1
|
Xuhong J, Wu N, Shi Q, Tian H, Peng Z, Jiang J, Zhang J, Qi X. Targeted multimodal synergistic therapy of drug-resistant HER2-positive breast cancer by pyrotinib-ICG self-assembled nanoparticles. Am J Cancer Res 2024; 14:3976-3993. [PMID: 39267659 PMCID: PMC11387853 DOI: 10.62347/jzrn6919] [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: 04/22/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024] Open
Abstract
Neoadjuvant targeted therapy combining targeted agents with chemotherapy significantly improve survival rates of patients suffering from human epidermal receptor (HER2)-positive breast cancer (BC) in early or locally advanced stages. However, approximately 50% of patients fail to achieve a pathological complete response. In response, targeted photothermal therapy (PTT) and photodynamic therapy (PDT) have emerged as effective strategies to bolster primary tumors treatment. In this context, we developed a novel nanodrug, referred to as "P/ICG", which comprised of a tyrosine-kinase inhibitor pyrotinib and the photosensitizer indocyanine green (ICG). This formulation was created for the targeted and multimodal synergistic therapy of HER2-positive BC. Upon irradiation with near-infrared light, ICG generates high levels of intracellular reactive oxygen species and elevated temperature, enhancing chemotherapy effects of pyrotinib. This synergistic action boosts a highly effective anticancer effect promoting the ferroptosis pathway, providing an efficient therapeutic strategy for treating HER2-positive BC.
Collapse
Affiliation(s)
- Juncheng Xuhong
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University Shigatse 857000, Xizang, China
| | - Nisha Wu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
| | - Qiyun Shi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
- The Eighth Medical Center, Chinese PLA General Hospital Beijing 100091, China
| | - Hao Tian
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
| | - Zaihui Peng
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
| | - Jun Jiang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
| | - Jing Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University Guangzhou 510515, Guangdong, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University Chongqing 400038, China
| |
Collapse
|
2
|
Liefaard MC, van der Voort A, van Seijen M, Thijssen B, Sanders J, Vonk S, Mittempergher L, Bhaskaran R, de Munck L, van Leeuwen-Stok AE, Salgado R, Horlings HM, Lips EH, Sonke GS. Tumor-infiltrating lymphocytes in HER2-positive breast cancer treated with neoadjuvant chemotherapy and dual HER2-blockade. NPJ Breast Cancer 2024; 10:29. [PMID: 38637568 PMCID: PMC11026378 DOI: 10.1038/s41523-024-00636-4] [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: 07/28/2023] [Accepted: 04/05/2024] [Indexed: 04/20/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) have been associated with outcomes in HER2-positive breast cancer patients treated with neoadjuvant chemotherapy and trastuzumab. However, it remains unclear if TILs could be a prognostic and/or predictive biomarker in the context of dual HER2-targeting treatment. In this study, we evaluated the association between TILs and pathological response (pCR) and invasive-disease free survival (IDFS) in 389 patients with stage II-III HER2 positive breast cancer who received neoadjuvant anthracycline-containing or anthracycline-free chemotherapy combined with trastuzumab and pertuzumab in the TRAIN-2 trial. Although no significant association was seen between TILs and pCR, patients with TIL scores ≥60% demonstrated an excellent 3-year IDFS of 100% (95% CI 100-100), regardless of hormone receptor status, nodal stage and attainment of pCR. Additionally, in patients with hormone receptor positive disease, TILs as a continuous variable showed a trend to a positive association with pCR (adjusted Odds Ratio per 10% increase in TILs 1.15, 95% CI 0.99-1.34, p = 0.070) and IDFS (adjusted Hazard Ratio per 10% increase in TILs 0.71, 95% CI 0.50-1.01, p = 0.058). We found no interactions between TILs and anthracycline treatment. Our results suggest that high TIL scores might be able to identify stage II-III HER2-positive breast cancer patients with a favorable prognosis.
Collapse
Affiliation(s)
- M C Liefaard
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A van der Voort
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M van Seijen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B Thijssen
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - S Vonk
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L Mittempergher
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - R Bhaskaran
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - L de Munck
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - A E van Leeuwen-Stok
- Dutch Breast Cancer Research Group, BOOG Study Center, Amsterdam, The Netherlands
| | - R Salgado
- Department of Pathology, GZA-ZNA Hospitals, Wilrijk, Antwerp, Belgium
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - E H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Dey S, Mondal A, Aash A, Mukherjee R, Kolay S, Murmu N, Murmu N, Giri B, Molla MR. Poly-β-thioester-Based Cross-Linked Nanocarrier for Cancer Cell Selectivity over Normal Cells and Cellular Apoptosis by Triggered Release of Parthenolide, an Anticancer Drug. ACS APPLIED BIO MATERIALS 2024; 7:1214-1228. [PMID: 38326023 DOI: 10.1021/acsabm.3c01121] [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: 02/09/2024]
Abstract
Breast cancer is the most prevalent and aggressive type of cancer, causing high mortality rates in women globally. Many drawbacks and side effects of the current chemotherapy force us to develop a robust chemotherapeutic system that can deal with off-target hazards and selectively combat cancer growth, invasiveness, and cancer-initiating cells. Here, a pH-responsive cross-linked nanocarrier (140-160 nm) endowed with poly-β-thioester functionality (CBAPTL) has been sketched and fabricated for noncovalent firm encapsulation of anticancer drug, parthenolide (PTL) at physiological pH (7.4), which enables sustain release of PTL at relevant endosomal pH (∼5.0-5.3). For this, a bolaamphiphilic molecule integrated with β-thioester and acrylate functionality was synthesized to fabricate the pH-responsive poly-β-thioester-based cross-linked nanocarrier via Michael addition click reactions in water. The poly-β-thioester functionality of CBAPTL hydrolyzes at endosomal acidic conditions, thus leading to the selective release of PTL inside the cancer cell. Cross-linked nanocarriers exhibit high serum stability, dilution insensitivity, and targeted cellular uptake at tumor microenvironment (TME), contrasting normal cells. In vitro study using human MCF-7 breast cancer cells demonstrated that CBAPTL exhibited selective cytotoxicity, reduced clonogenic potential, increased reactive oxygen species (ROS) generation, and arrested the progression of the cell cycle at the G0/G1 phase efficiently. CBAPTL induced apoptosis via downregulating pro-proliferative protein Bcl-2 and upregulating proapoptotic proteins p53, BAD, p21, and cleaved PARP-1. CBAPTL inhibited proliferating signaling by suppressing AKT phosphorylation and p38 expression. CBAPTL also blocked the invasion and migration of MCF-7 cells. CBAPTL effectively inhibits primary and secondary mammosphere formation, thereby preventing cancer-initiating cells' growth. Conversely, CBAPTL has negligible effect on human red blood cells (RBCs) and peripheral blood mononuclear cells (PBMCs). These findings highlight the superior efficacy of CBAPTL compared to PTL alone in suppressing cancer cell growth, inducing apoptosis, and preventing invasiveness of MCF-7 cells. Thus, CBAPTL could be considered a possible selective chemotherapeutic cargo against breast cancer without affecting normal cells.
Collapse
Affiliation(s)
- Sananda Dey
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Arun Mondal
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Asmita Aash
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Rimi Mukherjee
- Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, Kolkata 700026, West Bengal, India
| | - Soumya Kolay
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Nensina Murmu
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Nabendu Murmu
- Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, Kolkata 700026, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | | |
Collapse
|
4
|
Gamrani S, Akhouayri L, Boukansa S, Karkouri M, El Fatemi H. The Clinicopathological Features and Prognostic Significance of HER2-Low in Early Breast Tumors Patients Prognostic Comparison of HER-Low and HER2-Negative Breast Cancer Stratified by Hormone Receptor Status. Breast J 2023; 2023:6621409. [PMID: 38075551 PMCID: PMC10703525 DOI: 10.1155/2023/6621409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023]
Abstract
Introduction There has been increased interest in HER2-low breast tumors recently, as these tumors may have distinct clinical and molecular characteristics compared to HER2-negative and HER2-positive tumors. A new nomenclature has been proposed for HER2 1+ and HER2 2+ tumors that are confirmed negative according to fluorescence in situ hybridization (FISH). These tumors are now referred to as HER2-low, and it is thought that they may represent a distinct subtype of breast cancer that warrants further investigation. In this study, we aimed to evaluate the clinicopathological characteristics and prognostic impact of this particular subtype in a North-African context where HER2-low breast cancer is a relatively understudied subtype, particularly in non-Western populations. Methods We conducted a retrospective cohort study on 1955 breast tumors in Moroccan patients over 10 years, collected at the Pathology Department of Ibn Rochd University Hospital in Casablanca and at the pathology department of Hassan II University Hospital in Fes. We elaborated on their complete immunohistochemical profile based on the main breast cancer biomarkers: Ki-67, HER2, estrogen, and progesterone receptors. Their overall survival and disease free survival data were also retrieved from their respective records. Results Out of 1955 BC patients, 49.3% were classified as HER2-low; of which 80.7% and 19.2% were hormone receptors positive and negative, respectively. The clinicopathologic features indicate that HER2-low subtype tumors behave much more like HER2-positive than HER2-negative tumors. The survival analysis showed that the HER2-low subtype-belonging patients present significantly the poorest prognosis in disease-free survival (p = 0.003) in comparison with HER2-negative ones. When considering the hormonal status, hormonal-dependent tumors show a significant difference according to HER2 subtypes in disease-free survival (p < 0.001). Yet no significant difference was shown among hormonal negative tumors. Moreover, patients with hormonal positive tumors and simultaneously belonging to the HER2-low subgroup present a significantly good prognosis in overall survival compared to the ones with hormonal negative tumors (p = 0.008). Conclusion Our study has shown that the HER2-low phenotype is common among hormone-positive patients. The clinicopathological features and prognostic data indicate that the hormonal receptors effect and HER2 heterogeneity are crucial factors to consider. It is important to note that this particular subgroup is different from the HER2-negative one and should not be treated in the same way. Therefore, this study offers a new perspective in the management of HER2-low patients and can serve as a basis for future prospective analyses.
Collapse
Affiliation(s)
- Sanaa Gamrani
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Laila Akhouayri
- Dipartimento di Scienze Cliniche e Biologiche, Università Degli Studi Di Torino, Via Giuseppe Verdi, Torino, Italy
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
| | - Sara Boukansa
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mehdi Karkouri
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
- Department of Pathology, Ibn Rochd University Hospital, Rue des Hôpitaux, Casablanca, Morocco
| | - Hinde El Fatemi
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
5
|
Cataldo ML, De Placido P, Esposito D, Formisano L, Arpino G, Giuliano M, Bianco R, De Angelis C, Veneziani BM. The effect of the alpha-specific PI3K inhibitor alpelisib combined with anti-HER2 therapy in HER2+/PIK3CA mutant breast cancer. Front Oncol 2023; 13:1108242. [PMID: 37469415 PMCID: PMC10353540 DOI: 10.3389/fonc.2023.1108242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Background HER2 is amplified or overexpressed in around 20% of breast cancers (BC). HER2-targeted therapies have significantly improved the prognosis of patients with HER2+ BC, however, de novo and acquired resistance to anti-HER2 treatment is common. Activating mutations in the PIK3CA gene are reported in ∼30% of HER2+ BC and are associated with resistance to anti-HER2 therapies and a poor prognosis. Here, we investigated the in vitro and in vivo antitumor efficacy of the alpha-specific PI3K inhibitor alpelisib alone or in combination with anti-HER2 therapy using a panel of HER2+ BC cell lines. We also generated models of acquired resistance to alpelisib to investigate the mechanisms underlying resistance to alpha-specific PI3K inhibition. Materials and methods PIK3CA mutant (HCC1954, KPL4 and JMT1) and wild-type (BT474 and SKBR3) HER2+ BC cell lines were used. The HCC1954 and KPL4 cells were chronically exposed to increasing concentrations of alpelisib or to alpelisib + trastuzumab in order to generate derivatives with acquired resistance to alpelisib (AR) and to alpelisib + trastuzumab (ATR). The transcriptomic profiles of HCC1954, KPL4 and their AR and ATR derivatives were determined by RNA sequencing. Cell growth was assessed by MTT assay. Changes in the protein levels of key PI3K pathway components were assessed by Western blotting. Gene expression, cellular and patients' data from the Cancer Dependency Map (DepMap) and KMPlot datasets were interrogated. Results HER2+ BC cell lines harboring activating mutations in PIK3CA were less sensitive to single or dual anti-HER2 blockade compared to PIK3CA wild-type cells. Alpelisib treatment resulted in dose-dependent inhibition of the growth of cells with or without PIK3CA mutations and enhanced the antitumor efficacy of anti-HER2 therapies in vitro. In addition, alpelisib greatly delayed tumor growth of HCC1954 xenografts in vivo. Functional annotation of the significantly differentially expressed genes suggested the common activation of biological processes associated with oxidation reduction, cell proliferation, immune response and RNA synthesis in alpelisib-resistant models compared with native cells. Eight commonly upregulated genes (log2 fold-change >1, False Discovery Rate [FDR] <0.05) in models with acquired resistance to alpelisib or alpelisib + trastuzumab were identified. Among these, AKR1C1 was associated with alpelisib-resistance in vitro and with a poor prognosis in patients with HER2+ BC. Conclusions Our findings support the use of an alpha-selective PI3K inhibitor to overcome the therapeutic limitations associated with single or dual HER2 blockade in PIK3CA-mutant HER2+ breast cancer. Future studies are warranted to confirm the potential role of candidate genes/pathways in resistance to alpelisib.
Collapse
Affiliation(s)
- Maria Letizia Cataldo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Daniela Esposito
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
6
|
Liefaard MC, van der Voort A, van Ramshorst MS, Sanders J, Vonk S, Horlings HM, Siesling S, de Munck L, van Leeuwen AE, Kleijn M, Mittempergher L, Kuilman MM, Glas AM, Wesseling J, Lips EH, Sonke GS. BluePrint molecular subtypes predict response to neoadjuvant pertuzumab in HER2-positive breast cancer. Breast Cancer Res 2023; 25:71. [PMID: 37337299 DOI: 10.1186/s13058-023-01664-x] [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: 11/15/2022] [Accepted: 05/25/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND The introduction of pertuzumab has greatly improved pathological complete response (pCR) rates in HER2-positive breast cancer, yet effects on long-term survival have been limited and it is uncertain which patients derive most benefit. In this study, we determine the prognostic value of BluePrint subtyping in HER2-positive breast cancer. Additionally, we evaluate its use as a biomarker for predicting response to trastuzumab-containing neoadjuvant chemotherapy with or without pertuzumab. METHODS From a cohort of patients with stage II-III HER2-positive breast cancer who were treated with neoadjuvant chemotherapy and trastuzumab with or without pertuzumab, 836 patients were selected for microarray gene expression analysis, followed by readout of BluePrint standard (HER2, Basal and Luminal) and dual subtypes (HER2-single, Basal-single, Luminal-single, HER2-Basal, Luminal-HER2, Luminal-HER2-Basal). The associations between subtypes and pathological complete response (pCR), overall survival (OS) and breast cancer-specific survival (BCSS) were assessed, and pertuzumab benefit was evaluated within the BluePrint subgroups. RESULTS BluePrint results were available for 719 patients. In patients with HER2-type tumors, the pCR rate was 71.9% in patients who received pertuzumab versus 43.5% in patients who did not (adjusted Odds Ratio 3.43, 95% CI 2.36-4.96). Additionally, a significantly decreased hazard was observed for both OS (adjusted hazard ratio [aHR] 0.45, 95% CI 0.25-0.80) and BCSS (aHR 0.46, 95% CI 0.24-0.86) with pertuzumab treatment. Findings were similar in the HER2-single subgroup. No significant benefit of pertuzumab was seen in other subtypes. CONCLUSIONS In patients with HER2-type or HER2-single-type tumors, pertuzumab significantly improved the pCR rate and decreased the risk of breast cancer mortality, which was not observed in other subtypes. BluePrint subtyping may be valuable in future studies to identify patients that are likely to be highly sensitive to HER2-targeting agents.
Collapse
Affiliation(s)
- M C Liefaard
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A van der Voort
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M S van Ramshorst
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - S Vonk
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Core Facility Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - S Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
- Department of Health Technology and Services Research, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - L de Munck
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | - A E van Leeuwen
- Dutch Breast Cancer Research Group, BOOG Study Center, Amsterdam, The Netherlands
| | - M Kleijn
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - L Mittempergher
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - M M Kuilman
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - A M Glas
- Department of Research and Development, Agendia NV, Amsterdam, The Netherlands
| | - J Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - E H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Wang Z, Tan M, Su W, Huang W, Zhang J, Jia F, Cao G, Liu X, Song H, Ran H, Nie G, Wang H. Persistent Degradation of HER2 Protein by Hybrid nanoPROTAC for Programmed Cell Death. J Med Chem 2023; 66:6263-6273. [PMID: 37092695 DOI: 10.1021/acs.jmedchem.3c00013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Proteolysis-targeting chimera (PROTAC) has emerged as a promising strategy for degrading proteins of interest. Peptide-based PROTACs offer several advantages over small-molecule-based PROTACs, such as high specificity, low toxicity, and large protein-protein interaction surfaces. However, peptide-based PROTACs have several intrinsic shortcomings that strongly limit their application including poor cell permeability and low stability and potency. Herein, we designed a nanosized hybrid PROTAC (GNCTACs) to target and degrade human epidermal growth factor receptor 2 (HER2) in tumor cells. Gold nanoclusters (GNCs) were utilized to connect HER2-targeting peptides and cereblon (CRBN)-targeting ligands. GNCTACs could overcome the intrinsic barriers of peptide-based PROTACs, efficiently delivering HER2-targeting peptides in the cytoplasm and protecting them from degradation. Furthermore, a fasting-mimicking diet was applied to enhance the cellular uptake and proteasome activity. Consequently, more than 95% of HER2 in SKBR3 cells was degraded by GNCTACs, and the degradation lasted for at least 72 h, showing a catalytic-like reaction.
Collapse
Affiliation(s)
- Zhihang Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mixiao Tan
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing 400010, China
| | - Wen Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Wenping Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuhao Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoliang Cao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xinyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Haohao Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Haitao Ran
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing 400010, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Lander EM, Rappazzo KC, Huang LC, Hu JR, Chen H, Shyr Y, Abramson VG. Using the HER2/CEP17 FISH Ratio to Predict Pathologic Complete Response Following Neoadjuvant Anti-HER2 Doublet Therapy in HER2+ Breast Cancer. Oncologist 2023; 28:123-130. [PMID: 36495309 PMCID: PMC9907044 DOI: 10.1093/oncolo/oyac247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Clinical trials of HER2-directed therapy that omit neoadjuvant conventional chemotherapy for HER+ breast cancer demonstrate that a subset of patients still obtains a pCR. Identifying tumor characteristics which predict pCR may help select patients for de-escalated neoadjuvant dual HER2-targeted treatment without chemotherapy. This is the first study evaluating the HER2/CEP17 ratio by FISH as a biomarker to predict pCR among patients who received neoadjuvant anti-HER2 regimens without chemotherapy. PATIENTS AND METHODS Data from patients with locally advanced HER2+ breast cancer who received neoadjuvant dual HER2-targeted therapy without conventional chemotherapy from a single center was retrospectively reviewed. All patients were enrolled in one of 3 clinical trials evaluating chemotherapy de-escalation. Logistic regression modeling assessed for a relationship between the HER2/CEP17 FISH ratio obtained from baseline tissue biopsy and pCR based on pathology at the time of definitive breast surgery following neoadjuvant treatment. RESULTS Following neoadjuvant treatment with dual HER2-targeted therapies in 56 patients, the probability of pCR was 73% among patients with a HER2 ratio of 13.1 compared to a probability of 38% among patients with HER2 ratio of 5.5 (OR 4.14, 95% CI 1.44-11.89; P = .012). This positive association persisted after controlling for different treatment regimens administered (OR 2.87, 95% CI 0.9-9.18, P = .020). CONCLUSIONS These data found a positive association between the HER2/CEP17 FISH ratio and pCR following neoadjuvant dual HER2-targeted therapy without chemotherapy. Larger prospective studies are needed to validate the HER2 ratio as a biomarker to select patients for neoadjuvant dual anti-HER2 therapy without chemotherapy.
Collapse
Affiliation(s)
- Eric M Lander
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Katherine C Rappazzo
- Department of Hematology/Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Li-Ching Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Jiun-Ruey Hu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Vandana G Abramson
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
9
|
He L, Shen X, Liu Y, Gao L, Wu J, Yu C, Li G, Wang X, Shao X. The reversal of anti-HER2 resistance in advanced HER2-positive breast cancer using apatinib: two cases reports and literature review. Transl Cancer Res 2022; 11:4206-4217. [PMID: 36523304 PMCID: PMC9745359 DOI: 10.21037/tcr-22-2483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/15/2022] [Indexed: 09/29/2024]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2)-targeted treatment has yielded a notable clinical benefit in patients with HER2-positive breast cancer. However, nearly 50% of patients still suffer disease progression due to resistance to HER2-targeted therapy. After the failure of macromolecular monoclonal antibodies (mAbs) therapy, we can choose small molecule tyrosine kinase inhibitors (TKIs) to reverse HER2 resistance. When small molecule TKIs resistance, we can use mAb combined with small molecule TKI, or antibody-drug conjugates (ADCs) to reverse HER2 resistance. However, then due to the availability and price of ADCs, patients may not use them. Consequently, new therapeutic approaches are required to overcome HER2-targeted therapy resistance. Vascular endothelial growth factor and its receptors (VEGF/VEGFRs) promote tumor angiogenesis. They can also activate downstream signaling pathways to promote tumorigenesis. VEGFR is a key regulator of the tyrosine kinase signaling pathway and may be a potential target in HER2-positive breast cancer. Apatinib is a small molecule TKI that specifically binds to VEGFR2 and thus exerts an antitumor effect. Although there is no definite indication for apatinib in breast cancer, it has a good benefit in advanced gastric cancer. CASE DESCRIPTION The two patients we reported were both HER2-positive breast cancer who we followed for more than 10 years. After the failure of multi-line anti-HER2 treatment, apatinib combined with anti-HER2 treatment had PFS of 8.4 months and 10.6 months, respectively. One patient had grade 2 hand-foot syndrome. The other had grade 2 leukopenia and grade 2 thrombocytopenia, both of them improved after control. And the best response of them were PR and SD, respectively. CONCLUSIONS Our cases demonstrate that, in HER2-positive breast cancer patients with HER2-targeted resistance, apatinib may be able to reverse HER2 resistance. These two cases suggest an alternative method for clinical HER2-targeted treatment of drug-resistant breast cancer patients and provide new insights for future research.
Collapse
Affiliation(s)
- Libin He
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiabo Shen
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yiyuan Liu
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Gao
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayi Wu
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chang Yu
- Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Guangliang Li
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
10
|
Zhao M, Liu J, Tang Y, Zhang L, Ge X, Chen M, Wen Q, Zhu L, Ma Q. Hyaluronidase responsive second near-infrared fluorescent nanocomplex for combined HER2 blockade and chemotherapy of HER2+ breast cancer. BIOMATERIALS ADVANCES 2022; 141:213115. [PMID: 36115156 DOI: 10.1016/j.bioadv.2022.213115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/22/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
The human epidermal growth factor receptor-2-positive (HER2+) type is aggressive and has poor prognosis. Although anti-HER2 therapy alone or in combination with other treatment regimens showed significant improvement in survival outcomes, breast cancer patients are still suffering from tumor relapse and severe dose-limiting side effects. Thus, there is still an unmet challenge to develop effective therapeutic agents for HER2+ breast cancer treatment with minimized side effects. Herein, we produced a stimuli-responsive and tumor-targeted hyaluronic acid (HA) nanocomplex that combined HER2 blockade and chemotherapy for effective HER2+ breast cancer therapy. A hydrophobic NIR-II dye, IR1048, was covalently linked with HA to form a spherical HA-IR1048 nanoparticle (HINP), with Herceptin conjugated on the surface and paclitaxel (PTX) encapsulated inside. The fluorescent signals from the yielding Her-HINP/PTX are quenched originally, but a strong NIR-II signal is generated when HINP is degraded by the hyaluronidase that is overexpressed in breast tumors, thus allowing the tracking and visualization of Herceptin and PTX accumulation. Her-HINP/PTX peaked in HER2+ tumors at 24 h post injection as imaged by NIR-II fluorescent imaging. A significantly improved tumor growth inhibition effect was observed after five systemic treatments compared to single PTX (3.71 ± 0.41 times) or Herceptin (5.98 ± 0.51 times) treatment in a HER2-overexpressed breast cancer mouse model with prolonged survival. Collectively, the designed Her-HINP/PTX presents a new hyaluronidase-responsive and HER2 blockade nanoformulation that can visualize the accumulation of nanocomplexes and release drugs inside tumors for combined HER2+ breast cancer therapy with a great promise for translational study. STATEMENT OF SIGNIFICANCE: The high expressions of a protein called human epidermal growth factor receptor 2 (HER2) in breast tumors make this subtype of cancer aggressive. Currently, chemotherapy combined with a HER2 antibody, Herceptin, is a preferred approach for HER2-positive breast cancer therapy. However, these breast cancer patients still suffer from tumor relapse and severe side effects because various therapeutic agents have inherent different biodistributions, resulting in insufficient treatment effects and unfavorable normal organ uptake of these therapeutic agents. Herein, we produced a nanocomplex carrying both Herceptin and chemotherapy drug to simultaneously deliver two drugs into tumors for efficient HER2+ tumor treatment with minimized side effects, providing new insights for designing a combined therapy strategy.
Collapse
Affiliation(s)
- Min Zhao
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Junzhi Liu
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Lumeng Zhang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Xiaoguang Ge
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China; MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, China
| | - Minglong Chen
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Qiang Wen
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Lei Zhu
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Qingjie Ma
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| |
Collapse
|
11
|
The Prognosis and Predictive Value of Estrogen Negative/Progesterone Positive (ER−/PR+) Phenotype: Experience of 1159 Primary Breast Cancer from a Single Institute. Breast J 2022; 2022:9238804. [PMID: 35711896 PMCID: PMC9187286 DOI: 10.1155/2022/9238804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022]
Abstract
Breast cancer is a serious worldwide public health problem and is currently the most common cancer overall. Its endocrine therapy is related to the expression of the steroid hormones, estrogen receptor (ER), and progesterone receptor (PR). Breast cancers can be presented under multiple profiles of steroid hormones: ER(−)/PR(+), ER(+)/PR(−), double-positive/negative ER, and PR. 2–8% of all breast cancers express only PR (ER−/PR+) which is an abnormal phenotype, with less known about their behaviors and outcomes. Our study was performed on a large and well-characterized database of primary breast cancer from 2012 to 2019, up to 1159 cases. These cases were divided according to ER and PR expression, as we put all of our focus on ER-negative/PR-positive group, more specifically ER−/PR+/HER2+ and ER−/PR+/HER2− gene expressions, to highlight their features and find a pattern that links HR (hormone receptors) profiles and breast cancer subtypes. Out of the informative cases, 94 patients (8%) had ER−/PR+ breast cancers, while 676 (58.4%) had ER+/PR+, 88 (7.6%) had ER+/PR−, and 164 (14.2%) had ER−/PR− tumors. The ER−/PR+ group was statistically correlated with a high risk of recurrence and death in midway between the double-negative and double-positive HR. According to HER2 status, a low DFS was observed in patients ER−/PR+/HER2−, which is closer to the DFS of TNBC cases but worse than ER+/PR any. On the other side, the ER−/PR+/HER2+ showed also a poorer DFS closer to the HER2+ subgroup in between TNBC and ER+/PR any. The clinicopathological features of the ER−/PR+/HER2− and ER−/PR+ HER2+ have distinguished the patients into two groups with a difference in some clinicopathological characteristics: both groups had closer OS estimation, which was worse than ER−/PR any and better than TNBC and HER2. The ER−/PR+/HER2− seems to increase the risk of recurrence than ER−/PR+/HER2+ when compared to ER+/PR any. On the other hand, the ER−/PR+/HER2+ seems to increase the risk of death more than ER−/PR+/HER2− in comparison with ER+/PR any. Our results support that ER−/PR+ tumors really exist and are rare and clinically and biologically distinct subtypes of breast cancer. In addition, our analysis, which was based on dividing the groups according to HER2 expression, has revealed the existence of two distinct groups; this gave the ER−/PR+ subgroup a heterogeneity characterization. Moreover, this breast cancer subtype should not be treated as a luminal tumor but rather according to the HER2 expression status.
Collapse
|
12
|
Lüftner D, Peipp M. New Therapeutic Strategies in Advanced Nonoperable or Metastatic HER2-positive Breast Cancer. Geburtshilfe Frauenheilkd 2021; 81:666-678. [PMID: 34168380 PMCID: PMC8216785 DOI: 10.1055/a-1471-4063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/27/2021] [Indexed: 11/24/2022] Open
Abstract
Despite therapeutic gains in the treatment of HER2-positive (HER2: human epidermal growth factor receptor 2) advanced/metastatic breast cancer, there remains an urgent need for more effective treatment options. At present, there is no definitive approved standard therapy beyond second-line treatment. One of the major challenges is overcoming treatment resistance. Depending on the underlying resistance mechanism, different strategies are being pursued for new innovative treatment concepts in HER2-positive breast cancer. Specifically designed antibodies for targeted therapy are one important focus to successfully meet these challenges. Trastuzumab deruxtecan (T-DXd, DS-8201a), an optimised antibody drug conjugate (ADC) is in clinical trials, showing promising outcomes in patients with advanced, nonoperable or metastatic HER2-positive breast cancer who had already undergone intensive prior treatment. Based on this data, T-DXd has already been approved in the US and Japan for HER2-positive advanced nonoperable and metastatic breast cancer - in the US after at least two prior anti-HER2 targeted treatment lines and in Japan after prior chemotherapy. T-DXd represents successful "antibody engineering". Since the beginning of the year, T-DXd has also been approved in Europe as monotherapy for inoperable or metastatic HER2-positive breast cancer in patients who are pretreated with at least two anti-HER2 directed therapies. This paper presents strategies for improving treatment options in advanced nonoperable and metastatic HER2-positive breast cancer, with the development of T-DXd as an example.
Collapse
Affiliation(s)
- Diana Lüftner
- Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Peipp
- Sektion für Stammzelltransplantation und Immuntherapie, Dr. Mildred-Scheel-Haus, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
13
|
Ligorio F, Pellegrini I, Castagnoli L, Vingiani A, Lobefaro R, Zattarin E, Santamaria M, Pupa SM, Pruneri G, de Braud F, Vernieri C. Targeting lipid metabolism is an emerging strategy to enhance the efficacy of anti-HER2 therapies in HER2-positive breast cancer. Cancer Lett 2021; 511:77-87. [PMID: 33961924 DOI: 10.1016/j.canlet.2021.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022]
Abstract
De novo or acquired resistance of cancer cells to currently available Human Epidermal Growth Factor Receptor 2 (HER2) inhibitors represents a clinical challenge. Several resistance mechanisms have been identified in recent years, with lipid metabolism reprogramming, a well-established hallmark of cancer, representing the last frontier of preclinical and clinical research in this field. Fatty Acid Synthase (FASN), the key enzyme required for fatty acids (FAs) biosynthesis, is frequently overexpressed/activated in HER2-positive (HER2+) breast cancer (BC), and it crucially sustains HER2+ BC cell growth, proliferation and survival. After the synthesis of new, selective and well tolerated FASN inhibitors, clinical trials have been initiated to test if these compounds are able to re-sensitize cancer cells with acquired resistance to HER2 inhibition. More recently, the upregulation of FA uptake by cancer cells has emerged as a potentially new and targetable mechanism of resistance to anti-HER2 therapies in HER2+ BC, thus opening a new era in the field of targeting metabolic reprogramming in clinical setting. Here, we review the available preclinical and clinical evidence supporting the inhibition of FA biosynthesis and uptake in combination with anti-HER2 therapies in patients with HER2+ BC, and we discuss ongoing clinical trials that are investigating these combination approaches.
Collapse
Affiliation(s)
- Francesca Ligorio
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Ilaria Pellegrini
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Lorenzo Castagnoli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Andrea Vingiani
- Pathology Department, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy; Department of Oncology and Haematology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Emma Zattarin
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Marzia Santamaria
- IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, Italy
| | - Serenella M Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Giancarlo Pruneri
- Pathology Department, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy; Department of Oncology and Haematology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy; Department of Oncology and Haematology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy; IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, Italy.
| |
Collapse
|
14
|
McNamara KL, Caswell-Jin JL, Joshi R, Ma Z, Kotler E, Bean GR, Kriner M, Zhou Z, Hoang M, Beechem J, Zoeller J, Press MF, Slamon DJ, Hurvitz SA, Curtis C. Spatial proteomic characterization of HER2-positive breast tumors through neoadjuvant therapy predicts response. NATURE CANCER 2021; 2:400-413. [PMID: 34966897 PMCID: PMC8713949 DOI: 10.1038/s43018-021-00190-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The addition of HER2-targeted agents to neoadjuvant chemotherapy has dramatically improved pathological complete response (pCR) rates in early-stage, HER2-positive breast cancer. Nonetheless, up to 50% of patients have residual disease after treatment, while others are likely overtreated. Here, we performed multiplex spatial proteomic characterization of 122 samples from 57 HER2-positive breast tumors from the neoadjuvant TRIO-US B07 clinical trial sampled pre-treatment, after 14-21 d of HER2-targeted therapy and at surgery. We demonstrated that proteomic changes after a single cycle of HER2-targeted therapy aids the identification of tumors that ultimately undergo pCR, outperforming pre-treatment measures or transcriptomic changes. We further developed and validated a classifier that robustly predicted pCR using a single marker, CD45, measured on treatment, and showed that CD45-positive cell counts measured via conventional immunohistochemistry perform comparably. These results demonstrate robust biomarkers that can be used to enable the stratification of sensitive tumors early during neoadjuvant HER2-targeted therapy, with implications for tailoring subsequent therapy.
Collapse
Affiliation(s)
- Katherine L. McNamara
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L. Caswell-Jin
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rohan Joshi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhicheng Ma
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Eran Kotler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gregory R. Bean
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Zoey Zhou
- NanoString Technologies, Seattle, WA, USA
| | | | | | - Jason Zoeller
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Michael F. Press
- Department of Pathology Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dennis J. Slamon
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Sara A. Hurvitz
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christina Curtis
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Correspondence and requests for materials should be addressed to C.C.
| |
Collapse
|
15
|
Zubair M, Wang S, Ali N. Advanced Approaches to Breast Cancer Classification and Diagnosis. Front Pharmacol 2021; 11:632079. [PMID: 33716731 PMCID: PMC7952319 DOI: 10.3389/fphar.2020.632079] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
The International Agency for Research on Cancer (IARC) has recently reported a 66% increase in the global number of cancer deaths since 1960. In the US alone, about one in eight women is expected to develop invasive breast cancer(s) (breast cancer) at some point in their lifetime. Traditionally, a BC diagnosis includes mammography, ultrasound, and some high-end molecular bioimaging. Unfortunately, these techniques detect BC at a later stage. So early and advanced molecular diagnostic tools are still in demand. In the past decade, various histological and immuno-molecular studies have demonstrated that BC is highly heterogeneous in nature. Its growth pattern, cytological features, and expression of key biomarkers in BC cells including hormonal receptor markers can be utilized to develop advanced diagnostic and therapeutic tools. A cancer cell's progression to malignancy exhibits various vital biomarkers, many of which are still underrepresented in BC diagnosis and treatment. Advances in genetics have also enabled the development of multigene assays to detect genetic heterogeneity in BC. However, thus far, the FDA has approved only four such biomarkers-cancer antigens (CA); CA 15-3, CA 27-29, Human epidermal growth factor receptor 2 (HER2), and circulating tumor cells (CTC) in assessing BC in body fluids. An adequately structured portable-biosensor with its non-invasive and inexpensive point-of-care analysis can quickly detect such biomarkers without significantly compromising its specificity and selectivity. Such advanced techniques are likely to discriminate between BC and a healthy patient by accurately measuring the cell shape, structure, depth, intracellular and extracellular environment, and lipid membrane compositions. Presently, BC treatments include surgery and systemic chemo- and targeted radiation therapy. A biopsied sample is then subjected to various multigene assays to predict the heterogeneity and recurrence score, thus guiding a specific treatment by providing complete information on the BC subtype involved. Thus far, we have seven prognostic multigene signature tests for BC providing a risk profile that can avoid unnecessary treatments in low-risk patients. Many comparative studies on multigene analysis projected the importance of integrating clinicopathological information with genomic-imprint analysis. Current cohort studies such as MINDACT, TAILORx, Trans-aTTOM, and many more, are likely to provide positive impact on long-term patient outcome. This review offers consolidated information on currently available BC diagnosis and treatment options. It further describes advanced biomarkers for the development of state-of-the-art early screening and diagnostic technologies.
Collapse
Affiliation(s)
- M. Zubair
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - S. Wang
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - N. Ali
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, United States
| |
Collapse
|
16
|
Hussain Qureshi MF, Shah M, Lakhani M, Abubaker ZJ, Mohammad D, Farhan H, Zia I, Tafveez R, Khan ST, Rubina G, Shamim M, Ghulam H. Gene signatures of cyclin-dependent kinases: a comparative study in naïve early and advanced stages of lung metastasis breast cancer among pre- and post-menopausal women. Genes Cancer 2021; 12:1-11. [PMID: 33868579 PMCID: PMC8018704 DOI: 10.18632/genesandcancer.209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/03/2020] [Indexed: 11/25/2022] Open
Abstract
The Human epidermal growth factor receptor 2 positive (HER2+) breast cancer (BC) is a more aggressive tumor with 5 years median survival rates after metastasis. Despite successful treatment, unfortunately, the majority of affected patients die. Defects in cell cycle and transcription regulation phases which are governed by cyclin-dependent kinases (CDKs) are the hallmark of many cancers that underpinning the progression of the disease. Therefore, the current study looked at the alteration of six CDKs mRNA expression levels in pre- and postmenopausal lung metastasis BC groups; the majority were HER2+. Two hundred pre-and postmenopausal lung metastasis breast cancer and healthy control blood samples were taken for RNA isolation. Quantitative PCR was done for CDKs mRNA expressions. We observed overexpression of CDK11, CDK12, CDK17, CDK18, and CDK19 in both pre- and postmenopausal groups. However, CDK20 showed progressive downregulation from early to advanced stages in both groups of patients. Collectively, this data revealed that CDKs overexpression levels may predict BC disease progression and provide further rationale for novel anticancer strategies for HER2+ BC cancers.
Collapse
Affiliation(s)
| | - Muzna Shah
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | - Mahira Lakhani
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | | | - Danish Mohammad
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | - Hira Farhan
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | - Iman Zia
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | - Rida Tafveez
- Medical Students, Ziauddin University, Clifton, Karachi, Pakistan
| | | | - Ghani Rubina
- Department of Biochemistry, Sohail University, Karachi, Pakistan
| | - Mushtaq Shamim
- Department of Biochemistry, Ziauddin University, Clifton, Karachi, Pakistan
| | - Haider Ghulam
- Oncology Department, Jinnah Postgraduate Medical Center, Karachi, Pakistan
| |
Collapse
|
17
|
Di Modica M, Gargari G, Regondi V, Bonizzi A, Arioli S, Belmonte B, De Cecco L, Fasano E, Bianchi F, Bertolotti A, Tripodo C, Villani L, Corsi F, Guglielmetti S, Balsari A, Triulzi T, Tagliabue E. Gut Microbiota Condition the Therapeutic Efficacy of Trastuzumab in HER2-Positive Breast Cancer. Cancer Res 2021; 81:2195-2206. [PMID: 33483370 DOI: 10.1158/0008-5472.can-20-1659] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/17/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022]
Abstract
Emerging evidence indicates that gut microbiota affect the response to anticancer therapies by modulating the host immune system. In this study, we investigated the impact of gut microbiota on immune-mediated trastuzumab antitumor efficacy in preclinical models of HER2-positive breast cancer and in 24 patients with primary HER2-positive breast cancer undergoing trastuzumab-containing neoadjuvant treatment. In mice, the antitumor activity of trastuzumab was impaired by antibiotic administration or fecal microbiota transplantation from antibiotic-treated donors. Modulation of the intestinal microbiota was reflected in tumors by impaired recruitment of CD4+ T cells and granzyme B-positive cells after trastuzumab treatment. Antibiotics caused reductions in dendritic cell (DC) activation and the release of IL12p70 upon trastuzumab treatment, a mechanism that was necessary for trastuzumab effectiveness in our model. In patients, lower α-diversity and lower abundance of Lachnospiraceae, Turicibacteraceae, Bifidobacteriaceae, and Prevotellaceae characterized nonresponsive patients (NR) compared with those who achieved pathologic complete response (R), similar to antibiotic-treated mice. The transfer of fecal microbiota from R and NR into mice bearing HER2-positive breast cancer recapitulated the response to trastuzumab observed in patients. Fecal microbiota β-diversity segregated patients according to response and positively correlated with immune signature related to interferon (IFN) and NO2-IL12 as well as activated CD4+ T cells and activated DCs in tumors. Overall, our data reveal the direct involvement of the gut microbiota in trastuzumab efficacy, suggesting that manipulation of the gut microbiota is an optimal future strategy to achieve a therapeutic effect or to exploit its potential as a biomarker for treatment response. SIGNIFICANCE: Evidence of gut microbiota involvement in trastuzumab efficacy represents the foundation for new therapeutic strategies aimed at manipulating commensal bacteria to improve response in trastuzumab-resistant patients.See related commentary by Sharma, p. 1937 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2195/F1.large.jpg.
Collapse
Affiliation(s)
- Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgio Gargari
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arianna Bonizzi
- Department of Biomedical and Clinical Sciences "L. Sacco," Università degli Studi di Milano, Milan, Italy
| | - Stefania Arioli
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department PROMISE, Università degli Studi di Palermo, Palermo, Italy
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Fasano
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Bianchi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessia Bertolotti
- Department of Pathology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department PROMISE, Università degli Studi di Palermo, Palermo, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Laura Villani
- Pathology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences "L. Sacco," Università degli Studi di Milano, Milan, Italy.,Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Simone Guglielmetti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Biomedical Science for Health, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
18
|
Szymiczek A, Lone A, Akbari MR. Molecular intrinsic versus clinical subtyping in breast cancer: A comprehensive review. Clin Genet 2020; 99:613-637. [PMID: 33340095 DOI: 10.1111/cge.13900] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022]
Abstract
Breast cancer is a heterogeneous disease manifesting diversity at the molecular, histological and clinical level. The development of breast cancer classification was centered on informing clinical decisions. The current approach to the classification of breast cancer, which categorizes this disease into clinical subtypes based on the detection of estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, and proliferation marker Ki67, is not ideal. This is manifested as a heterogeneity of therapeutic responses and outcomes within the clinical subtypes. The newer classification model, based on gene expression profiling (intrinsic subtyping) informs about transcriptional responses downstream from IHC single markers, revealing deeper appreciation for the disease heterogeneity and capturing tumor biology in a more comprehensive way than an expression of a single protein or gene alone. While accumulating evidences suggest that intrinsic subtypes provide clinically relevant information beyond clinical surrogates, it is imperative to establish whether the current conventional immunohistochemistry-based clinical subtyping approach could be improved by gene expression profiling and if this approach has a potential to translate into clinical practice.
Collapse
Affiliation(s)
- Agata Szymiczek
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Amna Lone
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad R Akbari
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Prat A, Guarneri V, Paré L, Griguolo G, Pascual T, Dieci MV, Chic N, González-Farré B, Frassoldati A, Sanfeliu E, Cejalvo JM, Muñoz M, Bisagni G, Brasó-Maristany F, Urso L, Vidal M, Brandes AA, Adamo B, Musolino A, Miglietta F, Conte B, Oliveira M, Saura C, Pernas S, Alarcón J, Llombart-Cussac A, Cortés J, Manso L, López R, Ciruelos E, Schettini F, Villagrasa P, Carey LA, Perou CM, Piacentini F, D'Amico R, Tagliafico E, Parker JS, Conte P. A multivariable prognostic score to guide systemic therapy in early-stage HER2-positive breast cancer: a retrospective study with an external evaluation. Lancet Oncol 2020; 21:1455-1464. [PMID: 33152285 DOI: 10.1016/s1470-2045(20)30450-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/06/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND In early-stage HER2-positive breast cancer, escalation or de-escalation of systemic therapy is a controversial topic. As an aid to treatment decisions, we aimed to develop a prognostic assay that integrates multiple data types for predicting survival outcome in patients with newly diagnosed HER2-positive breast cancer. METHODS We derived a combined prognostic model using retrospective clinical-pathological data on stromal tumour-infiltrating lymphocytes, PAM50 subtypes, and expression of 55 genes obtained from patients who participated in the Short-HER phase 3 trial. The trial enrolled patients with newly diagnosed, node-positive, HER2-positive breast cancer or, if node negative, with at least one risk factor (ie, tumour size >2 cm, histological grade 3, lymphovascular invasion, Ki67 >20%, age ≤35 years, or hormone receptor negativity), and randomly assigned them to adjuvant anthracycline plus taxane-based combinations with either 9 weeks or 1 year of trastuzumab. Trastuzumab was administered intravenously every 3 weeks (8 mg/kg loading dose at first cycle, and 6 mg/kg thereafter) for 18 doses or weekly (4 mg/kg loading dose in the first week, and 2 mg/kg thereafter) for 9 weeks, starting concomitantly with the first taxane dose. Median follow-up was 91·4 months (IQR 75·1-105·6). The primary objective of our study was to derive and evaluate a combined prognostic score associated with distant metastasis-free survival (the time between randomisation and distant recurrence or death before recurrence), an exploratory endpoint in Short-HER. Patient samples in the training dataset were split into a training set (n=290) and a testing set (n=145), balancing for event and treatment group. The training set was further stratified into 100 iterations of Monte-Carlo cross validation (MCCV). Cox proportional hazard models were fit to MCCV training samples using Elastic-Net. A maximum of 92 features were assessed. The final prognostic model was evaluated in an independent combined dataset of 267 patients with early-stage HER2-positive breast cancer treated with different neoadjuvant and adjuvant anti-HER2-based combinations and from four other studies (PAMELA, CHER-LOB, Hospital Clinic, and Padova) with disease-free survival outcome data. FINDINGS From Short-HER, data from 435 (35%) of 1254 patients for tumour size (T1 vs rest), nodal status (N0 vs rest), number of tumour-infiltrating lymphocytes (continuous variable), subtype (HER2-enriched and basal-like vs rest), and 13 genes composed the final model (named HER2DX). HER2DX was significantly associated with distant metastasis-free survival as a continuous variable (p<0·0001). HER2DX median score for quartiles 1-2 was identified as the cutoff to identify low-risk patients; and the score that distinguished quartile 3 from quartile 4 was the cutoff to distinguish medium-risk and high-risk populations. The 5-year distant metastasis-free survival of the low-risk, medium-risk, and high-risk populations were 98·1% (95% CI 96·3-99·9), 88·9% (83·2-95·0), and 73·9% (66·0-82·7), respectively (low-risk vs high-risk hazard ratio [HR] 0·04, 95% CI 0·0-0·1, p<0·0001). In the evaluation cohort, HER2DX was significantly associated with disease-free survival as a continuous variable (HR 2·77, 95% CI 1·4-5·6, p=0·0040) and as group categories (low-risk vs high-risk HR 0·27, 0·1-0·7, p=0·005). 5-year disease-free survival in the HER2DX low-risk group was 93·5% (89·0-98·3%) and in the high-risk group was 81·1% (71·5-92·1). INTERPRETATION The HER2DX combined prognostic score identifies patients with early-stage, HER2-positive breast cancer who might be candidates for escalated or de-escalated systemic treatment. Future clinical validation of HER2DX seems warranted to establish its use in different scenarios, especially in the neoadjuvant setting. FUNDING Instituto Salud Carlos III, Save the Mama, Pas a Pas, Fundación Científica, Asociación Española Contra el Cáncer, Fundación SEOM, National Institutes of Health, Agenzia Italiana del Farmaco, International Agency for Research on Cancer, and the Veneto Institute of Oncology, and Italian Association for Cancer Research.
Collapse
Affiliation(s)
- Aleix Prat
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Laia Paré
- SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Tomás Pascual
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Lineberger Comprehensive Cancer Center, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria V Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Núria Chic
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Blanca González-Farré
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Antonio Frassoldati
- Clinical Oncology, Department of Morphology, Surgery and Experimental Medicine, S Anna University Hospital, Ferrara, Italy
| | - Esther Sanfeliu
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Juan M Cejalvo
- Department of Medical Oncology, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Montserrat Muñoz
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Giancarlo Bisagni
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Emilia-Romagna, Italy
| | - Fara Brasó-Maristany
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Loredana Urso
- Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Maria Vidal
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Alba A Brandes
- Medical Oncology, Azienda Unità Sanitaria Locale di Bologna-IRCCS Istituto delle Scienze Neurologiche, Bologna
| | - Barbara Adamo
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Antonino Musolino
- Department of Medicine and Surgery, and the Medical Oncology and Breast Unit, University Hospital of Parma, Piacenza, Italy
| | - Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Benedetta Conte
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital; Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Cristina Saura
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital; Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Sònia Pernas
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Institut Català d'Oncologia Hospitalet, Hospitalet de Llobregat, Spain
| | - Jesús Alarcón
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Hospital Universitario Son Espases, Carretera de Valldemossa, Palma de Mallorca, Spain
| | | | - Javier Cortés
- Department of Medical Oncology, Vall d'Hebron University Hospital; Vall d'Hebron Institute of Oncology, Barcelona, Spain; IOB Institute of Oncology, Quiron Group, Barcelona, Spain
| | - Luis Manso
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Rafael López
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Complejo Universitario de Santiago de Compostela-CIBERONC, Santiago de Compostela, Spain
| | - Eva Ciruelos
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Francesco Schettini
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, University of Naples Federico II, Naples, Italy
| | | | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Federico Piacentini
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pierfranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy; Department of Medical Oncology UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
20
|
Thanopoulou E, Khader L, Caira M, Wardley A, Ettl J, Miglietta F, Neven P, Guarneri V. Therapeutic Strategies for the Management of Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Positive (HR+/HER2+) Breast Cancer: A Review of the Current Literature. Cancers (Basel) 2020; 12:E3317. [PMID: 33182657 PMCID: PMC7696181 DOI: 10.3390/cancers12113317] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Enormous advances have been made in the understanding and treatment of human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) in the last 30 years that have resulted in survival gains for affected patients. A growing body of evidence suggests that hormone receptor-positive (HR+)/HER2+ BC and HR-negative (HR-)/HER2+ BC are biologically different, with complex molecular bidirectional crosstalk between the estrogen receptor and HER2 pathway potentially affecting sensitivity to both HER2-targeted and endocrine therapy in patients with HR+/HER2+ BC. Subgroup analyses from trials enrolling patients with HER2+ BC and the results of clinical trials specifically designed to evaluate therapy in patients with HR+/HER2+ BC are helping to guide treatment decisions. In this context, encouraging results with strategies aimed at delaying or reversing drug resistance, including extended adjuvant therapy and the addition of drugs targeting alternative pathways, such as cyclin-dependent kinase (CDK) 4 and 6 inhibitors, have recently emerged. We have reached the point where tailoring the treatment according to risk and biology has become the paradigm in early BC. However, further clinical trials are needed that integrate translational research principles and identify and consider specific patient subgroups and biomarkers.
Collapse
Affiliation(s)
- Eirini Thanopoulou
- Eli Lilly and Company Limited, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK;
| | - Leila Khader
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Morena Caira
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Andrew Wardley
- The NIHR Manchester Clinical Research Facility at The Christie NHS Foundation Trust, School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester M204BX, UK;
| | - Johannes Ettl
- Department of Obstetrics and Gynecology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany;
| | - Federica Miglietta
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| | - Patrick Neven
- Multidisciplinary Breast Center and Department of Gynecology and Obstetrics, UZ Leuven, 3000 Leuven, Belgium;
- Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| |
Collapse
|
21
|
Weyl A, Illac C, Delchier MC, Suc B, Cuellar E, Chantalat E. Splenic lesion mimicking breast metastasis: The first description of splenic parenchymal endometriosis. JOURNAL OF ENDOMETRIOSIS AND PELVIC PAIN DISORDERS 2020. [DOI: 10.1177/2284026520960846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction: Splenic parenchymal endometriosis has never been described to date. We report here the case of real parenchymal endometriosis of the spleen. Case description: In this case, a 54-year-old female patient presented a histologically proven metastatic recurrence of breast cancer in the internal breast chain. The CT-scan also detected a large cystic structure developed from the spleen, but non-suspected to be metastasis. The patient was treated with chemotherapy (paclitaxel) and a combination of targeted therapies (everolimus and trastuzumab). While a complete radiological and biological response was noted at 2 months, the splenic cyst gradually decreased over the years. When targeted therapies were stopped, a reincrease of the splenic lesion and de novo significant hypermetabolism of the splenic parenchyma on 18F-FDG PET scan were observed. A splenectomy was finally performed and revealed splenic parenchymal endometriosis. Conclusion: This case once again highlights the complexity of endometriosis disease, from a pathophysiological point of view, but also the difficulties of radiological characterisation, and diagnostic management.
Collapse
Affiliation(s)
- Ariane Weyl
- Department of Gynecologic surgery, University Hospital of Toulouse Rangueil, Toulouse, France
| | - Claire Illac
- Department of Pathology, Institut Universitaire du cancer de Toulouse Oncopole, Toulouse, France
| | | | - Bertrand Suc
- Department of Visceral Surgery, University Hospital of Toulouse Rangueil, Toulouse, France
| | - Emmanuel Cuellar
- Department of Visceral Surgery, University Hospital of Toulouse Rangueil, Toulouse, France
| | - Elodie Chantalat
- Department of Gynecologic surgery, University Hospital of Toulouse Rangueil, Toulouse, France
| |
Collapse
|
22
|
Chen JS, Chen J, Bhattacharjee S, Cao Z, Wang H, Swanson SD, Zong H, Baker JR, Wang SH. Functionalized nanoparticles with targeted antibody to enhance imaging of breast cancer in vivo. J Nanobiotechnology 2020; 18:135. [PMID: 32948179 PMCID: PMC7501678 DOI: 10.1186/s12951-020-00695-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Targeted contrast nanoparticles for breast tumor imaging facilitates early detection and improves treatment efficacy of breast cancer. This manuscript reports the development of an epidermal growth factor receptor-2 (HER-2) specific, bi-modal, dendrimer conjugate to enhance computed tomography (CT) and magnetic resonance imaging (MRI) of HER-2-positive breast cancer. This material employs generation 5 poly(amidoamine) dendrimers, encapsulated gold nanoparticles, chelated gadolinium, and anti-human HER-2 antibody to produce the nanoparticle contrast agent. RESULTS Testing in two mouse tumor models confirms this contrast agent's ability to image HER-2 positive tumors. Intravenous injection of this nanoparticle in mice bearing HER-2 positive mammary tumors significantly enhances MRI signal intensity by ~ 20% and improves CT resolution and contrast by two-fold. Results by flow cytometry and confocal microscopy validate the specific targeting of the conjugate and its internalization in human HER-2 positive cells. CONCLUSION These results demonstrate that this nanoparticle conjugate can efficiently target and image HER-2 positive tumors in vivo and provide a basis for the development of this diagnostic tool for early detection, metastatic assessment and therapeutic monitoring of HER-2 positive cancers.
Collapse
Affiliation(s)
- Jesse S Chen
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Jingwen Chen
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Somnath Bhattacharjee
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Zhengyi Cao
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Scott D Swanson
- Department of Radiology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Hong Zong
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - James R Baker
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Su He Wang
- Department of Internal Medicine, Division of Allergy, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Treatment selection for patients with equivocal HER2 status and in luminal versus HER2-enriched disease. Breast 2020; 48 Suppl 1:S49-S52. [PMID: 31839160 DOI: 10.1016/s0960-9776(19)31123-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Equivocal HER2 status has been variably defined in the past, and its clinical implications have long been debated. In the 2018 focused update, ASCO/CAP guidelines recommended that tumours with double-equivocal (by immunohistochemistry and in situ hybridization assays) HER2 status should be considered HER2-negative due to the lack of evidence for any benefit of HER2-targeted therapy. The biology and the response to systemic therapies of tumours co-expressing HR and HER2 is quite complex. There is an extensive bi-directional cross-talk between these 2 pathways, that may result in both intrinsic and acquired resistance to endocrine agents, as well as in lower sensitivity to HER2-targeted therapies. In fact, neoadjuvant studies indicate that pCR rates are significantly lower in HER2-positive/ER-positive than ER-negative tumours, regardless the type of HER2 targeted treatment. The recent identification of different subtypes of HER2-positive breast cancer, according to the co-expression of HR and/or the molecular (intrinsic) subtyping, has prompted a renewed interest for clinical studies aimed at better tailoring the systemic therapy for these patients. A subgroup of them might not need chemotherapy if treated with dual HER2 blockade, and this option has been tested in a number of neo-adjuvant trials. In addition, triple targeting of HR, HER2, and CDK4/6 pathways simultaneously may be an effective treatment and overcome the drug resistance mechanisms that are typical of the disease. Finally, HER2-positive breast cancer may well benefit from immunotherapeutic interventions with anti-programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) agents.
Collapse
|
24
|
Goutsouliak K, Veeraraghavan J, Sethunath V, De Angelis C, Osborne CK, Rimawi MF, Schiff R. Towards personalized treatment for early stage HER2-positive breast cancer. Nat Rev Clin Oncol 2020; 17:233-250. [PMID: 31836877 PMCID: PMC8023395 DOI: 10.1038/s41571-019-0299-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2019] [Indexed: 12/13/2022]
Abstract
Advances in HER2-targeted therapies have improved the survival of patients with HER2-positive breast cancer. The standard-of-care treatment for localized disease has been chemotherapy and 1 year of adjuvant HER2-targeted therapy, typically with the anti-HER2 antibody trastuzumab. Despite the effectiveness of this treatment, disease relapse occurs in a subset of patients; thus, focus has been placed on escalating treatment by either combining different HER2-targeted agents or extending the duration of HER2-targeted therapy. Indeed, dual HER2-targeted therapies and extended-duration anti-HER2 therapy, as well as adjuvant therapy with the anti-HER2 antibody-drug conjugate T-DM1, have all been approved for clinical use. Emerging evidence suggests, however, that some patients do not derive sufficient benefit from these additional therapies to offset the associated toxicities and/or costs. Similarly, the universal use of chemotherapy might not benefit all patients, and treatment de-escalation through omission of chemotherapy has shown promise in clinical trials and is currently being explored further. The future of precision medicine should therefore involve tailoring of therapy based on the genetics and biology of each tumour and the clinical characteristics of each patient. Predictive biomarkers that enable the identification of patients who will benefit from either escalated or de-escalated treatment will be crucial to this approach. In this Review, we summarize the available HER2-targeted agents and associated mechanisms of resistance, and describe the current therapeutic landscape of early stage HER2-positive breast cancer, focusing on strategies for treatment escalation or de-escalation.
Collapse
Affiliation(s)
- Kristina Goutsouliak
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jamunarani Veeraraghavan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vidyalakshmi Sethunath
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
25
|
Phenotypic changes of HER2-positive breast cancer during and after dual HER2 blockade. Nat Commun 2020; 11:385. [PMID: 31959756 PMCID: PMC6971277 DOI: 10.1038/s41467-019-14111-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
The HER2-enriched (HER2-E) subtype within HER2-positive (HER2+) breast cancer is highly addicted to the HER2 pathway. However, ∼20-60% of HER2+/HER2-E tumors do not achieve a complete response following anti-HER2 therapies. Here we evaluate gene expression data before, during and after neoadjuvant treatment with lapatinib and trastuzumab in HER2+/HER2-E tumors of the PAMELA trial and breast cancer cell lines. Our results reveal that dual HER2 blockade in HER2-E disease induces a low-proliferative Luminal A phenotype both in patient's tumors and in vitro models. These biological changes are more evident in hormone receptor-positive (HR+) disease compared to HR-negative disease. Interestingly, increasing the luminal phenotype with anti-HER2 therapy increased sensitivity to CDK4/6 inhibition. Finally, discontinuation of HER2-targeted therapy in vitro, or acquired resistance to anti-HER2 therapy, leads to restoration of the original HER2-E phenotype. Our findings support the use of maintenance anti-HER2 therapy and the therapeutic exploitation of subtype switching with CDK4/6 inhibition.
Collapse
|
26
|
Schettini F, Pascual T, Conte B, Chic N, Brasó-Maristany F, Galván P, Martínez O, Adamo B, Vidal M, Muñoz M, Fernández-Martinez A, Rognoni C, Griguolo G, Guarneri V, Conte PF, Locci M, Brase JC, Gonzalez-Farre B, Villagrasa P, De Placido S, Schiff R, Veeraraghavan J, Rimawi MF, Osborne CK, Pernas S, Perou CM, Carey LA, Prat A. HER2-enriched subtype and pathological complete response in HER2-positive breast cancer: A systematic review and meta-analysis. Cancer Treat Rev 2020; 84:101965. [PMID: 32000054 DOI: 10.1016/j.ctrv.2020.101965] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND HER2-positive (HER2+) breast cancer (BC) comprises all the four PAM50 molecular subtypes. Among these, the HER2-Enriched (HER2-E) appear to be associated with higher pathological complete response (pCR) rates following anti-HER2-based regimens. Here, we present a meta-analysis to validate the association of the HER2-E subtype with pCR following anti-HER2-based neoadjuvant treatments with or without chemotherapy (CT). METHODS A systematic literature search was performed in February 2019. The primary objective was to compare the association between HER2-E subtype (versus others) and pCR. Selected secondary objectives were to compare the association between 1) HER2-E subtype and pCR in CT-free studies, 2) HER2-E subtype within hormone receptor (HR)-negative and HR+ disease and 3) HR-negative disease (versus HR+) and pCR in all patients and within HER2-E subtype. A random-effect model was applied. The Higgins' I2 was used to quantify heterogeneity. RESULTS Sixteen studies were included, 5 of which tested CT-free regimens. HER2-E subtype was significantly associated with pCR in all patients (odds ratio [OR] = 3.50, p < 0.001, I2 = 33%), in HR+ (OR = 3.61, p < 0.001, I2 = 1%) and HR-negative tumors (OR = 2.28, p = 0.01, I2 = 47%). In CT-free studies, HER2-E subtype was associated with pCR in all patients (OR = 5.52, p < 0.001, I2 = 0%) and in HR + disease (OR = 4.08, p = 0.001, I2 = 0%). HR-negative status was significantly associated with pCR compared to HR + status in all patients (OR = 2.41, p < 0.001, I2 = 30%) and within the HER2-E subtype (OR = 1.76, p < 0.001, I2 = 0%). CONCLUSIONS The HER2-E biomarker identifies patients with a higher likelihood of achieving a pCR following neoadjuvant anti-HER2-based therapy beyond HR status and CT use. Future trial designs to escalate or de-escalate systemic therapy in HER2+ disease should consider this genomic biomarker.
Collapse
Affiliation(s)
- Francesco Schettini
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Benedetta Conte
- Department of Medical Oncology, Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Nuria Chic
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Olga Martínez
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Barbara Adamo
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Maria Vidal
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Montserrat Muñoz
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | | | - Carla Rognoni
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Milan, Italy
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto - IRCCSS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto - IRCCSS, Padova, Italy
| | - Pier Franco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto - IRCCSS, Padova, Italy
| | - Mariavittoria Locci
- Department of Neuroscience, Reproductive Medicine, Odontostomatology, University of Naples Federico II, Naples, Italy
| | | | | | | | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rachel Schiff
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jamunarani Veeraraghavan
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Kent Osborne
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sonia Pernas
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medical Oncology, Institut Català d'Oncologia-H. U. Bellvitge-IDIBELL, Barcelona, Spain
| | - Charles M Perou
- Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - Lisa A Carey
- Department of Medicine, University of North Carolina, Chapel Hill, USA
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
27
|
Prat A, Pascual T, De Angelis C, Gutierrez C, Llombart-Cussac A, Wang T, Cortés J, Rexer B, Paré L, Forero A, Wolff AC, Morales S, Adamo B, Brasó-Maristany F, Vidal M, Veeraraghavan J, Krop I, Galván P, Pavlick AC, Bermejo B, Izquierdo M, Rodrik-Outmezguine V, Reis-Filho JS, Hilsenbeck SG, Oliveira M, Dieci MV, Griguolo G, Fasani R, Nuciforo P, Parker JS, Conte P, Schiff R, Guarneri V, Osborne CK, Rimawi MF. HER2-Enriched Subtype and ERBB2 Expression in HER2-Positive Breast Cancer Treated with Dual HER2 Blockade. J Natl Cancer Inst 2020; 112:46-54. [PMID: 31037288 PMCID: PMC7850037 DOI: 10.1093/jnci/djz042] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/06/2019] [Accepted: 03/26/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Identification of HER2-positive breast cancers with high anti-HER2 sensitivity could help de-escalate chemotherapy. Here, we tested a clinically applicable RNA-based assay that combines ERBB2 and the HER2-enriched (HER2-E) intrinsic subtype in HER2-positive disease treated with dual HER2-blockade without chemotherapy. METHODS A research-based PAM50 assay was applied in 422 HER2-positive tumors from five II-III clinical trials (SOLTI-PAMELA, TBCRC023, TBCRC006, PER-ELISA, EGF104090). In SOLTI-PAMELA, TBCRC023, TBCRC006, and PER-ELISA, all patients had early disease and were treated with neoadjuvant lapatinib or pertuzumab plus trastuzumab for 12-24 weeks. Primary outcome was pathological complete response (pCR). In EGF104900, 296 women with advanced disease were randomized to receive either lapatinib alone or lapatinib plus trastuzumab. Progression-free survival (PFS), overall response rate (ORR), and overall survival (OS) were evaluated. RESULTS A total of 305 patients with early and 117 patients with advanced HER2-positive disease were analyzed. In early disease, HER2-E represented 83.8% and 44.7% of ERBB2-high and ERBB2-low tumors, respectively. Following lapatinib and trastuzumab, the HER2-E and ERBB2 (HER2-E/ERBB2)-high group showed a higher pCR rate compared to the rest (44.5%, 95% confidence interval [CI] = 35.4% to 53.9% vs 11.6%, 95% CI = 6.9% to 18.0%; adjusted odds ratio [OR] = 6.05, 95% CI = 3.10 to 11.80, P < .001). Similar findings were observed with neoadjuvant trastuzumab and pertuzumab (pCR rate of 66.7% in HER2-E/ERBB2-high, 95% CI = 22.3% to 95.7% vs 14.7% in others, 95% CI = 4.9% to 31.1%; adjusted OR = 11.60, 95% CI = 1.66 to 81.10, P = .01). In the advanced setting, the HER2-E/ERBB2-high group was independently associated with longer PFS (hazard ratio [HR] = 0.52, 95% CI = 0.35 to 0.79, P < .001); higher ORR (16.3%, 95% CI = 8.9% to 26.2% vs 3.7%, 95% CI = 0.8% to 10.3%, P = .02); and longer OS (HR = 0.66, 95% CI = 0.44 to 0.97, P = .01). CONCLUSIONS Combining HER2-E subtype and ERBB2 mRNA into a single assay identifies tumors with high responsiveness to HER2-targeted therapy. This biomarker could help de-escalate chemotherapy in approximately 40% of patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Aleix Prat
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Scientific Department, SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Tomás Pascual
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Scientific Department, SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Carmine De Angelis
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
| | - Carolina Gutierrez
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
| | | | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Javier Cortés
- IOB Institute of Oncology, Quironsalud Group, Madrid & Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Brent Rexer
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Laia Paré
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Scientific Department, SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | - Andres Forero
- Department of Medicine, University of Alabama-Birmingham, Birmingham, AL
| | | | - Serafín Morales
- Department of Medical Oncology, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Barbara Adamo
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Fara Brasó-Maristany
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Maria Vidal
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Jamunarani Veeraraghavan
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
| | - Ian Krop
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Patricia Galván
- Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Anne C Pavlick
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Begoña Bermejo
- Department of Medical Oncology, Hospital Clínico de Valencia, Valencia, Spain
| | | | | | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan G Hilsenbeck
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
| | - Mafalda Oliveira
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Maria Vittoria Dieci
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Gaia Griguolo
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Fasani
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Joel S Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Rachel Schiff
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - C Kent Osborne
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Mothaffar F Rimawi
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
28
|
O'Sullivan CC, Suman VJ, Goetz MP. The emerging role of CDK4/6i in HER2-positive breast cancer. Ther Adv Med Oncol 2019; 11:1758835919887665. [PMID: 31832106 PMCID: PMC6887797 DOI: 10.1177/1758835919887665] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022] Open
Abstract
Prior to the advent of the monoclonal antibody trastuzumab, human epidermal growth-factor receptor 2 (HER2)-positive (HER2+) breast cancer (BC) was associated with an aggressive clinical course and poor survival outcomes. In the era of effective HER2-directed therapies, median survival rates for patients with metastatic HER2+ BC now approach 5 years. Despite these improvements, the majority of affected patients unfortunately die from disease. Therapies to overcome treatment resistance are being actively pursued. One strategy has been to target the cyclin-dependent kinases 4/6 (CDK4/6), as they are downstream of HER2 and many of the cellular pathways driving resistance to HER2-targeted therapies, and play a key role in proliferation by controlling transition through the G1 restriction point to the S phase of the cell cycle. In this article, we review the published literature with regard to the rationale for CDK4/6-directed therapies in HER2+ BC and discuss ongoing clinical research and new challenges in the field.
Collapse
Affiliation(s)
- Ciara C O'Sullivan
- Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905-0002, USA
| | - Vera J Suman
- Department of Biostatistics, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
29
|
De Angelis C, Nagi C, Hoyt CC, Liu L, Roman K, Wang C, Zheng Y, Veeraraghavan J, Sethunath V, Nuciforo P, Wang T, Tsimelzon A, Mao S, Hilsenbeck SG, Trivedi MV, Cataldo ML, Pavlick A, Wolff AC, Weigelt B, Reis-Filho JS, Prat A, Gutierrez C, Osborne CK, Rimawi MF, Schiff R. Evaluation of the Predictive Role of Tumor Immune Infiltrate in Patients with HER2-Positive Breast Cancer Treated with Neoadjuvant Anti-HER2 Therapy without Chemotherapy. Clin Cancer Res 2019; 26:738-745. [PMID: 31653641 DOI: 10.1158/1078-0432.ccr-19-1402] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/17/2019] [Accepted: 10/21/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TIL) are associated with benefit to trastuzumab and chemotherapy in patients with early-stage HER2+ breast cancer. The predictive value of TILs, TIL subsets, and other immune cells in patients receiving chemotherapy-sparing lapatinib plus trastuzumab treatment is unclear.Experimental Design: Hematoxylin and eosin-stained slides (n = 59) were used to score stromal (s-)TILs from pretreatment biopsies of patients enrolled in the neoadjuvant TBCRC006 trial of 12-week lapatinib plus trastuzumab therapy (plus endocrine therapy for ER+ tumors). A 60% threshold was used to define lymphocyte-predominant breast cancer (LPBC). Multiplexed immunofluorescence (m-IF) staining (CD4, CD8, CD20, CD68, and FoxP3) and multispectral imaging were performed to characterize immune infiltrates in single formalin-fixed paraffin-embedded slides (n = 33). RESULTS The pathologic complete response (pCR) rate was numerically higher in patients with LPBC compared with patients with non-LPBC (50% vs. 19%, P = 0.057). Unsupervised hierarchical clustering of the five immune markers identified two patient clusters with different responses to lapatinib plus trastuzumab treatment (pCR = 7% vs. 50%, for cluster 1 vs. 2 respectively; P = 0.01). In multivariable analysis, cluster 2, characterized by high CD4+, CD8+, CD20+ s-TILs, and high CD20+ intratumoral TILs, was independently associated with a higher pCR rate (P = 0.03). Analysis of single immune subpopulations revealed a significant association of pCR with higher baseline infiltration by s-CD4, intratumoral (i-) CD4, and i-CD20+ TILs. CONCLUSIONS LPBC was marginally associated with higher pCR rate than non-LPBC in patients with lapatinib plus trastuzumab treated HER2+ breast cancer. Quantitative assessment of the immune infiltrate by m-IF is feasible and may help correlate individual immune cell subpopulations and immune cell profiles with treatment response.
Collapse
Affiliation(s)
- Carmine De Angelis
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Chandandeep Nagi
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | | | | | | | - Yi Zheng
- Akoya Biosciences, Hopkinton, Massachusetts
| | - Jamunarani Veeraraghavan
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Vidyalakshmi Sethunath
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Paolo Nuciforo
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tao Wang
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Sufeng Mao
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Meghana V Trivedi
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Maria Letizia Cataldo
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Anne Pavlick
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Antonio C Wolff
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clinic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Carolina Gutierrez
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Charles Kent Osborne
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Mothaffar F Rimawi
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rachel Schiff
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas. .,Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
30
|
Carbognin L, Miglietta F, Paris I, Dieci MV. Prognostic and Predictive Implications of PTEN in Breast Cancer: Unfulfilled Promises but Intriguing Perspectives. Cancers (Basel) 2019; 11:E1401. [PMID: 31546901 PMCID: PMC6769721 DOI: 10.3390/cancers11091401] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 12/31/2022] Open
Abstract
The characterization of tumor biology and consequently the identification of prognostic and predictive biomarkers represent key issues for the translational research in breast cancer (BC). Phosphatase and tensin homolog deleted on chromosome ten (PTEN), the negative regulator of the proto-oncogenic phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway, constitutes one of the most intriguing tumor suppressor genes involved in a series of biological processes, such as cell growth and survival, cellular migration and genomic stability. Loss of PTEN activity, due to protein, genetic or epigenetic alterations, was reported in up to almost half of BC cases. Recently, besides the role of PTEN in the pathogenesis of BC, investigated for over 20 years after the PTEN discovery, several retrospective and prospective translational studies, in the early and advanced setting, reported controversial results regarding the association between PTEN functional status and both clinical outcome and response to various BC treatments. This review explores the pre-clinical and clinical role of PTEN in BC with regard to the potential association of PTEN with prognosis and treatment response or resistance, underlying the complexity of the interpretation of available results and suggesting potential future perspectives.
Collapse
Affiliation(s)
- Luisa Carbognin
- Department of Medicine, University of Verona, 37126 Verona, Italy.
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| | - Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy.
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy.
| | - Ida Paris
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy.
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy.
| |
Collapse
|
31
|
Sethunath V, Hu H, De Angelis C, Veeraraghavan J, Qin L, Wang N, Simon LM, Wang T, Fu X, Nardone A, Pereira R, Nanda S, Griffith OL, Tsimelzon A, Shaw C, Chamness GC, Reis-Filho JS, Weigelt B, Heiser LM, Hilsenbeck SG, Huang S, Rimawi MF, Gray JW, Osborne CK, Schiff R. Targeting the Mevalonate Pathway to Overcome Acquired Anti-HER2 Treatment Resistance in Breast Cancer. Mol Cancer Res 2019; 17:2318-2330. [PMID: 31420371 DOI: 10.1158/1541-7786.mcr-19-0756] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022]
Abstract
Despite effective strategies, resistance in HER2+ breast cancer remains a challenge. While the mevalonate pathway (MVA) is suggested to promote cell growth and survival, including in HER2+ models, its potential role in resistance to HER2-targeted therapy is unknown. Parental HER2+ breast cancer cells and their lapatinib-resistant and lapatinib + trastuzumab-resistant derivatives were used for this study. MVA activity was found to be increased in lapatinib-resistant and lapatinib + trastuzumab-resistant cells. Specific blockade of this pathway with lipophilic but not hydrophilic statins and with the N-bisphosphonate zoledronic acid led to apoptosis and substantial growth inhibition of R cells. Inhibition was rescued by mevalonate or the intermediate metabolites farnesyl pyrophosphate or geranylgeranyl pyrophosphate, but not cholesterol. Activated Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) and mTORC1 signaling, and their downstream target gene product Survivin, were inhibited by MVA blockade, especially in the lapatinib-resistant/lapatinib + trastuzumab-resistant models. Overexpression of constitutively active YAP rescued Survivin and phosphorylated-S6 levels, despite blockade of the MVA. These results suggest that the MVA provides alternative signaling leading to cell survival and resistance by activating YAP/TAZ-mTORC1-Survivin signaling when HER2 is blocked, suggesting novel therapeutic targets. MVA inhibitors including lipophilic statins and N-bisphosphonates may circumvent resistance to anti-HER2 therapy warranting further clinical investigation. IMPLICATIONS: The MVA was found to constitute an escape mechanism of survival and growth in HER2+ breast cancer models resistant to anti-HER2 therapies. MVA inhibitors such as simvastatin and zoledronic acid are potential therapeutic agents to resensitize the tumors that depend on the MVA to progress on anti-HER2 therapies.
Collapse
Affiliation(s)
- Vidyalakshmi Sethunath
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Huizhong Hu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lanfang Qin
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Nicholas Wang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Portland, Oregon
| | - Lukas M Simon
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tao Wang
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xiaoyong Fu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Agostina Nardone
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Resel Pereira
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Sarmistha Nanda
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Obi L Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Anna Tsimelzon
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Chad Shaw
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Gary C Chamness
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura M Heiser
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Portland, Oregon
| | - Susan G Hilsenbeck
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Mothaffar F Rimawi
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Joe W Gray
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine, Portland, Oregon
| | - C Kent Osborne
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas. .,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
32
|
Veeraraghavan J, De Angelis C, Mao R, Wang T, Herrera S, Pavlick AC, Contreras A, Nuciforo P, Mayer IA, Forero A, Nanda R, Goetz MP, Chang JC, Wolff AC, Krop IE, Fuqua SAW, Prat A, Hilsenbeck SG, Weigelt B, Reis-Filho JS, Gutierrez C, Osborne CK, Rimawi MF, Schiff R. A combinatorial biomarker predicts pathologic complete response to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2+ breast cancer. Ann Oncol 2019; 30:927-933. [PMID: 30903140 PMCID: PMC6594453 DOI: 10.1093/annonc/mdz076] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND HER2-positive (+) breast cancers, defined by HER2 overexpression and/or amplification, are often addicted to HER2 to maintain their malignant phenotype. Yet, some HER2+ tumors do not benefit from anti-HER2 therapy. We hypothesize that HER2 amplification levels and PI3K pathway activation are key determinants of response to HER2-targeted treatments without chemotherapy. PATIENTS AND METHODS Baseline HER2+ tumors from patients treated with neoadjuvant lapatinib plus trastuzumab [with endocrine therapy for estrogen receptor (ER)+ tumors] in TBCRC006 (NCT00548184) were evaluated in a central laboratory for HER2 amplification by fluorescence in situ hybridization (FISH) (n = 56). HER2 copy number (CN) and FISH ratios, and PI3K pathway status, defined by PIK3CA mutations or PTEN levels by immunohistochemistry were available for 41 tumors. Results were correlated with pathologic complete response (pCR; no residual invasive tumor in breast). RESULTS Thirteen of the 56 patients (23%) achieved pCR. None of the 11 patients with HER2 ratio <4 and/or CN <10 achieved pCR, whereas 13/45 patients (29%) with HER2 ratio ≥4 and/or CN ≥10 attained pCR (P = 0.0513). Of the 18 patients with tumors expressing high PTEN or wild-type (WT) PIK3CA (intact PI3K pathway), 7 (39%) achieved pCR, compared with 1/23 (4%) with PI3K pathway alterations (P = 0.0133). Seven of the 16 patients (44%) with HER2 ratio ≥4 and intact PI3K pathway achieved pCR, whereas only 1/25 (4%) patients not meeting these criteria achieved pCR (P = 0.0031). CONCLUSIONS Our findings suggest that there is a clinical subtype in breast cancer with high HER2 amplification and intact PI3K pathway that is especially sensitive to HER2-targeted therapies without chemotherapy. A combination of HER2 FISH ratio and PI3K pathway status warrants validation to identify patients who may be treated with HER2-targeted therapy without chemotherapy.
Collapse
Affiliation(s)
- J Veeraraghavan
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center
| | - C De Angelis
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center
| | - R Mao
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center
| | - T Wang
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Departments of Medicine
| | - S Herrera
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Pathology, Baylor College of Medicine, Houston, USA
| | - A C Pavlick
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center
| | - A Contreras
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Pathology, Baylor College of Medicine, Houston, USA
| | - P Nuciforo
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Hospital Clinic de Barcelona, Barcelona, Spain
| | - I A Mayer
- Medicine, Hematology/Oncology, Vanderbilt University, Nashville
| | - A Forero
- Medicine, University of Alabama at Birmingham, Birmingham
| | - R Nanda
- Medicine, University of Chicago, Chicago
| | - M P Goetz
- Department of Oncology, Mayo Clinic, Rochester
| | - J C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston
| | - A C Wolff
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore
| | - I E Krop
- Department of Medicine, Dana-Farber Cancer Institute, Boston
| | - S A W Fuqua
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center
| | - A Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Hospital Clinic de Barcelona, Barcelona, Spain
| | - S G Hilsenbeck
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Departments of Medicine
| | - B Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - C Gutierrez
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Pathology, Baylor College of Medicine, Houston, USA
| | - C K Osborne
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Departments of Medicine; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA
| | - M F Rimawi
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Departments of Medicine
| | - R Schiff
- Lester and Sue Smith Breast Center; Dan L. Duncan Comprehensive Cancer Center; Departments of Medicine; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
33
|
Fararjeh AS, Chen LC, Ho YS, Cheng TC, Liu YR, Chang HL, Chang HW, Wu CH, Tu SH. Proteasome 26S Subunit, non-ATPase 3 (PSMD3) Regulates Breast Cancer by Stabilizing HER2 from Degradation. Cancers (Basel) 2019; 11:cancers11040527. [PMID: 31013812 PMCID: PMC6549480 DOI: 10.3390/cancers11040527] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 03/28/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022] Open
Abstract
It is well-known that human epidermal growth factor receptor 2 (HER2) is critical for breast cancer (BC) development and progression. Several studies have revealed the role of the ubiquitin/proteasome system (UPS) in cancer. In this study, we investigated the expression level of Proteasome 26S subunit, non-ATPase 3 (PSMD3) in BC using BC cell lines, human BC tissue samples, Oncomine, and TCGA databases and studied the PSMD3-HER2 protein interaction. PSMD3 was upregulated in BC, particularly in the HER2+ subtype. PSMD3 immunostaining was detected in the cytoplasm and nucleus of BC tumor tissues. Strong interaction between PSMD3 and HER2 at the protein level was observed. Knockdown of PSMD3 significantly impaired the stability of HER2, inhibited BC cell proliferation and colony formation, and induced cell apoptosis. Ubiquitination process was strongly enhanced after knockdown of PSMD3 in association with decreased HER2 level. Accumulation and Localization of LAMP-1 in the cell membrane with decreased HER2 immunostaining was observed after knockdown of PSMD3. High expression level of PSMD3 was associated with HER2 expression (p < 0.001), tumor size (p < 0.001), and clinical stage (p = 0.036). High expression level of PSMD3 predicted a short overall survival (OS), particularly for HER2+. Overall, we provide a novel function for PSMD3 in stabilizing HER2 from degradation in HER2+ BC, which suggests that PSMD3 is a novel target for HER2+ BC.
Collapse
Affiliation(s)
- Abdulfattah Salah Fararjeh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
| | - Li-Ching Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan.
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Tzu-Chun Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan.
| | - Hang-Lung Chang
- Department of Surgery, EnChu Kong Hospital, New Taipei City237, Taiwan.
| | - Hui-Wen Chang
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Chih-Hsiung Wu
- Department of Surgery, EnChu Kong Hospital, New Taipei City237, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Shih-Hsin Tu
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
34
|
Nuciforo P, Pascual T, Cortés J, Llombart-Cussac A, Fasani R, Paré L, Oliveira M, Galvan P, Martínez N, Bermejo B, Vidal M, Pernas S, López R, Muñoz M, Garau I, Manso L, Alarcón J, Martínez E, Rodrik-Outmezguine V, Brase JC, Villagrasa P, Prat A, Holgado E. A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade. Ann Oncol 2019; 29:170-177. [PMID: 29045543 DOI: 10.1093/annonc/mdx647] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background The presence of stromal tumor-infiltrating lymphocytes (TILs) is associated with increased pathologic complete response (pCR) and improved outcomes in HER2-positive early-breast cancer (BC) treated with anti-HER2-based chemotherapy. In the absence of chemotherapy, the association of TILs with pCR following anti-HER2 therapy-only is largely unknown. Patients and methods The PAMELA neoadjuvant trial treated 151 women with HER2-positive BC with lapatinib and trastuzumab [and hormonal therapy if hormone receptor (HR)-positive] for 18 weeks. Percentage of TILs and tumor cellularity were determined at baseline (N = 148) and at day 15 (D15) of treatment (N = 134). Associations of TILs and tumor cellularity with pCR in the breast were evaluated. A combined score based on tumor cellularity and TILs (CelTIL) measured at D15 was derived in PAMELA, and validated in D15 samples from 65 patients with HER2-positive disease recruited in the LPT109096 neoadjuvant trial, where anti-HER2 therapy-only was administer for 2 weeks, then standard chemotherapy was added for 24 weeks. Results In PAMELA, baseline and D15 TILs were significantly associated with pCR in univariate analysis. In multivariable analysis, D15 TILs, but not baseline TILs, were significantly associated with pCR. At D15, TILs and tumor cellularity were found independently associated with pCR. A combined score (CelTIL) taking into account both variables was derived. CelTIL at D15 as a continuous variable was significantly associated with pCR, and patients with CelTIL-low and CelTIL-high scores had a pCR rate of 0% and 33%, respectively. In LPT109096, CelTIL at D15 was found associated with pCR both as a continuous variable and as group categories using a pre-defined cut-off (75.0% versus 33.3%). Conclusions On-treatment TILs, but not baseline TILs, are independently associated with response following anti-HER2 therapy-only. A combined score of TILs and tumor cellularity measured at D15 provides independent predictive information upon completion of neoadjuvant anti-HER2-based therapy. Clinical trial number NCT01973660.
Collapse
Affiliation(s)
- P Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - T Pascual
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - J Cortés
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain.,Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Llombart-Cussac
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Valencia, Spain
| | - R Fasani
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - L Paré
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - M Oliveira
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Galvan
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - N Martínez
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - B Bermejo
- Department of Medical Oncology, Hospital Clínico de Valencia, Valencia, Spain
| | - M Vidal
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - S Pernas
- Department of Medical Oncology, Instituto Catalán de Oncología, Hospitalet, Spain
| | - R López
- Department of Medical Oncology, Complejo Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - M Muñoz
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - I Garau
- Department of Medical Oncology, Hospital de Son Llàtzer, Palma de Mallorca, Spain
| | - L Manso
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - J Alarcón
- Department of Medical Oncology, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - E Martínez
- Department of Medical Oncology, Hospital Provincial Centre de Castelló, Castelló de la Plana, Spain
| | | | - J C Brase
- Novartis Oncology, Basel, Switzerland
| | - P Villagrasa
- SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - A Prat
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - E Holgado
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain.,Department of Medical Oncology, Baselga Oncological Institute, Madrid, Spain.,Department of Medical Oncology, Baselga Oncological Institute, Barcelona, Spain
| |
Collapse
|
35
|
Bazzichetto C, Conciatori F, Pallocca M, Falcone I, Fanciulli M, Cognetti F, Milella M, Ciuffreda L. PTEN as a Prognostic/Predictive Biomarker in Cancer: An Unfulfilled Promise? Cancers (Basel) 2019; 11:cancers11040435. [PMID: 30925702 PMCID: PMC6520939 DOI: 10.3390/cancers11040435] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
Identifying putative biomarkers of clinical outcomes in cancer is crucial for successful enrichment, and for the selection of patients who are the most likely to benefit from a specific therapeutic approach. Indeed, current research in personalized cancer therapy focuses on the possibility of identifying biomarkers that predict prognosis, sensitivity or resistance to therapies. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor gene that regulates several crucial cell functions such as proliferation, survival, genomic stability and cell motility through both enzymatic and non-enzymatic activities and phosphatidylinositol 3-kinase (PI3K)-dependent and -independent mechanisms. Despite its undisputed role as a tumor suppressor, assessment of PTEN status in sporadic human tumors has yet to provide clinically robust prognostic, predictive or therapeutic information. This is possibly due to the exceptionally complex regulation of PTEN function, which involves genetic, transcriptional, post-transcriptional and post-translational events. This review shows a brief summary of the regulation and function of PTEN and discusses its controversial aspects as a prognostic/predictive biomarker.
Collapse
Affiliation(s)
- Chiara Bazzichetto
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
- Department of Molecular Medicine, University of Rome, La Sapienza, Rome 00185, Italy.
| | - Fabiana Conciatori
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Matteo Pallocca
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Italia Falcone
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Maurizio Fanciulli
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Francesco Cognetti
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona 37126, Italy.
| | - Ludovica Ciuffreda
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| |
Collapse
|
36
|
Tsai YF, Tseng LM, Lien PJ, Hsu CY, Lin YS, King KL, Wang YL, Chao TC, Liu CY, Chiu JH, Yang MH. HER2 immunohistochemical scores provide prognostic information for patients with HER2-type invasive breast cancer. Histopathology 2019; 74:578-586. [PMID: 30515868 DOI: 10.1111/his.13801] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/01/2018] [Indexed: 01/28/2023]
Abstract
AIMS Invasive breast cancer patients with human epidermal growth factor receptor 2 (HER2) immunohistochemical (IHC) scores of 3+ or 2+ with reflex in-situ hybridisation (ISH) positivity are suitable for anti-HER2 therapies. The aim of this study is to investigate whether the prognoses between IHC 3+ patients and IHC 2+/ISH+ patients are different. METHODS AND RESULTS We analysed the clinicopathological information of 886 consecutive cases of HER2-positive early breast cancer. The influences of the patients' age, cancer stage, hormone receptor status and anti-HER2 treatment were adjusted using a multivariate Cox regression model. Both HER2 copy numbers and HER2 ISH ratios of the IHC 3+ group were significantly higher than those of the IHC 2+/ISH+ group. The outcomes of IHC 3+ patients were significantly better than those of IHC 2+/ISH+ patients in the univariate and multivariate analyses. HER2 copy numbers of ≥8 represented the best prognostic value, and it was chosen to be the cut-off value. The reflex ISH for IHC 2+ patients with high HER2 copy numbers (≥8) predicted a better overall survival than that for those with low HER2 copy numbers. CONCLUSION HER2 IHC scores and HER2 copy numbers can provide prognostic information for patients with HER2-positive invasive breast cancer. Both IHC 3+ and IHC 2+ patients with high HER2 copy numbers had a better prognosis.
Collapse
Affiliation(s)
- Yi-Fang Tsai
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Ju Lien
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Yi Hsu
- Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,College of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Yen-Shu Lin
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuang-Liang King
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Ling Wang
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ta-Chung Chao
- Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Yu Liu
- Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jen-Hwey Chiu
- Comprehensive Breast Health Center and Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine and Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
37
|
Gavilá J, Oliveira M, Pascual T, Perez-Garcia J, Gonzàlez X, Canes J, Paré L, Calvo I, Ciruelos E, Muñoz M, Virizuela JA, Ruiz I, Andrés R, Perelló A, Martínez J, Morales S, Marín-Aguilera M, Martínez D, Quero JC, Llombart-Cussac A, Prat A. Safety, activity, and molecular heterogeneity following neoadjuvant non-pegylated liposomal doxorubicin, paclitaxel, trastuzumab, and pertuzumab in HER2-positive breast cancer (Opti-HER HEART): an open-label, single-group, multicenter, phase 2 trial. BMC Med 2019; 17:8. [PMID: 30621698 PMCID: PMC6325829 DOI: 10.1186/s12916-018-1233-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/10/2018] [Indexed: 01/06/2023] Open
Affiliation(s)
- Joaquín Gavilá
- Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Mafalda Oliveira
- Vall d' Hebron University Hospital/Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Tomás Pascual
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Hospital Clínic, Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Jose Perez-Garcia
- Vall d' Hebron University Hospital/Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Instituto Oncológico Baselga, Hospital Quirón, Barcelona, Spain
| | - Xavier Gonzàlez
- SOLTI Breast Cancer Research Group, Barcelona, Spain.,Institut Oncològic Rosell, Hospital General Catalunya, Barcelona, Spain
| | - Jordi Canes
- SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Laia Paré
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Isabel Calvo
- Centro Integral Oncológico Clara Campal, Madrid, Spain
| | | | - Montserrat Muñoz
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Hospital Clínic, Barcelona, Spain
| | | | - Isabel Ruiz
- Hospital Universitario Sant Joan De Reus, Reus, Spain
| | - Raquel Andrés
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | | | | | | | | | - Débora Martínez
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | | | - Aleix Prat
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain. .,Hospital Clínic, Barcelona, Spain. .,SOLTI Breast Cancer Research Group, Barcelona, Spain.
| |
Collapse
|
38
|
Escrivá-de-Romaní S, Arumí M, Zamora E, Bellet M. Neoadjuvant Model as a Platform for Research in Breast Cancer and Novel Targets under Development in this Field. Breast Care (Basel) 2018; 13:251-262. [PMID: 30319327 DOI: 10.1159/000492122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
For decades, the neoadjuvant setting has provided a useful scenario for research in breast cancer. Historically, neoadjuvant clinical trials, either hormone therapy-based or chemotherapy-based, have tried to recapitulate the results of their counterpart adjuvant studies, but with smaller patient numbers, more rapid outcomes (clinical response and/or pathologic complete response (pCR)), together with additional biologic information. As for neoadjuvant chemotherapy trials, the increase in pCR rates has been recently accepted as an appropriate surrogate marker to accelerate drug approval in high-risk breast cancer patients. In this setting, with the exception of luminal A tumors, pCR has been associated with improved long-term outcomes, particularly when the analysis is based on specific trials for each breast cancer subtype. For luminal tumors receiving neoadjuvant endocrine therapy, Ki67 at 2-4 weeks and the preoperative endocrine prognostic index score are the most accepted intermediate markers of efficacy, which will be validated in ongoing larger trials. In this review, we describe the different neoadjuvant designs: from the classical randomized trials in which treatment is delivered for 6 or more months to short non-therapeutic presurgical studies lasting just 2 or 3 weeks. We also review the main neoadjuvant trials, either ongoing or completed, for luminal, triple-negative, and HER2-positive breast cancer. The translational effort and research of biomarkers conducted in these studies will be particularly addressed.
Collapse
Affiliation(s)
- Santiago Escrivá-de-Romaní
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Miriam Arumí
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Esther Zamora
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Meritxell Bellet
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
39
|
Cejalvo JM, Pascual T, Fernández-Martínez A, Brasó-Maristany F, Gomis RR, Perou CM, Muñoz M, Prat A. Clinical implications of the non-luminal intrinsic subtypes in hormone receptor-positive breast cancer. Cancer Treat Rev 2018; 67:63-70. [PMID: 29763779 DOI: 10.1016/j.ctrv.2018.04.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022]
Abstract
Gene expression profiling has had a considerable impact on our understanding ofbreastcancer biology. Duringthelast decade, 4 intrinsic molecular subtypes of breast cancer (Luminal A, Luminal B, HER2-enriched [HER2-E] and Basal-like) have been identified and intensively studied. In this article, we review and discuss the clinical implications of the 2 non-luminal subtypes (i.e. HER2-E and Basal-like) identified within hormone receptor (HR)-positive disease. After reviewing 32 studies for a total of 13,091 samples, ∼8% and ∼ 15% of early and metastatic HR+/HER2-negative breast cancer, respectively, were found to be non-luminal. Clinically, HR+/HER2-negative/non-luminal subtypes have been associated with estrogen independence, chemo-sensitivity, resistance to CDK4/6 inhibition and poor outcome. Interestingly, EGFR/HER2 tyrosine kinase inhibition might be of value in the HR+/HER2-negative/HER2-E subtype. Finally, the HER2-E subtype within HR+/HER2 + disease represents ∼ 30% and has been associated with anti-HER2 sensitivity, chemo-sensitivity and resistance to CDK4/6 inhibition. In the upcoming years, retrospective and prospective clinical trials evaluating both biomarkers should lead to improvements in patient outcomes.
Collapse
Affiliation(s)
- Juan Miguel Cejalvo
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Oncology Program, Institute for Research in Biomedicine (IRB Barcelona) and CIBERONC, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clínic de Barcelona, Spain; SOLTI Breast Cancer Cooperative Group, Barcelona, Spain
| | | | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Roger R Gomis
- Oncology Program, Institute for Research in Biomedicine (IRB Barcelona) and CIBERONC, Spain; ICREA, Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain
| | - Charles M Perou
- Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - Montserrat Muñoz
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clínic de Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clínic de Barcelona, Spain; SOLTI Breast Cancer Cooperative Group, Barcelona, Spain.
| |
Collapse
|
40
|
HER2-positive breast cancer: Current and new therapeutic strategies. Breast 2018; 39:80-88. [PMID: 29631097 DOI: 10.1016/j.breast.2018.03.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/11/2018] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
Since the identification of the HER2 receptor amplification as an adverse prognostic factor that defined a special subtype of metastatic breast cancer, there has been a substantial improvement in survival of patients affected with this disease due to the development of anti-HER2 targeted therapies. The approval of trastuzumab and pertuzumab associated to a taxane in first line and subsequent treatment with the antibody-drug conjugate T-DM1 has certainly contributed to achieve these outcomes. The Tyrosine Kinase Inhibitor lapatinib was also approved in the basis of an improvement in progression free survival, becoming another commonly used treatment in combination with capecitabine. Inevitably, despite these therapeutic advances most patients progress on therapy due to primary or acquired resistance or because of an incorrect HER2 positivity assessment. Hence, it is crucial to correctly categorize HER2 amplified tumors and define mechanisms of resistance to design effective new treatment approaches. In addition, identifying biomarkers of response or resistance permits to tailor the therapeutic options for each patient sparing them from unnecessary toxicity as well as improving their outcomes. The aim of this review is to examine new strategies in development to treat HER2-positive metastatic breast cancer referring to the mechanisms of action of new drugs and new combinations including results reported so far.
Collapse
|
41
|
Nicolini A, Ferrari P, Diodati L, Carpi A. Recent Advances in Comprehending the Signaling Pathways Involved in the Progression of Breast Cancer. Int J Mol Sci 2017; 18:E2321. [PMID: 29099748 PMCID: PMC5713290 DOI: 10.3390/ijms18112321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022] Open
Abstract
This review describes recent advances in the comprehension of signaling pathways involved in breast cancer progression. Calcium sensing receptor (CaSR), caveolae signaling, signaling referred to hypoxia-inducing factors and disturbances in the apoptotic machinery are related to more general biological mechanisms and are considered first. The others refer to signaling pathways of more specific biological mechanisms, namely the heparin/heparin-sulfate interactome, over-expression of miRNA-378a-5p, restriction of luminal and basal epithelial cells, fatty-acid synthesis, molecular pathways related to epithelial to mesenchimal transition (EMT), HER-2/neu gene amplification and protein expression, and the expression of other members of the epithelial growth factor receptor family. This progress in basic research is fundamental to foster the ongoing efforts that use the new genotyping technologies, and aim at defining new prognostic and predictive biomarkers for a better personalized management of breast cancer disease.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Lucrezia Diodati
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
42
|
Adabor ES, Acquaah-Mensah GK. Machine learning approaches to decipher hormone and HER2 receptor status phenotypes in breast cancer. Brief Bioinform 2017; 20:504-514. [DOI: 10.1093/bib/bbx138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/27/2017] [Indexed: 11/14/2022] Open
Affiliation(s)
- Emmanuel S Adabor
- African Institute for Mathematical Sciences, Muizenberg, South Africa
| | - George K Acquaah-Mensah
- Massachusetts College of Pharmacy and Health Sciences, Pharmaceutical Sciences, Worcester, Massachusetts, United States
| |
Collapse
|