1
|
Jean J, Jochelson MS, Moo TA, Solomon SB, Bryce Y. Breast Cancer Recurrence after Cryoablation in Patients Who Are Poor Surgical Candidates or Who Refuse Surgery. J Vasc Interv Radiol 2025; 36:971-978. [PMID: 39909175 DOI: 10.1016/j.jvir.2025.01.048] [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: 03/21/2024] [Revised: 01/15/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025] Open
Abstract
PURPOSE To evaluate in-breast recurrence rates after cryoablation in patients with primary breast cancer who were poor surgical candidates or refused surgery. MATERIALS AND METHODS Patients with primary breast cancer who were poor surgical candidates or who refused surgery and were treated with cryoablation at a single academic cancer center between October 2018 and June 2023 were retrospectively reviewed. Of the 60 treated patients, 45 had invasive ductal carcinoma, 6 had invasive lobular carcinoma, 2 had multicentric ductal carcinoma in situ, and 7 had other histology. Tumor size ranged from 0.3 to 9 cm, with a mean of 2.7 cm. Recurrence was defined as new tumor or regrowth of residual tumor in the ipsilateral breast. RESULTS With a mean follow-up of 21 months and median follow-up of 9.8 months, there was a recurrence rate of 10% (6 of 60 patients). Patients in the recurrence group had more poorly differentiated disease than those in the nonrecurrence group (66.7% vs 22.2%; P = .038). Tumor size did not differ between nonrecurrence and recurrence groups (nonrecurrence group mean, 2.7 cm [SD ± 2.6]; recurrence group mean, 2.5 cm [SD ± 1.0]; P = .506). Patients who were treated with palliative intent rather than curative intent were significantly older (79.7 years [SD ± 12.2] vs 72.5 years [SD ± 11.3]; P = .032). CONCLUSIONS Cryoablation can be considered a treatment option in patients who are poor surgical candidates or who refuse surgery, with a 10% recurrence rate at a mean follow-up of 21 months in this retrospective review that included patients with tumors up to 9 cm, unfavorable pathology, and multicentric disease.
Collapse
MESH Headings
- Humans
- Cryosurgery/adverse effects
- Female
- Breast Neoplasms/surgery
- Breast Neoplasms/pathology
- Breast Neoplasms/diagnostic imaging
- Retrospective Studies
- Neoplasm Recurrence, Local
- Middle Aged
- Aged
- Treatment Outcome
- Time Factors
- Risk Factors
- Treatment Refusal
- Adult
- Risk Assessment
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Ductal, Breast/pathology
- Aged, 80 and over
- Carcinoma, Lobular/surgery
- Carcinoma, Lobular/pathology
- Patient Selection
- Tumor Burden
- Carcinoma, Intraductal, Noninfiltrating/surgery
- Carcinoma, Intraductal, Noninfiltrating/pathology
Collapse
Affiliation(s)
- Jolie Jean
- Radiology Department, New York-Presbyterian Hospital/Weill Cornell Imaging, New York, New York.
| | - Maxine S Jochelson
- Radiology Department, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tracy-Ann Moo
- Surgery Department, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephen B Solomon
- Radiology Department, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yolanda Bryce
- Radiology Department, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
2
|
Li L, Liu Z, Chen K, Li Y. Improving Prognostic Value in Invasive Triple Negative Breast Cancer Through a Combined Nomogram Approach. Clin Breast Cancer 2025; 25:e279-e287.e4. [PMID: 39674765 DOI: 10.1016/j.clbc.2024.11.013] [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: 04/06/2024] [Revised: 11/09/2024] [Accepted: 11/17/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVES To investigate the potential prognostic value of ultrasound (US) features in conjunction with pathological markers and to develop a preliminary working model for predicting poor outcomes in patients with invasive triple-negative breast cancer (TNBC). METHODS From January 2012 to December 2018, we enrolled 209 TNBC patients treated with standard therapy, systematically gathered data on US parameters, stromal tumor-infiltrating lymphocytes (TILs), lymphovascular invasion (LVI) status, and other relevant information, and recorded follow-up data. A nomogram combining AJCC staging with US score, stromal TILs, and LVI was constructed and validated to predict poor outcomes, defined as recurrence or death, in patients with invasive TNBC. RESULTS The US score of 4 was best related to poor outcomes in patients with TNBC (HR 3.87, P = .015). In the training set, the nomogram had a considerably greater prognostic value [area under the curve (AUC), 0.74 vs. 0.64, P = .045] than AJCC staging alone, and it was comparable to that of the validation set (AUC, 0.71 vs. 0.63, P = .804). An acceptable consistency between the nomogram-predicted and actual survival probabilities was found both in the training and validation sets, with Brier scores of 0.15 and 0.13, respectively. CONCLUSIONS The incorporation of AJCC stage with US score, stromal TILs, and LVI improved the model performance for predicting poor outcomes in patients with invasive TNBC compared to routine AJCC staging alone.
Collapse
Affiliation(s)
- Lian Li
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi Liu
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kairong Chen
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingjia Li
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Budzyń M, Kubicka A, Kaja E, Kycler W, Załuska-Kusz J, Brzeziński JJ, Sperling M, Bukowska A, Grupińska J. Significance of the monocyte CCR2-CCL2 axis in triple-negative breast cancer. Arch Med Res 2025; 56:103205. [PMID: 40073677 DOI: 10.1016/j.arcmed.2025.103205] [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: 09/10/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The monocyte CCR2-CCL2 axis appears to play a crucial role in the generation of tumor-associated macrophages (TAMs), which subsequently promotes tumor metastasis and resistance to therapy. AIMS Our study assessed the monocyte CCR2-CCL2 axis in triple-negative breast cancer (TNBC) and its ability to predict tumor response to neoadjuvant chemotherapy (NAC). METHODS The study included 42 female patients diagnosed with TNBC and eligible for NAC. Response to neoadjuvant chemotherapy was based on pathological complete response (pCR). Surface expression of CCR2 on monocytes was evaluated by flow cytometry. Circulating CCL2 was measured by Luminex X-Map technology. RESULTS Increased monocyte CCR2 expression and higher circulating CCL2 levels were observed in the patients with TNBC. After dividing the patients according to their response to NAC, a significant difference in CCL2 levels was found only between patients who achieved pCR and those who did not. ROC curves showed that the optimum diagnostic cut-off value of CCL2 ≤89.61 pg/mL better discriminated patients with TNBC who achieved pCR better than the Ki-67 index. Univariate analysis demonstrated that circulating. CCL2 ≤89.61 pg/mL was significantly associated with pCR. However, this correlation lost statistical significance in the multivariate model. CONCLUSIONS Our study demonstrated the activation of the monocyte CCR2-CCL2 axis in TNBC for the first time. This activation occurs mainly in patients who do not respond to NAC. Circulating CCL2 levels ≤89.61 pg/mL were found to predict, to some extent, the achievement of pCR in patients with TNBC receiving NAC.
Collapse
Affiliation(s)
- Magdalena Budzyń
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | - Agata Kubicka
- Departament of Cancer Pathology, Greater Poland Cancer Centre, Poznań, Poland; Departament of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, Poznań, Poland
| | - Elżbieta Kaja
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Witold Kycler
- Department of Oncological Surgery of Gastrointestinal Diseases, Greater Poland Cancer Centre, Poznań, Poland
| | - Joanna Załuska-Kusz
- Department of Clinical Oncology and Immuno-oncology, Greater Poland Cancer Center, Poznań, Poland
| | - Jacek J Brzeziński
- Department of Oncological Surgery of Gastrointestinal Diseases, Greater Poland Cancer Centre, Poznań, Poland
| | - Marcelina Sperling
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Alicja Bukowska
- Medical Analysis Laboratory, Regional Blood Center, Poznań, Poland
| | - Joanna Grupińska
- Chair and Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Poznań, Poland; Hospital Pharmacy, Greater Poland Cancer Center, Poznań, Poland
| |
Collapse
|
4
|
Manoochehri H, Farrokhnia M, Sheykhhasan M, Mahaki H, Tanzadehpanah H. Key target genes related to anti-breast cancer activity of ATRA: A network pharmacology, molecular docking and experimental investigation. Heliyon 2024; 10:e34300. [PMID: 39108872 PMCID: PMC11301165 DOI: 10.1016/j.heliyon.2024.e34300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 01/07/2025] Open
Abstract
All-trans retinoic acid (ATRA) has promising activity against breast cancer. However, the exact mechanisms of ATRA's anticancer effects remain complex and not fully understood. In this study, a network pharmacology and molecular docking approach was applied to identify key target genes related to ATRA's anti-breast cancer activity. Gene/disease enrichment analysis for predicted ATRA targets was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID), the Comparative Toxicogenomics Database (CTD), and the Gene Set Cancer Analysis (GSCA) database. Protein-Protein Interaction Network (PPIN) generation and analysis was conducted via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and cytoscape, respectively. Cancer-associated genes were evaluated using MyGeneVenn from the CTD. Differential expression analysis was conducted using the Tumor, Normal, and Metastatic (TNM) Plot tool and the Human Protein Atlas (HPA). The Glide docking program was used to predict ligand-protein binding. Treatment response predication and clinical profile assessment were performed using Receiver Operating Characteristic (ROC) Plotter and OncoDB databases, respectively. Cytotoxicity and gene expression were measured using MTT/fluorescent assays and Real-Time PCR, respectively. Molecular functions of ATRA targets (n = 209) included eicosanoid receptor activity and transcription factor activity. Some enriched pathways included inclusion body myositis and nuclear receptors pathways. Network analysis revealed 35 hub genes contributing to 3 modules, with 16 of them were associated with breast cancer. These genes were involved in apoptosis, cell cycle, androgen receptor pathway, and ESR-mediated signaling, among others. CCND1, ESR1, MMP9, MDM2, NCOA3, and RARA were significantly overexpressed in tumor samples. ATRA showed a high affinity towards CCND1/CDK4 and MMP9. CCND1, ESR1, and MDM2 were associated with poor treatment response and were downregulated after treatment of the breast cancer cell line with ATRA. CCND1 and ESR1 exhibited differential expression across breast cancer stages. Therefore, some part of ATRA's anti-breast cancer activity may be exerted through the CCND1/CDK4 complex.
Collapse
Affiliation(s)
- Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Farrokhnia
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Dent R, André F, Gonçalves A, Martin M, Schmid P, Schütz F, Kümmel S, Swain SM, Bilici A, Loirat D, Villalobos Valencia R, Im SA, Park YH, De Laurentis M, Colleoni M, Guarneri V, Bianchini G, Li H, Kirchmayer Machackova Z, Mouta J, Deurloo R, Gan X, Fan M, Mani A, Swat A, Cortés J. IMpassion132 double-blind randomised phase III trial of chemotherapy with or without atezolizumab for early relapsing unresectable locally advanced or metastatic triple-negative breast cancer. Ann Oncol 2024; 35:630-642. [PMID: 38755096 DOI: 10.1016/j.annonc.2024.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors improve the efficacy of first-line chemotherapy for patients with programmed death-ligand 1 (PD-L1)-positive unresectable locally advanced/metastatic triple-negative breast cancer (aTNBC), but randomised data in rapidly relapsing aTNBC are scarce. PATIENTS AND METHODS IMpassion132 (NCT03371017) enrolled patients with aTNBC relapsing <12 months after last chemotherapy dose (anthracycline and taxane required) or surgery for early TNBC. PD-L1 status was centrally assessed using SP142 before randomisation. Initially patients were enrolled irrespective of PD-L1 status. From August 2019, enrolment was restricted to PD-L1-positive (tumour immune cell ≥1%) aTNBC. Patients were randomised 1:1 to placebo or atezolizumab 1200 mg every 21 days with investigator-selected chemotherapy until disease progression or unacceptable toxicity. Stratification factors were chemotherapy regimen (carboplatin plus gemcitabine or capecitabine monotherapy), visceral (lung and/or liver) metastases and (initially) PD-L1 status. The primary endpoint was overall survival (OS), tested hierarchically in patients with PD-L1-positive tumours and then, if positive, in the modified intent-to-treat (mITT) population (all-comer patients randomised pre-August 2019). Secondary endpoints included progression-free survival (PFS), objective response rate (ORR) and safety. RESULTS Among 354 patients with rapidly relapsing PD-L1-positive aTNBC, 68% had a disease-free interval of <6 months and 73% received carboplatin/gemcitabine. The OS hazard ratio was 0.93 (95% confidence interval 0.73-1.20, P = 0.59; median 11.2 months with placebo versus 12.1 months with atezolizumab). mITT and subgroup results were consistent. Median PFS was 4 months across treatment arms and populations. ORRs were 28% with placebo versus 40% with atezolizumab. Adverse events (predominantly haematological) were similar between arms and as expected with atezolizumab plus carboplatin/gemcitabine or capecitabine following recent chemotherapy exposure. CONCLUSIONS OS, which is dismal in patients with TNBC relapsing within <12 months, was not improved by adding atezolizumab to chemotherapy. A biology-based definition of intrinsic resistance to immunotherapy in aTNBC is urgently needed to develop novel therapies for these patients in next-generation clinical trials.
Collapse
Affiliation(s)
- R Dent
- Division of Medical Oncology, National Cancer Center, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| | - F André
- Gustave Roussy, Université Paris Saclay, Villejuif
| | - A Gonçalves
- Aix Marseille University, CNRS, INSERM, Department of Medical Oncology, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - M Martin
- Hospital General Universitario Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - P Schmid
- Barts Cancer Institute, Centre for Experimental Cancer Medicine, London, UK
| | - F Schütz
- University Breast Unit, National Center for Tumor Diseases, Heidelberg
| | - S Kümmel
- Breast Unit, Kliniken Essen-Mitte, Essen; Department of Gynecology with Breast Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - S M Swain
- Georgetown University Medical Center and MedStar Health, Washington, USA
| | - A Bilici
- Department of Medical Oncology, Istanbul Medipol University Medical Faculty, Istanbul, Turkey
| | - D Loirat
- Medical Oncology Department, Institut Curie, Paris, France
| | | | - S-A Im
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul National University, Seoul
| | - Y H Park
- Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - M De Laurentis
- Department of Breast Oncology, IRCCS Istituto Nazionale Tumori Fondazione Giovanni Pascale, Napoli
| | - M Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology, IRCCS, Milan
| | - V Guarneri
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova; Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova
| | - G Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan; Università Vita-Salute San Raffaele, Milan, Italy. https://twitter.com/BianchiniGP
| | - H Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Z Kirchmayer Machackova
- Global Product Development/Medical Affairs Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - J Mouta
- Global Product Development/Medical Affairs Oncology, Roche Farmacêutica Química Lda, Amadora, Portugal
| | - R Deurloo
- Translational Medicine Oncology gRED, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - X Gan
- Product Development Safety, Roche (China) Holding Ltd, Shanghai, China
| | - M Fan
- Data Science, Hoffmann-La Roche Limited, Mississauga, Canada
| | - A Mani
- Global Product Development, Genentech/Roche, South San Francisco, USA
| | - A Swat
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - J Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona; Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid; IOB Madrid, Hospital Beata Maria Ana, Madrid, Spain
| |
Collapse
|
6
|
Ferreira Almeida C, Correia-da-Silva G, Teixeira N, Amaral C. Influence of tumor microenvironment on the different breast cancer subtypes and applied therapies. Biochem Pharmacol 2024; 223:116178. [PMID: 38561089 DOI: 10.1016/j.bcp.2024.116178] [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: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Despite the significant improvements made in breast cancer therapy during the last decades, this disease still has increasing incidence and mortality rates. Different targets involved in general processes, like cell proliferation and survival, have become alternative therapeutic options for this disease, with some of them already used in clinic, like the CDK4/6 inhibitors for luminal A tumors treatment. Nevertheless, there is a demand for novel therapeutic strategies focused not only on tumor cells, but also on their microenvironment. Tumor microenvironment (TME) is a very complex and dynamic system that, more than surrounding and supporting tumor cells, actively participates in tumor development and progression. During the last decades, it has become clear that the cellular and acellular components of TME differ between the various breast cancer subtypes and shape the differences regarding their severity and prognosis. The pivotal role of the TME in controlling tumor growth and influencing responses to therapy represents a potential source for novel targets and therapeutic strategies. In this review, we present a description of the multiple therapeutic options used for different breast cancer subtypes, as well as the influence that the TME may exert on the development of the disease and on the response to the distinct therapies, which in some cases may explain their failure by the occurrence of relapses and resistance. Furthermore, the ongoing studies focused on the use of TME components for developing potential cancer treatments are described.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Natércia Teixeira
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|