1
|
Chen JH, Addanki S, Roy D, Bassett R, Kalashnikova E, Spickard E, Kuerer HM, Meas S, Sarli VN, Korkut A, White JB, Rauch GM, Tripathy D, Arun BK, Barcenas CH, Yam C, Sethi H, Rodriguez AA, Liu MC, Moulder SL, Lucci A. Monitoring response to neoadjuvant chemotherapy in triple negative breast cancer using circulating tumor DNA. BMC Cancer 2024; 24:1016. [PMID: 39148033 PMCID: PMC11328413 DOI: 10.1186/s12885-024-12689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an aggressive subtype with poor prognosis. We aimed to determine whether circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) could predict response and long-term outcomes to neoadjuvant chemotherapy (NAC). METHODS Patients with TNBC were enrolled between 2017-2021 at The University of Texas MD Anderson Cancer Center (Houston, TX). Serial plasma samples were collected at four timepoints: pre-NAC (baseline), 12-weeks after NAC (mid-NAC), after NAC/prior to surgery (post-NAC), and one-year after surgery. ctDNA was quantified using a tumor-informed ctDNA assay (SignateraTM, Natera, Inc.) and CTC enumeration using CellSearch. Wilcoxon and Fisher's exact tests were used for comparisons between groups and Kaplan-Meier analysis used for survival outcomes. RESULTS In total, 37 patients were enrolled. The mean age was 50 and majority of patients had invasive ductal carcinoma (34, 91.9%) with clinical T2, (25, 67.6%) node-negative disease (21, 56.8%). Baseline ctDNA was detected in 90% (27/30) of patients, of whom 70.4% (19/27) achieved ctDNA clearance by mid-NAC. ctDNA clearance at mid-NAC was significantly associated with pathologic complete response (p = 0.02), whereas CTC clearance was not (p = 0.52). There were no differences in overall survival (OS) and recurrence-free survival (RFS) with positive baseline ctDNA and CTC. However, positive ctDNA at mid-NAC was significantly associated with worse OS and RFS (p = 0.0002 and p = 0.0034, respectively). CONCLUSIONS Early clearance of ctDNA served as a predictive and prognostic marker in TNBC. Personalized ctDNA monitoring during NAC may help predict response and guide treatment.
Collapse
Affiliation(s)
- Jennifer H Chen
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Sridevi Addanki
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Dhruvajyoti Roy
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Roland Bassett
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | - Henry M Kuerer
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Salyna Meas
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Vanessa N Sarli
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jason B White
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Gaiane M Rauch
- Abdominal Imaging Department, MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Banu K Arun
- Breast Medical Oncology and Clinical Cancer Genetics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Carlos H Barcenas
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Clinton Yam
- Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Stacy L Moulder
- Medical Oncology, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, FCT 7.6000, Unit 1484,, Houston, TX, 77030, US.
| |
Collapse
|
2
|
Tarantino P, Tolaney SM. Challenging immune exhaustion in early recurrent triple-negative breast cancer: pitfalls and hopes. Ann Oncol 2024; 35:579-581. [PMID: 38910014 DOI: 10.1016/j.annonc.2024.05.536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/25/2024] Open
Affiliation(s)
- P Tarantino
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - S M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Panet F, Papakonstantinou A, Borrell M, Vivancos J, Vivancos A, Oliveira M. Use of ctDNA in early breast cancer: analytical validity and clinical potential. NPJ Breast Cancer 2024; 10:50. [PMID: 38898045 PMCID: PMC11187121 DOI: 10.1038/s41523-024-00653-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating free tumor DNA (ctDNA) analysis is gaining popularity in precision oncology, particularly in metastatic breast cancer, as it provides non-invasive, real-time tumor information to complement tissue biopsies, allowing for tailored treatment strategies and improved patient selection in clinical trials. Its use in early breast cancer has been limited so far, due to the relatively low sensitivity of available techniques in a setting characterized by lower levels of ctDNA shedding. However, advances in sequencing and bioinformatics, as well as the use of methylome profiles, have led to an increasing interest in the application of ctDNA analysis in early breast cancer, from screening to curative treatment evaluation and minimal residual disease (MRD) detection. With multiple prospective clinical trials in this setting, ctDNA evaluation may become useful in clinical practice. This article reviews the data regarding the analytical validity of the currently available tests for ctDNA detection and the clinical potential of ctDNA analysis in early breast cancer.
Collapse
Affiliation(s)
- François Panet
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Lady Davis Institute, Jewish General Hospital, Montréal, QC, Canada
| | - Andri Papakonstantinou
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Breast, Endocrine Tumors and Sarcomas, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Borrell
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Joan Vivancos
- Cancer Genomics Group, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Mafalda Oliveira
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain.
| |
Collapse
|
4
|
Zhang X, Feng R, Xu Y, Yang L, Xie F, Yang H, Wang S, Peng Y, Liu M, Wang C, Wang S. Baseline circulating tumor DNA predicts long-term survival outcomes for patients with early breast cancer. Gland Surg 2024; 13:684-696. [PMID: 38845832 PMCID: PMC11150192 DOI: 10.21037/gs-24-115] [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: 04/11/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024]
Abstract
Background Circulating tumor DNA (ctDNA) is a potential biomarker not only capable of monitoring the treatment response during neoadjuvant therapy (NAT) or rescue therapy, but also identifying minimal residual disease (MRD) and detecting early relapses after primary treatment. However, it remains uncertain whether the detection of ctDNA at diagnosis, before any treatment, can predict the prognosis for patients with early breast cancer. The objective of our study was to evaluate the predictive value of baseline ctDNA for prognosis in patients with early breast cancer. Methods A total of 90 patients with early breast cancer and 24 healthy women were recruited between August 2016 and October 2016. Peripheral blood samples were collected from patients at diagnosis, before any treatment. Blood samples were processed and subjected to targeted deep sequencing with a next-generation sequencing (NGS) panel of 1,021 cancer-related genes. The recurrence-free survival (RFS) and invasive disease-free survival (iDFS) were reported. Results The 90 patients with breast cancer included 6 patients with ductal carcinoma in situ (DCIS) and 84 patients with invasive breast cancer. Within the cohort of patients with invasive breast cancer, ctDNA were detected in 57 patients, with a ctDNA detection rate of 67.9%. Meanwhile, no ctDNA was detected in DCIS patients. Among 84 patients with invasive breast cancer, patients with high-level ctDNA had a significantly lower RFS compared to patients with low-level ctDNA (log-rank P=0.0036). Conclusions Our study suggested that ctDNA at diagnosis, before any treatment, could potentially serve as a biomarker to predict the prognosis for patients with early breast cancer. However, further follow-up and more studies with large sample sizes are required to confirm these findings.
Collapse
Affiliation(s)
- Xiangui Zhang
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Rui Feng
- Breast Surgery, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University Affiliated Maternity Hospital, Tianjin, China
| | - Yaqian Xu
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Liu Yang
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Fei Xie
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Houpu Yang
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Siyuan Wang
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Yuan Peng
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Miao Liu
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Chaobin Wang
- Breast Center, Peking University People’s Hospital, Beijing, China
| | - Shu Wang
- Breast Center, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
5
|
Martín M, Yoder R, Salgado R, del Monte-Millán M, Álvarez EL, Echavarría I, Staley JM, O’Dea AP, Nye LE, Stecklein SR, Bueno C, Jerez Y, Cebollero M, Bueno O, Saenz JÁG, Moreno F, Bohn U, Gómez H, Massarrah T, Khan QJ, Godwin AK, López-Tarruella S, Sharma P. Tumor-Infiltrating Lymphocytes Refine Outcomes in Triple-Negative Breast Cancer Treated with Anthracycline-Free Neoadjuvant Chemotherapy. Clin Cancer Res 2024; 30:2160-2169. [PMID: 38466643 PMCID: PMC11096004 DOI: 10.1158/1078-0432.ccr-24-0106] [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: 01/09/2024] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
PURPOSE Stromal tumor-infiltrating lymphocytes (sTIL) are associated with pathologic complete response (pCR) and long-term outcomes for triple-negative breast cancer (TNBC) in the setting of anthracycline-based chemotherapy. The impact of sTILs on refining outcomes beyond prognostic information provided by pCR in anthracycline-free neoadjuvant chemotherapy (NAC) is not known. EXPERIMENTAL DESIGN This is a pooled analysis of two studies where patients with stage I (T>1 cm)-III TNBC received carboplatin (AUC 6) plus docetaxel (75 mg/m2; CbD) NAC. sTILs were evaluated centrally on pre-treatment hematoxylin and eosin slides using standard criteria. Cox regression analysis was used to examine the effect of variables on event-free survival (EFS) and overall survival (OS). RESULTS Among 474 patients, 44% had node-positive disease. Median sTILs were 5% (range, 1%-95%), and 32% of patients had ≥30% sTILs. pCR rate was 51%. On multivariable analysis, T stage (OR, 2.08; P = 0.007), nodal status (OR, 1.64; P = 0.035), and sTILs (OR, 1.10; P = 0.011) were associated with pCR. On multivariate analysis, nodal status (HR, 0.46; P = 0.008), pCR (HR, 0.20; P < 0.001), and sTILs (HR, 0.95; P = 0.049) were associated with OS. At 30% cut-point, sTILs stratified outcomes in stage III disease, with 5-year OS 86% versus 57% in ≥30% versus <30% sTILs (HR, 0.29; P = 0.014), and numeric trend in stage II, with 5-year OS 93% versus 89% in ≥30% versus <30% sTILs (HR, 0.55; P = 0.179). Among stage II-III patients with pCR, EFS was better in those with ≥30% sTILs (HR, 0.16; P, 0.047). CONCLUSIONS sTILs density was an independent predictor of OS beyond clinicopathologic features and pathologic response in patients with TNBC treated with anthracycline-free CbD chemotherapy. Notably, sTILs density stratified outcomes beyond tumor-node-metastasis (TNM) stage and pathologic response. These findings highlight the role of sTILs in patient selection and stratification for neo/adjuvant escalation and de-escalation strategies.
Collapse
Affiliation(s)
- Miguel Martín
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Rachel Yoder
- The University of Kansas Cancer Center, Westwood, KS, USA
| | | | - María del Monte-Millán
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Enrique L. Álvarez
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Isabel Echavarría
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | | | - Anne P. O’Dea
- University of Kansas Medical Center, Westwood, KS, USA
| | - Lauren E. Nye
- University of Kansas Medical Center, Westwood, KS, USA
| | | | | | - Yolanda Jerez
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - María Cebollero
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Oscar Bueno
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Fernando Moreno
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain
| | - Uriel Bohn
- Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Canary Islands
| | - Henry Gómez
- Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Tatiana Massarrah
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Qamar J. Khan
- University of Kansas Medical Center, Westwood, KS, USA
| | | | - Sara López-Tarruella
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | | |
Collapse
|
6
|
Roussot N, Constantin G, Desmoulins I, Bergeron A, Arnould L, Beltjens F, Mayeur D, Kaderbhai C, Hennequin A, Jankowski C, Padeano MM, Costaz H, Jacinto S, Michel E, Amet A, Coutant C, Costa B, Jouannaud C, Deblock M, Levy C, Ferrero JM, Kerbrat P, Brain E, Mouret-Reynier MA, Coudert B, Bertaut A, Ladoire S. Prognostic stratification ability of the CPS+EG scoring system in HER2-low and HER2-zero early breast cancer treated with neoadjuvant chemotherapy. Eur J Cancer 2024; 202:114037. [PMID: 38554542 DOI: 10.1016/j.ejca.2024.114037] [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: 12/29/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND The CPS+EG scoring system was initially described in unselected early breast cancer (eBC) patients treated with neoadjuvant chemotherapy (NAC), leading to refined prognostic stratification, and thus helping to select patients for additional post-NAC treatments. It remains unknown whether the performance is the same in new biological breast cancer entities such as the HER2-low subtype. PATIENTS AND METHODS Outcomes (disease-free (DFS) and overall survival OS)) of 608 patients with HER2-non amplified eBC and treated with NAC were retrospectively analyzed according to CPS-EG score. We compared the prognostic stratification abilities of the CPS+EG in HER2-low and HER2-0 eBC, analyzing ER+ and ER- tumors separately. RESULTS In ER+ eBC, the CPS+EG scoring system seems to retain a prognostic value, both in HER2-low and HER2-0 tumors, by distinguishing populations with significantly different outcomes (good: score 0-1, poor: score 2-3, and very poor: score 4-5). Using C-indices for DFS and OS, CPS+EG provided the highest prognostic information in ER+ eBC, especially in HER2-0 tumors. In contrast, in ER- eBC, the CPS+EG does not appear to be able to distinguish different outcome groups, either in HER2-low or HER2-0 tumors. In ER- eBC, C-indices for DFS and OS were highest for pathological stage, reflecting the predominant prognostic importance of residual disease in this subtype. CONCLUSIONS HER2-low status does not influence the prognostic performance of the CPS+EG score. Our results confirm the usefulness of the CPS+EG score in stratifying the prognosis of ER+ eBC after NAC, for both HER2-0 and HER2-low tumors. For ER- eBC, HER2-low status does not influence the performance of the CPS+EG score, which was lower than that of the pathological stage alone.
Collapse
Affiliation(s)
- Nicolas Roussot
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France; Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center, Dijon, France; INSERM U1231, 21000 Dijon, France
| | - Guillaume Constantin
- Unit of Methodology and Biostatistics, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Anthony Bergeron
- Department of Biology and Pathology of tumors, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Laurent Arnould
- Department of Biology and Pathology of tumors, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Françoise Beltjens
- Department of Biology and Pathology of tumors, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Didier Mayeur
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Courèche Kaderbhai
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Clémentine Jankowski
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Marie Martine Padeano
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Hélène Costaz
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Sarah Jacinto
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Eloise Michel
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Alix Amet
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Charles Coutant
- Department of Surgical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France; University of Burgundy-Franche Comté, 21000 Dijon, France
| | - Brigitte Costa
- Department of Medical Oncology, Institut Jean Godinot, Reims, France
| | | | - Mathilde Deblock
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - Pierre Kerbrat
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Etienne Brain
- Department of Medical Oncology, Institut Curie, Centre René Huguenin, Saint-Cloud, France
| | | | - Bruno Coudert
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Aurélie Bertaut
- Unit of Methodology and Biostatistics, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France; Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center, Dijon, France; University of Burgundy-Franche Comté, 21000 Dijon, France; INSERM U1231, 21000 Dijon, France.
| |
Collapse
|
7
|
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
|
8
|
Nicolò E, Gianni C, Pontolillo L, Serafini MS, Munoz-Arcos LS, Andreopoulou E, Curigliano G, Reduzzi C, Cristofanilli M. Circulating tumor cells et al.: towards a comprehensive liquid biopsy approach in breast cancer. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:10. [PMID: 38751670 PMCID: PMC11093063 DOI: 10.21037/tbcr-23-55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/21/2024] [Indexed: 05/18/2024]
Abstract
Liquid biopsy has emerged as a crucial tool in managing breast cancer (BC) patients, offering a minimally invasive approach to detect circulating tumor biomarkers. Until recently, the majority of the studies in BC focused on evaluating a single liquid biopsy analyte, primarily circulating tumor DNA and circulating tumor cells (CTCs). Despite the proven prognostic and predictive value of CTCs, their low abundance when detected using enrichment methods, especially in the early stages, poses a significant challenge. It is becoming evident that combining diverse circulating biomarkers, each representing different facets of tumor biology, has the potential to enhance the management of patients with BC. This article emphasizes the importance of considering these biomarkers as complementary/synergistic rather than competitive, recognizing their ability to contribute to a comprehensive disease profile. The review provides an overview of the clinical significance of simultaneously analyzing CTCs and other biomarkers, including cell-free circulating DNA, extracellular vesicles, non-canonical CTCs, cell-free RNAs, and non-malignant cells. Such a comprehensive liquid biopsy approach holds promise not only in BC but also in other cancer types, offering opportunities for early detection, prognostication, and therapy monitoring. However, addressing associated challenges, such as refining detection methods and establishing standardized protocols, is crucial for realizing the full potential of liquid biopsy in transforming our understanding and approach to BC. As the field evolves, collaborative efforts will be instrumental in unlocking the revolutionary impact of liquid biopsy in BC research and management.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Caterina Gianni
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Letizia Pontolillo
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Medical Oncology Department, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mara Serena Serafini
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Laura Sofia Munoz-Arcos
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Eleni Andreopoulou
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Carolina Reduzzi
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
9
|
Pankotai-Bodó G, Oláh-Németh O, Sükösd F, Pankotai T. Routine molecular applications and recent advances in breast cancer diagnostics. J Biotechnol 2024; 380:20-28. [PMID: 38122830 DOI: 10.1016/j.jbiotec.2023.12.005] [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: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
Cancer stands as one of the most common and lethal diseases, imposing a substantial burden on global mortality rates. Breast cancer is distinct from other forms of cancer in which it is the primary cause of death for women. Early detection of breast cancer can significantly lower the risk of mortality, improving the prognosis for those who are affected. The death rate of breast cancer has been steadily rising, according to epidemiological data, especially since the COVID-19 pandemic. This emphasizes the necessity of sensitive and precise technologies that can be utilized in early breast cancer diagnosis. In this process, biomarkers play a pivotal role by facilitating the early detection and diagnosis of breast cancer. Currently, a wide variety of cancer biomarkers have been identified, improving the accuracy of cancer diagnosis. These biomarkers can be applied in liquid biopsies as well as on solid tissues. In the context of breast cancer, biomarkers are particularly valuable for determining who is predisposed to the disease, predicting prognosis at the time of diagnosis, and selecting the best course of therapy. This review comprehensively explores the recently developed gene-based biomarkers from biofluids that are used in the context of breast cancer, as well as the conventional and cutting-edge techniques that have been employed for breast cancer diagnosis.
Collapse
Affiliation(s)
- Gabriella Pankotai-Bodó
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
| | - Orsolya Oláh-Németh
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary; Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, Budapesti út 9, Szeged H-6728, Hungary
| | - Farkas Sükösd
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
| | - Tibor Pankotai
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary; Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, Budapesti út 9, Szeged H-6728, Hungary; Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér 13, Szeged H-6720, Hungary.
| |
Collapse
|
10
|
Lopez-Gonzalez L, Sanchez Cendra A, Sanchez Cendra C, Roberts Cervantes ED, Espinosa JC, Pekarek T, Fraile-Martinez O, García-Montero C, Rodriguez-Slocker AM, Jiménez-Álvarez L, Guijarro LG, Aguado-Henche S, Monserrat J, Alvarez-Mon M, Pekarek L, Ortega MA, Diaz-Pedrero R. Exploring Biomarkers in Breast Cancer: Hallmarks of Diagnosis, Treatment, and Follow-Up in Clinical Practice. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:168. [PMID: 38256428 PMCID: PMC10819101 DOI: 10.3390/medicina60010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Breast cancer is a prevalent malignancy in the present day, particularly affecting women as one of the most common forms of cancer. A significant portion of patients initially present with localized disease, for which curative treatments are pursued. Conversely, another substantial segment is diagnosed with metastatic disease, which has a worse prognosis. Recent years have witnessed a profound transformation in the prognosis for this latter group, primarily due to the discovery of various biomarkers and the emergence of targeted therapies. These biomarkers, encompassing serological, histological, and genetic indicators, have demonstrated their value across multiple aspects of breast cancer management. They play crucial roles in initial diagnosis, aiding in the detection of relapses during follow-up, guiding the application of targeted treatments, and offering valuable insights for prognostic stratification, especially for highly aggressive tumor types. Molecular markers have now become the keystone of metastatic breast cancer diagnosis, given the diverse array of chemotherapy options and treatment modalities available. These markers signify a transformative shift in the arsenal of therapeutic options against breast cancer. Their diagnostic precision enables the categorization of tumors with elevated risks of recurrence, increased aggressiveness, and heightened mortality. Furthermore, the existence of therapies tailored to target specific molecular anomalies triggers a cascade of changes in tumor behavior. Therefore, the primary objective of this article is to offer a comprehensive review of the clinical, diagnostic, prognostic, and therapeutic utility of the principal biomarkers currently in use, as well as of their clinical impact on metastatic breast cancer. In doing so, our goal is to contribute to a more profound comprehension of this complex disease and, ultimately, to enhance patient outcomes through more precise and effective treatment strategies.
Collapse
Affiliation(s)
- Laura Lopez-Gonzalez
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Alicia Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Cristina Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | | | - Javier Cassinello Espinosa
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Tatiana Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Oscar Fraile-Martinez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Cielo García-Montero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Ana María Rodriguez-Slocker
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - Soledad Aguado-Henche
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Jorge Monserrat
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Melchor Alvarez-Mon
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Leonel Pekarek
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Miguel A. Ortega
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| |
Collapse
|
11
|
Zaikova E, Cheng BYC, Cerda V, Kong E, Lai D, Lum A, Bates C, den Brok W, Kono T, Bourque S, Chan A, Feng X, Fenton D, Gurjal A, Levasseur N, Lohrisch C, Roberts S, Shenkier T, Simmons C, Taylor S, Villa D, Miller R, Aguirre-Hernandez R, Aparicio S, Gelmon K. Circulating tumour mutation detection in triple-negative breast cancer as an adjunct to tissue response assessment. NPJ Breast Cancer 2024; 10:3. [PMID: 38182588 PMCID: PMC10770342 DOI: 10.1038/s41523-023-00607-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/02/2023] [Indexed: 01/07/2024] Open
Abstract
Circulating tumour DNA (ctDNA) detection via liquid biopsy is an emerging alternative to tissue biopsy, but its potential in treatment response monitoring and prognosis in triple negative breast cancer (TNBC) is not yet well understood. Here we determined the prevalence of actionable mutations detectable in ctDNA using a clinically validated cancer gene panel assay in patients with TNBC, without recurrence at the time of study entry. Sequencing of plasma DNA and validation of variants from 130 TNBC patients collected within 7 months of primary treatment completion revealed that 7.7% had detectable residual disease with a hotspot panel. Among neoadjuvant treated patients, we observed a trend where patients with incomplete pathologic response and positive ctDNA within 7 months of treatment completion were at much higher risk of reduced progression free survival. We propose that a high risk subset of early TNBC patients treated in neoadjuvant therapy protocols may be identifiable by combining tissue response and sensitive ctDNA detection.
Collapse
Affiliation(s)
- Elena Zaikova
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Brian Y C Cheng
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Viviana Cerda
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Esther Kong
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Daniel Lai
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Amy Lum
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Cherie Bates
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Wendie den Brok
- Medical Oncology, BC Cancer, 600 W10th Avenue, Vancouver, Canada
| | - Takako Kono
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada
| | - Sylvie Bourque
- Medical Oncology, BC Cancer, 13750 96 Ave, Surrey, Canada
| | - Angela Chan
- Medical Oncology, BC Cancer, 13750 96 Ave, Surrey, Canada
| | - Xioalan Feng
- Medical Oncology, BC Cancer, 2410 Lee Ave, Victoria, Canada
| | - David Fenton
- Medical Oncology, BC Cancer, 2410 Lee Ave, Victoria, Canada
| | - Anagha Gurjal
- Medical Oncology, BC Cancer, 32900 Marshall Rd, Abbotsford, Canada
| | | | | | - Sarah Roberts
- Medical Oncology, BC Cancer, 1215 Lethbridge St, Prince George, Canada
| | - Tamara Shenkier
- Medical Oncology, BC Cancer, 600 W10th Avenue, Vancouver, Canada
| | | | - Sara Taylor
- Medical Oncology, BC Cancer, 399 Royal Ave, Kelowna, Canada
| | - Diego Villa
- Medical Oncology, BC Cancer, 600 W10th Avenue, Vancouver, Canada
| | - Ruth Miller
- Imagia Canexia Health, 204-2389 Health Sciences Mall, Vancouver, Canada
| | | | - Samuel Aparicio
- Molecular Oncology, BC Cancer, 675 W10th Avenue, Vancouver, Canada.
| | - Karen Gelmon
- Medical Oncology, BC Cancer, 600 W10th Avenue, Vancouver, Canada.
| |
Collapse
|
12
|
Agostinetto E, Nader-Marta G, Ignatiadis M. Circulating tumor DNA in breast cancer: a biomarker for patient selection. Curr Opin Oncol 2023; 35:426-435. [PMID: 37551949 DOI: 10.1097/cco.0000000000000964] [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: 08/09/2023]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of circulating tumor DNA (ctDNA) as a biomarker for patient selection in breast cancer. We describe the current evidence and the main ongoing trials both in the early and metastatic setting. RECENT FINDINGS In the metastatic setting, the analysis of ctDNA can identify specific genetic alterations amenable of molecularly targeted treatments. Several assays are now approved for the detection of genetic alterations in plasma cell-free DNA to guide treatment decision (e.g., PIK3CA mutations for PI3K inhibitors, and ESR1 mutations for the selective estrogen receptor degrader elacestrant). In the early setting, emerging evidence is demonstrating that ctDNA can identify a disease relapse with a lead-time of approximately 10 months before imaging. This could help select patients who may benefit from escalation treatment strategy, although this hypothesis needs to be first prospectively validated. SUMMARY Liquid biopsy for ctDNA detection represents an exciting new field in rapid evolution. Several trials are ongoing to validate the clinical utility of ctDNA in daily practice in the early setting and to expand its current indications in the metastatic one.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | | | | |
Collapse
|
13
|
Matou-Nasri S, Aldawood M, Alanazi F, Khan AL. Updates on Triple-Negative Breast Cancer in Type 2 Diabetes Mellitus Patients: From Risk Factors to Diagnosis, Biomarkers and Therapy. Diagnostics (Basel) 2023; 13:2390. [PMID: 37510134 PMCID: PMC10378597 DOI: 10.3390/diagnostics13142390] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is usually the most malignant and aggressive mammary epithelial tumor characterized by the lack of expression for estrogen receptors and progesterone receptors, and the absence of epidermal growth factor receptor (HER)2 amplification. Corresponding to 15-20% of all breast cancers and well-known by its poor clinical outcome, this negative receptor expression deprives TNBC from targeted therapy and makes its management therapeutically challenging. Type 2 diabetes mellitus (T2DM) is the most common ageing metabolic disorder due to insulin deficiency or resistance resulting in hyperglycemia, hyperinsulinemia, and hyperlipidemia. Due to metabolic and hormonal imbalances, there are many interplays between both chronic disorders leading to increased risk of breast cancer, especially TNBC, diagnosed in T2DM patients. The purpose of this review is to provide up-to-date information related to epidemiology and clinicopathological features, risk factors, diagnosis, biomarkers, and current therapy/clinical trials for TNBC patients with T2DM compared to non-diabetic counterparts. Thus, in-depth investigation of the diabetic complications on TNBC onset, development, and progression and the discovery of biomarkers would improve TNBC management through early diagnosis, tailoring therapy for a better outcome of T2DM patients diagnosed with TNBC.
Collapse
Affiliation(s)
- Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Maram Aldawood
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Post Graduate and Zoology Department, King Saud University, Riyadh 12372, Saudi Arabia
| | - Fatimah Alanazi
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Abdul Latif Khan
- Tissue Biobank, KAIMRC, MNG-HA, Riyadh 11481, Saudi Arabia
- Pathology and Clinical Laboratory Medicine, King Abdulaziz Medical City (KAMC), Riyadh 11564, Saudi Arabia
| |
Collapse
|
14
|
Tran J, Thaper A, Lopetegui-Lia N, Ali A. Locoregional recurrence in triple negative breast cancer: past, present, and future. Expert Rev Anticancer Ther 2023; 23:1085-1093. [PMID: 37750222 DOI: 10.1080/14737140.2023.2262760] [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: 07/17/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is a rare but aggressive biological subtype of breast cancer associated with higher locoregional and distant recurrence rates and lower overall survival despite advancements in diagnostic and treatment strategies. AREAS COVERED This review explores the evolving landscape of locoregional recurrence (LRR) in TNBC with improved surgical and radiation therapy delivery techniques including salvage breast conserving surgery (SBCS), re-irradiation, and thermo-radiation. We review current retrospective and prospective, albeit limited, clinical data highlighting the optimal management of locoregionally recurrent TNBC. We also discuss tumor genomic profiling and transcriptome analysis and review potential investigational directions. EXPERT OPINION Significant progress has been made in the prevention of LRR but rates remain suboptimal, particularly in the TNBC population, and outcomes following LRR are poor. Further prospective studies are needed to identify the most effective and safest systemic therapy regimens and to whom it should be offered. There has been growing interest in the role of molecular markers, genomic signatures, and tumor microenvironment in predicting outcomes and guiding LRR treatment. Transcriptome analyses and biomarker-driven investigations are currently being studied and represent a promising era of development in the management of LRR.
Collapse
Affiliation(s)
- Jennifer Tran
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, USA
| | - Arushi Thaper
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Nerea Lopetegui-Lia
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, USA
| | - Azka Ali
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, USA
| |
Collapse
|