared to HER2-zero BC patients, and its prognostic value is consistent among different HR status patients. Whether HER2-low breast cancer is an independent subtype of breast cancer is still a subject of ongoing research, and more studies are needed to fully understand the molecular and clinical features of this subtype.
Collapse
Affiliation(s)
- Chengxin Li
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Qianqian Yuan
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Tong Deng
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Gaoran Xu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Jinxuan Hou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Lewei Zheng
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Abstract
Breast carcinomas classified based on traditional morphologic assessment provide useful prognostic information. Although morphology is still the gold standard of classification, recent advances in molecular technologies have enabled the classification of these tumors into four distinct subtypes based on its intrinsic molecular profile that provide both predictive and prognostic information. This article describes the association between the different molecular subtypes with the histologic subtypes of breast cancer and illustrates how these subtypes may affect the appearance of tumors on imaging studies.
Collapse
Affiliation(s)
- Madhuchhanda Roy
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, B1761 WIMR, 1111 Highland Avenue, Madison, WI 53705, USA.
| | - Amy M Fowler
- Department of Radiology, Section of Breast Imaging and Intervention, University of Wisconsin - Madison, 600 Highland Avenue, Madison, WI 53792-3252, USA; Department of Medical Physics, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI 53792-3252, USA
| | - Gary A Ulaner
- Hoag Family Cancer Institute, 16105 Sand Canyon Avenue, Ste 215, Irvine, CA 92618, USA; Department of Radiology, Department of Translational Genomics, University of Southern California, Los Angeles, CA 90007, USA
| | - Aparna Mahajan
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, B1781 WIMR, 1111 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
10
|
Stålhammar G, Grossniklaus HE. Overrepresentation of human epidermal growth factor receptor 2 positive- and Luminal B breast cancer metastases in the eyes and orbit. Eye (Lond) 2023; 37:2499-2504. [PMID: 36517577 PMCID: PMC10397265 DOI: 10.1038/s41433-022-02363-1] [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: 07/18/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Breast cancer is the most common cancer to spread to the choroid and orbit. Depending on a set of prognostic and predictive biomarkers, breast cancer can be divided into at least four distinct subtypes with separate treatment and clinical course. SUBJECTS Thirty-two patients with metastases to the eye and periocular area diagnosed between 2005 and 2020, of which 11 also had primary tumour tissue available. Expression levels of oestrogen- (ER) and progesterone receptors (PR), Human epidermal growth factor receptor 2 (HER2) and the proliferation marker Ki67 were analysed. RESULTS Twenty-five of 32 patients (78%) had a history of primary breast cancer, whereas the remaining 7 (22%) presented with metastatic disease. Of available metastases, 83% were positive for ER, 37% for PR, 54% for HER2, and 50% for Ki67. Metastases had significantly lower proportions of PR-positive cells than primary tumours, and the distribution of the Luminal A, Luminal B, HER2 enriched and triple-negative subtypes differed between primary tumours and metastases (P = 0.012): Six of 9 patients with a full set of biomarkers on both primary tumours and metastases switched subtype (67%), and 23 of 32 metastases (77%) were of the Luminal B subtype. CONCLUSIONS Nearly 4 in 5 breast cancer metastases in the eyes and orbit are of the Luminal B subtype, and a majority are HER2 positive. The breast cancer subtype frequently switches between primary tumours and metastases. Future studies should evaluate these results in larger cohorts.
Collapse
Affiliation(s)
- Gustav Stålhammar
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
- St. Erik Eye Hospital, Stockholm, Sweden.
| | - Hans E Grossniklaus
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
11
|
Yap YS. Outcomes in breast cancer-does ethnicity matter? ESMO Open 2023; 8:101564. [PMID: 37290358 DOI: 10.1016/j.esmoop.2023.101564] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 06/10/2023] Open
Abstract
Ethnic or racial differences in breast cancer (BC) survival outcomes have been reported, but current data are largely restricted to comparisons between African Americans and non-Hispanic whites. Most analyses have traditionally been based on self-reported race which may not always be accurate, or are oversimplified in their classification. With increasing globalization, quantification of the genetic ancestry from genomic data may offer a solution to infer the complex makeup from admixture of races. Focusing on the larger and the latest studies, we will discuss recent findings on the differing host and tumor biology that may be driving these disparities, in addition to the extrinsic environmental or lifestyle factors. Socioeconomic disparities with lower cancer literacy may lead to late presentation, poorer adherence to treatment, and other lifestyle factors such as unhealthy diet, obesity, and inadequate physical activity. These hardships may also result in greater allostatic load, which is in turn associated with aggressive BC features in disadvantaged populations. Epigenetic reprogramming may mediate the effects of the environment or lifestyle factors on gene expression, with ensuing differences in BC characteristics and outcome. There is increasing evidence that germline genetics can influence somatic gene alterations or expression, as well as modulate the tumor or immune microenvironment. Although the precise mechanisms remain elusive, this may account for the varying distribution of different BC subtypes across ethnicities. These gaps in our knowledge highlight the need to interrogate the multiomics landscape of BC in diverse populations, ideally in large-scale collaborative settings with standardized methodology for the comparisons to be statistically robust. Together with improving BC awareness and access to good quality health care, a holistic approach with insights of the biological underpinnings is much needed to eradicate ethnic disparities in BC outcomes.
Collapse
Affiliation(s)
- Y-S Yap
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Oncology Academic Clinical Programme, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore.
| |
Collapse
|
12
|
Thill M, Zahn MO, Welt A, Stickeler E, Nusch A, Fietz T, Rauh J, Wetzel N, Kruggel L, Jänicke M, Marschner N, Harbeck N, Wöckel A, Decker T. Treatment and outcome in metastatic lobular breast cancer in the prospective German research platform OPAL. Breast Cancer Res Treat 2023; 198:545-553. [PMID: 36807725 PMCID: PMC10036409 DOI: 10.1007/s10549-023-06882-7] [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: 12/07/2022] [Accepted: 02/01/2023] [Indexed: 02/20/2023]
Abstract
PURPOSE Evidence about routine treatment and outcome of patients with invasive lobular cancer (ILC) is limited, especially regarding metastatic disease. Here we present prospective real-world data of patients with metastatic ILC (mILC) as compared to patients with metastatic invasive ductal cancer (mIDC) receiving systemic therapy in routine care in Germany. METHODS Prospective data on patient and tumor characteristics, treatments, and outcomes of patients with mILC (n = 466) and mIDC (n = 2100), recruited between 2007 and 2021 into the Tumor Registry Breast Cancer/OPAL were analyzed. RESULTS Compared to mIDCs, patients with mILC were older at start of first-line treatment (median 69 vs. 63 years) and had more often lower grade (G1/G2: 72.8% vs. 51.2%), hormone receptor (HR)-positive (83.7% vs. 73.2%) and less often HER2-positive (14.2% vs. 28.6%) tumors, which metastasized more frequently to the bone (19.7% vs. 14.5%) or peritoneum (9.9% vs. 2.0%), and less frequently to the lungs (0.9% vs. 4.0%). Median OS of patients with mILC (n = 209) and mIDC (n = 1158) was 30.2 months [95% confidence interval (CI) 25.3, 36.0] and 33.7 months [95% CI 30.3, 37.9], respectively. Multivariate survival analysis did not show a significant prognostic impact of the histological subtype [HR mILC vs. mIDC 1.18 (95% CI 0.97-1.42)]. CONCLUSION Overall, our real-world data confirm clinicopathological differences between mILC and mIDC breast cancer patients. Despite patients with mILC presenting with some favorable prognostic factors, ILC histopathology was not associated with a better clinical outcome in multivariate analysis, suggesting the need for more tailored treatment strategies for patients with the lobular subtype.
Collapse
Affiliation(s)
- M Thill
- Agaplesion Markus Krankenhaus, Frankfurt, Germany
| | - M-O Zahn
- MVZ Onkologische Kooperation Harz, Goslar, Germany
| | - A Welt
- Universitätsklinikum Essen, Essen, Germany
| | - E Stickeler
- Universitätsklinikum Aachen, Aachen, Germany
| | - A Nusch
- Praxis für Hämatologie und internistische Onkologie, Ratingen, Germany
| | - T Fietz
- Schwerpunktpraxis für Hämatologie und internistische Onkologie, Singen, Germany
| | - J Rauh
- Gemeinschaftspraxis Innere Medizin, Witten, Germany
| | | | | | | | - N Marschner
- Praxis für interdisziplinäre Onkologie und Hämatologie, Freiburg, Germany
| | - N Harbeck
- Brustzentrum, Frauenklinik LMU Klinikum, Munich, Germany
| | - A Wöckel
- Universitätsklinikum Würzburg, Würzburg, Germany
| | - T Decker
- Onkologie Ravensburg, Praxis für Hämatologie und Onkologie GbR, Ravensburg, Germany.
| | | |
Collapse
|
13
|
Voorwerk L, Isaeva OI, Horlings HM, Balduzzi S, Chelushkin M, Bakker NAM, Champanhet E, Garner H, Sikorska K, Loo CE, Kemper I, Mandjes IAM, de Maaker M, van Geel JJL, Boers J, de Boer M, Salgado R, van Dongen MGJ, Sonke GS, de Visser KE, Schumacher TN, Blank CU, Wessels LFA, Jager A, Tjan-Heijnen VCG, Schröder CP, Linn SC, Kok M. PD-L1 blockade in combination with carboplatin as immune induction in metastatic lobular breast cancer: the GELATO trial. NATURE CANCER 2023; 4:535-549. [PMID: 37038006 PMCID: PMC10132987 DOI: 10.1038/s43018-023-00542-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 03/08/2023] [Indexed: 04/12/2023]
Abstract
Invasive lobular breast cancer (ILC) is the second most common histological breast cancer subtype, but ILC-specific trials are lacking. Translational research revealed an immune-related ILC subset, and in mouse ILC models, synergy between immune checkpoint blockade and platinum was observed. In the phase II GELATO trial ( NCT03147040 ), patients with metastatic ILC were treated with weekly carboplatin (area under the curve 1.5 mg ml-1 min-1) as immune induction for 12 weeks and atezolizumab (PD-L1 blockade; triweekly) from the third week until progression. Four of 23 evaluable patients had a partial response (17%), and 2 had stable disease, resulting in a clinical benefit rate of 26%. From these six patients, four had triple-negative ILC (TN-ILC). We observed higher CD8+ T cell infiltration, immune checkpoint expression and exhausted T cells after treatment. With this GELATO trial, we show that ILC-specific clinical trials are feasible and demonstrate promising antitumor activity of atezolizumab with carboplatin, particularly for TN-ILC, and provide insights for the design of highly needed ILC-specific trials.
Collapse
Affiliation(s)
- Leonie Voorwerk
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Olga I Isaeva
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hugo M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sara Balduzzi
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maksim Chelushkin
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Noor A M Bakker
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Elisa Champanhet
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hannah Garner
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Karolina Sikorska
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Claudette E Loo
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Inge Kemper
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid A M Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michiel de Maaker
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jasper J L van Geel
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jorianne Boers
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Maaike de Boer
- Department of Medical Oncology, GROW, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Marloes G J van Dongen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Karin E de Visser
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton N Schumacher
- Oncode Institute, Utrecht, the Netherlands
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Christian U Blank
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Vivianne C G Tjan-Heijnen
- Department of Medical Oncology, GROW, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marleen Kok
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Mortazavipour MM, Mohamadalizadeh-Hanjani Z, Geranpayeh L, Mahdian R, Shahbazi S. K-Ras4A Plays a More Significant Role than K-Ras4B in Ductal Carcinoma of Breast. IRANIAN BIOMEDICAL JOURNAL 2023; 27:126-35. [PMID: 37070675 PMCID: PMC10314762 DOI: 10.61186/ibj.3857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/28/2022] [Indexed: 12/17/2023]
Abstract
Background K-Ras mutations rarely occur in breast cancer. However, studies have supported that K-Ras upregulation is involved in breast cancer pathogenesis. Two main K-Ras transcript variants; K-Ras4A and K-Ras4B, originate from the alternative splicing of exon 4. In this study, we aimed to evaluate variations in the expression of K-Ras4A and K-Ras4B and their role in breast ductal carcinoma. Methods Total RNA was extracted from breast tumors, and the NATs obtained via mastectomy. Patients were selected from new cases of breast cancer with no prior history of chemotherapy. Relative mRNA expression was calculated based on a pairwise comparison between the tumors and the NATs following normalization to the internal control gene. Predictive values of the transcript variants were examined by ROC curve analysis. Results A statistically significant increase was found in the K-Ras4A and K-Ras4B expression with the mean fold changes of 7.58 (p = 0.01) and 2.47 (p = 0.001), respectively. The K-Ras4A/K-Ras4B ratio was lower in the tumors than that of the normal tissues. ROC curve analysis revealed the potential of K-Ras4A (AUC: 0.769) and K-Ras4B (AUC: 0.688) in breast cancer prediction. There was also a significant association between K-Ras4B expression and HER2 statues (p = 0.04). Furthermore, a significant link was detected between K-Ras4A expression and pathological prognostic stages (p = 0.04). Conclusion Our findings revealed that expression levels of K-Ras4A and K-Ras4B is higher in the tumor compared to the normal breast tissues. Increase in K-Ras4A expression was more significant than that of K-Ras4B.
Collapse
Affiliation(s)
| | | | - Loabat Geranpayeh
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Mahdian
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
van Ommen-Nijhof A, Steenbruggen TG, Capel L, Vergouwen M, Vrancken Peeters MJT, Wiersma TG, Sonke GS. Survival and prognostic factors in oligometastatic breast cancer. Breast 2022; 67:14-20. [PMID: 36549169 PMCID: PMC9795523 DOI: 10.1016/j.breast.2022.12.007] [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: 10/21/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Guidelines for oligometastatic breast cancer (OMBC) propagate multimodality treatment including polychemotherapy and local ablative treatment (LAT) of all lesions. The aim of this approach is prolonged disease remission, or even cure. Long-term outcomes in OMBC and factors associated with prognosis are largely unknown, due to the rarity of this condition. We report overall survival (OS), event-free survival (EFS), and prognostic factors in a large real-world cohort of patients with OMBC. METHODS Patients with breast cancer and 1-3 distant metastatic lesions, treated in the Netherlands Cancer Institute between 1997 and 2020, were identified via text mining of medical files. We collected patient, tumor and treatment characteristics. The Kaplan-Meier method was used to calculate OS and EFS estimates, and Cox regression analyses to assess prognostic factors. RESULTS The cohort included 239 patients, of whom 54% had ERpos/HER2neg, 20% HER2pos and 20% triple negative disease. Median follow-up was 88.0 months (95% confidence interval (CI) 82.9-93.1) during which 107 patients died and 139 developed disease progression/recurrence; median OS was 93.0 months (95%CI 66.2-119.8). Factors associated with OS in multivariable analysis were subtype, disease-free interval and radiologic response to first-line systemic therapy; LAT was associated with EFS, but not OS. CONCLUSIONS In this large real-world cohort of patients with OMBC, OS and EFS compare favorably to survival in the general MBC population. Radiologic complete response to first-line systemic therapy was associated with favorable OS and EFS, indicating the importance of early optimal systemic therapy. The value of LAT in OMBC requires further study.
Collapse
Affiliation(s)
- Annemiek van Ommen-Nijhof
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands
| | - Tessa G. Steenbruggen
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands,Department of Internal Medicine, St Antonius Hospital, PO Box 2500, 3430 EM, Nieuwegein, the Netherlands
| | - Laura Capel
- Department of Internal Medicine, St Antonius Hospital, PO Box 2500, 3430 EM, Nieuwegein, the Netherlands
| | - Michel Vergouwen
- Department of Biometrics, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands
| | - Marie-Jeanne T. Vrancken Peeters
- Department of Surgical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands,Department of Surgery, Amsterdam University Medical Center, PO Box 22660, 1100 DD, Amsterdam, the Netherlands
| | - Terry G. Wiersma
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands
| | - Gabe S. Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands,Department of Medical Oncology, Amsterdam University Medical Center, PO Box 22660, 1100 DD, Amsterdam, the Netherlands,Corresponding author. Department of Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, PO Box 90203, 1006 BE, Amsterdam, the Netherlands. @annemiekvon
| |
Collapse
|
16
|
Gilardi L, Airò Farulla LS, Ceci F. Imaging for illuminating actionable pathways in breast cancer. Curr Opin Oncol 2022; 34:606-613. [PMID: 36093891 PMCID: PMC10852035 DOI: 10.1097/cco.0000000000000908] [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: 11/26/2022]
Abstract
PURPOSE OF REVIEW Nuclear medicine has the potential to explore and illuminate several pathways in breast cancer (BC) offering different radiopharmaceuticals for positron emission tomography (PET) designed to target specific tumor characteristics. The aim of this critical review is to give an overview about emerging opportunities in PET imaging, underlining the future potential contribution in the management of BC patients. RECENT FINDINGS Beside 2-deoxy-2-[ 18 F]-fluoro- d -glucose (FDG), new generation tracers for PET imaging have been recently proposed to investigate specific characteristics in breast cancer, both targeting tumor cells and the tumor micro-environment (TME). SUMMARY FDG-PET is a procedure that received extensive clinical validation. However, its role in BC is still suboptimal due to the low-FDG avidity of specific tumor subtypes. Human epidermal growth receptor-2 and integrin targeted PET radiotracers might provide useful information selecting patients more likely to respond to target therapy. FluoroEstradiol (FES) is a FDA-approved PET radiotracer targeting the estrogen receptor (ER), useful to investigate metastatic ER+ patients, to assess in vivo ER heterogeneity and to evaluate hormonal therapy efficacy. Inhibitors of the fibroblast activation protein (FAPi) targeting the cancer-associated fibroblast can explore the TME with PET imaging. FAPi is also proposed a theranostic agent for radio-ligand therapy.
Collapse
Affiliation(s)
- Laura Gilardi
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
| | - Lighea Simona Airò Farulla
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Liu Y, Gan Y, AiErken N, Chen W, Zhang S, Ouyang J, Zeng L, Tang D. Combining Organoid Models with Next-Generation Sequencing to Reveal Tumor Heterogeneity and Predict Therapeutic Response in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:9390912. [PMID: 36046364 PMCID: PMC9423951 DOI: 10.1155/2022/9390912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022]
Abstract
Estrogen receptor-positive (ER+) breast cancer (BC) is a common subtype of BC with a relatively good prognosis. However, recurrence and death from ER+ BC occur because of tumor heterogeneity. This study aimed to explore tumor heterogeneity using next-generation sequencing (NGS) and tumor-organoid models to promote BC precise therapy. We collected needle biopsy, surgical excision, and cerebrospinal fluid (CSF) samples to establish tumor organoids. We found that the histological characteristics of organoids were consistent with original lesions and recapitulated their heterogenicity. In addition, the NGS results showed that PIK3CA and TP53 genes had detrimental mutations. BAP1, RET, AXIN2, and PPP2R2A genes had mutations with unknown function. The score for homologous recombination deficiency (HRD) of genome was 56, indicating that the tumor was likely sensitive to PARPi. The mutant-allele tumor heterogeneity (MATH) value of the tumor genome was 68.03, indicating high tumor heterogeneity. At last, we performed a drug screening on organoids. The toxicity of different drugs toward BC organoids originated from needle biopsy and surgical excision was tested, respectively. The IC50 values in the needle biopsy groups were paclitaxel 2.83 μM, carboplatin 61.47 μM, neratinib 0.8 μM, lapatinib >100 μM; in the surgical excision groups: trastuzumab >100 μM, docetaxel 0.036 μM, tamoxifen 20.54 μM, olaparib 5.478 μM, BYL719 < 0.1 μM. The toxicity data showed that the BC organoids could show dynamic characteristics of tumor progression and reflect the heterogeneity of BC. Our study demonstrates that the combined use of tumor organoids and NGS is a potential way to test tumor heterogeneity and predict drug response in ER + BC, which contributes to the development of personalized therapy.
Collapse
Affiliation(s)
- Yuhong Liu
- The Seventh Affiliated Hospital of Sun Yat-Sen University, General Surgery, Shenzhen 518107, China
| | - Yixiang Gan
- School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - NiJiati AiErken
- The Seventh Affiliated Hospital of Sun Yat-Sen University, General Surgery, Shenzhen 518107, China
| | - Wei Chen
- The Seventh Affiliated Hospital, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen 518107, China
| | - Shiwei Zhang
- The Seventh Affiliated Hospital, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen 518107, China
| | - Jie Ouyang
- Department of Breast Surgery, Dongguan Tungwah Hospital, Dongguan 518107, China
| | - Leli Zeng
- The Seventh Affiliated Hospital, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen 518107, China
| | - Di Tang
- The Seventh Affiliated Hospital of Sun Yat-Sen University, General Surgery, Shenzhen 518107, China
| |
Collapse
|
18
|
Agostinetto E, Nader-Marta G, Paesmans M, Ameye L, Veys I, Buisseret L, Neven P, Taylor D, Fontaine C, Duhoux FP, Canon JL, Denys H, Coussy F, Chakiba C, Ribeiro JM, Piccart M, Desmedt C, Ignatiadis M, Aftimos P. ROSALINE: a phase II, neoadjuvant study targeting ROS1 in combination with endocrine therapy in invasive lobular carcinoma of the breast. Future Oncol 2022; 18:2383-2392. [PMID: 35695563 DOI: 10.2217/fon-2022-0358] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is the most common histologic subtype of breast cancer after invasive ductal carcinoma (i.e., no special type [NST]). ILC differs from NST in clinical presentation, site-specific metastases and response to conventional therapies. Loss of E-cadherin protein expression, due to alterations in its encoding gene CDH1, is the most frequent oncogenic event in ILC. Synthetic lethality approaches have shown promising antitumor effects of ROS1 inhibitors in models of E-cadherin-defective breast cancer in in vivo studies and provide the rationale for testing their clinical activity in patients with ILC. Entrectinib is a tyrosine kinase inhibitor targeting TRK, ROS1 and ALK tyrosine kinases. Here, the authors present ROSALINE (NCT04551495), a phase II study testing neoadjuvant entrectinib and endocrine therapy in women with estrogen receptor-positive, HER2-negative early ILC.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium.,Humanitas University, Department of Biomedical Sciences, via Rita Levi Montalcini 4, 20090 Pieve Emanuele - Milan, Italy
| | | | - Marianne Paesmans
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | - Lieveke Ameye
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Veys
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Buisseret
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | - Martine Piccart
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Desmedt
- Department of Oncology, Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, Belgium
| | - Michail Ignatiadis
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Institut Jules Bordet & l'Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Hildebrandt MG, Naghavi-Behzad M, Vogsen M. A role of FDG-PET/CT for response evaluation in metastatic breast cancer? Semin Nucl Med 2022; 52:520-530. [PMID: 35525631 DOI: 10.1053/j.semnuclmed.2022.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 01/19/2023]
Abstract
Breast cancer prognosis is steadily improving due to early detection of primary cancer in screening programs and revolutionizing treatment development. In the metastatic setting, therapy improvements render breast cancer a chronic disease. Although FDG-PET/CT has emerged as a highly accurate method for staging metastatic breast cancer, there has been no change in response evaluation methods for decades. FDG-PET/CT has proven high prognostic values in patients with metastatic breast cancer when using quantitative PET methods. It has also shown a higher predictive value than conventional CT when applying the respective response evaluation criteria, RECIST and PERCIST. Response categorization using FDG-PET/CT is more sensitive in detecting progressive and regressive disease, while conventional imaging such as CT and bone scintigraphy deem stable disease more often. These findings reflect the higher accuracy of FDG-PET/CT for response evaluation in this patient group. But does the higher accuracy of FDG-PET/CT translate into a patient benefit when implementing it for monitoring response to palliative treatment? We have evidence of survival benefit from a retrospective study indicating the superiority of using FDG-PET/CT compared with conventional imaging for response evaluation in metastatic breast cancer patients. The survival benefit seems to result from earlier detection of progression with FDG-PET/CT than conventional imaging, leading to an earlier change in treatment with potentially better efficacy of the subsequent treatment line. FDG-PET/CT can be used semiquantitatively as suggested in PERCIST. However, we still need to improve clinically applicable methods based on neural network modeling to better integrate the quantitative information in a smart and standardized way, enabling relevant comparability between scans, patients, and institutions. Such innovation is warranted to support imaging specialists in diagnostic response assessment. Prospective multicenter studies analyzing patients' survival, quality of life, societal and patient costs of replacing conventional imaging with FDG-PET/CT are needed before firm conclusions can be drawn on which type of scan to recommend in future clinical guidelines.
Collapse
Affiliation(s)
- Malene Grubbe Hildebrandt
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Center for Personalized Response Monitoring in Oncology, PREMIO, Odense University Hospital, Odense, Denmark; Center for Innovative Medical Technology, CIMT, Odense University Hospital, Odense, Denmark.
| | - Mohammad Naghavi-Behzad
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Center for Personalized Response Monitoring in Oncology, PREMIO, Odense University Hospital, Odense, Denmark
| | - Marianne Vogsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Center for Personalized Response Monitoring in Oncology, PREMIO, Odense University Hospital, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark
| |
Collapse
|