1
|
Singh SR, Bhaskar R, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Mladenov M, Hadzi-Petrushev N, Stojchevski R, Sinha JK, Avtanski D. Exploring the Genetic Orchestra of Cancer: The Interplay Between Oncogenes and Tumor-Suppressor Genes. Cancers (Basel) 2025; 17:1082. [PMID: 40227591 PMCID: PMC11988167 DOI: 10.3390/cancers17071082] [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: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is complex because of the critical imbalance in genetic regulation as characterized by both the overexpression of oncogenes (OGs), mainly through mutations, amplifications, and translocations, and the inactivation of tumor-suppressor genes (TSGs), which entail the preservation of genomic integrity by inducing apoptosis to counter the malignant growth. Reviewing the intricate molecular interplay between OGs and TSGs draws attention to their cell cycle, apoptosis, and cancer metabolism regulation. In the present review, we discuss seminal discoveries, such as Knudson's two-hit hypothesis, which framed the field's understanding of cancer genetics, leading to the next breakthroughs with next-generation sequencing and epigenetic profiling, revealing novel insights into OG and TSG dysregulation with opportunities for targeted therapy. The key pathways, such as MAPK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin, are presented in the context of tumor progression. Importantly, we further highlighted the advances in therapeutic strategies, including inhibitors of KRAS and MYC and restoration of TSG function, despite which mechanisms of resistance and tumor heterogeneity pose daunting challenges. A high-level understanding of interactions between OG-TSGs forms the basis for effective, personalized cancer treatment-something to strive for in better clinical outcomes. This synthesis should integrate foundational biology with translation and, in this case, contribute to the ongoing effort against cancer.
Collapse
Affiliation(s)
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, India
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
2
|
Giacoletto CJ, Valente LJ, Brown L, Patterson S, Gokhale R, Mockus SM, Grody WW, Deng HW, Rotter JI, Schiller MR. New Gain-of-Function Mutations Prioritize Mechanisms of HER2 Activation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323043. [PMID: 40093211 PMCID: PMC11908269 DOI: 10.1101/2025.03.03.25323043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
ERBB2 (HER2) is a well-studied oncogene with several driver mutations apart from the well-known amplification defect in some breast cancers. We used the GigaAssay to test the functional effect of HER2 missense mutations on its receptor tyrosine kinase function. The GigaAssay is a modular high-throughput one-pot assay system for simultaneously measuring molecular function of thousands of genetic variants at very high accuracy. The activities of 5,886 mutations were classified, significantly more than mutants previously reported. These variants include 112 new in vitro, 10 known, and 9 new in vivo gain-of-function (GOF) mutations. Many of the GOFs spatially cluster in sequence and structure, supporting the activation mechanisms of heterodimerization with EGFR and release of kinase inhibition by the juxtamembrane domain. Retrospective analysis of patient outcomes from the Genomic Data Commons predicts increased survival with the newly identified HER2 GOF variants.
Collapse
Affiliation(s)
- Christopher J Giacoletto
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
| | - Liz J Valente
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
| | - Lancer Brown
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
| | - Sara Patterson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032
| | - Rewatee Gokhale
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032
| | | | | | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Department of Deming Department of Medicine, Tulane University, New Orleans, 70112 USA
| | - Jerome I Rotter
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Martin R Schiller
- Heligenics Inc., 10530 Discovery Dr., Las Vegas, NV 89135 USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
- School of Life Sciences, University of Nevada, Las Vegas, 4505 S. Maryland Parkway, Las Vegas, Nevada, 89154 USA
| |
Collapse
|
3
|
Manrai PA, McHenry A, Sun T, Santin AD, Ratner E, Lin DI, Elvin JA, Hui P, Buza N. Targetable ERBB2/HER2 Mutations in Gynecologic Malignancies: Clinicopathological, Immunohistochemical, and Molecular Correlations. Int J Gynecol Pathol 2025; 44:144-154. [PMID: 38914011 DOI: 10.1097/pgp.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Targeted anti-HER2 therapy has been recently added to the standard treatment recommendations in endometrial serous carcinoma. Current eligibility requires testing for HER2 overexpression and/or gene amplification by immunohistochemistry and by fluorescence in situ hybridization. However, clinical trials have also demonstrated the efficacy of anti-HER2 drugs against activating ERBB2/HER2 mutations in a variety of solid tumor types, and fam-trastuzumab deruxtecan is now approved by the US Food and Drug Administration for HER2 -mutant non-small cell lung cancer. This study aimed at evaluating the detailed clinical, histomorphological, immunohistochemical, and molecular characteristics of gynecologic malignancies with ERBB2/HER2 mutations. We identified 16 tumors with 19 ERBB2/HER2 mutations in our departmental archives: 11 endometrial primaries, 2 endocervical adenocarcinomas, 1 ovarian mucinous adenocarcinoma, 1 tubo-ovarian undifferentiated carcinoma, and 1 high-grade endometrioid adenocarcinoma of Mullerian origin. ERBB2/HER2 mutations most often involved the tyrosine kinase domain (52.6%), and the most frequent specific mutation was R678Q (31.6%), involving the juxtamembrane domain. More than half (54.5%) of endometrial carcinomas and half of all tumors were MMR-deficient, resulting from MSH6 loss in all but 2 tumors. None of the tumors (0%) were POLE- mutated, while 18.8% were TP53 -mutated. HER2 IHC was negative (score 0 or 1+) in 12 tumors (67%) and equivocal (score 2+) in 4 tumors (33%), whereas none of the tumors were scored as HER2 3+. Score 2+ was associated with R678Q, L755S, I767M mutations, and ERBB2/HER2 rearrangement with a breakpoint in exon 23. Concurrent ERBB2/HER2 amplification was identified in 2 endometrial carcinomas, with HER2/CEP17 ratios of 3.1 and 3.5. We also queried the cBioportal database, which revealed 70 ERBB2/HER2 -mutant gynecologic tumors with a total of 77 ERBB2/HER2 mutations, most often involving the active site of the tyrosine kinase domain (n=36; 46.8%), and the most common specific mutation was S310F (n=20; 26%), located in the extracellular domain. Our results provide important details regarding the clinicopathological and molecular associations of potentially actionable ERBB2/HER2 mutations in endometrial carcinoma and other gynecological cancer types and contribute to addressing clinical treatment needs and improving pathology testing recommendations in the future.
Collapse
Affiliation(s)
| | - Austin McHenry
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Tong Sun
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale School of Medicine, New Haven, CT
| | - Elena Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale School of Medicine, New Haven, CT
| | | | | | - Pei Hui
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Natalia Buza
- Department of Pathology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
4
|
Venturini J, Massaro G, Lavacchi D, Rossini D, Pillozzi S, Caliman E, Pellegrini E, Antonuzzo L. The emerging HER2 landscape in colorectal cancer: the key to unveil the future treatment algorithm? Crit Rev Oncol Hematol 2024; 204:104515. [PMID: 39304034 DOI: 10.1016/j.critrevonc.2024.104515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Colorectal cancer (CRC) represents a global health threat, standing as the second leading cause of cancer-related death worldwide. Targeted therapies brought new hope for the metastatic stage, which historically bore a very poor prognosis. Human epidermal growth receptor 2 (HER2) overexpression concerns about 5 % of the metastatic CRC (mCRC) patients, including both gene amplifications and point mutations. Albeit its controversial prognostic role, preclinical and clinical data indicate HER2 as a negative predictive biomarker of response to anti-EGFR therapies. Tissue and plasma-based NGS testing, could permit a precise identification of this resistance mechanism both at baseline and during treatment, thus guiding decision-making. Furthermore, promising results come from completed and ongoing randomized trials, testing HER2 as an actionable target. In this review, we discuss the available evidence on HER2 targeting in advanced CRC, analyzing its possible future role in the treatment algorithm.
Collapse
Affiliation(s)
- Jacopo Venturini
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Giulia Massaro
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Rossini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Department of Health Science, University of Florence, Florence 50139, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Elisa Pellegrini
- Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy.
| |
Collapse
|
5
|
Miligy IM, Badr N, Stevens A, Spooner D, Awasthi R, Mir Y, Khurana A, Sharma V, Chandaran U, Rakha EA, Maurice Y, Kearns D, Oweis R, Asar A, Ironside A, Shaaban AM. Pathological Changes Following Neoadjuvant Endocrine Therapy (NAET): A Multicentre Study of 391 Breast Cancers. Int J Mol Sci 2024; 25:7381. [PMID: 39000487 PMCID: PMC11242101 DOI: 10.3390/ijms25137381] [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: 04/19/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Oestrogen receptor (ER)-positive breast cancer (BC) is generally well responsive to endocrine therapy. Neoadjuvant endocrine therapy (NAET) is increasingly being used for downstaging ER-positive tumours. This study aims to analyse the effect of NAET on a well-characterised cohort of ER-positive BC with particular emphasis on receptor expression. This is a retrospective United Kingdom (UK) multicentre study of 391 patients who received NAET between October 2012 and October 2020. Detailed analyses of the paired pre- and post-NAET morphological changes and hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) expression were performed. The median duration of NAET was 86 days, with median survival and overall survival rates of 380 days and 93.4%, respectively. A total of 90.3% of cases achieved a pathological partial response, with a significantly higher rate of response in the HER2-low cancers. Following NAET, BC displayed some pathological changes involving the tumour stroma including central scarring and an increase in tumour infiltrating lymphocytes (TILs) and tumour cell morphology. Significant changes associated with the duration of NAET were observed in tumour grade (30.6% of cases), with downgrading identified in 19.3% of tumours (p < 0.001). The conversion of ER status from positive to low or negative was insignificant. The conversion of progesterone receptor (PR) and HER2 status to negative status was observed in 31.3% and 38.1% of cases, respectively (p < 0.001). HER2-low breast cancer decreased from 63% to 37% following NAET in the paired samples. Significant morphological and biomarker changes involving PR and HER2 expression occurred following NAET. The findings support biomarker testing on pre-treatment core biopsies and post-treatment residual carcinoma.
Collapse
Affiliation(s)
- Islam M. Miligy
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
- Histopathology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom 11352, Egypt;
| | - Nahla Badr
- Histopathology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom 11352, Egypt;
| | - Andrea Stevens
- Oncology Department, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (A.S.); (D.S.)
| | - David Spooner
- Oncology Department, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (A.S.); (D.S.)
| | - Rachna Awasthi
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
| | - Yasmeen Mir
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Anuj Khurana
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Vijay Sharma
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Usha Chandaran
- Histopathology Department, Salford Royal Hospital, Salford M6 8HD, UK;
| | - Emad A. Rakha
- Histopathology Department, Nottingham City Hospital, Nottingham NG5 1PB, UK;
| | - Yasmine Maurice
- Histopathology Department, Heartlands General Hospital, Birmingham B9 5SS, UK;
| | - Daniel Kearns
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
| | - Rami Oweis
- Histopathology Department, Rotherham Foundation Trust, Rotherham S60 2UD, UK; (R.O.); (A.A.)
| | - Amal Asar
- Histopathology Department, Rotherham Foundation Trust, Rotherham S60 2UD, UK; (R.O.); (A.A.)
| | | | - Abeer M. Shaaban
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| |
Collapse
|
6
|
Bon G, Di Lisa FS, Filomeno L, Arcuri T, Krasniqi E, Pizzuti L, Barba M, Messina B, Schiavoni G, Sanguineti G, Botti C, Cappelli S, Pelle F, Cavicchi F, Puccica I, Costantini M, Perracchio L, Maugeri-Saccà M, Ciliberto G, Vici P. HER2 mutation as an emerging target in advanced breast cancer. Cancer Sci 2024; 115:2147-2158. [PMID: 38715247 PMCID: PMC11247561 DOI: 10.1111/cas.16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 07/13/2024] Open
Abstract
HER2 activating mutations have emerged as oncogenic drivers and therapeutic targets in a variety of human tumors. In breast cancer, these deregulations occur at low frequency, and are mostly detected in HER2-nonamplified, metastatic disease. Preclinical evidence has clarified the role of hotspot mutations in HER2 constitutive activation, defining them as an alternative mechanism to HER2 gene amplification. Furthermore, recent clinical studies have indicated the emergence of newly acquired HER2 deregulations in significant proportions of breast cancer patients who experience disease progression following both endocrine and HER2-targeted therapies. As the involvement of HER2 mutation in therapy resistance may profoundly impact patient outcomes on successive therapies, several clinical trials are currently investigating the efficacy of various HER2-targeted drugs in HER2-mutant breast cancer. In this review, we firstly summarize the structural organization of the HER2 oncogene and its historical impact on breast cancer prognosis and therapeutic advancement. Then, we provide an overview of the frequencies and functional relevance of clinically recurrent HER2 mutations in breast cancer with a special focus on their role in therapeutic resistance. Finally, we provide a collection of the clinical trials that are currently exploring novel therapeutic approaches for this patient subset and discuss the related perspectives and challenges.
Collapse
Affiliation(s)
- Giulia Bon
- Department of Research, Cellular Network and Molecular Therapeutic Target Unit, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Lorena Filomeno
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Teresa Arcuri
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Radiological, Medical Oncology A, Policlinico Umberto I; Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Eriseld Krasniqi
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Beatrice Messina
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Schiavoni
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sonia Cappelli
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabio Pelle
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Flavia Cavicchi
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ilaria Puccica
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maurizio Costantini
- Department of Plastic and Reconstructive Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Letizia Perracchio
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Department of Research, Biostatistics and Bioinformatics Unit, Clinical Trial Center, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizia Vici
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
7
|
Krystel-Whittemore M, Tan PH, Wen HY. Predictive and prognostic biomarkers in breast tumours. Pathology 2024; 56:186-191. [PMID: 38212230 PMCID: PMC10949537 DOI: 10.1016/j.pathol.2023.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 01/13/2024]
Abstract
In the age of precision medicine, extensive research has investigated tumour biomarkers to predict the behaviour of cancer and/or response to treatment in order to better understand the prognosis and treatment of disease. In breast cancer, significant progress has been made to categorise a common disease into subtypes defined by intrinsic tumour biology, measured by tumour biomarkers. This review encompasses the established biomarkers within breast cancer with the most up-to-date information regarding their understanding and clinical use as predictive and/or prognostic markers of breast cancer.
Collapse
Affiliation(s)
| | | | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
8
|
Tozbikian G, Krishnamurthy S, Bui MM, Feldman M, Hicks DG, Jaffer S, Khoury T, Wei S, Wen H, Pohlmann P. Emerging Landscape of Targeted Therapy of Breast Cancers With Low Human Epidermal Growth Factor Receptor 2 Protein Expression. Arch Pathol Lab Med 2024; 148:242-255. [PMID: 37014972 DOI: 10.5858/arpa.2022-0335-ra] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 04/06/2023]
Abstract
CONTEXT.— Human epidermal growth factor receptor 2 (HER2) status in breast cancer is currently classified as negative or positive for selecting patients for anti-HER2 targeted therapy. The evolution of the HER2 status has included a new HER2-low category defined as an HER2 immunohistochemistry score of 1+ or 2+ without gene amplification. This new category opens the door to a targetable HER2-low breast cancer population for which new treatments may be effective. OBJECTIVE.— To review the current literature on the emerging category of breast cancers with low HER2 protein expression, including the clinical, histopathologic, and molecular features, and outline the clinical trials and best practice recommendations for identifying HER2-low-expressing breast cancers by immunohistochemistry. DATA SOURCES.— We conducted a literature review based on peer-reviewed original articles, review articles, regulatory communications, ongoing and past clinical trials identified through ClinicalTrials.gov, and the authors' practice experience. CONCLUSIONS.— The availability of new targeted therapy potentially effective for patients with breast cancers with low HER2 protein expression requires multidisciplinary recognition. In particular, pathologists need to recognize and identify this category to allow the optimal selection of patients for targeted therapy.
Collapse
Affiliation(s)
- Gary Tozbikian
- From the Department of Pathology, The Ohio State University, Wexner Medical Center, Columbus (Tozbikian)
| | - Savitri Krishnamurthy
- the Department of Pathology (Krishnamurthy), The University of Texas MD Anderson Cancer Center, Houston
| | - Marilyn M Bui
- the Department of Pathology, Moffitt Cancer Center & Research Institute, Tampa, Florida (Bui)
| | - Michael Feldman
- the Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia (Feldman)
| | - David G Hicks
- the Department of Pathology, University of Rochester Medical Center, Rochester, New York (Hicks)
| | - Shabnam Jaffer
- the Department of Pathology, Mount Sinai Medical Center, New York, New York (Jaffer)
| | - Thaer Khoury
- the Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, New York (Khoury)
| | - Shi Wei
- the Department of Pathology, University of Kansas Medical Center; Kansas City (Wei)
| | - Hannah Wen
- the Department of Pathology, Memorial Sloan Kettering Cancer Center; New York, New York (Wen)
| | - Paula Pohlmann
- the Department of Breast Medical Oncology (Pohlmann), The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
9
|
Lee H, Cho YA, Kim DG, Cho EY. Next-Generation Sequencing in Breast Cancer Patients: Real-World Data for Precision Medicine. Cancer Res Treat 2024; 56:149-161. [PMID: 37591784 PMCID: PMC10789952 DOI: 10.4143/crt.2023.800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
PURPOSE Breast cancer is one of the most common causes of cancer-related death in females. Numerous drug-targetable biomarkers and predictive biomarkers have been developed. Some researchers have expressed doubts about the need for next-generation sequencing (NGS) studies in daily practice. This study analyzed the results of NGS studies on breast cancer at a single institute and evaluated the real-world applications of NGS data to precision medicine for breast cancer. MATERIALS AND METHODS We retrospectively collected the results of NGS studies and analyzed the histopathologic features and genetic profiles of patients treated for breast cancer from 2010 to 2021. Seventy cases had data from CancerSCAN, a customized panel of 375 cancer-associated genes, and 110 cases had data from TruSight Oncology 500. RESULTS The most frequently detected single nucleotide variant was the TP53 mutation (123/180, 68.3%), followed by PIK3CA mutations (51/180, 28.3%). Estrogen receptor 1 (ESR1) mutation was detected in 11 patients (6.1%), of whom 10 had hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer, and two had no history of prior endocrine therapy. Based on their NGS study results, 13 patients (7.2%) received target therapy. Among them, four patients had a BRCA1 or BRCA2 germline mutation, and nine patients had a PIK3CA mutation. CONCLUSION NGS can provide information about predictive biomarkers and drug-targetable biomarkers that can enable treatment and participation in clinical trials based on precision medicine. Further studies should be conducted to excavate novel drug-targetable biomarkers and develop additional target therapies.
Collapse
Affiliation(s)
- Hyunwoo Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoon Ah Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Deok Geun Kim
- Department of Digital Health, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Clinical Genomic Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Yoon Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Mu Y, Meng Y, Du Y, Liu X, Zhang J. Clinical characteristics and treatment outcomes of HER2 mutation and HER2 fusion in 22 patients with advanced breast cancer. Thorac Cancer 2023; 14:3381-3388. [PMID: 37863840 PMCID: PMC10693943 DOI: 10.1111/1759-7714.15130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND The clinical characteristics and efficacy of human epidermal growth factor receptor-2 (HER-2)-directed agents against HER2 mutations and HER2 fusions in breast cancer are obscure due to their low frequency. METHODS We conducted a retrospective study in patients with advanced breast cancer harboring HER2 mutations and/or HER2 fusions between January 1, 2017 and January 1, 2021. RESULTS Among a total of 22 patients, 17 HER2 mutations were detected, including L755S, S310F, R100=, V777L, R897W, T862A, 440-17C > G, H878Y, V842I, 73 + 9G > C, T278fs, E1069K, L755P, 226-11C > T, 574 + 12C>T, L114V and P128L. The majority of patients had ductal carcinoma, which mostly coexisted with HER2 amplification/overexpression. The median progression-free survival (PFS) of the 22 patients was 6.9 months (95% CI: 4.7, 9.1) in the first-line setting. The median PFS of patients who received first-line trastuzumab-based regimens was significantly longer than that of patients who received a first-line tyrosine kinase inhibitor (TKI) (10.8 months [95% CI: 2.9, 18.7] vs. 1.9 months [95% CI: 0.8, 3.0], p < 0.005). A total of 14 patients were treated with anti-HER2 antibody-drug conjugate (ADC), among whom the median treatment line of first-time of administration of anti-HER2 ADC was 4.5 (range, 1-10). Anti-HER2 ADC reached an objective response rate (ORR) of 42.9%, a disease control rate (DCR) of 85.7% and a median PFS of 7.3 months (95% CI: 4.4-10.1) from the first-time of administration. CONCLUSION Our data demonstrated the clinical benefit of anti-HER2 treatment in Chinese breast cancer patients harboring HER2 mutation and/or HER2 fusion. The value of immunotherapy and treatment selection among individual HER2 variants needs further study.
Collapse
Affiliation(s)
- Yuxin Mu
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Yanchun Meng
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Yiqun Du
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Xiaojun Liu
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Jian Zhang
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| |
Collapse
|
11
|
Jhaveri K, Eli LD, Wildiers H, Hurvitz SA, Guerrero-Zotano A, Unni N, Brufsky A, Park H, Waisman J, Yang ES, Spanggaard I, Reid S, Burkard ME, Vinayak S, Prat A, Arnedos M, Bidard FC, Loi S, Crown J, Bhave M, Piha-Paul SA, Suga JM, Chia S, Saura C, Garcia-Saenz JÁ, Gambardella V, de Miguel MJ, Gal-Yam EN, Rapael A, Stemmer SM, Ma C, Hanker AB, Ye D, Goldman JW, Bose R, Peterson L, Bell JSK, Frazier A, DiPrimeo D, Wong A, Arteaga CL, Solit DB. Neratinib + fulvestrant + trastuzumab for HR-positive, HER2-negative, HER2-mutant metastatic breast cancer: outcomes and biomarker analysis from the SUMMIT trial. Ann Oncol 2023; 34:885-898. [PMID: 37597578 PMCID: PMC11335023 DOI: 10.1016/j.annonc.2023.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND HER2 mutations are targetable alterations in patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC). In the SUMMIT basket study, patients with HER2-mutant MBC received neratinib monotherapy, neratinib + fulvestrant, or neratinib + fulvestrant + trastuzumab (N + F + T). We report results from 71 patients with HR+, HER2-mutant MBC, including 21 (seven in each arm) from a randomized substudy of fulvestrant versus fulvestrant + trastuzumab (F + T) versus N + F + T. PATIENTS AND METHODS Patients with HR+ HER2-negative MBC with activating HER2 mutation(s) and prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) therapy received N + F + T (oral neratinib 240 mg/day with loperamide prophylaxis, intramuscular fulvestrant 500 mg on days 1, 15, and 29 of cycle 1 then q4w, intravenous trastuzumab 8 mg/kg then 6 mg/kg q3w) or F + T or fulvestrant alone. Those whose disease progressed on F + T or fulvestrant could cross-over to N + F + T. Efficacy endpoints included investigator-assessed objective response rate (ORR), clinical benefit rate (RECIST v1.1), duration of response, and progression-free survival (PFS). Plasma and/or formalin-fixed paraffin-embedded tissue samples were collected at baseline; plasma was collected during and at end of treatment. Extracted DNA was analyzed by next-generation sequencing. RESULTS ORR for 57 N + F + T-treated patients was 39% [95% confidence interval (CI) 26% to 52%); median PFS was 8.3 months (95% CI 6.0-15.1 months). No responses occurred in fulvestrant- or F + T-treated patients; responses in patients crossing over to N + F + T supported the requirement for neratinib in the triplet. Responses were observed in patients with ductal and lobular histology, 1 or ≥1 HER2 mutations, and co-occurring HER3 mutations. Longitudinal circulating tumor DNA sequencing revealed acquisition of additional HER2 alterations, and mutations in genes including PIK3CA, enabling further precision targeting and possible re-response. CONCLUSIONS The benefit of N + F + T for HR+ HER2-mutant MBC after progression on CDK4/6is is clinically meaningful and, based on this study, N + F + T has been included in the National Comprehensive Cancer Network treatment guidelines. SUMMIT has improved our understanding of the translational implications of targeting HER2 mutations with neratinib-based therapy.
Collapse
Affiliation(s)
- K Jhaveri
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York; Weill Cornell Medical College, New York.
| | - L D Eli
- Clinical Development, Puma Biotechnology, Los Angeles, USA
| | - H Wildiers
- University Hospitals Leuven, Leuven, Belgium
| | - S A Hurvitz
- David Geffen School of Medicine, UCLA, Los Angeles, Santa Monica, USA
| | - A Guerrero-Zotano
- Medical Oncology Department, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - N Unni
- UT Southwestern Medical Center, Dallas
| | - A Brufsky
- Magee-Womens Hospital of UPMC, Pittsburgh
| | - H Park
- Washington University School of Medicine, St. Louis
| | - J Waisman
- City of Hope Comprehensive Cancer Center, Duarte
| | - E S Yang
- University of Alabama at Birmingham, Birmingham, USA
| | - I Spanggaard
- Department of Oncology, Rigshospitalet - Copenhagen University Hospital, Copenhagen, Denmark
| | - S Reid
- Division of Hematology/Oncology (Breast Oncology), The Vanderbilt-Ingram Cancer Center, Nashville
| | - M E Burkard
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - S Vinayak
- Seattle Cancer Care Alliance, Seattle, USA
| | - A Prat
- Hospital Clínic de Barcelona, Barcelona, Spain
| | - M Arnedos
- Department of Medical Oncology, Gustave Roussy, Villejuif
| | - F-C Bidard
- Department of Medical Oncology, UVSQ/Paris-Saclay University, Institut Curie, Saint Cloud, France
| | - S Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne; The Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | - J Crown
- St. Vincent's University Hospital, Dublin, Ireland
| | - M Bhave
- Department of Hematology/Oncology, Emory University, Winship Cancer Institute, Atlanta
| | - S A Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston
| | - J M Suga
- Kaiser Permanente, Department of Medical Oncology, Vallejo, USA
| | - S Chia
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - C Saura
- Medical Oncology Service, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona
| | - J Á Garcia-Saenz
- Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), CIBERONC, Madrid
| | - V Gambardella
- Hospital Clínico de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia
| | - M J de Miguel
- START Madrid - Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - E N Gal-Yam
- Institute of Breast Oncology, Sheba Medical Center, Ramat Gan
| | - A Rapael
- Sourasky Medical Center, Tel Aviv
| | - S M Stemmer
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva; Tel Aviv University, Tel Aviv, Israel
| | - C Ma
- Division of Medical Oncology, Department of Medicine and Siteman Cancer Center, Washington University, St. Louis
| | - A B Hanker
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas
| | - D Ye
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas
| | | | - R Bose
- Division of Medical Oncology, Department of Medicine and Siteman Cancer Center, Washington University, St. Louis
| | - L Peterson
- Division of Medical Oncology, Department of Medicine and Siteman Cancer Center, Washington University, St. Louis
| | | | - A Frazier
- Clinical Development, Puma Biotechnology, Los Angeles, USA
| | - D DiPrimeo
- Clinical Development, Puma Biotechnology, Los Angeles, USA
| | - A Wong
- Clinical Development, Puma Biotechnology, Los Angeles, USA
| | - C L Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas
| | - D B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
12
|
Marín A, Al Mamun A, Patel H, Akamatsu H, Ye D, Sudhan DR, Eli L, Marcelain K, Brown BP, Meiler J, Arteaga CL, Hanker AB. Acquired Secondary HER2 Mutations Enhance HER2/MAPK Signaling and Promote Resistance to HER2 Kinase Inhibition in Breast Cancer. Cancer Res 2023; 83:3145-3158. [PMID: 37404061 PMCID: PMC10530374 DOI: 10.1158/0008-5472.can-22-3617] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 07/06/2023]
Abstract
HER2 mutations drive the growth of a subset of breast cancers and are targeted with HER2 tyrosine kinase inhibitors (TKI) such as neratinib. However, acquired resistance is common and limits the durability of clinical responses. Most HER2-mutant breast cancers progressing on neratinib-based therapy acquire secondary mutations in HER2. It is unknown whether these secondary HER2 mutations, other than the HER2T798I gatekeeper mutation, are causal to neratinib resistance. Herein, we show that secondary acquired HER2T862A and HER2L755S mutations promote resistance to HER2 TKIs via enhanced HER2 activation and impaired neratinib binding. While cells expressing each acquired HER2 mutation alone were sensitive to neratinib, expression of acquired double mutations enhanced HER2 signaling and reduced neratinib sensitivity. Computational structural modeling suggested that secondary HER2 mutations stabilize the HER2 active state and reduce neratinib binding affinity. Cells expressing double HER2 mutations exhibited resistance to most HER2 TKIs but retained sensitivity to mobocertinib and poziotinib. Double-mutant cells showed enhanced MEK/ERK signaling, which was blocked by combined inhibition of HER2 and MEK. Together, these findings reveal the driver function of secondary HER2 mutations in resistance to HER2 inhibition and provide a potential treatment strategy to overcome acquired resistance to HER2 TKIs in HER2-mutant breast cancer. SIGNIFICANCE HER2-mutant breast cancers acquire secondary HER2 mutations that drive resistance to HER2 tyrosine kinase inhibitors, which can be overcome by combined inhibition of HER2 and MEK.
Collapse
Affiliation(s)
- Arnaldo Marín
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Doctoral Program in Medical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
- These authors contributed equally: Arnaldo Marin, Abdullah Al Mamun
| | - Abdullah Al Mamun
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- These authors contributed equally: Arnaldo Marin, Abdullah Al Mamun
| | - Hima Patel
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
| | - Hiroaki Akamatsu
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Current Address: Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Dan Ye
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Dhivya R. Sudhan
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Lisa Eli
- Puma Biotechnology, Inc., Los Angeles, CA 90024, USA
| | - Katherine Marcelain
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
| | - Benjamin P. Brown
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, 04103, Germany
| | - Carlos L. Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ariella B. Hanker
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
13
|
Mugoni V, Ciani Y, Quaini O, Tomasini S, Notarangelo M, Vannuccini F, Marinelli A, Leonardi E, Pontalti S, Martinelli A, Rossetto D, Pesce I, Mansy SS, Barbareschi M, Ferro A, Caffo O, Attard G, Di Vizio D, D'Agostino VG, Nardella C, Demichelis F. Integrating extracellular vesicle and circulating cell-free DNA analysis using a single plasma aliquot improves the detection of HER2 positivity in breast cancer patients. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e108. [PMID: 38046436 PMCID: PMC10688391 DOI: 10.1002/jex2.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 12/05/2023]
Abstract
Multi-analyte liquid biopsies represent an emerging opportunity for non-invasive cancer assessment. We developed ONCE (One Aliquot for Circulating Elements), an approach for the isolation of extracellular vesicles (EV) and cell-free DNA (cfDNA) from a single aliquot of blood. We assessed ONCE performance to classify HER2-positive early-stage breast cancer (BrCa) patients by combining EV-associated RNA (EV-RNA) and cfDNA signals on n = 64 healthy donors (HD) and non-metastatic BrCa patients. Specifically, we isolated EV-enriched samples by a charge-based (CB) method and investigated EV-RNA and cfDNA by next-generation sequencing (NGS) and by digital droplet PCR (ddPCR). Sequencing of cfDNA and EV-RNA from HER2- and HER2+ patients demonstrated concordance with in situ molecular analyses of matched tissues. Combined analysis of the two circulating analytes by ddPCR showed increased sensitivity in ERBB2/HER2 detection compared to single nucleic acid components. Multi-analyte liquid biopsy prediction performance was comparable to tissue-based sequencing results from TCGA. Also, imaging flow cytometry analysis revealed HER2 protein on the surface of EV isolated from the HER2+ BrCa plasma, thus corroborating the potential relevance of studying EV as companion analyte to cfDNA. This data confirms the relevance of combining cfDNA and EV-RNA for HER2 cancer assessment and supports ONCE as a valuable tool for multi-analytes liquid biopsies' clinical implementation.
Collapse
Affiliation(s)
- Vera Mugoni
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Yari Ciani
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Orsetta Quaini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Simone Tomasini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Michela Notarangelo
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Federico Vannuccini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Alessia Marinelli
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Elena Leonardi
- Unit of Surgical Pathology, Santa Chiara Hospital, APSSTrentoItaly
| | - Stefano Pontalti
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | - Angela Martinelli
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Daniele Rossetto
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Isabella Pesce
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Sheref S. Mansy
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | | | - Antonella Ferro
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | - Orazio Caffo
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | - Caterina Nardella
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| |
Collapse
|
14
|
Tian H, Qu M, Zhang G, Yuan L, Shi Q, Wang Y, Yang Y, Zhang Y, Qi X. Dramatic Response to Pyrotinib and T-DM1 in HER2-Negative Metastatic Breast Cancer With 2 Activating HER2 Mutations. Oncologist 2023:7147272. [PMID: 37120151 DOI: 10.1093/oncolo/oyad122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023] Open
Abstract
HER2 signaling is activated in response to somatic HER2 mutations, which are often found in invasive lobular breast cancer (ILC) and are associated with poor prognosis. Tyrosine kinase inhibitors (TKIs) have demonstrated considerable antitumor activity in patients with HER2-mutated advanced breast cancer (BC). Further, several clinical trials have indicated that HER2-targeted antibody-drug conjugates (ADCs) exhibit promising efficacy in lung cancer with HER2 mutations, and the efficacy of ADCs against HER2-mutated BC is currently being evaluated. Several preclinical studies have demonstrated that the therapeutic efficacy of ADCs in HER2-mutated cancer can be enhanced by the addition of irreversible TKIs, but the potential of such a combined treatment regimen for the treatment of HER2-mutated BC has not been reported. Herein, we describe a case in which a patient with estrogen receptor-positive/HER2-negative metastatic ILC with 2 activating HER2 mutations (D769H and V777L) exhibited a significant and durable response to anti-HER2 treatment with pyrotinib (an irreversible TKI) in combination with ado-trastuzumab emtansine, which was administered after multiple lines of therapy that had resulted in disease progression. Further, based on the evidence from the present case, TKI plus ADC seems to be a promising combination anti-HER2 regimen for patients with HER2-negative/HER2-mutated advanced BC, although further rigorous studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Hao Tian
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Man Qu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Guozhi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Long Yuan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qiyun Shi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yinhuan Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ying Yang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
15
|
Collins SJ, Guo J, Rizzo RC, Miller WT. Inhibition of mutationally activated HER2. Chem Biol Drug Des 2023; 101:87-102. [PMID: 36029027 PMCID: PMC9879383 DOI: 10.1111/cbdd.14125] [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: 04/25/2022] [Revised: 06/24/2022] [Accepted: 07/30/2022] [Indexed: 01/28/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is an oncogenic driver and key therapeutic target for human cancers. Current therapies targeting HER2 are primarily based on overexpression of the wild-type form of HER2. However, kinase domain mutations have been identified that can increase the activity of HER2 even when expressed at basal levels. Using purified enzymes, we confirmed the hyperactivity of two HER2 mutants (D769Y and P780insGSP). To identify small molecule inhibitors against these cancer-associated variants, we used a combined approach consisting of biochemical testing, similarity-based searching, and in silico modeling. These approaches resulted in the identification of a candidate molecule that inhibits mutant forms of HER2 in vitro and in cell-based assays. Our structural model predicts that the compound takes advantage of water-mediated interactions in the HER2 kinase binding pocket.
Collapse
Affiliation(s)
- Stephen J. Collins
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA
| | - Jiaye Guo
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York, USA
| | - Robert C. Rizzo
- Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York, USA,Laufer Center for Physical & Quantitative Biology, Stony Brook University, Stony Brook, New York, USA,Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA,Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York, USA,Department of Veterans Affairs Medical Center, Northport, New York, USA
| |
Collapse
|
16
|
Bourbour F, Pourtaheri A, Abbasi K, Hasanpour Ardekanizadeh N, Gholamalizadeh M, Hajipour A, Abdollahi S, Bagheri SE, Ahmadzadeh M, Doaei S, Haghighian A. Interactions dietary components with expression level of breast cancer-related genes. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00375-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract
Background
Dietary components can influence the effects of genetic background in breast cancer (BC). This review study aimed to investigate the effect of dietary components on the expression level of BC-related genes.
Methods
In this narrative review, Embase, PubMed, PsycInfo, and the Cochrane databases were used to collect the related papers with interactions of BC, genetics, and dietary intake. Appropriate keywords such as BC, gene expression, mutation, nutrient, and diet (alone and together) were applied for data collection.
Results
The association of BC with some genes including the BC1 gene (BRCA1), the human epidermal growth factor receptor 2 (HER2), and the fat mass and obesity-associated (FTO) gene can be affected by dietary components. Moderate B12 supplementation may be protective against BC in people with the inherited mutation of BRCA. The olive oil may have a protective effect against BC through several mechanisms such as suppressing HER-2 expression. Furthermore, high glycemic index foods may increase the risk of BC by the activation of the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway and the up-regulation of FTO gene expression.
Conclusion
There are interactions between BC, BC-related genes, and dietary intake. Dietary components such as macronutrients, micronutrients, and phytochemicals may regulate the expression level of BC-related genes. Further longitudinal studies are needed to confirm the associations between BC-related genes and diet and to discover the underlying mechanisms.
Collapse
|
17
|
The role of HER2 alterations in clinicopathological and molecular characteristics of breast cancer and HER2-targeted therapies: a comprehensive review. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:210. [PMID: 36175719 DOI: 10.1007/s12032-022-01817-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is the most common malignancy in women and one of the leading causes of cancer mortality, despite significant treatment advancements over the last decades. Human epidermal growth factor receptor-2 (HER2) is a member of the ERBB family of receptor tyrosine kinases which have long been known to mediate cancer cell growth and invasion through constitutive activation of oncogenic downstream signaling, such as PI3K/Akt/mTOR and MAPK. Overexpression/amplification of HER2 in various tumors, especially BC, offers the possible therapeutic potential for target therapies. HER2-targeted therapies, either with a combination of chemotherapy or through multi-anti-HER2 therapies without chemotherapy, have significantly improved the prognosis of HER2-positive tumors. In recent years, novel anti-HER2 agents and combination therapies have garnered much attention, especially for heavily treated advanced or metastatic BCs. HER2-positive BC is biologically a heterogeneous group depending on HER2 activation mechanisms, hormone receptor status, genome variations, tumor heterogeneity, and treatment resistance, which affect the treatment benefit and patients' outcomes. This review will discuss HER2 alternations (gene amplification or receptor overexpression) in BC, their correlation with clinicopathological characteristics and molecular characteristics, and HER2-based therapies in tumors with HER2 overexpression/amplification.
Collapse
|
18
|
Eli LD, Kavuri SM. Mechanisms of neratinib resistance in HER2-mutant metastatic breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:873-881. [PMID: 36627899 PMCID: PMC9771739 DOI: 10.20517/cdr.2022.48] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 01/13/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a major drug target and clinical biomarker in breast cancer treatment. Targeting HER2 gene amplification is one of the greatest successes in oncology, resulting in the use of a wide array of HER2-directed agents in the clinic. The discovery of HER2-activating mutations as novel therapeutic targets in breast and other cancers marked a significant advance in the field, which led to the metastatic breast and other solid tumor trials MutHER (NCT01670877), SUMMIT (NCT01953926), and one arm of plasmaMATCH (NCT03182634). These trials reported initial clinical benefit followed by eventual relapse ascribed to either primary or acquired resistance. These resistance mechanisms are mediated by additional secondary genomic alterations within HER2 itself and via hyperactivation of oncogenic signaling within the downstream signaling axis.
Collapse
Affiliation(s)
- Lisa D. Eli
- Translational Medicine and Diagnostics, Puma Biotechnology, Inc., Los Angeles, CA 90024, USA
| | - Shyam M. Kavuri
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
19
|
Kalra R, Chen CH, Wang J, Salam AB, Dobrolecki LE, Lewis A, Sallas C, Yates CC, Gutierrez C, Karanam B, Anurag M, Lim B, Ellis MJ, Kavuri SM. Poziotinib Inhibits HER2-Mutant-Driven Therapeutic Resistance and Multiorgan Metastasis in Breast Cancer. Cancer Res 2022; 82:2928-2939. [PMID: 35736563 PMCID: PMC9379360 DOI: 10.1158/0008-5472.can-21-3106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/01/2022] [Accepted: 06/14/2022] [Indexed: 01/07/2023]
Abstract
The pan-HER tyrosine kinase inhibitor (TKI) neratinib is therapeutically active against metastatic breast cancers harboring activating HER2 mutations, but responses are variable and often not durable. Here we demonstrate that recurrent HER2 mutations have differential effects on endocrine therapy responsiveness, metastasis, and pan-HER TKI therapeutic sensitivity. The prevalence and prognostic significance may also depend on whether the HER2 mutant has arisen in the context of lobular versus ductal histology. The most highly recurrent HER2 mutant, L755S, was particularly resistant to neratinib but sensitive to the pan-HER TKI poziotinib, alone or in combination with fulvestrant. Poziotinib reduced tumor growth, diminished multiorgan metastasis, and inhibited mTOR activation more effectively than neratinib. Similar therapeutic effects of poziotinib were observed in both an engineered HER2L755S MCF7 model and a patient-derived xenograft harboring a HER2G778_P780dup mutation. Overall, these findings support the need for clinical evaluation of poziotinib for the treatment of HER2-mutant metastatic breast cancer. SIGNIFICANCE Evaluation of the functional impact of HER2 mutations on therapy-induced resistance and metastasis identifies robust antitumor activity of poziotinib and supports the clinical evaluation of poziotinib in ER+ HER2 mutant breast cancer.
Collapse
Affiliation(s)
- Rashi Kalra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ching Hui Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Junkai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ahmad Bin Salam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Alaina Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Christina Sallas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Clayton C. Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Carolina Gutierrez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Balasubramanyam Karanam
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Bora Lim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Corresponding Authors: Shyam M. Kavuri, Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. Phone: 1314-651-3876; E-mail: ; and Matthew J. Ellis, Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. E-mail:
| | - Shyam M. Kavuri
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Corresponding Authors: Shyam M. Kavuri, Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. Phone: 1314-651-3876; E-mail: ; and Matthew J. Ellis, Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030. E-mail:
| |
Collapse
|
20
|
LeNoue-Newton ML, Chen SC, Stricker T, Hyman DM, Blauvelt N, Bedard PL, Meric-Bernstam F, Punglia RS, Schrag D, Lepisto EM, Andre F, Smyth L, Dogan S, Yu C, Wathoo C, Levy M, Eli LD, Xu F, Mann G, Lalani AS, Ye F, Micheel CM, Arnedos M. Natural History and Characteristics of ERBB2-mutated Hormone Receptor-positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case-control Study from AACR Project GENIE. Clin Cancer Res 2022; 28:2118-2130. [PMID: 35190802 DOI: 10.1158/1078-0432.ccr-21-0885] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/21/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE We wanted to determine the prognosis and the phenotypic characteristics of hormone receptor-positive advanced breast cancer tumors harboring an ERBB2 mutation in the absence of a HER2 amplification. EXPERIMENTAL DESIGN We retrospectively collected information from the American Association of Cancer Research-Genomics Evidence Neoplasia Information Exchange registry database from patients with hormone receptor-positive, HER2-negative, ERBB2-mutated advanced breast cancer. Phenotypic and co-mutational features, as well as response to treatment and outcome were compared with matched control cases ERBB2 wild type. RESULTS A total of 45 ERBB2-mutant cases were identified for 90 matched controls. The presence of an ERBB2 mutation was not associated with worse outcome determined by overall survival (OS) from first metastatic relapse. No significant differences were observed in phenotypic characteristics apart from higher lobular infiltrating subtype in the ERBB2-mutated group. ERBB2 mutation did not seem to have an impact in response to treatment or time-to-progression (TTP) to endocrine therapy compared with ERBB2 wild type. In the co-mutational analyses, CDH1 mutation was more frequent in the ERBB2-mutated group (FDR < 1). Although not significant, fewer co-occurring ESR1 mutations and more KRAS mutations were identified in the ERBB2-mutated group. CONCLUSIONS ERBB2-activating mutation was not associated with a worse OS from time of first metastatic relapse, or differences in TTP on treatment as compared with a series of matched controls. Although not significant, differences in coexisting mutations (CDH1, ESR1, and KRAS) were noted between the ERBB2-mutated and the control group.
Collapse
Affiliation(s)
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Thomas Stricker
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Rinaa S Punglia
- Department of Radiation Oncology, DFCI, Harvard Medical School, Boston, Massachusetts
| | - Deborah Schrag
- Division of Population Sciences and the Department of Medical Oncology, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Eva M Lepisto
- Division of Population Sciences and the Department of Medical Oncology, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Fabrice Andre
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lillian Smyth
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Semih Dogan
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Celeste Yu
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Chetna Wathoo
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Mia Levy
- Departments of Biomedical Informatics and Medicine, Division of Hematology/Oncology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa D Eli
- PUMA Biotechnology, Los Angeles, California
| | - Feng Xu
- PUMA Biotechnology, Los Angeles, California
| | - Grace Mann
- PUMA Biotechnology, Los Angeles, California
| | | | - Fei Ye
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Micheel
- Department of Medicine, Division of Hematology/Oncology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM Unit, U981, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
21
|
Shishido SN, Masson R, Xu L, Welter L, Prabakar RK, D' Souza A, Spicer D, Kang I, Jayachandran P, Hicks J, Lu J, Kuhn P. Disease characterization in liquid biopsy from HER2-mutated, non-amplified metastatic breast cancer patients treated with neratinib. NPJ Breast Cancer 2022; 8:22. [PMID: 35181666 PMCID: PMC8857263 DOI: 10.1038/s41523-022-00390-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 01/14/2022] [Indexed: 12/18/2022] Open
Abstract
Metastatic breast cancer (mBC) patients have a high risk of progression and face poor prognosis overall, with about one third (34%) surviving five years or more. In rare instances (2-4% of cases) patients with mBC have ERBB2 (HER2) activating mutations but are ERBB2 non-amplified. Neratinib is a potent, irreversible inhibitor that binds HER2 and inhibits downstream signaling. We used the previously validated high-definition single cell assay (HDSCA) workflow to investigate the clinical significance of the liquid biopsy in ERBB2 mutant, non-amplified, post-menopausal mBC patients starting neratinib and fulvestrant combination therapy. Characterization with a comprehensive liquid biopsy methodology (HDSCA) included genomic analysis of both the cell-free DNA (cfDNA) and single circulating tumor cells (CTCs) to monitor tumor evolution and identify potential mutational variants unique to the patient's clinical response. A limited series of five sequentially enrolled patients presented here were from the MutHER ( https://www.clinicaltrials.gov , NCT01670877) or SUMMIT ( https://www.clinicaltrials.gov , NCT01953926) trials. Patients had an average of 5.4 lines of therapy before enrollment, variable hormone receptor status, and ERBB2 mutations at diagnosis and during treatment. CTC enumeration alone was not sufficient to predict clinical response. Treatment pressure was shown to lead to an observable change in CTC morphology and genomic instability (GI), suggesting these parameters may inform prognosis. Single cell copy number alteration (CNA) analysis indicated that the persistence or development of a clonal population of CTCs during treatment was associated with a worse response. Hierarchical clustering analysis of the single cells across all patients and timepoints identified distinct aberrant regions shared among patients, comprised of 26 genes that are similarly affected and may be related to drug resistance. Additionally, the genomic analysis of the cfDNA, identified new mutations in ERBB2, PIK3CA, and TP53 that arose likely due to treatment pressure in a patient with poor response, further providing insights on the dynamics of the cancer genome over the course of therapy. The data presented in this small cohort study demonstrates the feasibility of real-time molecular profiling of the cellular and acellular fractions of the liquid biopsy using the HDSCA methodology. Additional studies are necessary to determine the potential use of morphometric and genomic analysis as a prognostic tool to advance personalized oncology.
Collapse
Affiliation(s)
- Stephanie N Shishido
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Rahul Masson
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Liya Xu
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Lisa Welter
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Rishvanth Kaliappan Prabakar
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Anishka D' Souza
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, NTT-3440, Los Angeles, CA, 90033, USA
| | - Darcy Spicer
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, NTT-3440, Los Angeles, CA, 90033, USA
| | - Irene Kang
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, NTT-3440, Los Angeles, CA, 90033, USA
| | - Priya Jayachandran
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, NTT-3440, Los Angeles, CA, 90033, USA
| | - James Hicks
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA
| | - Janice Lu
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Ave, NTT-3440, Los Angeles, CA, 90033, USA
| | - Peter Kuhn
- Convergent Science Institute in Cancer (CSI-Cancer), Michelson Center for Convergent Bioscience, University of Southern California,1002 Childs Way, MCB 220, Los Angeles, CA, 90089, USA.
| |
Collapse
|
22
|
Sturgill EG, Misch A, Lachs R, Jones CC, Schlauch D, Jones SF, Shastry M, Yardley DA, Burris HA, Spigel DR, Hamilton EP, McKenzie AJ. Next-Generation Sequencing of Patients With Breast Cancer in Community Oncology Clinics. JCO Precis Oncol 2022; 5:1297-1311. [PMID: 34994634 DOI: 10.1200/po.20.00469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Molecular biomarkers informing disease diagnosis, prognosis, and treatment decisions in patients with breast cancer are being uncovered by next-generation sequencing (NGS) technologies. In this study, we survey how NGS is used for patients with breast cancer in real-world settings with a focus on physician behaviors and sequencing results. METHODS We conducted a retrospective analysis of patients with breast cancer who received NGS testing from commercial vendors as part of standard of care from 2014 to 2019. A total of 2,635 NGS reports from 2,316 unique breast cancer patients were assessed. Hormone receptor and human epidermal growth factor receptor 2 statuses were abstracted from patient medical records. Comparative gene amplification and mutation frequencies were analyzed using Pearson's correlation and Lin's concordance statistics. RESULTS The number of physicians ordering NGS tests for patients with breast cancer increased more than six-fold from 2014 to 2019. Tissue- and plasma-based tests were ordered roughly equally by 2019, with plasma-based testing ordered most frequently in hormone receptor-positive subtypes. Patients with triple-negative breast cancer were most likely to receive NGS testing. Gene amplifications including ERBB2 were detected less frequently in our real-world data set as compared to previous genomic landscape studies, whereas the opposite was true for gene mutations including ESR1. Pathogenic mutations in the PI3K pathway (38.6%) and DNA damage repair pathway (11.0%) were frequently reported. Alterations were also reported across other cellular pathways. CONCLUSION Overall, we found that an increasing number of physicians in community settings are adopting NGS in the care of patients with breast cancer. Discrepancies between our real-world NGS data and previous genomic landscape studies are likely owed to the prevalence of plasma-based testing in community oncology clinics, as the reference data were from tissue-based NGS alone.
Collapse
Affiliation(s)
| | - Amanda Misch
- Sarah Cannon Research Institute, Nashville, TN.,Genospace, Boston, MA
| | - Rebecca Lachs
- Sarah Cannon Research Institute, Nashville, TN.,Genospace, Boston, MA
| | | | - Dan Schlauch
- Sarah Cannon Research Institute, Nashville, TN.,Genospace, Boston, MA
| | | | | | - Denise A Yardley
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, Nashville, TN
| | - Howard A Burris
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, Nashville, TN
| | - David R Spigel
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, Nashville, TN
| | - Erika P Hamilton
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, Nashville, TN
| | | |
Collapse
|
23
|
Diwanji D, Trenker R, Thaker TM, Wang F, Agard DA, Verba KA, Jura N. Structures of the HER2-HER3-NRG1β complex reveal a dynamic dimer interface. Nature 2021; 600:339-343. [PMID: 34759323 PMCID: PMC9298180 DOI: 10.1038/s41586-021-04084-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/29/2021] [Indexed: 02/04/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) and HER3 form a potent pro-oncogenic heterocomplex1-3 upon binding of growth factor neuregulin-1β (NRG1β). The mechanism by which HER2 and HER3 interact remains unknown in the absence of any structures of the complex. Here we isolated the NRG1β-bound near full-length HER2-HER3 dimer and, using cryo-electron microscopy, reconstructed the extracellulardomain module, revealing unexpected dynamics at the HER2-HER3 dimerization interface. We show that the dimerization arm of NRG1β-bound HER3 is unresolved because the apo HER2 monomer does not undergo a ligand-induced conformational change needed to establish a HER3 dimerization arm-binding pocket. In a structure of the oncogenic extracellular domain mutant HER2(S310F), we observe a compensatory interaction with the HER3 dimerization arm that stabilizes the dimerization interface. Both HER2-HER3 and HER2(S310F)-HER3 retain the capacity to bind to the HER2-directed therapeutic antibody trastuzumab, but the mutant complex does not bind to pertuzumab. Our structure of the HER2(S310F)-HER3-NRG1β-trastuzumab Fab complex reveals that the receptor dimer undergoes a conformational change to accommodate trastuzumab. Thus, similar to oncogenic mutations, therapeutic agents exploit the intrinsic dynamics of the HER2-HER3 heterodimer. The unique features of a singly liganded HER2-HER3 heterodimer underscore the allosteric sensing of ligand occupancy by the dimerization interface and explain why extracellular domains of HER2 do not homo-associate via a canonical active dimer interface.
Collapse
Affiliation(s)
- Devan Diwanji
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA,Medical Scientist Training Program, University of California San Francisco, San Francisco, CA 94158, USA
| | - Raphael Trenker
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Tarjani M. Thaker
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA,Department of Chemistry and Biochemistry, The University of Arizona, AZ 85721, USA
| | - Feng Wang
- Department of Biochemistry and Biophysics, University of California San Francisco, CA 94158, USA
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California San Francisco, CA 94158, USA
| | - Kliment A. Verba
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA 94158, USA,Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA,Correspondence should be addressed to K.A.V. () or N.J. ()
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA. .,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Biomarkers Changes after Neoadjuvant Chemotherapy in Breast Cancer: A Seven-Year Single Institution Experience. Diagnostics (Basel) 2021; 11:diagnostics11122249. [PMID: 34943486 PMCID: PMC8700750 DOI: 10.3390/diagnostics11122249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
The adoption of neoadjuvant chemotherapy (NACT) for breast cancer (BC) is increasing. The need to repeat the biomarkers on a residual tumor after NACT is still a matter of debate. We verified estrogen receptors (ER), progesterone receptors (PR), Ki67 and human epidermal growth factor receptor 2 (HER2) status changes impact in a retrospective monocentric series of 265 BCs undergoing NACT. All biomarkers changed with an overall tendency toward a reduced expression. Changes in PR and Ki67 were statistically significant (p = 0.001). Ki67 changed in 114/265 (43.0%) cases, PR in 44/265 (16.6%), ER in 31/265 (11.7%) and HER2 in 26/265 (9.8%). Overall, intrinsic subtype changed in 72/265 (27.2%) cases after NACT, and 10/265 (3.8%) cases switched to a different adjuvant therapy accordingly. Luminal subtypes changed most frequently (66/175; 31.7%) but with less impact on therapy (5/175; 2.8%). Only 3 of 58 triple-negative BCs (5.2%) changed their intrinsic subtype, but all of them switched treatment. No correlation was found between intrinsic subtype changes and clinicopathological features. To conclude, biomarkers changes with prognostic implications occurred in all BC intrinsic subtypes, albeit they impacted therapy mostly in HER2 negative and/or hormone receptors negative BCs. Biomarkers retesting after NACT is important to improve both tailored adjuvant therapies and prognostication of patients.
Collapse
|
25
|
Creeden JF, Nanavaty NS, Einloth KR, Gillman CE, Stanbery L, Hamouda DM, Dworkin L, Nemunaitis J. Homologous recombination proficiency in ovarian and breast cancer patients. BMC Cancer 2021; 21:1154. [PMID: 34711195 PMCID: PMC8555001 DOI: 10.1186/s12885-021-08863-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023] Open
Abstract
Homologous recombination and DNA repair are important for genome maintenance. Genetic variations in essential homologous recombination genes, including BRCA1 and BRCA2 results in homologous recombination deficiency (HRD) and can be a target for therapeutic strategies including poly (ADP-ribose) polymerase inhibitors (PARPi). However, response is limited in patients who are not HRD, highlighting the need for reliable and robust HRD testing. This manuscript will review BRCA1/2 function and homologous recombination proficiency in respect to breast and ovarian cancer. The current standard testing methods for HRD will be discussed as well as trials leading to approval of PARPi's. Finally, standard of care treatment and synthetic lethality will be reviewed.
Collapse
Affiliation(s)
- Justin Fortune Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Nisha S Nanavaty
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Katelyn R Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cassidy E Gillman
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | | - Danae M Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | |
Collapse
|
26
|
Lopes C, Piairo P, Chícharo A, Abalde-Cela S, Pires LR, Corredeira P, Alves P, Muinelo-Romay L, Costa L, Diéguez L. HER2 Expression in Circulating Tumour Cells Isolated from Metastatic Breast Cancer Patients Using a Size-Based Microfluidic Device. Cancers (Basel) 2021; 13:4446. [PMID: 34503260 PMCID: PMC8431641 DOI: 10.3390/cancers13174446] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
HER2 is a prognostic and predictive biomarker in breast cancer, normally assessed in tumour biopsy and used to guide treatment choices. Circulating tumour cells (CTCs) escape the primary tumour and enter the bloodstream, exhibiting great metastatic potential and representing a real-time snapshot of the tumour burden. Liquid biopsy offers the unique opportunity for low invasive sampling in cancer patients and holds the potential to provide valuable information for the clinical management of cancer patients. This study assesses the performance of the RUBYchip™, a microfluidic system for CTC capture based on cell size and deformability, and compares it with the only FDA-approved technology for CTC enumeration, CellSearch®. After optimising device performance, 30 whole blood samples from metastatic breast cancer patients were processed with both technologies. The expression of HER2 was assessed in isolated CTCs and compared to tissue biopsy. Results show that the RUBYchipTM was able to isolate CTCs with higher efficiency than CellSearch®, up to 10 times more, averaging all samples. An accurate evaluation of different CTC subpopulations, including HER2+ CTCs, was provided. Liquid biopsy through the use of the RUBYchipTM in the clinic can overcome the limitations of histological testing and evaluate HER2 status in patients in real-time, helping to tailor treatment during disease evolution.
Collapse
Affiliation(s)
- Cláudia Lopes
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Paulina Piairo
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Alexandre Chícharo
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Sara Abalde-Cela
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Liliana R. Pires
- RUBYnanomed Lda, Praça Conde de Agrolongo 123, 4700-312 Braga, Portugal;
| | - Patrícia Corredeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
| | - Patrícia Alves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706 Santiago de Compostela, Spain;
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Calle de Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Luís Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
- Oncology Division, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| |
Collapse
|
27
|
Hanker AB, Brown BP, Meiler J, Marín A, Jayanthan HS, Ye D, Lin CC, Akamatsu H, Lee KM, Chatterjee S, Sudhan DR, Servetto A, Brewer MR, Koch JP, Sheehan JH, He J, Lalani AS, Arteaga CL. Co-occurring gain-of-function mutations in HER2 and HER3 modulate HER2/HER3 activation, oncogenesis, and HER2 inhibitor sensitivity. Cancer Cell 2021; 39:1099-1114.e8. [PMID: 34171264 PMCID: PMC8355076 DOI: 10.1016/j.ccell.2021.06.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/28/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
Activating mutations in HER2 (ERBB2) drive the growth of a subset of breast and other cancers and tend to co-occur with HER3 (ERBB3) missense mutations. The HER2 tyrosine kinase inhibitor neratinib has shown clinical activity against HER2-mutant tumors. To characterize the role of HER3 mutations in HER2-mutant tumors, we integrate computational structural modeling with biochemical and cell biological analyses. Computational modeling predicts that the frequent HER3E928G kinase domain mutation enhances the affinity of HER2/HER3 and reduces binding of HER2 to its inhibitor neratinib. Co-expression of mutant HER2/HER3 enhances HER2/HER3 co-immunoprecipitation and ligand-independent activation of HER2/HER3 and PI3K/AKT, resulting in enhanced growth, invasiveness, and resistance to HER2-targeted therapies, which can be reversed by combined treatment with PI3Kα inhibitors. Our results provide a mechanistic rationale for the evolutionary selection of co-occurring HER2/HER3 mutations and the recent clinical observations that HER3 mutations are associated with a poor response to neratinib in HER2-mutant cancers.
Collapse
MESH Headings
- Aminopyridines/administration & dosage
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gain of Function Mutation
- Humans
- Mice, Nude
- Molecular Docking Simulation
- Molecular Dynamics Simulation
- Morpholines/administration & dosage
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors/administration & dosage
- Protein Multimerization
- Quinolines/administration & dosage
- Quinolines/chemistry
- Quinolines/metabolism
- Quinolines/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/chemistry
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Trastuzumab/pharmacology
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Ariella B Hanker
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Benjamin P Brown
- Chemical and Physical Biology Program, Center for Structural Biology, and Medical Scientist Training Program, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA; Institute for Drug Discovery, Leipzig University Medical School, Leipzig, SAC 04103, Germany
| | - Arnaldo Marín
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Doctoral Program in Medical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Harikrishna S Jayanthan
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Dan Ye
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Chang-Ching Lin
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Hiroaki Akamatsu
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Kyung-Min Lee
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04736, Republic of Korea
| | - Sumanta Chatterjee
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Dhivya R Sudhan
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Alberto Servetto
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA
| | - Monica Red Brewer
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - James P Koch
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan H Sheehan
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie He
- Foundation Medicine, Cambridge, MA 02141, USA
| | | | - Carlos L Arteaga
- UTSW Simmons Comprehensive Cancer Center, Dallas, 5323 Harry Hines Boulevard, TX 75390, USA; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
28
|
Clinicopathologic features and treatment advances in cancers with HER2 alterations. Biochim Biophys Acta Rev Cancer 2021; 1876:188605. [PMID: 34358635 DOI: 10.1016/j.bbcan.2021.188605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022]
Abstract
HER2 is one of the most important proteins of the epidermal growth factor receptor (EGFR) family, whose alterations include amplification, overexpression and gene mutation. Growing attention has been given to HER2 as a biomarker for prognosis, an indicator for treatment response and a target for new drugs. Tumors with HER2 alterations have been well studied in multiple locations as distinct entities for treatment, especially breast cancer, gastric cancer, lung cancer and colorectal cancer. These four cancers are the leading causes of cancer incidence and cancer-related death worldwide. The present study details the landscape of HER2 amplification/overexpression and mutations and gives an up-to-date analysis of current clinical trials in the four cancers mentioned above. Different HER2-altered cancers not only respond differently to HER2-targeting therapies but also display diverse survival outcomes. Even in the same type of cancer, HER2 amplification/overexpression differs from HER2 mutation in terms of clinicopathologic features and treatment strategies. As an emerging strategy in cancer treatment, immune checkpoint inhibitors demonstrate distinct outcomes in HER2-altered breast cancer, gastric cancer and lung cancer.
Collapse
|
29
|
Timbres J, Moss C, Mera A, Haire A, Gillett C, Van Hemelrijck M, Sawyer E. Survival Outcomes in Invasive Lobular Carcinoma Compared to Oestrogen Receptor-Positive Invasive Ductal Carcinoma. Cancers (Basel) 2021; 13:cancers13123036. [PMID: 34207042 PMCID: PMC8234044 DOI: 10.3390/cancers13123036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/19/2021] [Accepted: 06/07/2021] [Indexed: 01/19/2023] Open
Abstract
Invasive lobular breast cancer (ILC) accounts for 10-15% of breast cancers and has distinct characteristics compared with the more common invasive ductal carcinoma (IDC). Studies have shown that ILC may be less sensitive to chemotherapy than IDC, with lower rates of complete pathological response after neo-adjuvant chemotherapy, but it is not clear how this affects long-term survival. Patients at Guy's and St Thomas' NHS Foundation Trust between 1975 and 2016 diagnosed with ER+ IDC or ER+ ILC were eligible for inclusion. Kaplan-Meier plots and Cox proportional-hazards regression models were used for analysis. There was no difference in overall survival comparing ER+ ILC to ER+ IDC (OR: 0.94, 95% CI: 0.83, 1.04) with a median follow-up time of 8.3 years compared to 8.4 years in IDC. However, ER+HER2- ILC had worse survival compared to ER+HER2- IDC in those that received chemotherapy (OR: 1.46, 95% CI: 1.06, 2.01). Here, median follow-up time was 7.0 years in ILC compared to 8.1 years in IDC. These results indicate worse overall survival after chemotherapy (neo-adjuvant and adjuvant) in ILC compared to ER+HER2- IDC even when correcting for tumour grade, age, size, and nodal involvement, but validation is needed in a larger study population.
Collapse
Affiliation(s)
- Jasmine Timbres
- Breast Cancer Genetics, King’s College London, London SE1 9RT, UK;
- Correspondence:
| | - Charlotte Moss
- Translational Oncology and Urology Research, King’s College London, London SE1 9RT, UK; (C.M.); (A.H.); (M.V.H.)
| | - Anca Mera
- Guy’s & St. Thomas’ Hospital, London SE1 9RT, UK;
| | - Anna Haire
- Translational Oncology and Urology Research, King’s College London, London SE1 9RT, UK; (C.M.); (A.H.); (M.V.H.)
| | - Cheryl Gillett
- KHP Cancer Biobank, King’s College London, London SE1 9RT, UK;
| | - Mieke Van Hemelrijck
- Translational Oncology and Urology Research, King’s College London, London SE1 9RT, UK; (C.M.); (A.H.); (M.V.H.)
| | - Elinor Sawyer
- Breast Cancer Genetics, King’s College London, London SE1 9RT, UK;
| |
Collapse
|
30
|
Uchida S, Kojima T, Sugino T. Clinicopathological Features, Tumor Mutational Burden, and Tumour-Infiltrating Lymphocyte Interplay in ERBB2-Mutated Breast Cancer: In Silico Analysis. Pathol Oncol Res 2021; 27:633243. [PMID: 34257600 PMCID: PMC8262144 DOI: 10.3389/pore.2021.633243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/01/2021] [Indexed: 01/01/2023]
Abstract
Recent evidence suggests that somatic mutations in ERBB2 activate ERBB2 signaling. These mutations occur at a frequency of approximately 3% in breast cancer (BC). ERBB2 mutations indicate poor prognosis as they are associated with recurrence and metastasis. This study aimed to evaluate the clinicopathological features, immune infiltration levels, tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs) in ERBB2-mutated breast cancer (ERBB2-mutated BC) using a bioinformatic approach and publicly available datasets (i.e., TCGA-BRCA and TIMER2.0). ERBB2-mutated BCs were associated with a high histological grade. ERBB2-mutated BCs comprised invasive breast carcinoma of no special type (21/35, 60%), classic invasive lobular carcinoma (12/35, 34.3%), and pleomorphic invasive lobular carcinoma (2/35, 5.7%). A Kaplan-Meier survival curve demonstrated that ERBB2-mutated BC was associated with a significantly worse prognosis compared to ERBB2 non-mutated BC (p < 0.01). Furthermore, 40% (14/35) of the patients with ERBB2-mutated BC harbored CDH1 mutations. Mutations at L755 and V777 accounted for 30.5% of these mutations in ERBB2-mutated BC, suggesting that these sites are mutational hot spots in BC, particularly in invasive lobular carcinoma. Of the ERBB2-mutated BCs, 8.6% were classified as TIL-high, whereas 77.1% were TILs-low; TMB significantly correlated with TILs (p < 0.05). CD8+ T cell infiltration levels were significantly higher in ERBB2 non-mutated BC. Among ERBB2-mutated BCs, 22.9% were classified as TMB-high, which was significantly higher than the rate in the ERBB2 non-mutated BC (p < 0.01). These findings provide evidence for a link between ERBB2 mutations and high TMB in BC.
Collapse
Affiliation(s)
- Shiro Uchida
- Division of Diagnostic Pathology, Kikuna Memorial Hospital, Yokohama, Japan.,Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan.,Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takaaki Kojima
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
31
|
Qu Y, Liu Y, Ding K, Li Y, Hong X, Zhang H. Partial Response to Pyrotinib Plus Capecitabine in an Advanced Breast Cancer Patient with HER2 Amplification and R157W Mutation After Anti- HER2 Treatment: A Case Report and Literature Review. Onco Targets Ther 2021; 14:1581-1588. [PMID: 33688205 PMCID: PMC7936716 DOI: 10.2147/ott.s289876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/07/2021] [Indexed: 01/08/2023] Open
Abstract
Human epidermal growth factor receptor2 (HER2) overexpression/amplification is associated with high malignancy, rapid disease progression and poor overall survival in breast cancer. The application of anti-HER2 drugs has greatly improved the survival of patients with HER2-positive breast cancer, but drug resistance issues affect the long-term efficacy. The HER2 mutation is considered to be one of the reasons for resistance to anti-HER2 therapy, and there is currently no standard treatment. We report for the first time the detection of HER2 amplification with R157W mutation by second-generation sequencing (NGS) in a 57-year-old hormone receptor-negative, HER2-positive woman with advanced breast cancer who was resistant to multi-line anti-HER2 therapies. She subsequently received pyrotinib combined with capecitabine treatment and achieved partial response. The small-molecule pan-HER family irreversible inhibitor pyrotinib combined with capecitabine has shown a promising effect in the treatment of HER2 mutation-induced resistance, but the molecular mechanism and efficacy need to be further verified.
Collapse
Affiliation(s)
- Yanchun Qu
- Department of Oncology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, People's Republic of China.,The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yufeng Liu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Kailin Ding
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yong Li
- Department of Oncology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, People's Republic of China
| | - Xiaoyu Hong
- Nanjing Geneseeq Technology Inc, Nanjing, People's Republic of China
| | - Haibo Zhang
- Department of Oncology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, People's Republic of China
| |
Collapse
|
32
|
Kawaji H, Kubo M, Yamashita N, Yamamoto H, Kai M, Kajihara A, Yamada M, Kurata K, Kaneshiro K, Harada Y, Hayashi S, Shimazaki A, Mori H, Akiyoshi S, Oki E, Oda Y, Baba E, Mori M, Nakamura M. Comprehensive molecular profiling broadens treatment options for breast cancer patients. Cancer Med 2020; 10:529-539. [PMID: 33274848 PMCID: PMC7877356 DOI: 10.1002/cam4.3619] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/12/2020] [Accepted: 10/31/2020] [Indexed: 12/30/2022] Open
Abstract
Precision oncology with next generation sequencing (NGS) using tumor tissue with or without blood has begun in Japan. Tumor molecular profiling tests are available, including the OncoGuide™ NCC Oncopanel System and FoundationOne® CDx (F1CDx). Our purpose was to identify potentially actionable genetic alterations in breast cancer with this comprehensive tumor profiling test. We enrolled 115 patients with pathologically diagnosed advanced or metastatic breast cancer. Comprehensive tumor genomic profiling, microsatellite instability, and tumor mutational burden (TMB) were determined using F1CDx. Testing was successful in 109/115 cases (94.8%). Clinically actionable alterations were identified in 76% of advanced breast cancer patients. The most frequent short variants were in TP53 (48.6%), PIK3CA (38.5%), GATA3 (11.0%), PTEN (11.0%), and BRCA1 (10.1%), and structural variants were in ERBB2 (24.8%), MYC (21.1%), RAD21 (21.1%), CCND1 (11.9%), FGF19 (10.1%), and PTEN (10.1%). Regarding human epidermal growth factor receptor (HER)2 status, 106/109 samples (97.2%) were concordant between F1CDx and HER2 testing with immunohistochemistry/fluorescence in situ hybridization. However, ERBB2 amplification was newly detected in four samples and ERBB2 mutations were detected in five HER2‐negative breast cancer samples. Oncogenic BRCA mutations were found in three samples with F1CDx among 27 germline testing‐negative samples. The mean TMB in all samples was 6.28 mut/Mb and tended to be higher in luminal B and triple‐negative breast cancer (mean = 8.1 and 5.9 mut/Mb, respectively) compared with other subtypes. In conclusion, we established a system for precision oncology and obtained preliminary data with NGS as the first step. The information in this clinical sequencing panel will help guide the development of new treatments for breast cancer patients.
Collapse
Affiliation(s)
- Hitomi Kawaji
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Kubo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nami Yamashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaya Kai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsuko Kajihara
- Foundation Medicine Business Department, Foundation Medicine Unit, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Mai Yamada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kanako Kurata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhisa Kaneshiro
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yurina Harada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Saori Hayashi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Shimazaki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hitomi Mori
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sayuri Akiyoshi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
33
|
Exman P, Garrido-Castro AC, Hughes ME, Freedman RA, Li T, Trippa L, Bychkovsky BL, Barroso-Sousa R, Di Lascio S, Mackichan C, Lloyd MR, Krevalin M, Cerami E, Merrill MS, Santiago R, Crowley L, Kuhnly N, Files J, Lindeman NI, MacConaill LE, Kumari P, Tolaney SM, Krop IE, Bose R, Johnson BE, Ma CX, Dillon DA, Winer EP, Wagle N, Lin NU. Identifying ERBB2 Activating Mutations in HER2-Negative Breast Cancer: Clinical Impact of Institute-Wide Genomic Testing and Enrollment in Matched Therapy Trials. JCO Precis Oncol 2020; 3:1900087. [PMID: 32923853 DOI: 10.1200/po.19.00087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2019] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The yield of comprehensive genomic profiling in recruiting patients to molecular-based trials designed for small subgroups has not been fully evaluated. We evaluated the likelihood of enrollment in a clinical trial that required the identification of a specific genomic change based on our institute-wide genomic tumor profiling. PATIENTS AND METHODS Using genomic profiling from archived tissue samples derived from patients with metastatic breast cancer treated between 2011 and 2017, we assessed the impact of systematic genomic characterization on enrollment in an ongoing phase II trial (ClinicalTrials.gov identifier: NCT01670877). Our primary aim was to describe the proportion of patients with a qualifying ERBB2 mutation identified by our institutional genomic panel (OncoMap or OncoPanel) who enrolled in the trial. Secondary objectives included median time from testing result to trial registration, description of the spectrum of ERBB2 mutations, and survival. Associations were calculated using Fisher's exact test. RESULTS We identified a total of 1,045 patients with metastatic breast cancer without ERBB2 amplification who had available genomic testing results. Of these, 42 patients were found to have ERBB2 mutation and 19 patients (1.8%) were eligible for the trial on the basis of the presence of an activating mutation, 18 of which were identified by OncoPanel testing. Fifty-eight percent of potentially eligible patients were approached, and 33.3% of eligible patients enrolled in the trial guided exclusively by OncoPanel testing. CONCLUSION More than one half of eligible patients were approached for trial participation and, significantly, one third of those were enrolled in NCT01670877. Our data illustrate the ability to enroll patients in trials of rare subsets in routine clinical practice and highlight the need for these broadly based approaches to effectively support the success of these studies.
Collapse
Affiliation(s)
| | | | | | | | - Tianyu Li
- Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ian E Krop
- Dana-Farber Cancer Institute, Boston, MA
| | - Ron Bose
- Washington University School of Medicine, St. Louis, MO
| | | | - Cynthia X Ma
- Washington University School of Medicine, St. Louis, MO
| | - Deborah A Dillon
- Dana-Farber Cancer Institute, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | | | | | | |
Collapse
|
34
|
Ivanova E, Ward A, Wiegmans AP, Richard DJ. Circulating Tumor Cells in Metastatic Breast Cancer: From Genome Instability to Metastasis. Front Mol Biosci 2020; 7:134. [PMID: 32766277 PMCID: PMC7378584 DOI: 10.3389/fmolb.2020.00134] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of clinical resistance in repeatedly treated cancers extends from the primary tumor's capability to exploit genome instability to adapt, escape, and progress. Triple negative breast cancer serves as a good example of such a response demonstrating poor clinical outcome due to a high rate of cellular heterogeneity resulting in metastatic relapse. The capability to effectively track the emergence of therapeutic resistance in real-time and adapt the clinical response is the holy grail for precision medicine and has yet to be realized. In this review we present liquid biopsy using CTCs and ctDNA as a potential replacement and/or addition to the current diagnostic tests to deliver personalized therapies to patients with advanced breast cancer. We outline current uses of liquid biopsy in the metastatic breast cancer setting and discuss their limitations. In addition, we provide a detailed overview of common genome instability events in patients with metastatic breast cancer and how these can be tracked using liquid biopsy.
Collapse
Affiliation(s)
- Ekaterina Ivanova
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia.,Centre for Tumour and Immune Biology (ZTI), Philipps University Marburg, Marburg, Germany
| | - Ambber Ward
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston, QLD, Australia
| | - Adrian P Wiegmans
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia
| | - Derek John Richard
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolongabba, QLD, Australia
| |
Collapse
|
35
|
Gaibar M, Beltrán L, Romero-Lorca A, Fernández-Santander A, Novillo A. Somatic Mutations in HER2 and Implications for Current Treatment Paradigms in HER2-Positive Breast Cancer. JOURNAL OF ONCOLOGY 2020; 2020:6375956. [PMID: 32256585 PMCID: PMC7081042 DOI: 10.1155/2020/6375956] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
In one of every four or five cases of breast cancer, the human epidermal growth factor receptor-2 (HER2) gene is overexpressed. These carcinomas are known as HER2-positive. HER2 overexpression is linked to an aggressive phenotype and a lower rate of disease-free and overall survival. Drugs such as trastuzumab, pertuzumab, lapatinib, neratinib, and the more recent afatinib target the deregulation of HER2 expression. Some authors have attributed somatic mutations in HER2, a role in resistance to anti-HER2 therapy as differential regulation of HER2 has been observed among patients. Recently, studies in metastatic ER + tumors suggest that some HER2 mutations emerge as a mechanism of acquired resistance to endocrine therapy. In an effort to identify possible biomarkers of the efficacy of anti-HER2 therapy, we here review the known single-nucleotide polymorphisms (SNPs) of the HER2 gene found in HER2-positive breast cancer patients and their relationship with clinical outcomes. Information was recompiled on 11 somatic HER2 SNPs. Seven polymorphisms are located in the tyrosine kinase domain region of the gene contrasting with the low number of mutations found in extracellular and transmembrane areas. HER2-positive patients carrying S310F, S310Y, R678Q, D769H, or I767M mutations seem good candidates for anti-HER2 therapy as they show favorable outcomes and a good response to current pharmacological treatments. Carrying the L755S or D769Y mutation could also confer benefits when receiving neratinib or afatinib. By contrast, patients with mutations L755S, V842I, K753I, or D769Y do not seem to benefit from trastuzumab. Resistance to lapatinib has been reported in patients with L755S, V842I, and K753I. These data suggest that exploring HER2 SNPs in each patient could help individualize anti-HER2 therapies. Advances in our understanding of the genetics of the HER2 gene and its relations with the efficacy of anti-HER2 treatments are needed to improve the outcomes of patients with this aggressive breast cancer.
Collapse
Affiliation(s)
- Maria Gaibar
- Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C/Tajo, S/N, 28670 Villaviciosa de Odón, Madrid, Spain
| | - Laura Beltrán
- Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C/Tajo, S/N, 28670 Villaviciosa de Odón, Madrid, Spain
| | - Alicia Romero-Lorca
- Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C/Tajo, S/N, 28670 Villaviciosa de Odón, Madrid, Spain
| | - Ana Fernández-Santander
- Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C/Tajo, S/N, 28670 Villaviciosa de Odón, Madrid, Spain
| | - Apolonia Novillo
- Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C/Tajo, S/N, 28670 Villaviciosa de Odón, Madrid, Spain
| |
Collapse
|
36
|
Li A, Schleicher SM, Andre F, Mitri ZI. Genomic Alteration in Metastatic Breast Cancer and Its Treatment. Am Soc Clin Oncol Educ Book 2020; 40:1-14. [PMID: 32213086 DOI: 10.1200/edbk_280463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Metastatic breast cancer (mBC) remains responsible for the majority of breast cancer deaths. Whereas clinical outcomes have improved with the development of novel therapies, resistance almost inevitably develops, indicating the need for novel therapeutic approaches for the treatment of mBC. Recent investigations into mBC genomic alterations have revealed novel and potential therapeutic targets. Most notably, therapies against PIK3CA mutation and germline BRCA1/2 mutations have solidified the role of targeted therapy in mBC, with treatments against these alterations now approved by the U.S. Food and Drug Administration (FDA) on the basis of clinical benefit for patients with mBC. Familiarity with relevant genomic alterations in mBC, technologies for mutation detection, methods of interpreting genomic alterations, and an understanding of their clinical impact will aid practicing clinicians in the treatment of mBC as the field of breast oncology moves toward the era of precision medicine.
Collapse
Affiliation(s)
- Allen Li
- Department of Hematology Oncology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | | | - Fabrice Andre
- Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Zahi I Mitri
- Department of Hematology Oncology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| |
Collapse
|
37
|
Marchiò C, Annaratone L, Marques A, Casorzo L, Berrino E, Sapino A. Evolving concepts in HER2 evaluation in breast cancer: Heterogeneity, HER2-low carcinomas and beyond. Semin Cancer Biol 2020; 72:123-135. [PMID: 32112814 DOI: 10.1016/j.semcancer.2020.02.016] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/16/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
The human epidermal growth factor receptor 2 (HER2) is a well-known negative prognostic factor in breast cancer and a target of the monoclonal antibody trastuzumab as well as of other anti-HER2 compounds. Pioneering works on HER2-positive breast cancer in the 90s' launched a new era in clinical research and oncology practice that has reshaped the natural history of this disease. In diagnostic pathology the HER2 status is routinely assessed by using a combination of immunohistochemistry (IHC, to evaluate HER2 protein expression levels) and in situ hybridization (ISH, to assess HER2 gene status). For this purpose, international recommendations have been developed by a consensus of experts in the field, which have changed over the years according to new experimental and clinical data. In this review article we will document the changes that have contributed to a better evaluation of the HER2 status in clinical practice, furthermore we will discuss HER2 heterogeneity defined by IHC and ISH as well as by transcriptomic analysis and we will critically describe the complexity of HER2 equivocal results. Finally, we will introduce the clinical impact of HER2 mutations and we will define the upcoming category of HER2-low breast cancer with respect to emerging clinical data on the efficacy of specific anti-HER2 agents in subgroups of breast carcinomas lacking the classical oncogene addition dictated by HER2 amplification.
Collapse
Affiliation(s)
- Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Ana Marques
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Pathology Unit, Centro Hospitalar São João, Porto, Portugal
| | - Laura Casorzo
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Enrico Berrino
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
38
|
Chen Z, Sun T, Yang Z, Zheng Y, Yu R, Wu X, Yan J, Shao YW, Shao X, Cao W, Wang X. Monitoring treatment efficacy and resistance in breast cancer patients via circulating tumor DNA genomic profiling. Mol Genet Genomic Med 2019; 8:e1079. [PMID: 31867841 PMCID: PMC7005625 DOI: 10.1002/mgg3.1079] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/16/2019] [Accepted: 10/23/2019] [Indexed: 01/03/2023] Open
Abstract
Background One of the major challenges in managing invasive breast cancer (BC) is the lack of reliable biomarkers to track response. Circulating tumor DNA (ctDNA) from liquid biopsy, as a candidate biomarker, provides a valuable assessment of BC patients. In this retrospective study, we evaluated the utility of ctDNA to reflect the efficacy of treatment and to monitor resistance mechanisms. Methods Targeted next‐generation sequencing (NGS) of 416 cancer‐relevant genes was performed on 41 plasma biopsy samples of 19 HER2+ and 12 HER2‐ BC patients. Longitudinal ctDNA samples were analyzed in three BC patients over the treatment course for detecting acquired mutations. Results In HER2+ BC patients, ERBB2 somatic copy numbers in ctDNA samples were significantly higher in patients progressed on HER2‐targeted therapy than those who were still responding to the treatment. Recurrent acquired mutations were detected in genes including ERBB2, TP53, EGFR, NF1, and SETD2, which may contribute to trastuzumab resistance. In longitudinal analyses, the observed mutation allele frequencies were tracked closely in concordance with treatment responses. A novel ERBB2 p.(Leu869Arg) mutation was acquired in one patient upon resistant to trastuzumab therapy, which was further validated as an oncogenic mutation in vitro and contributed to resistance. In HER2‐ BC patients with chemotherapy resistance, genetic alterations on TP53, PIK3CA, and DNA damage repair genes were frequently observed. Conclusions In summary, ctDNA monitoring, particularly longitudinal analyses, provides valuable insights into the assessment of targeted therapy efficacy and gene alterations underlying trastuzumab resistance and chemotherapy resistance in HER2+ and HER2‐ BC patients, respectively.
Collapse
Affiliation(s)
- Zhanhong Chen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Tian Sun
- Translational Medicine Research Institute, Geneseeq Technology Inc, Toronto, Ontario, Canada
| | - Ziyan Yang
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, China
| | - Yabing Zheng
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ruoying Yu
- Translational Medicine Research Institute, Geneseeq Technology Inc, Toronto, Ontario, Canada
| | - Xue Wu
- Translational Medicine Research Institute, Geneseeq Technology Inc, Toronto, Ontario, Canada
| | - Junrong Yan
- Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yang W Shao
- Nanjing Geneseeq Technology Inc., Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiying Shao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wenming Cao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiaojia Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
39
|
Smyth LM, Piha-Paul SA, Won HH, Schram AM, Saura C, Loi S, Lu J, Shapiro GI, Juric D, Mayer IA, Arteaga CL, de la Fuente MI, Brufksy AM, Spanggaard I, Mau-Sørensen M, Arnedos M, Moreno V, Boni V, Sohn J, Schwartzberg LS, Gonzàlez-Farré X, Cervantes A, Bidard FC, Gorelick AN, Lanman RB, Nagy RJ, Ulaner GA, Chandarlapaty S, Jhaveri K, Gavrila EI, Zimel C, Selcuklu SD, Melcer M, Samoila A, Cai Y, Scaltriti M, Mann G, Xu F, Eli LD, Dujka M, Lalani AS, Bryce R, Baselga J, Taylor BS, Solit DB, Meric-Bernstam F, Hyman DM. Efficacy and Determinants of Response to HER Kinase Inhibition in HER2-Mutant Metastatic Breast Cancer. Cancer Discov 2019; 10:198-213. [PMID: 31806627 DOI: 10.1158/2159-8290.cd-19-0966] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/23/2019] [Accepted: 12/02/2019] [Indexed: 11/16/2022]
Abstract
HER2 mutations define a subset of metastatic breast cancers with a unique mechanism of oncogenic addiction to HER2 signaling. We explored activity of the irreversible pan-HER kinase inhibitor neratinib, alone or with fulvestrant, in 81 patients with HER2-mutant metastatic breast cancer. Overall response rate was similar with or without estrogen receptor (ER) blockade. By comparison, progression-free survival and duration of response appeared longer in ER+ patients receiving combination therapy, although the study was not designed for direct comparison. Preexistent concurrent activating HER2 or HER3 alterations were associated with poor treatment outcome. Similarly, acquisition of multiple HER2-activating events, as well as gatekeeper alterations, were observed at disease progression in a high proportion of patients deriving clinical benefit from neratinib. Collectively, these data define HER2 mutations as a therapeutic target in breast cancer and suggest that coexistence of additional HER signaling alterations may promote both de novo and acquired resistance to neratinib. SIGNIFICANCE: HER2 mutations define a targetable breast cancer subset, although sensitivity to irreversible HER kinase inhibition appears to be modified by the presence of concurrent activating genomic events in the pathway. These findings have implications for potential future combinatorial approaches and broader therapeutic development for this genomically defined subset of breast cancer.This article is highlighted in the In This Issue feature, p. 161.
Collapse
Affiliation(s)
- Lillian M Smyth
- Memorial Sloan Kettering Cancer Center, New York, New York.,St. Vincent's University Hospital, Dublin, Ireland
| | | | - Helen H Won
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Janice Lu
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | | | - Dejan Juric
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Carlos L Arteaga
- The University of Texas Southwestern Medical Center Harold C. Simmons Comprehensive Cancer Center, Dallas, Texas
| | | | - Adam M Brufksy
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | | | | | | | - Valentina Boni
- START Madrid Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Joohyuk Sohn
- Yonsei Cancer Center, University College of Medicine, Seoul, Korea
| | | | | | - Andrés Cervantes
- CIBERONC, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | | | | | | | | | - Gary A Ulaner
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | - Myra Melcer
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Yanyan Cai
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Grace Mann
- Puma Biotechnology, Inc., Los Angeles, California
| | - Feng Xu
- Puma Biotechnology, Inc., Los Angeles, California
| | - Lisa D Eli
- Puma Biotechnology, Inc., Los Angeles, California
| | | | | | | | - José Baselga
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
40
|
Shin JW, Kim S, Ha S, Choi B, Kim S, Im SA, Yoon TY, Chung J. The HER2 S310F Mutant Can Form an Active Heterodimer with the EGFR, Which Can Be Inhibited by Cetuximab but Not by Trastuzumab as well as Pertuzumab. Biomolecules 2019; 9:E629. [PMID: 31635022 PMCID: PMC6843359 DOI: 10.3390/biom9100629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
G309 or S310 mutations on the HER2 extracellular domain II induce receptor activation. Clinically, S310F is most frequent among HER2 extracellular domain mutations and patients with the S310F mutation without HER2 amplification responded to trastuzumab with or without the pertuzumab combination. However, the ability of S310F mutant to form homodimers or heterodimers with wild-type HER2 and other HER receptors, or their reactivity to trastuzumab and pertuzumab treatments, has not been reported. We overexpressed S310F as well as G309A, G309E and S310Y HER2 mutants and tested their reactivity to trastuzumab and pertuzumab. All mutants reacted to trastuzumab, but S310F mutant did not react to pertuzumab along with S310Y or G309E mutants. Thereafter, we tested the effects of trastuzumab and pertuzumab on 5637 cell line expressing both wild-type HER2 and S310F mutant. The ligand-independent HER2 homodimerization blocking antibody, trastuzumab, did not inhibit the activation of the HER2 receptor, suggesting that the S310F HER2 mutant did not form homodimers or heterodimers with wild-type HER2. Because 5637 cells overexpressed the EGFR, the effects of cetuximab and gefitinib were determined, and both inhibited the activation of HER2 and significantly reduced cell growth. Because pertuzumab did not inhibit the phosphorylation of HER2 while it bound to wild-type HER2, EGFR-mediated phosphorylation is expected to occur on the S310F mutant. To confirm whether the S310F mutant HER2 retained its affinity to the EGFR, single molecule interaction analyses using TIRF microscopy were performed, which showed that S310F mutant successfully formed complexes with EGFR. In conclusion, HER2 S310F mutant can form an active heterodimer with the EGFR and it can be inhibited by cetuximab, but not by trastuzumab in combination with pertuzumab.
Collapse
Affiliation(s)
- Jung Won Shin
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Soohyun Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
- Department of Cancer Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Suji Ha
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
- Department of Cancer Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Byungsan Choi
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| | - Seongyeong Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Tae-Young Yoon
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea.
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|
41
|
Gene Expression and miRNAs Profiling: Function and Regulation in Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Breast Cancer. Cancers (Basel) 2019; 11:cancers11050646. [PMID: 31083383 PMCID: PMC6562440 DOI: 10.3390/cancers11050646] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the second most common cause of cancer-related deaths among women worldwide. It is a heterogeneous disease with four major molecular subtypes. One of the subtypes, human epidermal growth factor receptor 2 (HER2)-enriched (HER2-positive) is characterized by the absence of estrogen and progesterone receptors and overexpression of HER2 receptor, and accounts for 15–20% of all breast cancers. Despite the anti-HER2 and cytotoxic chemotherapy, HER2 subtype is an aggressive disease with significant mortality. Recent advances in molecular biology techniques, including gene expression profiling, proteomics, and microRNA analysis, have been extensively used to explore the underlying mechanisms behind human breast carcinogenesis and metastasis including HER2-positive breast cancer, paving the way for developing new targeted therapies. This review focuses on recent advances on gene expression and miRNA status in HER2-positive breast cancer.
Collapse
|
42
|
Lei L, Ye WW, Zheng LF, Huang P, Shi L, Huang J, Zheng YB, Chen ZH, Wang XJ, Wang X. A significant response to a combination of trastuzumab and vinorelbine in HER2-negative metastatic breast cancer with HER2 V777L mutation. Onco Targets Ther 2019; 12:2931-2936. [PMID: 31118664 PMCID: PMC6475092 DOI: 10.2147/ott.s199931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Metastatic breast cancer (MBC) is the most life-threatening disease in women worldwide. HER2-mutated breast carcinoma has been reported to benefit from HER2-targeted tyrosine kinase inhibitors recently. Here, we presented a heavy pretreated and harbored HER2 V777L mutation de novo stage IV Luminal B (HER2 unamplified) breast cancer patient who achieved an unexpected good response to trastuzumab combined with vinorelbine therapy. Although HER2-unamplified MBC patients do not regularly benefit from anti-HER2 target therapy, HER2 V777L mutation detected by next-generation sequencing from ctDNA may present as a predictive biomarker for anti-HER2-based strategy therapy in HER2-negative MBC patients.
Collapse
Affiliation(s)
- Lei Lei
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China, .,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Wei-Wu Ye
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Lin-Feng Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Ping Huang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Lei Shi
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Jian Huang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Ya-Bing Zheng
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Zhan-Hong Chen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Xiao-Jia Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China,
| |
Collapse
|
43
|
Budiarto BR, Pohan PU, Desriani. Nucleic acid amplification-based HER2 molecular detection for breast cancer. JOURNAL OF ONCOLOGICAL SCIENCES 2019. [DOI: 10.1016/j.jons.2018.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
44
|
Abstract
Neratinib (Nerlynx™) is an oral, irreversible inhibitor of the human epidermal growth factor receptors HER1 (EGFR), HER2 and HER4. The drug originally arose from research by Wyeth (now Pfizer) and is now being developed by Puma Biotechnology primarily for the treatment of HER2-positive (HER+) breast cancer. Neratinib is approved in the USA for the extended adjuvant treatment of patients with HER2+ early-stage breast cancer who have been previously treated with a trastuzumab-based adjuvant regimen, and is in the preregistration phase for this indication in the EU. Neratinib, as monotherapy and/or combination therapy, is also in phase 3 development for metastatic breast cancer and in phase 1/2 development for advanced breast cancer and other solid tumours, including non-small cell lung cancer, colorectal cancer and glioblastoma. This article summarizes the milestones in the development of neratinib leading to this first approval for breast cancer.
Collapse
Affiliation(s)
- Emma D Deeks
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
45
|
Zhang Y, Yang W, Li D, Yang JY, Guan R, Yang MQ. Toward the precision breast cancer survival prediction utilizing combined whole genome-wide expression and somatic mutation analysis. BMC Med Genomics 2018; 11:104. [PMID: 30454048 DOI: 10.1109/bibm.2017.8217762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND Breast cancer is the most common type of invasive cancer in woman. It accounts for approximately 18% of all cancer deaths worldwide. It is well known that somatic mutation plays an essential role in cancer development. Hence, we propose that a prognostic prediction model that integrates somatic mutations with gene expression can improve survival prediction for cancer patients and also be able to reveal the genetic mutations associated with survival. METHOD Differential expression analysis was used to identify breast cancer related genes. Genetic algorithm (GA) and univariate Cox regression analysis were applied to filter out survival related genes. DAVID was used for enrichment analysis on somatic mutated gene set. The performance of survival predictors were assessed by Cox regression model and concordance index(C-index). RESULTS We investigated the genome-wide gene expression profile and somatic mutations of 1091 breast invasive carcinoma cases from The Cancer Genome Atlas (TCGA). We identified 118 genes with high hazard ratios as breast cancer survival risk gene candidates (log rank p < 0.0001 and c-index = 0.636). Multiple breast cancer survival related genes were found in this gene set, including FOXR2, FOXD1, MTNR1B and SDC1. Further genetic algorithm (GA) revealed an optimal gene set consisted of 88 genes with higher c-index (log rank p < 0.0001 and c-index = 0.656). We validated this gene set on an independent breast cancer data set and achieved a similar performance (log rank p < 0.0001 and c-index = 0.614). Moreover, we revealed 25 functional annotations, 15 gene ontology terms and 14 pathways that were significantly enriched in the genes that showed distinct mutation patterns in the different survival risk groups. These functional gene sets were used as new features for the survival prediction model. In particular, our results suggested that the Fanconi anemia pathway had an important role in breast cancer prognosis. CONCLUSIONS Our study indicated that the expression levels of the gene signatures remain the effective indicators for breast cancer survival prediction. Combining the gene expression information with other types of features derived from somatic mutations can further improve the performance of survival prediction. The pathways that were associated with survival risk suggested by our study can be further investigated for improving cancer patient survival.
Collapse
Affiliation(s)
- Yifan Zhang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - William Yang
- Department of Computer Science, Carnegie Mellon University School of Computer Science, 5000 Forbes Ave, Pittsburgh, 24105, USA
| | - Dan Li
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Jack Y Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Renchu Guan
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Mary Qu Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA.
| |
Collapse
|
46
|
Zhang Y, Yang W, Li D, Yang JY, Guan R, Yang MQ. Toward the precision breast cancer survival prediction utilizing combined whole genome-wide expression and somatic mutation analysis. BMC Med Genomics 2018; 11:104. [PMID: 30454048 PMCID: PMC6245494 DOI: 10.1186/s12920-018-0419-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Breast cancer is the most common type of invasive cancer in woman. It accounts for approximately 18% of all cancer deaths worldwide. It is well known that somatic mutation plays an essential role in cancer development. Hence, we propose that a prognostic prediction model that integrates somatic mutations with gene expression can improve survival prediction for cancer patients and also be able to reveal the genetic mutations associated with survival. Method Differential expression analysis was used to identify breast cancer related genes. Genetic algorithm (GA) and univariate Cox regression analysis were applied to filter out survival related genes. DAVID was used for enrichment analysis on somatic mutated gene set. The performance of survival predictors were assessed by Cox regression model and concordance index(C-index). Results We investigated the genome-wide gene expression profile and somatic mutations of 1091 breast invasive carcinoma cases from The Cancer Genome Atlas (TCGA). We identified 118 genes with high hazard ratios as breast cancer survival risk gene candidates (log rank p < 0.0001 and c-index = 0.636). Multiple breast cancer survival related genes were found in this gene set, including FOXR2, FOXD1, MTNR1B and SDC1. Further genetic algorithm (GA) revealed an optimal gene set consisted of 88 genes with higher c-index (log rank p < 0.0001 and c-index = 0.656). We validated this gene set on an independent breast cancer data set and achieved a similar performance (log rank p < 0.0001 and c-index = 0.614). Moreover, we revealed 25 functional annotations, 15 gene ontology terms and 14 pathways that were significantly enriched in the genes that showed distinct mutation patterns in the different survival risk groups. These functional gene sets were used as new features for the survival prediction model. In particular, our results suggested that the Fanconi anemia pathway had an important role in breast cancer prognosis. Conclusions Our study indicated that the expression levels of the gene signatures remain the effective indicators for breast cancer survival prediction. Combining the gene expression information with other types of features derived from somatic mutations can further improve the performance of survival prediction. The pathways that were associated with survival risk suggested by our study can be further investigated for improving cancer patient survival. Electronic supplementary material The online version of this article (10.1186/s12920-018-0419-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yifan Zhang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - William Yang
- Department of Computer Science, Carnegie Mellon University School of Computer Science, 5000 Forbes Ave, Pittsburgh, 24105, USA
| | - Dan Li
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Jack Y Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Renchu Guan
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Mary Qu Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA.
| |
Collapse
|
47
|
Identification, clinical-pathological characteristics and treatment outcomes of patients with metastatic breast cancer and somatic human epidermal growth factor receptor 2 (ERBB2) mutations. Breast Cancer Res Treat 2018; 174:55-63. [PMID: 30456437 DOI: 10.1007/s10549-018-5049-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 11/13/2018] [Indexed: 01/25/2023]
Abstract
PURPOSE The human epidermal growth factor receptor 2 (ERBB2) may harbour somatic mutations that drive breast tumorigenesis. Here, we study prevalence, tumour characteristics and disease outcome of ERBB2 mutations in a large unselected cohort of metastatic breast cancer (mBC) patients. METHODS We retrospectively included all mBC patients with sufficient primary breast tumour, diagnosed between 2000 and 2015 (n = 775). Genomic DNA was subjected to a targeted-resequencing assay to identify hotspot mutations in exon 8, 17, 19, 20, and 21 of ERBB2. We studied demographics, tumour characteristics, median distant disease-free survival (DDFS), using a time-to-event analysis and time to progression (TTP) and overall survival (OS) upon metastasis, using Kaplan-Meier and log-rank statistics to assess differences between ERBB2-mutation statuses. RESULTS ERBB2 mutations were observed in 1.8% of the samples (13/721). Patient and tumour characteristics were independent of ERBB2 mutations. Luminal ERBB2-mutated (ERBB2mut+) cases (n = 5) had a shorter DDFS than ERBB2mut- cases (median DDFS 0.8 vs. > 4.0 years, p = 0.02). ER-positive ERBB2mut+ patients who received an aromatase inhibitor (AI) as first-line treatment (stage IV disease) had a worse TTP vs. ERBB2mut- patients (n = 3 vs. 156; median TTP 103 vs. 311 days, p = 0.04). OS for all subtypes was lower for ERBB2mut+ vs. ERBB2mut- cases (n = 11 vs. 669; median OS 1.1 vs. 2.3 years, p = 0.46). CONCLUSION ERBB2mut+ are rare in patients in whom mBC developed and no evidence was found for an association with specific types of BC or patient characteristics, although outcomes of ERBB2mut+ carriers might be worse. The latter, however, needs to be validated in larger populations.
Collapse
|
48
|
Toss A, Piacentini F, Cortesi L, Artuso L, Bernardis I, Parenti S, Tenedini E, Ficarra G, Maiorana A, Iannone A, Omarini C, Moscetti L, Cristofanilli M, Federico M, Tagliafico E. Genomic alterations at the basis of treatment resistance in metastatic breast cancer: clinical applications. Oncotarget 2018; 9:31606-31619. [PMID: 30167082 PMCID: PMC6114971 DOI: 10.18632/oncotarget.25810] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/12/2018] [Indexed: 12/17/2022] Open
Abstract
The standard of care for breast cancer has gradually evolved from empirical treatments based on clinical-pathological characteristics to the use of targeted approaches based on the molecular profile of the tumor. Consequently, an increasing number of molecularly targeted drugs have been developed. These drugs target specific alterations, called driver mutations, which confer a survival advantage to cancer cells. To date, the main challenge remains the identification of predictive biomarkers for the selection of the optimal treatment. On this basis, we evaluated a panel of 25 genes involved in the mechanisms of targeted treatment resistance, in 16 primary breast cancers and their matched recurrences, developed during treatment. Overall, we found a detection rate of mutations higher than that described in the literature. In particular, the most frequently mutated genes were ERBB2 and those involved in the PI3K/AKT/mTOR and the MAPK signaling pathways. The study revealed substantial discordances between primary tumors and metastases, stressing the need for analysis of metastatic tissues at recurrence. We observed that 85.7% of patients with an early-stage or locally advanced primary tumor showed at least one mutation in the primary tumor. This finding could explain the subsequent relapse and might therefore justify more targeted adjuvant treatments. Finally, the mutations detected in 50% of relapsed tissues could have guided subsequent treatment choices in a different way. This study demonstrates that mutation events may be present at diagnosis or arise during cancer treatment. As a result, profiling primary and metastatic tumor tissues may be a major step in defining optimal treatments.
Collapse
Affiliation(s)
- Angela Toss
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Federico Piacentini
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Laura Cortesi
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Lucia Artuso
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Isabella Bernardis
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Parenti
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Tenedini
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Guido Ficarra
- Department of Pathology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Antonino Maiorana
- Department of Pathology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Anna Iannone
- Department of Diagnostics, Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudia Omarini
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Luca Moscetti
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Massimo Federico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Department of Diagnostics, Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
49
|
Mishra R, Hanker AB, Garrett JT. Genomic alterations of ERBB receptors in cancer: clinical implications. Oncotarget 2017; 8:114371-114392. [PMID: 29371993 PMCID: PMC5768410 DOI: 10.18632/oncotarget.22825] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/09/2017] [Indexed: 12/28/2022] Open
Abstract
The ERBB family of receptor tyrosine kinases has been implicated in carcinogenesis for over three decades with rigorous attention to EGFR and HER2. ERBB receptors, consisting of EGFR, HER2, HER3, and HER4 are part of a complicated signaling network that activates downstream signaling pathways including PI3K/AKT, Ras/Raf/MAPK, JAK/STAT and PKC. It is well established that EGFR is amplified and/or mutated in gliomas and non-small-cell lung carcinoma while HER2 is amplified and/or over-expressed in breast, gastric, ovarian, non-small cell lung carcinoma, and several other tumor types. With the advent of next generation sequencing and large scale efforts to explore the entire spectrum of genomic alterations involved in human cancer progression, it is now appreciated that somatic ERBB receptor mutations occur at relatively low frequencies across multiple tumor types. Some of these mutations may represent oncogenic driver events; clinical studies are underway to determine whether tumors harboring these alterations respond to small molecule EGFR/HER2 inhibitors. Recent evidence suggests that some somatic ERBB receptor mutations render resistance to FDA-approved EGFR and HER2 inhibitors. In this review, we focus on the landscape of genomic alterations of EGFR, HER2, HER3 and HER4 in cancer and the clinical implications for patients harboring these alterations.
Collapse
Affiliation(s)
- Rosalin Mishra
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, U.S.A
| | - Ariella B Hanker
- Department of Medicine, Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, U.S.A
| | - Joan T Garrett
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, U.S.A
| |
Collapse
|
50
|
Connell CM, Doherty GJ. Activating HER2 mutations as emerging targets in multiple solid cancers. ESMO Open 2017; 2:e000279. [PMID: 29209536 PMCID: PMC5708307 DOI: 10.1136/esmoopen-2017-000279] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) family of transmembrane receptor tyrosine kinases activates signalling pathways regulating cellular proliferation and survival. HER2 is a non-ligand-binding member of this family and exerts its activity through heterodimerisation with other EGFR family members. HER2 functional activation promotes oncogenesis, leading to the investigation of HER2-directed agents in cancers with HER2 alterations. This has been best characterised in the context of HER2 gene amplification in breast and gastro-oesophageal cancers, for which HER2-directed drugs form part of standard treatment regimens. More recently, somatic HER2 gene mutations have been detected in a range of human cancer types. Preclinical data suggest that functionally activating HER2 mutations may drive and maintain cancers in a manner analogous to HER2 gene amplification and that HER2 mutations may similarly confer sensitivity to HER2-directed drugs. Here, we critically review the emerging roles for HER2-directed drugs in HER2 mutant cancers. We review data from experimental models, where our knowledge of the underlying biology of HER2 mutational activation remains incomplete. We discuss clinical data from Phase I and II clinical trials which evaluate HER2-directed agents (tyrosine kinase inhibitors and antibody-based drugs) in several cancer types. We highlight the heterogeneity of HER2 mutations in human cancers, differences in the clinical efficacy of HER2-directed drugs between cancer types and possible mechanisms of primary and acquired resistance, in order to guide clinical practice and future drug development.
Collapse
Affiliation(s)
- Claire M Connell
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Gary J Doherty
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|