1
|
Altman JE, Olex AL, Zboril EK, Walker CJ, Boyd DC, Myrick RK, Hairr NS, Koblinski JE, Puchalapalli M, Hu B, Dozmorov MG, Chen XS, Chen Y, Perou CM, Lehmann BD, Visvader JE, Harrell JC. Single-cell transcriptional atlas of human breast cancers and model systems. Clin Transl Med 2024; 14:e70044. [PMID: 39417215 PMCID: PMC11483560 DOI: 10.1002/ctm2.70044] [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: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Breast cancer's complex transcriptional landscape requires an improved understanding of cellular diversity to identify effective treatments. The study of genetic variations among breast cancer subtypes at single-cell resolution has potential to deepen our insights into cancer progression. METHODS In this study, we amalgamate single-cell RNA sequencing data from patient tumours and matched lymph metastasis, reduction mammoplasties, breast cancer patient-derived xenografts (PDXs), PDX-derived organoids (PDXOs), and cell lines resulting in a diverse dataset of 117 samples with 506 719 total cells. These samples encompass hormone receptor positive (HR+), human epidermal growth factor receptor 2 positive (HER2+), and triple-negative breast cancer (TNBC) subtypes, including isogenic model pairs. Herein, we delineated similarities and distinctions across models and patient samples and explore therapeutic drug efficacy based on subtype proportions. RESULTS PDX models more closely resemble patient samples in terms of tumour heterogeneity and cell cycle characteristics when compared with TNBC cell lines. Acquired drug resistance was associated with an increase in basal-like cell proportions within TNBC PDX tumours as defined with SCSubtype and TNBCtype cell typing predictors. All patient samples contained a mixture of subtypes; compared to primary tumours HR+ lymph node metastases had lower proportions of HER2-Enriched cells. PDXOs exhibited differences in metabolic-related transcripts compared to PDX tumours. Correlative analyses of cytotoxic drugs on PDX cells identified therapeutic efficacy was based on subtype proportion. CONCLUSIONS We present a substantial multimodel dataset, a dynamic approach to cell-wise sample annotation, and a comprehensive interrogation of models within systems of human breast cancer. This analysis and reference will facilitate informed decision-making in preclinical research and therapeutic development through its elucidation of model limitations, subtype-specific insights and novel targetable pathways. KEY POINTS Patient-derived xenografts models more closely resemble patient samples in tumour heterogeneity and cell cycle characteristics when compared with cell lines. 3D organoid models exhibit differences in metabolic profiles compared to their in vivo counterparts. A valuable multimodel reference dataset that can be useful in elucidating model differences and novel targetable pathways.
Collapse
Affiliation(s)
- Julia E. Altman
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Amy L. Olex
- C. Kenneth and Diane Wright Center for Clinical and Translational ResearchVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Emily K. Zboril
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of BiochemistryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Carson J. Walker
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - David C. Boyd
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Rachel K. Myrick
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Nicole S. Hairr
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jennifer E. Koblinski
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Madhavi Puchalapalli
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Bin Hu
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mikhail G. Dozmorov
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - X. Steven Chen
- Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Yunshun Chen
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Charles M. Perou
- Lineberger Comprehensive Cancer CenterUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Brian D. Lehmann
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jane E. Visvader
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - J. Chuck Harrell
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
- Center for Pharmaceutical EngineeringVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
2
|
Jernström H, Rydén L. Into the twilight zone - should ER-low breast cancer be treated as triple negative breast cancer? THE LANCET REGIONAL HEALTH. EUROPE 2024; 40:100896. [PMID: 38590941 PMCID: PMC10999462 DOI: 10.1016/j.lanepe.2024.100896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024]
Affiliation(s)
- Helena Jernström
- Department of Clinical Sciences Lund, Oncology, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC) and Skåne University Hospital Comprehensive Cancer Center (SUHCC), Sweden
| | - Lisa Rydén
- Lund University Cancer Centre (LUCC) and Skåne University Hospital Comprehensive Cancer Center (SUHCC), Sweden
- Department of Clinical Sciences Lund, Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| |
Collapse
|
3
|
Bertucci F, Lerebours F, Ceccarelli M, Guille A, Syed N, Finetti P, Adélaïde J, Van Laere S, Goncalves A, Viens P, Birnbaum D, Mamessier E, Callens C, Bedognetti D. Mutational landscape of inflammatory breast cancer. J Transl Med 2024; 22:374. [PMID: 38637846 PMCID: PMC11025259 DOI: 10.1186/s12967-024-05198-4] [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: 03/08/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Inflammatory breast cancer (IBC) is the most pro-metastatic form of BC. Better understanding of its enigmatic pathophysiology is crucial. We report here the largest whole-exome sequencing (WES) study of clinical IBC samples. METHODS We retrospectively applied WES to 54 untreated IBC primary tumor samples and matched normal DNA. The comparator samples were 102 stage-matched non-IBC samples from TCGA. We compared the somatic mutational profiles, spectra and signatures, copy number alterations (CNAs), HRD and heterogeneity scores, and frequencies of actionable genomic alterations (AGAs) between IBCs and non-IBCs. The comparisons were adjusted for the molecular subtypes. RESULTS The number of somatic mutations, TMB, and mutational spectra were not different between IBCs and non-IBCs, and no gene was differentially mutated or showed differential frequency of CNAs. Among the COSMIC signatures, only the age-related signature was more frequent in non-IBCs than in IBCs. We also identified in IBCs two new mutational signatures not associated with any environmental exposure, one of them having been previously related to HIF pathway activation. Overall, the HRD score was not different between both groups, but was higher in TN IBCs than TN non-IBCs. IBCs were less frequently classified as heterogeneous according to heterogeneity H-index than non-IBCs (21% vs 33%), and clonal mutations were more frequent and subclonal mutations less frequent in IBCs. More than 50% of patients with IBC harbored at least one high-level of evidence (LOE) AGA (OncoKB LOE 1-2, ESCAT LOE I-II), similarly to patients with non-IBC. CONCLUSIONS We provide the largest mutational landscape of IBC. Only a few subtle differences were identified with non-IBCs. The most clinically relevant one was the higher HRD score in TN IBCs than in TN non-IBCs, whereas the most intriguing one was the smaller intratumor heterogeneity of IBCs.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France.
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France.
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
- Department of Public Health Sciences, University of Miami, Miami, USA
| | - Arnaud Guille
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Najeeb Syed
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Pascal Finetti
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - José Adélaïde
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Anthony Goncalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Daniel Birnbaum
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Emilie Mamessier
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Céline Callens
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Davide Bedognetti
- Tumor Biology and Immunology Laboratory, Research Branch, Sidra Medicine, Doha, Qatar
| |
Collapse
|
4
|
Drula R, Pardini B, Fu X, De los Santos MC, Jurj A, Pang L, El-Daly SM, Fabris L, Knutsen E, Dragomir MP, Bayraktar R, Li Y, Chen M, Del Vecchio F, Berland L, Dae J, Fan D, Shimizu M, Tran AM, Barzi M, Pioppini C, Gutierrez AM, Ivan C, Meas S, Hall CS, Alahari SK, Berindan-Neagoe I, Fabbri M, Lucci A, Arun B, Anfossi S, Calin GA. 17β-estradiol promotes extracellular vesicle release and selective miRNA loading in ERα-positive breast cancer. Proc Natl Acad Sci U S A 2023; 120:e2122053120. [PMID: 37252969 PMCID: PMC10266002 DOI: 10.1073/pnas.2122053120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/14/2023] [Indexed: 06/01/2023] Open
Abstract
The causes and consequences of abnormal biogenesis of extracellular vesicles (EVs) are not yet well understood in malignancies, including in breast cancers (BCs). Given the hormonal signaling dependence of estrogen receptor-positive (ER+) BC, we hypothesized that 17β-estradiol (estrogen) might influence EV production and microRNA (miRNA) loading. We report that physiological doses of 17β-estradiol promote EV secretion specifically from ER+ BC cells via inhibition of miR-149-5p, hindering its regulatory activity on SP1, a transcription factor that regulates the EV biogenesis factor nSMase2. Additionally, miR-149-5p downregulation promotes hnRNPA1 expression, responsible for the loading of let-7's miRNAs into EVs. In multiple patient cohorts, we observed increased levels of let-7a-5p and let-7d-5p in EVs derived from the blood of premenopausal ER+ BC patients, and elevated EV levels in patients with high BMI, both conditions associated with higher levels of 17β-estradiol. In brief, we identified a unique estrogen-driven mechanism by which ER+ BC cells eliminate tumor suppressor miRNAs in EVs, with effects on modulating tumor-associated macrophages in the microenvironment.
Collapse
Affiliation(s)
- Rares Drula
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- The Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337Cluj Napoca, Romania
| | - Barbara Pardini
- Italian Institute for Genomic Medicine, c/o FPO-IRCCS Candiolo, 10060Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060Candiolo, Italy
| | - Xiao Fu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi, China
| | - Mireia Cruz De los Santos
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, 17164Solna, Sweden
| | - Ancuta Jurj
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- The Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337Cluj Napoca, Romania
| | - Lan Pang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Sherien M. El-Daly
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Medical Biochemistry Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo12622, Egypt
| | - Linda Fabris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Erik Knutsen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Medical Biology, Faculty of Health Sciences, UiT, The Artic University of Norway, N-9037Tromso, Norway
- Centre for Clinical Research and Education, University Hospital of North Norway, N-9037Tromso, Norway
| | - Mihnea P. Dragomir
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10178Berlin, Germany
- German Cancer Research Center (DKFZ), Partner Site Berlin, and German Cancer Consortium (DKTK), 69120Heidelberg, Germany
- Berlin Institute of Health, 10178Berlin, Germany
| | - Recep Bayraktar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Yongfeng Li
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022 Zhejiang, P.R. China
| | - Meng Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Filippo Del Vecchio
- University of Hawaii Cancer Center, Cancer Biology Program, Honolulu, HI96813
| | - Léa Berland
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Research Imaging, Dana Farber Cancer Institute, Boston, MA02215
| | - Jessica Dae
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- College of Natural Sciences, The University of Texas at Austin, Austin, TX78712
| | - Daniel Fan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- College of Natural Sciences, The University of Texas at Austin, Austin, TX78712
| | - Masayoshi Shimizu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Anh M. Tran
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Chemistry, Mount Holyoke College, South Hadley, MA01075
| | - Mercedes Barzi
- Department of Pediatrics, Duke University, Durham, NC27708
| | - Carlotta Pioppini
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117Berlin, Germany
| | - Angelica M. Gutierrez
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Salyna Meas
- Breast Surgical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Carolyn S. Hall
- Breast Surgical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Suresh K. Alahari
- Department of Biochemistry and Molecular Biology, Stanley S Scott Cancer Center, Louisiana State University School of Medicine, New Orleans, LA70112
| | - Ioana Berindan-Neagoe
- The Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337Cluj Napoca, Romania
| | - Muller Fabbri
- University of Hawaii Cancer Center, Cancer Biology Program, Honolulu, HI96813
- Center for Cancer and Immunology Research, Children’s National Hospital, WashingtonDC20010
| | - Anthony Lucci
- Breast Surgical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Banu Arun
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Simone Anfossi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| |
Collapse
|
5
|
Panella R, Cotton CA, Maymi VA, Best S, Berry KE, Lee S, Batalini F, Vlachos IS, Clohessy JG, Kauppinen S, Paolo Pandolfi P. Targeting of microRNA-22 Suppresses Tumor Spread in a Mouse Model of Triple-Negative Breast Cancer. Biomedicines 2023; 11:biomedicines11051470. [PMID: 37239141 DOI: 10.3390/biomedicines11051470] [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: 10/27/2022] [Revised: 01/21/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
microRNA-22 (miR-22) is an oncogenic miRNA whose up-regulation promotes epithelial-mesenchymal transition (EMT), tumor invasion, and metastasis in hormone-responsive breast cancer. Here we show that miR-22 plays a key role in triple negative breast cancer (TNBC) by promoting EMT and aggressiveness in 2D and 3D cell models and a mouse xenograft model of human TNBC, respectively. Furthermore, we report that miR-22 inhibition using an LNA-modified antimiR-22 compound is effective in reducing EMT both in vitro and in vivo. Importantly, pharmacologic inhibition of miR-22 suppressed metastatic spread and markedly prolonged survival in mouse xenograft models of metastatic TNBC highlighting the potential of miR-22 silencing as a new therapeutic strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Riccardo Panella
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Center for Genomic Medicine, Desert Research Institute, Reno, NV 89512, USA
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Cody A Cotton
- Center for Genomic Medicine, Desert Research Institute, Reno, NV 89512, USA
| | - Valerie A Maymi
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sachem Best
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kelsey E Berry
- Center for Genomic Medicine, Desert Research Institute, Reno, NV 89512, USA
| | - Samuel Lee
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Felipe Batalini
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ioannis S Vlachos
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, 2450 Copenhagen, Denmark
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10154 Turin, Italy
- Renown Institute for Cancer, Nevada System of Higher Education, Reno, NV 89502, USA
| |
Collapse
|
6
|
Yang Z, Liu Y, Huang Y, Chen Z, Zhang H, Yu Y, Wang X, Cao X. The regrouping of Luminal B (HER2 negative), a better discriminator of outcome and recurrence score. Cancer Med 2022; 12:2493-2504. [PMID: 35909232 PMCID: PMC9939104 DOI: 10.1002/cam4.5089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/02/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Breast cancer (BC) remains the leading cause of cancer-related deaths worldwide. High recurrence risk Luminal BC receives adjuvant chemotherapy in addition to standard hormone therapy. Considering the heterogeneity of Luminal B BC, a more accurate classification model is urgently needed. METHODS In this study, we retrospectively reviewed the data of 1603 patients who were diagnosed with HER2-negative breast invasive ductal carcinoma. According to the expression level of PR and Ki-67 index, the Luminal B (HER2-negative) BCs were divided into three groups: ER+PR-Ki67low (ER-positive, PR-negative, and Ki-67 index <20%), ER+PR+Ki67high (ER-positive, PR-positive, and Ki-67 index ≥20%), and ER+PR-Ki67high (ER-positive, PR-negative, and Ki-67 index ≥20%). The cox proportional hazards regression model was used to evaluate the correlation between each variable and outcomes. Besides, discriminatory accuracy of the models was compared using the area under the receiver operating characteristic curve and log-rank χ2 value. RESULTS The analysis results showed that there was a significant correlation between subtypes using this newly defined classification and overall survival (p < 0.001) and disease-free survival (DFS) (p < 0.001). Interestingly, patients in the ER+PR-Ki67high subgroup have the worst survival outcome in Luminal B (HER2-negative) subtype, similar to Triple-negative patients. Besides, the ER+PR+Ki67high has worse 5-year DFS compared with Luminal A group. There was a significant relationship between the regrouping subtype and the recurrence score index (RI) (p < 0.001). Moreover, the results showed that patients in ER+PR-Ki67high subtype were more likely to have high RI for distance recurrence (RI-DR) and local recurrence (RI-LRR). Our newly defined classification has a better discrimination ability to predict survival outcome and recurrence score of Luminal B (HER2-negative) BC patients, which may help in clinical decision-making for individual treatment.
Collapse
Affiliation(s)
- Zheng‐Jun Yang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Yu‐Xiao Liu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Yue Huang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Zu‐Jin Chen
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Hao‐Zhi Zhang
- Key Laboratory of Cancer Prevention and TherapyTianjinChina,Department of Thyroid and Neck CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Yue Yu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Xin Wang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Xu‐Chen Cao
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| |
Collapse
|
7
|
Vathiotis IA, Trontzas I, Gavrielatou N, Gomatou G, Syrigos NK, Kotteas EA. Immune Checkpoint Blockade in Hormone Receptor-Positive Breast Cancer: Resistance Mechanisms and Future Perspectives. Clin Breast Cancer 2022; 22:642-649. [PMID: 35906130 DOI: 10.1016/j.clbc.2022.06.004] [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: 11/17/2021] [Revised: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Anti-programmed cell death protein 1 immunotherapy has been incorporated in the treatment algorithm of triple-negative breast cancer (TNBC). However, clinical trial results for patients with hormone receptor (HR)-positive disease appear less compelling. HR-positive tumors exhibit lower levels of programmed death-ligand 1 expression in comparison with their triple-negative counterparts. Moreover, signaling through estrogen receptor alters the immune microenvironment, rendering such tumors immunologically "cold." To explain differential responses to immune checkpoint blockade, this review interrogates differences between HR-positive and TNBC. Starting from distinct genomic features, we further present disparities concerning the tumor microenvironment and finally, we summarize early-phase clinical trial results on promising novel immunotherapy combinations.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece; Department of Pathology, Yale University School of Medicine, New Haven, CT.
| | - Ioannis Trontzas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Niki Gavrielatou
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Georgia Gomatou
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Nikolaos K Syrigos
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Elias A Kotteas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| |
Collapse
|
8
|
De Silva F, Alcorn J. A Tale of Two Cancers: A Current Concise Overview of Breast and Prostate Cancer. Cancers (Basel) 2022; 14:2954. [PMID: 35740617 PMCID: PMC9220807 DOI: 10.3390/cancers14122954] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is a global issue, and it is expected to have a major impact on our continuing global health crisis. As populations age, we see an increased incidence in cancer rates, but considerable variation is observed in survival rates across different geographical regions and cancer types. Both breast and prostate cancer are leading causes of morbidity and mortality worldwide. Although cancer statistics indicate improvements in some areas of breast and prostate cancer prevention, diagnosis, and treatment, such statistics clearly convey the need for improvements in our understanding of the disease, risk factors, and interventions to improve life span and quality of life for all patients, and hopefully to effect a cure for people living in developed and developing countries. This concise review compiles the current information on statistics, pathophysiology, risk factors, and treatments associated with breast and prostate cancer.
Collapse
Affiliation(s)
- Franklyn De Silva
- Drug Discovery & Development Research Group, College of Pharmacy and Nutrition, 104 Clinic Place, Health Sciences Building, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada
| | - Jane Alcorn
- Drug Discovery & Development Research Group, College of Pharmacy and Nutrition, 104 Clinic Place, Health Sciences Building, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada
| |
Collapse
|
9
|
Whitworth PW, Beitsch PD, Pellicane JV, Baron PL, Lee LA, Dul CL, Murray MK, Gittleman MA, Budway RJ, Rahman RL, Kelemen PR, Dooley WC, Rock DT, Cowan KH, Lesnikoski BA, Barone JL, Ashikari AY, Dupree BB, Wang S, Menicucci AR, Yoder EB, Finn C, Corcoran K, Blumencranz LE, Audeh W. Distinct Neoadjuvant Chemotherapy Response and 5-Year Outcome in Patients With Estrogen Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Tumors That Reclassify as Basal-Type by the 80-Gene Signature. JCO Precis Oncol 2022; 6:e2100463. [PMID: 35476550 PMCID: PMC9200401 DOI: 10.1200/po.21.00463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The 80-gene molecular subtyping signature (80-GS) reclassifies a proportion of immunohistochemistry (IHC)-defined luminal breast cancers (estrogen receptor–positive [ER+], human epidermal growth factor receptor 2–negative [HER2–]) as Basal-Type. We report the association of 80-GS reclassification with neoadjuvant treatment response and 5-year outcome in patients with breast cancer. Identity exposed: genomic assay unmasks TNBC-like breast cancer tumors disguised as HR+ #NBRST![]()
Collapse
Affiliation(s)
- Pat W Whitworth
- Nashville Breast Center, Nashville, TN.,Targeted Medical Education, Cupertino, CA
| | - Peter D Beitsch
- Targeted Medical Education, Cupertino, CA.,Dallas Surgical Group, Dallas, TX
| | | | - Paul L Baron
- Breast and Melanoma Specialist of Charleston, Charleston, SC.,Lenox Hill Hospital/Northwell Health, New York, NY
| | - Laura A Lee
- Comprehensive Cancer Center, Palm Springs, CA
| | - Carrie L Dul
- Ascension St John Hospital Great Lakes Cancer Management Specialists, Grosse Pointe Woods, MI
| | - Mary K Murray
- Akron General Medical Center, Akron, OH.,Cleveland Clinic Akron General, Akron, OH
| | | | | | | | - Pond R Kelemen
- Ashikari Breast Center, Sleepy Hollow, NY.,Zucker School of Medicine, Hofstra University, Hempstead, NY
| | - William C Dooley
- Breast Institute, University of Oklahoma Health Sciences, Oklahoma City, OK.,Stephenson Cancer Center, Oklahoma City, OK
| | - David T Rock
- Regional Breast Care, Fort Myers, FL.,Genesis Care, Fort Myers, FL
| | - Kenneth H Cowan
- Fred and Pamela Buffet Cancer Center and Eppley Institute for Research in Cancer at University of Nebraska Medical Center, Omaha, NE
| | - Beth-Ann Lesnikoski
- The Breast Institute at JFK Medical Center, Atlantis, FL.,Baptist MD Anderson Cancer Center, Jacksonville, FL
| | - Julie L Barone
- Exempla Saint Joseph Hospital, Denver, CO.,Vail Health, Vail, CO
| | - Andrew Y Ashikari
- Zucker School of Medicine, Hofstra University, Hempstead, NY.,Northwell Health Physician Partners, Mount Kisco, NY
| | - Beth B Dupree
- St Mary Medical Alliance Cancer Specialists, Langhorne, PA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Bergamino MA, Morani G, Parker J, Schuster EF, Leal MF, López-Knowles E, Tovey H, Bliss JM, Robertson JF, Smith IE, Dowsett M, Cheang MC. Impact of Duration of Neoadjuvant Aromatase Inhibitors on Molecular Expression Profiles in Estrogen Receptor-positive Breast Cancers. Clin Cancer Res 2022; 28:1217-1228. [PMID: 34965950 PMCID: PMC7612503 DOI: 10.1158/1078-0432.ccr-21-2718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/18/2021] [Accepted: 12/16/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Aromatase inhibitor (AI) treatment is the standard of care for postmenopausal women with primary estrogen receptor-positive breast cancer. The impact of duration of neoadjuvant endocrine therapy (NET) on molecular characteristics is still unknown. We evaluated and compared changes of gene expression profiles under short-term (2-week) versus longer-term neoadjuvant AIs. EXPERIMENTAL DESIGN Global gene expression profiles from the PeriOperative Endocrine Therapy for Individualised Care (POETIC) trial (137 received 2 weeks of AIs and 47 received no treatment) and targeted gene expression from 80 patients with breast cancer treated with NET for more than 1 month (NeoAI) were assessed. Intrinsic subtyping, module scores covering different cancer pathways and immune-related genes were calculated for pretreated and posttreated tumors. RESULTS The differences in intrinsic subtypes after NET were comparable between the two cohorts, with most Luminal B (90.0% in the POETIC trial and 76.3% in NeoAI) and 50.0% of HER2 enriched at baseline reclassified as Luminal A or normal-like after NET. Downregulation of proliferative-related pathways was observed after 2 weeks of AIs. However, more changes in genes from cancer-signaling pathways such as MAPK and PI3K/AKT/mTOR and immune response/immune-checkpoint components that were associated with AI-resistant tumors and differential outcome were observed in the NeoAI study. CONCLUSIONS Tumor transcriptional profiles undergo bigger changes in response to longer NET. Changes in HER2-enriched and Luminal B subtypes are similar between the two cohorts, thus AI-sensitive intrinsic subtype tumors associated with good survival might be identified after 2 weeks of AI. The changes of immune-checkpoint component expression in early AI resistance and its impact on survival outcome warrants careful investigation in clinical trials.
Collapse
Affiliation(s)
- Milana A. Bergamino
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Gabriele Morani
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Joel Parker
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | - Holly Tovey
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Judith M. Bliss
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - John F.R. Robertson
- Faculty of Medicine & Health Sciences, Queen's Medical Centre, Nottingham, United Kingdom
| | | | - Mitch Dowsett
- Royal Marsden Hospital, London, United Kingdom.,Breast Cancer Now Research Centre, The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Maggie C.U. Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.,Corresponding Author: Maggie C.U. Cheang, Clinical Trials and Statistics Unit (ICR-CTSU), The Institute of Cancer Research, 15 Cotswold Rd, Sutton SM2 5NG, United Kingdom. Phone: 4420-8722-4552; E-mail:
| |
Collapse
|
11
|
de Nonneville A, Finetti P, Mamessier E, Bertucci F. RE: NDRG1 in Aggressive Breast Cancer Progression and Brain Metastasis. J Natl Cancer Inst 2022; 114:1046-1047. [PMID: 35148398 PMCID: PMC9275762 DOI: 10.1093/jnci/djac031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/12/2022] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alexandre de Nonneville
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - François Bertucci
- Correspondence to: François Bertucci, MD, PhD, Department of Medical Oncology, Laboratory of Predictive Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009 Marseille, France (e-mail: )
| |
Collapse
|
12
|
Liu Q, Palmgren VA, Danen EHJ, Le Dévédec SE. Acute vs. chronic vs. intermittent hypoxia in breast Cancer: a review on its application in in vitro research. Mol Biol Rep 2022; 49:10961-10973. [PMID: 36057753 PMCID: PMC9618509 DOI: 10.1007/s11033-022-07802-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
Abstract
Hypoxia has been linked to elevated instances of therapeutic resistance in breast cancer. The exposure of proliferating cancer cells to hypoxia has been shown to induce an aggressive phenotype conducive to invasion and metastasis. Regions of the primary tumors in the breast may be exposed to different types of hypoxia including acute, chronic or intermittent. Intermittent hypoxia (IH), also called cyclic hypoxia, is caused by exposure to cycles of hypoxia and reoxygenation (H-R cycles). Importantly, there is currently no consensus amongst the scientific community on the total duration of hypoxia, the oxygen level, and the possible presence of H-R cycles. In this review, we discuss current methods of hypoxia research, to explore how exposure regimes used in experiments are connected to signaling by different hypoxia inducible factors (HIFs) and to distinct cellular responses in the context of the hallmarks of cancer. We highlight discrepancies in the existing literature on hypoxia research within the field of breast cancer in particular and propose a clear definition of acute, chronic, and intermittent hypoxia based on HIF activation and cellular responses: (i) acute hypoxia is when the cells are exposed for no more than 24 h to an environment with 1% O2 or less; (ii) chronic hypoxia is when the cells are exposed for more than 48 h to an environment with 1% O2 or less and (iii) intermittent hypoxia is when the cells are exposed to at least two rounds of hypoxia (1% O2 or less) separated by at least one period of reoxygenation by exposure to normoxia (8.5% O2 or higher). Our review provides for the first time a guideline for definition of hypoxia related terms and a clear foundation for hypoxia related in vitro (breast) cancer research.
Collapse
Affiliation(s)
- Qiuyu Liu
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Victoria A.C. Palmgren
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Erik HJ Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
13
|
Asleh K, Tu D, Gao D, Bramwell V, Levine MN, Pritchard KI, Shepherd LE, Nielsen TO. Predictive Significance of an Optimized Panel for Basal-like Breast Cancer: Results from the Canadian Cancer Trials Group MA.5 and MA.12 Phase III Clinical Trials. Clin Cancer Res 2021; 27:6570-6579. [PMID: 34615722 DOI: 10.1158/1078-0432.ccr-21-1942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Accurate IHC biomarkers incorporating nestin positivity or inositol polyphosphate-4-phosphate (INPP4B) loss have recently been optimized to identify the basal-like intrinsic breast cancer subtype regardless of estrogen, progesterone, or Her2 status. We examined the predictive capacity of these basal biomarkers in the CCTG MA.5 chemotherapy and MA.12 endocrine therapy trials. EXPERIMENTAL DESIGN Formalin-fixed paraffin embedded blocks of primary tumors from patients randomized in the two trials were used to build tissue microarrays. IHC staining for nestin and INPP4B followed published methods and REMARK criteria. A prespecified statistical plan tested the hypothesis that patients with basal breast cancer (nestin+ or INPP4B-) would not benefit from anthracycline substitution in MA.5 or from tamoxifen in MA.12. RESULTS Nestin positivity or INPP4B loss was observed in 110/453 (24%) interpretable samples from MA.5 and 47/366 (13%) from MA.12, and was associated with high grade, younger age, estrogen receptor negativity, triple-negative, core basal, and PAM50 basal-like subtypes. In the MA.5 trial, patients assigned as basal experienced lower benefit from anthracycline versus nonanthracycline adjuvant chemotherapy [HR, 1.49; 95% confidence interval (CI), 0.72-3.10] when compared with non-basal (nestin- and INPP4B+) cases where there was a higher benefit from anthracyclines (HR, 0.75; 95% CI, 0.54-1.04; P interaction = 0.01). In the MA.12 trial, patients assigned as basal did not demonstrate a benefit from adjuvant tamoxifen versus placebo (HR, 0.48; 95% CI, 0.12-1.86; P = 0.29), whereas nonbasal cases displayed significant benefit (HR, 0.66; 95% CI, 0.45-0.98; P = 0.04), although the interaction test was not significant. CONCLUSIONS The nestin/INPP4B IHC panel identifies women with basal breast cancers who benefit from nonanthracycline chemotherapy but not endocrine adjuvant treatments.
Collapse
Affiliation(s)
- Karama Asleh
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vivien Bramwell
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Mark N Levine
- Department of Medical Oncology, McMaster University, Hamilton, Ontario, Canada
| | | | - Lois E Shepherd
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Torsten O Nielsen
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
14
|
Villodre ES, Hu X, Larson R, Finetti P, Gomez K, Balema W, Stecklein SR, Santiago‐Sanchez G, Krishnamurthy S, Song J, Su X, Ueno NT, Tripathy D, Van Laere S, Bertucci F, Vivas‐Mejía P, Woodward WA, Debeb BG. Lipocalin 2 promotes inflammatory breast cancer tumorigenesis and skin invasion. Mol Oncol 2021; 15:2752-2765. [PMID: 34342930 PMCID: PMC8486564 DOI: 10.1002/1878-0261.13074] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive form of primary breast cancer characterized by rapid onset and high risk of metastasis and poor clinical outcomes. The biological basis for the aggressiveness of IBC is still not well understood and no IBC-specific targeted therapies exist. In this study, we report that lipocalin 2 (LCN2), a small secreted glycoprotein belonging to the lipocalin superfamily, is expressed at significantly higher levels in IBC vs non-IBC tumors, independently of molecular subtype. LCN2 levels were also significantly higher in IBC cell lines and in their culture media than in non-IBC cell lines. High expression was associated with poor-prognosis features and shorter overall survival in IBC patients. Depletion of LCN2 in IBC cell lines reduced colony formation, migration, and cancer stem cell populations in vitro and inhibited tumor growth, skin invasion, and brain metastasis in mouse models of IBC. Analysis of our proteomics data showed reduced expression of proteins involved in cell cycle and DNA repair in LCN2-silenced IBC cells. Our findings support that LCN2 promotes IBC tumor aggressiveness and offer a new potential therapeutic target for IBC.
Collapse
Affiliation(s)
- Emilly S. Villodre
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoding Hu
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Richard Larson
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Pascal Finetti
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Kristen Gomez
- Department of Biological SciencesThe University of Texas at BrownsvilleTXUSA
| | - Wintana Balema
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Shane R. Stecklein
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ginette Santiago‐Sanchez
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Savitri Krishnamurthy
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of PathologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Juhee Song
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoping Su
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Naoto T. Ueno
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Debu Tripathy
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Steven Van Laere
- Center for Oncological Research (CORE)Integrated Personalized and Precision Oncology Network (IPPON)University of AntwerpBelgium
| | - François Bertucci
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Pablo Vivas‐Mejía
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Wendy A. Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Bisrat G. Debeb
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
15
|
de Nonneville A, Finetti P, Birnbaum D, Mamessier E, Bertucci F. WEE1 Dependency and Pejorative Prognostic Value in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101030. [PMID: 34227743 PMCID: PMC8425927 DOI: 10.1002/advs.202101030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/19/2021] [Indexed: 06/12/2023]
Abstract
The WEE1 G2 checkpoint kinase acts as a negative cell cycle regulator for entry into mitosis (G2-to-M transition). This comment extends a recent Advanced Science paper by reporting higher WEE1-dependency of triple negative breast cancer (TNBC) cell lines, pejorative prognostic value of WEE1 expression in TNBC clinical samples as well as higher expression of biomarkers of sensitivity to WEE1 inhibitor.
Collapse
Affiliation(s)
- Alexandre de Nonneville
- Laboratory of Predictive OncologyInstitut Paoli‐CalmettesComprehensive Cancer CenterAix‐Marseille UnivMarseille13009France
| | - Pascal Finetti
- Laboratory of Predictive OncologyInstitut Paoli‐CalmettesComprehensive Cancer CenterAix‐Marseille UnivMarseille13009France
| | - Daniel Birnbaum
- Laboratory of Predictive OncologyInstitut Paoli‐CalmettesComprehensive Cancer CenterAix‐Marseille UnivMarseille13009France
| | - Emilie Mamessier
- Laboratory of Predictive OncologyInstitut Paoli‐CalmettesComprehensive Cancer CenterAix‐Marseille UnivMarseille13009France
| | - François Bertucci
- Laboratory of Predictive OncologyInstitut Paoli‐CalmettesComprehensive Cancer CenterAix‐Marseille UnivMarseille13009France
| |
Collapse
|
16
|
Schuler LA, Murdoch FE. Endogenous and Therapeutic Estrogens: Maestro Conductors of the Microenvironment of ER+ Breast Cancers. Cancers (Basel) 2021; 13:3725. [PMID: 34359625 PMCID: PMC8345134 DOI: 10.3390/cancers13153725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) marks heterogeneous breast cancers which display a repertoire of somatic genomic mutations and an immune environment that differs from other breast cancer subtypes. These cancers also exhibit distinct biological behaviors; despite an overall better prognosis than HER2+ or triple negative breast cancers, disseminated dormant cells can lead to disease recurrence decades after the initial diagnosis and treatment. Estrogen is the best studied driver of these cancers, and antagonism or reduction of estrogen activity is the cornerstone of therapeutic approaches. In addition to reducing proliferation of ERα+ cancer cells, these treatments also alter signals to multiple other target cells in the environment, including immune cell subpopulations, cancer-associated fibroblasts, and endothelial cells via several distinct estrogen receptors. In this review, we update progress in our understanding of the stromal cells populating the microenvironments of primary and metastatic ER+ tumors, the effects of estrogen on tumor and stromal cells to modulate immune activity and the extracellular matrix, and net outcomes in experimental and clinical studies. We highlight new approaches that will illuminate the unique biology of these cancers, provide the foundation for developing new treatment and prevention strategies, and reduce mortality of this disease.
Collapse
Affiliation(s)
- Linda A. Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | | |
Collapse
|
17
|
Yu KD, Cai YW, Wu SY, Shui RH, Shao ZM. Estrogen receptor-low breast cancer: Biology chaos and treatment paradox. Cancer Commun (Lond) 2021; 41:968-980. [PMID: 34251757 PMCID: PMC8504145 DOI: 10.1002/cac2.12191] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/23/2021] [Accepted: 06/19/2021] [Indexed: 02/03/2023] Open
Abstract
Hormone receptor testing mainly serves the purpose of guiding treatment choices for breast cancer patients. Patients with estrogen receptor (ER)‐positive breast cancers show significant response to endocrine therapy. However, the methods to define ER status and eligibility for treatment remain controversial. Despite recent guidelines considering staining ≥1% of tumor nuclei by immunohistology as ER‐positive, it has raised concerns on the benefit of endocrine therapy for tumors with ER 1%‐10% expression, termed “ER‐low positive”. This subgroup accounts for 3% to 9% of all patients and is likely to have unique molecular features, and therefore distinct therapeutic response to endocrine therapy compared with ER‐high positive tumors. The latest guidelines did not provide detailed descriptions for those patients, resulting in inconsistent treatment strategies. Consequently, we aimed to resolve this dilemma comprehensively. This review discusses molecular traits and recent ER‐low positive breast cancer innovations, highlighting molecular‐targeted treatment rather than traditional unified endocrine therapy for future basic and clinical research.
Collapse
Affiliation(s)
- Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China.,Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China.,Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Song-Yang Wu
- Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Ruo-Hong Shui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China.,Shanghai Key Laboratory of Breast Cancer, Shanghai, 200032, P. R. China
| |
Collapse
|
18
|
Núñez-Iglesias MJ, Novio S, García C, Pérez-Muñuzuri ME, Martínez MC, Santiago JL, Boso S, Gago P, Freire-Garabal M. Co-Adjuvant Therapy Efficacy of Catechin and Procyanidin B2 with Docetaxel on Hormone-Related Cancers In Vitro. Int J Mol Sci 2021; 22:7178. [PMID: 34281228 PMCID: PMC8268784 DOI: 10.3390/ijms22137178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/16/2023] Open
Abstract
Prostate (PC) and breast cancer (BC) are heterogeneous hormonal cancers. Treatment resistance and adverse effects are the main limitations of conventional chemotherapy treatment. The use of sensitizing agents could improve the effectiveness of chemotherapeutic drugs as well as obviate these limitations. This study analyzes the effect of single catechin (CAT), procyanidin B2 (ProB2) treatment as well as the co-adjuvant treatment of each of these compounds with docetaxel (DOCE). We used PC- and BC-derived cell lines (PC3, DU-145, T47D, MCF-7 and MDA-MB-231). The short and long-term pro-apoptotic, anti-proliferative and anti-migratory effects were analyzed. RT-qPCR was used to discover molecular bases of the therapeutic efficacy of these compounds. ProB2 treatment induced a two- to five-fold increase in anti-proliferative and pro-apoptotic effects compared to single DOCE treatment, and also had a more sensitizing effect than DOCE on DU145 cells. Regarding BC cells, ProB2- and CAT-mediated sensitization to DOCE anti-proliferative and pro-apoptotic effects was cell-independent and cell-dependent, respectively. Combined treatment led to high-efficacy effects on MCF-7 cells, which were associated to the up-regulation of CDKN1A, BAX, caspase 9 and E-cadherin mRNA under combined treatment compared to single DOCE treatment. CAT and ProB2 can enhance the efficacy of DOCE therapy on PC and BC cells by the sensitizing mechanism.
Collapse
Affiliation(s)
- Mª Jesús Núñez-Iglesias
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Silvia Novio
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Carlota García
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Mª Elena Pérez-Muñuzuri
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - María-Carmen Martínez
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - José-Luis Santiago
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Susana Boso
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Pilar Gago
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Manuel Freire-Garabal
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| |
Collapse
|
19
|
Prat A, Chaudhury A, Solovieff N, Paré L, Martinez D, Chic N, Martínez-Sáez O, Brasó-Maristany F, Lteif A, Taran T, Babbar N, Su F. Correlative Biomarker Analysis of Intrinsic Subtypes and Efficacy Across the MONALEESA Phase III Studies. J Clin Oncol 2021; 39:1458-1467. [PMID: 33769862 PMCID: PMC8196091 DOI: 10.1200/jco.20.02977] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The prognostic and predictive value of intrinsic subtypes in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer treated with endocrine therapy and ribociclib (RIB) is currently unknown. We evaluated the association of intrinsic subtypes with progression-free survival (PFS) in the MONALEESA trials. METHODS A retrospective and exploratory PAM50-based analysis of tumor samples from the phase III MONALEESA-2, MONALEESA-3, and MONALEESA-7 trials was undertaken. The prognostic relationship of PAM50-based subtypes with PFS and risk of disease progression by subtype and treatment were evaluated using a multivariable Cox proportional hazards model, adjusting for age, prior chemotherapy, performance status, visceral disease, bone-only metastases, histological grade, number of metastatic sites, prior endocrine therapy, and de novo metastatic disease. RESULTS Overall, 1,160 tumors from the RIB (n = 672) and placebo (n = 488) cohorts were robustly profiled. Subtype distribution was luminal A (LumA), 46.7%; luminal B (LumB), 24.0%; normal-like, 14.0%; HER2-enriched (HER2E), 12.7%; and basal-like, 2.6% and was generally consistent across treatment arms and trials. The associations between subtypes and PFS were statistically significant in both arms (P < .001). The risks of disease progression for LumB, HER2E, and basal-like subtypes were 1.44, 2.31, and 3.96 times higher compared with those for LumA, respectively. All subtypes except basal-like demonstrated significant PFS benefit with RIB. HER2E (hazard ratio [HR], 0.39; P < .0001), LumB (HR, 0.52; P < .0001), LumA (HR, 0.63; P = .0007), and normal-like (HR, 0.47; P = .0005) subtypes derived benefit from RIB. Patients with basal-like subtype (n = 30) did not derive benefit from RIB (HR, 1.15; P = .77). CONCLUSION In this retrospective exploratory analysis of hormone receptor-positive and human epidermal growth factor receptor 2-negative advanced breast cancer, each intrinsic subtype exhibited a consistent PFS benefit with RIB, except for basal-like.
Collapse
Affiliation(s)
- Aleix Prat
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain,SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Department of Medicine, University of Barcelona, Barcelona, Spain,Institute of Oncology (IOB) Quiron, Barcelona, Spain,Aleix Prat, MD, PhD, Hospital Clínic of Barcelona, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Villarroel 170, 08035 Barcelona, Spain; e-mail:
| | | | | | - Laia Paré
- SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Débora Martinez
- SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Chic
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain,SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Olga Martínez-Sáez
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain,SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fara Brasó-Maristany
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain,SOLTI Breast Cancer Research Group, Barcelona, Spain,Translational Genomics and Targeted Therapies in Solid Tumors, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Agnes Lteif
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | | | - Naveen Babbar
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Fei Su
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| |
Collapse
|
20
|
Sheng IY, Barata P, Alameddine R, Garcia JA. Volume matters and intensification is needed: emerging trends in the management of advanced prostate cancer. Drugs Context 2021; 10:2020-10-2. [PMID: 33796138 PMCID: PMC7968923 DOI: 10.7573/dic.2020-10-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/07/2020] [Indexed: 11/21/2022] Open
Abstract
Significant changes in the management of patients with de novo metastatic prostate cancer have led to the use of novel oral agents and docetaxel-based chemotherapy earlier in the natural history of their disease. Our main challenge is the lack of prospective randomized data comparing these regimens. It is clear that treatment intensification is needed. Yet, the heterogeneity of this patient population coupled with the lack of understanding of the specific biology for a given individual makes treatment selection challenging. The aim of this narrative review is to discuss the importance of defining advanced disease by volume, the necessity for treatment intensification, and the current and future landscape of metastatic hormone-sensitive prostate cancer management.
Collapse
Affiliation(s)
- Iris Y Sheng
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Pedro Barata
- Department of Internal Medicine, Section of Hematology Oncology, Tulane University Medical School, New Orleans, LA, USA
| | - Raafat Alameddine
- Department of Hematology Oncology, University Hospital Cleveland Medical Center, Cleveland, OH, USA
| | - Jorge A Garcia
- Department of Hematology Oncology, University Hospital Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
21
|
Torrisi R, Marrazzo E, Agostinetto E, De Sanctis R, Losurdo A, Masci G, Tinterri C, Santoro A. Neoadjuvant chemotherapy in hormone receptor-positive/HER2-negative early breast cancer: When, why and what? Crit Rev Oncol Hematol 2021; 160:103280. [PMID: 33667658 DOI: 10.1016/j.critrevonc.2021.103280] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 02/17/2021] [Accepted: 02/27/2021] [Indexed: 12/13/2022] Open
Abstract
Indication for neoadjuvant chemotherapy (NACT) in HR+/HER2-negative tumors is controversial. Pathological complete response (pCR) rates range from 0 to 18 % while breast-conserving surgery (BCS) is achievable in up to 60 % of tumors. No pathological feature definitely predicts pCR; lobular and molecular luminal A tumors are less likely to achieve pCR although experiencing better outcomes. Luminal B subtype, high proliferation, lack of progesterone receptor, high tumor-infiltrating lymphocytes are positively associated with increased pCR rates but worse outcomes and the prognostic role of pCR is inconsistent across studies. Molecular intrinsic subtyping and genomic signatures appear as more accurate predictors of benefit from NACT, but larger studies are needed. Anthracycline and taxane-based chemotherapy remains the standard NACT; however, CDK 4/6 inhibitors and immune checkpoint inhibitors are under evaluation. In conclusion, NACT may be proposed for luminal tumors requiring downsizing for BCS after multidisciplinary evaluation, provided that other contraindications to BCS are excluded.
Collapse
Affiliation(s)
- Rosalba Torrisi
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy.
| | - Emilia Marrazzo
- IRCCS Humanitas Research Hospital, Breast Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Elisa Agostinetto
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| | - Rita De Sanctis
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| | - Agnese Losurdo
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Giovanna Masci
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Corrado Tinterri
- IRCCS Humanitas Research Hospital, Breast Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Armando Santoro
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| |
Collapse
|
22
|
Li L, Patil D, Petruncio G, Harnden KK, Somasekharan JV, Paige M, Wang LV, Salvador-Morales C. Integration of Multitargeted Polymer-Based Contrast Agents with Photoacoustic Computed Tomography: An Imaging Technique to Visualize Breast Cancer Intratumor Heterogeneity. ACS NANO 2021; 15:2413-2427. [PMID: 33464827 PMCID: PMC8106867 DOI: 10.1021/acsnano.0c05893] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
One of the primary challenges in breast cancer diagnosis and treatment is intratumor heterogeneity (ITH), i.e., the coexistence of different genetically and epigenetically distinct malignant cells within the same tumor. Thus, the identification of ITH is critical for designing better treatments and hence to increase patient survival rates. Herein, we report a noninvasive hybrid imaging technology that integrates multitargeted and multiplexed patchy polymeric photoacoustic contrast agents (MTMPPPCAs) with single-impulse panoramic photoacoustic computed tomography (SIP-PACT). The target specificity ability of MTMPPPCAs to distinguish estrogen and progesterone receptor-positive breast tumors was demonstrated through both fluorescence and photoacoustic measurements and validated by tissue pathology analysis. This work provides the proof-of-concept of the MTMPPPCAs/SIP-PACT system to identify ITH in nonmetastatic tumors, with both high molecular specificity and real-time detection capability.
Collapse
Affiliation(s)
- Lei Li
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering and Department of Electrical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Deepanjali Patil
- Department of Chemistry & Biochemistry, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA
| | - Greg Petruncio
- Department of Chemistry & Biochemistry, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA
| | | | - Jisha V. Somasekharan
- Research and Post Graduate Department of Chemistry, MES Keveeyam College, Valanchery, Kerala 676552, India
| | - Mikell Paige
- Department of Chemistry & Biochemistry, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA
| | - Lihong V. Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering and Department of Electrical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Carolina Salvador-Morales
- Department of Chemistry & Biochemistry, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA
| |
Collapse
|
23
|
Davey MG, Ryan ÉJ, McAnena PF, Boland MR, Barry MK, Sweeney KJ, Malone CM, McLaughlin RJ, Lowery AJ, Kerin MJ. Disease recurrence and oncological outcome of patients treated surgically with curative intent for estrogen receptor positive, lymph node negative breast cancer. Surg Oncol 2021; 37:101531. [PMID: 33545657 DOI: 10.1016/j.suronc.2021.101531] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/10/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The molecular era has identified four breast cancer subtypes. Luminal A breast cancer (LABC) is defined by estrogen-receptor positive (ER+), progesterone-receptor positive (PgR+) and human epidermal growth factor receptor-2 negative (HER2-) tumours; these cancers are the most common and carry favourable prognoses. AIMS To describe clinicopathologic features, oncological outcome and relapse patterns in LABC. METHODS Consecutive female patients diagnosed with ER/PgR+/HER2-, lymph node negative (LN-) breast cancer between 2005 and 2015 were included. Clinicopathological and recurrence data was recorded using descriptive statistics. Oncological outcome was determined using Kaplan-Meier and Cox-regression analyses. RESULTS Analysis was performed for 849 patients with median follow-up of 102.1 months. Mean disease-free (DFS) and overall survival (OS) were 85.8% and 91.8%. Seventy patients died during this study (8.2%), while 58 patients had recurrence; 7 had local recurrence (0.8%) and 51 had distant recurrence (DDR) (6.0%). Patients developing DDR were likely to be postmenopausal (P = 0.028), present symptomatically (P < 0.001) and have larger tumours (P < 0.001). The mean time to DDR was 65.7 months, with fatal recurrence occurring in 66.6% of patients with DDR (34/51). Systemic chemotherapy prescription did not influence DDR (P = 0.053). Age >65 (hazards ratio (HR):1.66, 95% Confidence Interval (CI):1.07-2.55, P = 0.022), presenting symptomatically (HR:2.28, 95%CI:1.21-4.29, P = 0.011) and tumour size >20 mm (HR:1.81, 95%CI:1.25-2.62, P = 0.002) predicted DFS, while age>65 (HR:2.60, 95%CI:1.49-4.53, P = 0.001) and being postmenopausal at diagnosis (HR:3.13, 95%CI:1.19-8.22, P = 0.020) predicted OS. CONCLUSION Our series demonstrated excellent survival outcomes for patients diagnosed with LN- LABC after almost a decade of follow-up. However, following DDR, fatal progression is often imminent.
Collapse
Affiliation(s)
- M G Davey
- The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland; Department of Surgery, Galway University Hospitals, Galway, Ireland.
| | - É J Ryan
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - P F McAnena
- The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland; Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - M R Boland
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - M K Barry
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - K J Sweeney
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - C M Malone
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - R J McLaughlin
- Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - A J Lowery
- The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland; Department of Surgery, Galway University Hospitals, Galway, Ireland
| | - M J Kerin
- The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland; Department of Surgery, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
24
|
Clinical implications of intrinsic molecular subtypes of breast cancer for sentinel node status. Sci Rep 2021; 11:2259. [PMID: 33500440 PMCID: PMC7838175 DOI: 10.1038/s41598-021-81538-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 11/08/2022] Open
Abstract
Axillary lymph node status is an important prognostic factor for breast cancer patients and sentinel lymph node biopsy (SLNB) is a less invasive surgical proxy. We examined if consecutively derived molecular subtypes from primary breast cancers provide additional predictive value for SLNB status. 1556 patients with a breast cancer > 10 mm underwent primary surgical procedure including SLNB and tumor specimens were assigned with a transcriptomics-based molecular subtype. 1020 patients had a negative sentinel node (SN) and 536 a positive. A significant association between tumor size and SN status (p < 0.0001) was found across all samples, but no association between size and SN status (p = 0.14) was found for BasL tumors. A BasL subtype was a predictor of an SN-negative status (p = 0.001, OR 0.58, 95% CI 0.38;0.90) and among the BasL, postmenopausal status was a predictor for SN-negative status (p = 0.01). Overall survival was significantly lower (p = 0.02) in patients with BasL tumors and a positive SN. Interestingly, we identified a significant correlation between hormone receptor activity and SN status within the BasL subtype. Taken together, molecular subtypes and hormone receptor activity of breast cancers add predictive value for SLNB status.
Collapse
|
25
|
Chattu V, Umakanthan S, Bukelo M, Maharaj R, Khan N, Keane K, Khadoo N, Khan A, Khan A, Kong R, Korkmaz S, Kovoor A. Breast cancer in Trinidad and Tobago: Etiopathogenesis, histopathology and receptor study. J Family Med Prim Care 2021; 10:4438-4445. [PMID: 35280639 PMCID: PMC8884328 DOI: 10.4103/jfmpc.jfmpc_627_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/09/2022] Open
Abstract
Background: Breast Carcinoma (BCa) is the leading cause of cancer among females in Trinidad and Tobago (TnT). This twin-island has a diversified population of 1.3 million individuals that display and are exposed to a variety of lifestyle choices that have been linked to the development of BCa. Therefore, this study aimed to identify the risk factors that influence the development of BCa, analyze the common histopathological details, and categorize BCa based on receptor study. Methods: Cancer information for 120 BCa cases at Eric Williams Medical Sciences Complex from 2012 to 2019 was retrieved, analyzed, and statistically estimated. The clinical details were categorized based on data tabulations, and histological assessment was performed to identify specific features. The receptor analysis was classified based on estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor-2 (HER-2neu) staining intensity. A descriptive data analysis and comparison were statistically evaluated in all these cases. Results: Epidemiological factors influencing the development of BCa were age with a peak of 56–65 years 27.5% (n = 33), ethnicity predominated in Indo-Trinidadians 48.33% (n = 58), and marital status primarily in unmarried/single/widowed patients 55% (n = 66). Infiltrating ductal carcinoma was the principal histopathological type 91.66% (n = 110). Receptor analysis revealed ER/PR + HER-2neu as the most common type 40% (n = 18) for therapeutic surveillance. Conclusion: This study highlights various epidemiological factors that influence the development of BCa among females in TnT. Histopathological analysis and receptor studies would provide a useful link between the tumor behavior and its prognosis.
Collapse
|
26
|
Autenshlyus A, Davletova K, Varaksin N, Marinkin I, Lyakhovich V. Cytokines in various molecular subtypes of breast cancer. Int J Immunopathol Pharmacol 2021; 35:20587384211034089. [PMID: 34399595 PMCID: PMC8375341 DOI: 10.1177/20587384211034089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 06/28/2021] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION Breast cancer is a heterogeneous disease that has multiple molecular and morphological subtypes. Nonetheless, the relation between various molecular subtypes and functional characteristics of a tumor in terms of cytokine secretion remains unknown. METHODS We studied spontaneous and mitogen-induced cytokine secretion by invasive breast carcinoma of no special type (IBC NST; cultured tumors and cultured peripheral blood cells), depending on a molecular tumor subtype (where "mitogens" means "polyclonal activators" (PA): phytohemagglutinin p, phytohemagglutinin M, concanavalin A, and Escherichia coli lipopolysaccharide). Enzyme-linked immunosorbent assays were used to determine concentrations of IL-6, IL-8, IL-10, IL-17, IL-18, IL-1β, IL-1Ra, TNF-α, IFN-γ, G-CSF, GM-CSF, VEGF, and MCP-1 in culture supernatants of the tumors and peripheral blood cells. RESULTS The luminal B HER2-positive molecular subtype of IBC NST was found to feature the highest spontaneous secretion of IL-6 and IL-8 and the highest mitogen-induced secretion of IL-6, IL-8, IL-1Ra, and TNF-α by tumors; the highest mitogen-induced secretion of IL-2, IL-6, IL-8, IL-1β, TNF-α, IFN-γ, and G-CSF by peripheral blood cells; and the highest cytokine-producing potential (the ratio of mitogen-induced to spontaneous secretion) of peripheral blood cells for the secretion of IL-6, IL-8, and IL-1Ra as compared to other molecular subtypes. The triple-negative subtype of IBC NST was characterized by the lowest cytokine-producing potential of tumors for the secretion of IL-6 and IL-8 as compared to other molecular subtypes as well as a lower "stimulation index of polyclonal activators" (calculated as (cytokine secretion after incubation with PA)/(spontaneous cytokine secretion)) for IL-18 secretion as compared to luminal subtypes. The XYZ correlated with a suppressive effect of PA on cytokine secretion by tumors of the triple-negative molecular subtype. CONCLUSION Therefore, our findings indicate that in IBC NST of luminal B HER2-positive and triple-negative molecular subtypes, the cytokine network has distinctive functional features.
Collapse
Affiliation(s)
- Alexsander Autenshlyus
- Federal Publicly Funded Institution of Higher Education, Novosibirsk State Medical University, Novosibirsk, Russian Federation
- Federal Publicly Funded Scientific Institution Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Novosibirsk, Russian Federation
| | - Kristina Davletova
- Federal Publicly Funded Institution of Higher Education, Novosibirsk State Medical University, Novosibirsk, Russian Federation
- Federal Publicly Funded Scientific Institution Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Novosibirsk, Russian Federation
| | - Nikolay Varaksin
- AO Vector-Best, Koltsovo, Novosibirsk Oblast, Russian Federation
| | - Igor Marinkin
- Federal Publicly Funded Institution of Higher Education, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Vyacheslav Lyakhovich
- Federal Publicly Funded Scientific Institution Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Novosibirsk, Russian Federation
| |
Collapse
|
27
|
Szymiczek A, Lone A, Akbari MR. Molecular intrinsic versus clinical subtyping in breast cancer: A comprehensive review. Clin Genet 2020; 99:613-637. [PMID: 33340095 DOI: 10.1111/cge.13900] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022]
Abstract
Breast cancer is a heterogeneous disease manifesting diversity at the molecular, histological and clinical level. The development of breast cancer classification was centered on informing clinical decisions. The current approach to the classification of breast cancer, which categorizes this disease into clinical subtypes based on the detection of estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, and proliferation marker Ki67, is not ideal. This is manifested as a heterogeneity of therapeutic responses and outcomes within the clinical subtypes. The newer classification model, based on gene expression profiling (intrinsic subtyping) informs about transcriptional responses downstream from IHC single markers, revealing deeper appreciation for the disease heterogeneity and capturing tumor biology in a more comprehensive way than an expression of a single protein or gene alone. While accumulating evidences suggest that intrinsic subtypes provide clinically relevant information beyond clinical surrogates, it is imperative to establish whether the current conventional immunohistochemistry-based clinical subtyping approach could be improved by gene expression profiling and if this approach has a potential to translate into clinical practice.
Collapse
Affiliation(s)
- Agata Szymiczek
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Amna Lone
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad R Akbari
- Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|