1
|
Bakhtiar H, Sharifi MN, Helzer KT, Shi Y, Bootsma ML, Shang TA, Chrostek MR, Berg TJ, Carson Callahan S, Carreno V, Blitzer GC, West MT, O'Regan RM, Wisinski KB, Sjöström M, Zhao SG. A phenocopy signature of TP53 loss predicts response to chemotherapy. NPJ Precis Oncol 2024; 8:220. [PMID: 39358429 PMCID: PMC11447220 DOI: 10.1038/s41698-024-00722-7] [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: 04/02/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
In preclinical studies, p53 loss of function impacts chemotherapy response, but this has not been consistently validated clinically. We trained a TP53-loss phenocopy gene expression signature from pan-cancer clinical samples in the TCGA. In vitro, the TP53-loss phenocopy signature predicted chemotherapy response across cancer types. In a clinical dataset of 3003 breast cancer samples treated with neoadjuvant chemotherapy, the TP53-loss phenocopy samples were 56% more likely to have a pathologic complete response (pCR), with a significant association between TP53-loss phenocopy and pCR in both ER positive and ER negative tumors. In an independent clinical validation in the I-SPY2 trial (N = 987), we confirmed the association with neoadjuvant chemotherapy pCR and found higher rates of chemoimmunotherapy response in TP53-loss phenocopy tumors compared to non-TP53-loss phenocopy tumors (64% vs. 28%). The TP53-loss phenocopy signature predicts chemotherapy response across cancer types in vitro, and in a proof-of-concept clinical validation is associated with neoadjuvant chemotherapy response across multiple clinical breast cancer cohorts.
Collapse
Affiliation(s)
- Hamza Bakhtiar
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Marina N Sharifi
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Kyle T Helzer
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Yue Shi
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Matthew L Bootsma
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Tianfu A Shang
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | | | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - S Carson Callahan
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Viridiana Carreno
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
| | - Grace C Blitzer
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Malinda T West
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
| | - Ruth M O'Regan
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Kari B Wisinski
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Martin Sjöström
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA.
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
2
|
Huang M, Fasching PA, Haiderali A, Xue W, Pan W, Karantza V, Yang F, Truscott J, Xin Y, O'Shaughnessy J. Association between event-free survival and overall survival in early-stage triple-negative breast cancer. Future Oncol 2024; 20:335-348. [PMID: 37602372 DOI: 10.2217/fon-2023-0315] [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] [Indexed: 08/22/2023] Open
Abstract
Aim: This study evaluated event-free survival (EFS) as a surrogate outcome for overall survival (OS) in neoadjuvant therapy for early-stage triple-negative breast cancer (eTNBC). Methods: Meta-regression analyses based on a targeted literature review were used to evaluate the individual- and trial-level associations between EFS and OS. Results: In the individual-level analyses, 3-year EFS was a significant predictor of 5-year OS (p < 0.01; coefficient of determinations [R2]: 0.82 [95% CI: 0.68-0.91]). Additionally, there was a statistically significant association between the treatment effect on EFS and OS at the trial level (p < 0.001; R2: 0.64 [95% CI: 0.45-0.82]). Conclusion: This study demonstrates significant associations between EFS and OS and suggests that EFS is a valid surrogate for OS following neoadjuvant therapy for eTNBC.
Collapse
Affiliation(s)
| | - Peter A Fasching
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Department of Gynecology & Obstetrics, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology & US Oncology, Dallas, TX, USA
| |
Collapse
|
3
|
Aguilar-Mahecha A, Alirezaie N, Lafleur J, Bareke E, Przybytkowski E, Lan C, Cavallone L, Salem M, Pelmus M, Aleynikova O, Greenwood C, Lovato A, Ferrario C, Boileau JF, Mihalcioiu C, Roy JA, Marcus E, Discepola F, Majewski J, Basik M. The Mutational Spectrum of Pre- and Post-Neoadjuvant Chemotherapy Triple-Negative Breast Cancers. Genes (Basel) 2023; 15:27. [PMID: 38254917 PMCID: PMC10815241 DOI: 10.3390/genes15010027] [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: 11/04/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
The response of triple-negative breast cancer (TNBC) patients to pre-operative (neoadjuvant chemotherapy) is a critical factor of their outcome. To determine the effects of chemotherapy on the tumor genome and to identify mutations associated with chemoresistance and sensitivity, we performed whole exome sequencing on pre/post-chemotherapy tumors and matched lymphocytes from 26 patients. We observed great inter-tumoral heterogeneity with no gene mutated recurrently in more than four tumors besides TP53. Although the degree of response to chemotherapy in residual tumors was associated with more subclonal changes during chemotherapy, there was minimal evolution between pre/post-tumors. Indeed, gene sets enriched for mutations in pre- and post-chemotherapy tumors were very similar and reflected genes involved in the biological process of neurogenesis. Somatically mutated genes present in chemosensitive tumors included COL1A2, PRMD15, APOBEC3B, PALB2 and histone protein encoding genes, while BRCA1, ATR, ARID1A, XRCC3 and genes encoding for tubulin-associated proteins were present in the chemoresistant tumors. We also found that the mutational spectrum of post-chemotherapy tumors was more reflective of matching metastatic tumor biopsies than pre-chemotherapy samples. These findings support a portrait of modest ongoing genomic instability with respect to single-nucleotide variants induced by or selected for by chemotherapy in TNBCs.
Collapse
Affiliation(s)
- Adriana Aguilar-Mahecha
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Najmeh Alirezaie
- Department of Human Genetics, McGill University, Montreal, QC H3A 1A4, Canada; (N.A.); (J.M.)
| | - Josiane Lafleur
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Eric Bareke
- Department of Human Genetics, McGill University, Montreal, QC H3A 1A4, Canada; (N.A.); (J.M.)
| | - Ewa Przybytkowski
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Cathy Lan
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Luca Cavallone
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Myriam Salem
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Manuela Pelmus
- Department of Pathology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Olga Aleynikova
- Department of Pathology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Celia Greenwood
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (C.G.)
| | - Amanda Lovato
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (C.G.)
| | - Cristiano Ferrario
- Department of Oncology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | | | | | - Josée-Anne Roy
- Hôpital du Sacré-Cœur de Montréal, Montreal, QC H4J 1C5, Canada;
| | | | - Federico Discepola
- Department of Radiology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC H3A 1A4, Canada; (N.A.); (J.M.)
| | - Mark Basik
- Cancer Genomics and Translational Research Laboratory, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Department of Oncology, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- McGill University Health Center, Montreal, QC H3A 3J1, Canada
| |
Collapse
|
4
|
Wang C, Li K, An J, Lv X, Ma W, Wang Y, Meng N, Yun T, Zhao T. ASPP1/2 positive patients with invasive breast cancers have good prognosis. Heliyon 2023; 9:e20613. [PMID: 37886763 PMCID: PMC10597814 DOI: 10.1016/j.heliyon.2023.e20613] [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: 08/02/2022] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
Although the expression of ASPP family members in multiple tumors has been studied, especially in various cell lines of breast cancer (BC), but the expressions pattern of ASPP family members in invasive BC tissues are not clear. We studied the expression and expression pattern of ASPPs family member in BCs, the relationship between ASPP family members and clinic-pathologic features of BCs was also analyzed. The results showed that the expression of ASPP1, ASPP2 and iASPP was observed on AE1/AE3+ tumor cells, and not on infiltrated lymphocytes and capillaries. The relationship between ASPP1 expression and pTNM stage has statistical difference (p<0.01). The relationship between expression of ASPP2 and SBR grade has statistical difference (p<0.05). The relationship between expression of iASPP and clinic-pathologic feature of patients has no statistical difference (p>0.05). The patients with positive expression of ASPP1 and the patients with negative expression of ASPP1 have statistical difference in 3-year survival rate and 5-year survival rate (χ2 = 4.49, P = 0.03; χ2 = 3.79, P = 0.048). Overall, our work demonstrated that the expression of ASPP1/2 contributes to predict the prognosis of patients with BC.
Collapse
Affiliation(s)
- Changsong Wang
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Ke Li
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Junling An
- Department of Pathology and Pathophysiology, School of Basic Medicine, Henan University of Science and Technology, Luoyang, Henan, PR China
| | - Xuexia Lv
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Wenfeng Ma
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Yaxi Wang
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Nianlong Meng
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Tian Yun
- Department of Pathology, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| | - Ting Zhao
- Department of Colorectal Surgery, People's Liberation Army Joint Logistic Support Force 989th Hospital, Luoyang, Henan, PR China
| |
Collapse
|
5
|
Graff BT, Palanivel C, Jenkins CB, Baranowska-Kortylewicz J, Yan Y. Benzimidazole carbamate induces cytotoxicity in breast cancer cells via two distinct cell death mechanisms. Cell Death Discov 2023; 9:162. [PMID: 37179350 PMCID: PMC10183037 DOI: 10.1038/s41420-023-01454-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Metastatic breast cancer (mBC) is responsible for >90% of breast cancer-related deaths. Microtubule-targeting agents (MTAs) are the front-line treatment for mBC. However, the effectiveness of MTAs is frequently limited by the primary or acquired resistance. Furthermore, recurrent mBC derived from cancer cells that survived MTA treatment are typically more chemoresistant. The overall response rates for the second- and third-line MTAs in mBC patients previously treated with MTAs are 12-35%. Thus, there is an ongoing search for novel MTAs with a distinct mode of action that can circumvent chemoresistance mechanisms. Our results show that methyl N-(6-benzoyl-1H-benzimidazol-2-yl)carbamate (BCar), a microtubule-disrupting anthelmintic that binds to the colchicine binding site separate from the binding sites of clinically used MTAs, has the potential to treat MTA-resistant mBC. We have comprehensively evaluated the cellular effects of BCar in a panel of human breast cancer (BC) cell lines and normal breast cells. BCar effects on the clonogenic survival, cell cycle, apoptosis, autophagy, senescence, and mitotic catastrophe were measured. Approximately 25% of BCs harbor mutant p53. For this reason, the p53 status was included as a variable. The results show that BC cells are >10x more sensitive to BCar than normal mammary epithelial cells (HME). p53-mutant BC cells are significantly more sensitive to BCar treatment than p53 wild-type BC cells. Furthermore, BCar appears to kill BC cells primarily via either p53-dependent apoptosis or p53-independent mitotic catastrophe. When compared to docetaxel and vincristine, two clinical MTAs, BCar is fairly innocuous in HME cells, providing a much wider therapeutic window than docetaxel and vincristine. Together, the results strongly support the notion that BCar-based therapeutics may serve as a new line of MTAs for mBC treatment.
Collapse
Affiliation(s)
- Brendan T Graff
- Department of Radiation Oncology, College of Medicine University of Nebraska Medical Center Omaha, Nebraska, USA
| | - Chitra Palanivel
- Department of Radiation Oncology, College of Medicine University of Nebraska Medical Center Omaha, Nebraska, USA
| | - Christopher B Jenkins
- Department of Radiation Oncology, College of Medicine University of Nebraska Medical Center Omaha, Nebraska, USA
| | - Janina Baranowska-Kortylewicz
- Department of Pharmaceutical Sciences, College of Pharmacy University of Nebraska Medical Center Omaha, Nebraska, USA.
| | - Ying Yan
- Department of Radiation Oncology, College of Medicine University of Nebraska Medical Center Omaha, Nebraska, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine University of Nebraska Medical Center Omaha, Nebraska, USA.
| |
Collapse
|
6
|
Park JH, Kwon MJ, Seo J, Kim HY, Min SK, Kim LS. Somatic Mutations of TP53 Identified by Targeted Next-Generation Sequencing Are Poor Prognostic Factors for Primary Operable Breast Cancer: A Single-Center Study. J Breast Cancer 2022; 25:379-386. [PMID: 36265889 PMCID: PMC9629967 DOI: 10.4048/jbc.2022.25.e41] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/12/2022] [Accepted: 09/18/2022] [Indexed: 11/28/2022] Open
Abstract
Few studies have reported on the clinical utility of targeted next-generation sequencing (NGS) for breast cancer in Korea. We retrospectively reviewed the targeted NGS data of 219 patients with breast cancer who underwent surgical resection between August 2018 and April 2021. Here, we described the mutational profiles of breast cancer and examined their prognostic implications. The most frequently mutated gene was PIK3CA (n = 97/219, 44.3%), followed by TP53 (n = 79/219, 36.1%), AKT1 (n = 23/219, 10.5%), and GATA3 (n = 20/219, 9.1%). TP53 mutations were associated with aggressive histologic features. We followed up for 31 (range, 1-39) months and observed 11 (5.0%) recurrences: nine were TP53 mutant and two were TP53 wild-type. Multivariable analysis revealed that TP53 mutation was an independent prognostic factor for recurrence (p = 0.012). Although no drug is currently available for TP53 mutations, it is valuable to know the mutational status of TP53 for the precise management of breast cancer.
Collapse
Affiliation(s)
- Jung Ho Park
- Division of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Jinwon Seo
- Department of Pathology, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Ho Young Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Soo Kee Min
- Department of Pathology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea
| | - Lee Su Kim
- Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea
| |
Collapse
|
7
|
Clonal evolution in primary breast cancers under sequential epirubicin and docetaxel monotherapy. Genome Med 2022; 14:86. [PMID: 35948919 PMCID: PMC9367103 DOI: 10.1186/s13073-022-01090-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 07/13/2022] [Indexed: 11/30/2022] Open
Abstract
Background Subclonal evolution during primary breast cancer treatment is largely unexplored. We aimed to assess the dynamic changes in subclonal composition of treatment-naïve breast cancers during neoadjuvant chemotherapy. Methods We performed whole exome sequencing of tumor biopsies collected before, at therapy switch, and after treatment with sequential epirubicin and docetaxel monotherapy in 51 out of 109 patients with primary breast cancer, who were included in a prospectively registered, neoadjuvant single-arm phase II trial. Results There was a profound and differential redistribution of subclones during epirubicin and docetaxel treatment, regardless of therapy response. While truncal mutations and main subclones persisted, smaller subclones frequently appeared or disappeared. Reassessment of raw data, beyond formal mutation calling, indicated that the majority of subclones seemingly appearing during treatment were in fact present in pretreatment breast cancers, below conventional detection limits. Likewise, subclones which seemingly disappeared were still present, below detection limits, in most cases where tumor tissue remained. Tumor mutational burden (TMB) dropped during neoadjuvant therapy, and copy number analysis demonstrated specific genomic regions to be systematically lost or gained for each of the two chemotherapeutics. Conclusions Sequential epirubicin and docetaxel monotherapy caused profound redistribution of smaller subclones in primary breast cancer, while early truncal mutations and major subclones generally persisted through treatment. Trial registration ClinicalTrials.gov, NCT00496795, registered on July 4, 2007. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01090-2.
Collapse
|
8
|
Integrative, In Silico and Comparative Analysis of Breast Cancer Secretome Highlights Invasive-Ductal-Carcinoma-Grade Progression Biomarkers. Cancers (Basel) 2022; 14:cancers14163854. [PMID: 36010848 PMCID: PMC9406168 DOI: 10.3390/cancers14163854] [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: 07/06/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Globally, BC is the most frequently diagnosed cancer in women. The aim of this study was to identify novel secreted biomarkers that may indicate progression to high-grade BC malignancies and therefore predict metastatic potential. A total of 33 studies of breast cancer and 78 of other malignancies were screened via a systematic review for eligibility, yielding 26 datasets, 8 breast cancer secretome datasets, and 18 of other cancers that were included in the comparative secretome analysis. Sequential bioinformatic analysis using online resources enabled the identification of enriched GO_terms, overlapping clusters, and pathway reconstruction. This study identified putative predictors of IDC grade progression and their association with breast cancer patient mortality outcomes, namely, HSPG2, ACTG1, and LAMA5 as biomarkers of in silico pathway prediction, offering a putative approach by which the abovementioned proteins may mediate their effects, enabling disease progression. This study also identified ITGB1, FBN1, and THBS1 as putative pan-cancer detection biomarkers. The present study highlights novel, putative secretome biomarkers that may provide insight into the tumor biology and could inform clinical decision making in the context of IDC management in a non-invasive manner.
Collapse
|
9
|
Singhal SK, Byun JS, Yan T, Yancey R, Caban A, Gil Hernandez S, Bufford S, Hewitt SM, Winfield J, Pradhan JS, Mustkov V, McDonald JA, Pérez-Stable EJ, Napoles AM, Vohra N, De Siervi A, Yates C, Davis MB, Yang M, Tsai YC, Weissman AM, Gardner K. Protein expression of the gp78 E3-ligase predicts poor breast cancer outcome based on race. JCI Insight 2022; 7:157465. [PMID: 35639484 PMCID: PMC9310521 DOI: 10.1172/jci.insight.157465] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Women of African ancestry suffer higher rates of breast cancer mortality compared to all other groups in the United States. Though the precise reasons for these disparities remain unclear, many recent studies have implicated a role for differences in tumor biology. Using an epitope-validated antibody against the endoplasmic reticulum-associated degradation (ERAD) E3 ubiquitin ligase, gp78, we show that elevated levels of gp78 in patient breast cancer cells predict poor survival. Moreover, high levels of gp78 are associated with poor outcomes in both ER-positive and ER-negative tumors, and breast cancers expressing elevated amounts of gp78 protein are enriched in gene expression pathways that influence cell cycle, metabolism, receptor-mediated signaling, and cell stress response pathways. In multivariate analysis adjusted for subtype and grade, gp78 protein is an independent predictor of poor outcomes in women of African ancestry. Furthermore, gene expression signatures, derived from patients stratified by gp78 protein expression, are strong predictors of recurrence and pathological complete response in retrospective clinical trial data and share many common features with gene sets previously identified to be overrepresented in breast cancers based on race. These findings implicate a prominent role for gp78 in tumor progression and offer new insights into our understanding of racial differences in breast cancer outcomes.
Collapse
Affiliation(s)
- Sandeep K Singhal
- Department of Pathology, University of North Dakota, Grand Forks, United States of America
| | - Jung S Byun
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Tingfen Yan
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Ryan Yancey
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Ambar Caban
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Sara Gil Hernandez
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Sediqua Bufford
- Masters of Science Biotechnology, Morehouse School of Medicine, Atlanta, United States of America
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, United States of America
| | - Joy Winfield
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Jaya Sarin Pradhan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Vesco Mustkov
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Jasmine A McDonald
- Department of Epidemiology, Columbia University Medical Center, New York, United States of America
| | - Eliseo J Pérez-Stable
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Anna Maria Napoles
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Nasreen Vohra
- Brody School of Medicine, East Carolina University, Greenville, United States of America
| | - Adriana De Siervi
- Directora del Laboratorio de Oncología Molecular y Nuevos Blancos Terapéut, CONICET, Buenos Aiers, Argentina
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, United States of America
| | - Melissa B Davis
- Department of Surgery (Breast Surgery & Oncology), Weill Cornell Medicine, New York, United States of America
| | - Mei Yang
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Yien Che Tsai
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Allan M Weissman
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Kevin Gardner
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| |
Collapse
|
10
|
Sammut SJ, Crispin-Ortuzar M, Chin SF, Provenzano E, Bardwell HA, Ma W, Cope W, Dariush A, Dawson SJ, Abraham JE, Dunn J, Hiller L, Thomas J, Cameron DA, Bartlett JMS, Hayward L, Pharoah PD, Markowetz F, Rueda OM, Earl HM, Caldas C. Multi-omic machine learning predictor of breast cancer therapy response. Nature 2022; 601:623-629. [PMID: 34875674 PMCID: PMC8791834 DOI: 10.1038/s41586-021-04278-5] [Citation(s) in RCA: 209] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/23/2021] [Indexed: 11/09/2022]
Abstract
Breast cancers are complex ecosystems of malignant cells and the tumour microenvironment1. The composition of these tumour ecosystems and interactions within them contribute to responses to cytotoxic therapy2. Efforts to build response predictors have not incorporated this knowledge. We collected clinical, digital pathology, genomic and transcriptomic profiles of pre-treatment biopsies of breast tumours from 168 patients treated with chemotherapy with or without HER2 (encoded by ERBB2)-targeted therapy before surgery. Pathology end points (complete response or residual disease) at surgery3 were then correlated with multi-omic features in these diagnostic biopsies. Here we show that response to treatment is modulated by the pre-treated tumour ecosystem, and its multi-omics landscape can be integrated in predictive models using machine learning. The degree of residual disease following therapy is monotonically associated with pre-therapy features, including tumour mutational and copy number landscapes, tumour proliferation, immune infiltration and T cell dysfunction and exclusion. Combining these features into a multi-omic machine learning model predicted a pathological complete response in an external validation cohort (75 patients) with an area under the curve of 0.87. In conclusion, response to therapy is determined by the baseline characteristics of the totality of the tumour ecosystem captured through data integration and machine learning. This approach could be used to develop predictors for other cancers.
Collapse
Affiliation(s)
- Stephen-John Sammut
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mireia Crispin-Ortuzar
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Elena Provenzano
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Helen A Bardwell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Wenxin Ma
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Wei Cope
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Ali Dariush
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Institute of Astronomy, University of Cambridge, Cambridge, UK
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Centre of Cancer Research and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Jean E Abraham
- Department of Oncology, University of Cambridge, Cambridge, UK
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Janet Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Jeremy Thomas
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
- Q2 Laboratory Solutions, Livingston, UK
| | - David A Cameron
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
| | - John M S Bartlett
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Larry Hayward
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
| | - Paul D Pharoah
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Oscar M Rueda
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Helena M Earl
- Department of Oncology, University of Cambridge, Cambridge, UK
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK.
- Department of Oncology, University of Cambridge, Cambridge, UK.
- CRUK Cambridge Centre, Cambridge Experimental Cancer Medicine Centre (ECMC) and NIHR Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
11
|
Conforti F, Pala L, Sala I, Oriecuia C, De Pas T, Specchia C, Graffeo R, Pagan E, Queirolo P, Pennacchioli E, Colleoni M, Viale G, Bagnardi V, Gelber RD. Evaluation of pathological complete response as surrogate endpoint in neoadjuvant randomised clinical trials of early stage breast cancer: systematic review and meta-analysis. BMJ 2021; 375:e066381. [PMID: 34933868 PMCID: PMC8689398 DOI: 10.1136/bmj-2021-066381] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To evaluate pathological complete response as a surrogate endpoint for disease-free survival and overall survival in regulatory neoadjuvant trials of early stage breast cancer. DESIGN Systematic review and meta-analysis. DATA SOURCES Medline, Embase, and Scopus to 1 December 2020. ELIGIBILITY CRITERIA FOR STUDY SELECTION Randomised clinical trials that tested neoadjuvant chemotherapy given alone or combined with other treatments, including anti-human epidermal growth factor 2 (anti-HER2) drugs, targeted treatments, antivascular agents, bisphosphonates, and immune checkpoint inhibitors. DATA EXTRACTION AND SYNTHESIS Trial level associations between the surrogate endpoint pathological complete response and disease-free survival and overall survival. METHODS A weighted regression analysis was performed on log transformed treatment effect estimates (hazard ratio for disease-free survival and overall survival and relative risk for pathological complete response), and the coefficient of determination (R2) was used to quantify the association. The secondary objective was to explore heterogeneity of results in preplanned subgroups analysis, stratifying trials according treatment type in the experimental arm, definition used for pathological complete response (breast and lymph nodes v breast only), and biological features of the disease (HER2 positive or triple negative breast cancer). The surrogate threshold effect was also evaluated, indicating the minimum value of the relative risk for pathological complete response necessary to confidently predict a non-null effect on hazard ratio for disease-free survival or overall survival. RESULTS 54 randomised clinical trials comprising a total of 32 611 patients were included in the analysis. A weak association was observed between the log(relative risk) for pathological complete response and log(hazard ratio) for both disease-free survival (R2=0.14, 95% confidence interval 0.00 to 0.29) and overall survival (R2 =0.08, 0.00 to 0.22). Similar results were found across all subgroups evaluated, independently of the definition used for pathological complete response, treatment type in the experimental arm, and biological features of the disease. The surrogate threshold effect was 5.19 for disease-free survival but was not estimable for overall survival. Consistent results were confirmed in three sensitivity analyses: excluding small trials (<200 patients enrolled), excluding trials with short median follow-up (<24 months), and replacing the relative risk for pathological complete response with the absolute difference of pathological complete response rates between treatment arms. CONCLUSION A lack of surrogacy of pathological complete response was identified at trial level for both disease-free survival and overall survival. The findings suggest that pathological complete response should not be used as primary endpoint in regulatory neoadjuvant trials of early stage breast cancer.
Collapse
Affiliation(s)
- Fabio Conforti
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Laura Pala
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Isabella Sala
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Chiara Oriecuia
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Tommaso De Pas
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rossella Graffeo
- Breast Unit of Southern Switzerland, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Paola Queirolo
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Elisabetta Pennacchioli
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Viale
- Department of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Richard D Gelber
- Medical School, Harvard T H Chan School of Public Health, and Frontier Science and Technology Research Foundation, Boston, MA, USA
| |
Collapse
|
12
|
Patterns of genomic change in residual disease after neoadjuvant chemotherapy for estrogen receptor-positive and HER2-negative breast cancer. Br J Cancer 2021; 125:1356-1364. [PMID: 34480095 PMCID: PMC8575785 DOI: 10.1038/s41416-021-01526-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 07/04/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Treatment of patients with residual disease after neoadjuvant chemotherapy for breast cancer is an unmet clinical need. We hypothesised that tumour subclones showing expansion in residual disease after chemotherapy would contain mutations conferring drug resistance. METHODS We studied oestrogen receptor and/or progesterone receptor-positive, HER2-negative tumours from 42 patients in the EORTC 10994/BIG 00-01 trial who failed to achieve a pathological complete response. Genes commonly mutated in breast cancer were sequenced in pre and post-treatment samples. RESULTS Oncogenic driver mutations were commonest in PIK3CA (38% of tumours), GATA3 (29%), CDH1 (17%), TP53 (17%) and CBFB (12%); and amplification was commonest for CCND1 (26% of tumours) and FGFR1 (26%). The variant allele fraction frequently changed after treatment, indicating that subclones had expanded and contracted, but there were changes in both directions for all of the commonly mutated genes. CONCLUSIONS We found no evidence that expansion of clones containing recurrent oncogenic driver mutations is responsible for resistance to neoadjuvant chemotherapy. The persistence of classic oncogenic mutations in pathways for which targeted therapies are now available highlights their importance as drug targets in patients who have failed chemotherapy but provides no support for a direct role of driver oncogenes in resistance to chemotherapy. CLINICALTRIALS.GOV: EORTC 10994/BIG 1-00 Trial registration number NCT00017095.
Collapse
|
13
|
Tian JH, Liu SH, Yu CY, Wu LG, Wang LB. The Role of Non-Coding RNAs in Breast Cancer Drug Resistance. Front Oncol 2021; 11:702082. [PMID: 34589423 PMCID: PMC8473733 DOI: 10.3389/fonc.2021.702082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is one of the commonly occurring malignancies in females worldwide. Despite significant advances in therapeutics, the mortality and morbidity of BC still lead to low survival and poor prognosis due to the drug resistance. There are certain chemotherapeutic, endocrine, and target medicines often used for BC patients, including anthracyclines, taxanes, docetaxel, cisplatin, and fluorouracil. The drug resistance mechanisms of these medicines are complicated and have not been fully elucidated. It was reported that non-coding RNAs (ncRNAs), such as micro RNAs (miRNA), long-chain non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) performed key roles in regulating tumor development and mediating therapy resistance. However, the mechanism of these ncRNAs in BC chemotherapeutic, endocrine, and targeted drug resistance was different. This review aims to reveal the mechanism and potential functions of ncRNAs in BC drug resistance and to highlight the ncRNAs as a novel target for achieving improved treatment outcomes for BC patients.
Collapse
Affiliation(s)
- Jin-Hai Tian
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Shi-Hai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuan-Yang Yu
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Li-Gang Wu
- Department of Oncology, General Hospital of Ningxia Medical University, Yingchuan, China
| | - Li-Bin Wang
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
14
|
Bai H, Yu J, Jia S, Liu X, Liang X, Li H. Prognostic Value of the TP53 Mutation Location in Metastatic Breast Cancer as Detected by Next-Generation Sequencing. Cancer Manag Res 2021; 13:3303-3316. [PMID: 33889023 PMCID: PMC8057094 DOI: 10.2147/cmar.s298729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/19/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose The status of TP53 mutations was measured in cell-free DNA from patients with metastatic breast cancer (MBC) to investigate disease characteristics and the prognostic role of different locations of the TP53 mutation site. Patients and Methods Blood samples were taken from a total of 187 patients diagnosed with MBC who were treated at the Department of Breast Oncology, Peking University Cancer Hospital between January 2013 and March 2020. Next-generation sequencing was used to investigate the TP53 mutation spectra of circulating free DNA in these blood samples. Results Among the 187 MBC patients, TP53-mutated patients had a significantly shorter median disease-free survival (DFS) and overall survival (OS) compared with TP53 wild-type patients (P=0.001 and P=0.006, respectively). Additionally, in hormone receptor positive/HER2 negative (HR+/HER2-) and triple negative (TNBC) cohorts, TP53-mutated patients had a significantly shorter median DFS than TP53 wild-type patients (P=0.038 and P=0.023, respectively). The 79 patients with TP53 mutations carried 87 somatic TP53 mutations, of which most (77.0%) mapped to the DNA-binding domain (DBD) of the protein encoded by TP53 exons 5–8. In patients with TP53 mutations, those occurring in the non-DBD had a significantly shorter median DFS and OS than TP53 wild type (P<0.001 and P=0.001, respectively). Additionally, patients with non-missense mutations in the DBD had a significantly shorter median DFS and OS than TP53 wild-type patients (P=0.001 and P<0.001, respectively). TP53-mutated patients had a significantly shorter DFS than TP53 wild-type patients in the adjuvant endocrine therapy sensitive group (P=0.008), but differences in the endocrine therapy resistant group were not significant. Conclusion TP53-mutated MBC patients had a significantly worse outcome than TP53 wild-type patients especially those in HR+/HER2– and TNBC cohorts. Of TP53-mutated patients, those with non-missense mutations in the DBD had worse breast cancer-related survival. TP53 mutations were also associated with endocrine resistance.
Collapse
Affiliation(s)
- Han Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, People's Republic of China
| | - Jianjun Yu
- Huidu Shanghai Medical Sciences, Shanghai, 201499, People's Republic of China
| | - Shidong Jia
- Huidu Shanghai Medical Sciences, Shanghai, 201499, People's Republic of China
| | - Xiaoran Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, People's Republic of China
| | - Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, People's Republic of China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, People's Republic of China
| |
Collapse
|
15
|
Yu KD, Ge JY, Liu XY, Mo M, He M, Shao ZM. Cyclophosphamide-free Adjuvant Chemotherapy for Ovarian Protection in Young Women with Breast Cancer: a Randomized Phase 3 Trial. J Natl Cancer Inst 2021; 113:1352-1359. [PMID: 33822134 PMCID: PMC8486325 DOI: 10.1093/jnci/djab065] [Citation(s) in RCA: 3] [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: 12/17/2020] [Revised: 01/08/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chemotherapy-induced premature menopause leads to some consequences, including infertility. We initiated this randomized phase 3 trial to determine whether a cyclophosphamide-free adjuvant chemotherapy regimen would increase the likelihood of menses resumption and improve survival outcomes. METHODS Young women with operable ER-positive HER2-negative breast cancer after definitive surgery were randomized to receive adjuvant epirubicin/cyclophosphamide followed by weekly paclitaxel (EC-wP) or epirubicin/paclitaxel followed by weekly paclitaxel (EP-wP). All patients received at least 5-year adjuvant endocrine therapy after chemotherapy. Two coprimary endpoints were the rate of menstrual resumption at 12 months after chemotherapy and 5-year disease-free survival (DFS) in the intention-to-treat population. This study is registered at ClinicalTrials.gov (NCT01026116). All statistical tests were 2-sided. RESULTS Between Jan 2011 and Dec 2016, 521 patients (median age = 34 years; interquartile range = 31-38 years) were enrolled, with 261 in the EC-wP group and 260 in the EP-wP group. The rate of menstrual resumption at 12 months after chemotherapy was 48.3% in EC-wP (95% confidence interval [CI] = 42.2% to 54.3%) and 63.1% in EP-wP (95% CI = 57.2% to 68.9%), with an absolute difference of 14.8% (95% CI = 6.37% to 23.2%, P < 0.001). The post-hoc exploratory analysis by patient-reported outcome questionnaires indicated that pregnancy might occur in fewer women in the EC-wP group than in the EP-wP group. At a median follow-up of 62 months, the 5-year DFS was 78.3% (95% CI = 72.2% to 83.3%) in EC-wP and 84.7% (95% CI = 79.3% to 88.8%) in EP-wP (stratified log-rank P = 0.07). The safety data were consistent with the known safety profiles of relevant drugs. CONCLUSIONS The cyclophosphamide-free chemotherapy regimen might be associated with a higher probability of menses resumption.
Collapse
Affiliation(s)
- Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Correspondence to: Zhi-Ming Shao, MD, Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Rd, Shanghai 200032, China (e-mail: ) and Ke-Da Yu, MD, PhD, Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Rd, Shanghai 200032, China (e-mail: )
| | - Jing-Yu Ge
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi-Yu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Miao Mo
- Department of Cancer Prevention and Clinical Statistics Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Min He
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai, China
| | | |
Collapse
|
16
|
O’Leary B, Cutts RJ, Huang X, Hrebien S, Liu Y, André F, Loibl S, Loi S, Garcia-Murillas I, Cristofanilli M, Bartlett CH, Turner NC. Circulating Tumor DNA Markers for Early Progression on Fulvestrant With or Without Palbociclib in ER+ Advanced Breast Cancer. J Natl Cancer Inst 2021; 113:309-317. [PMID: 32940689 PMCID: PMC7936069 DOI: 10.1093/jnci/djaa087] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/25/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND There are no established molecular biomarkers for patients with breast cancer receiving combination endocrine and CDK4/6 inhibitor (CDK4/6i). We aimed to determine whether genomic markers in circulating tumor DNA (ctDNA) can identify patients at higher risk of early progression on fulvestrant therapy with or without palbociclib, a CDK4/6i. METHODS PALOMA-3 was a phase III, multicenter, double-blind randomized controlled trial of palbociclib plus fulvestrant (n = 347) vs placebo plus fulvestrant (n = 174) in patients with endocrine-pretreated estrogen receptor-positive (ER+) breast cancer. Pretreatment plasma samples from 459 patients were analyzed for mutations in 17 genes, copy number in 14 genes, and circulating tumor fraction. Progression-free survival (PFS) was compared in patients with circulating tumor fraction above or below a prespecified cutoff of 10% and with or without a specific genomic alteration. All statistical tests were 2-sided. RESULTS Patients with high ctDNA fraction had worse PFS on both palbociclib plus fulvestrant (hazard ratio [HR] = 1.62, 95% confidence interval [CI] = 1.17 to 2.24; P = .004) and placebo plus fulvestrant (HR = 1.77, 95% CI = 1.21 to 2.59; P = .004). In multivariable analysis, high-circulating tumor fraction was associated with worse PFS (HR = 1.20 per 10% increase in tumor fraction, 95% CI = 1.09 to 1.32; P < .001), as was TP53 mutation (HR = 1.84, 95% CI = 1.27 to 2.65; P = .001) and FGFR1 amplification (HR = 2.91, 95% CI = 1.61 to 5.25; P < .001). No interaction with treatment randomization was observed. CONCLUSIONS Pretreatment ctDNA identified a group of high-risk patients with poor clinical outcome despite the addition of CDK4/6 inhibition. These patients might benefit from inclusion in future trials of escalating treatment, with therapies that may be active in these genomic contexts.
Collapse
Affiliation(s)
- Ben O’Leary
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, Royal Marsden Hospital, London, UK
| | - Rosalind J Cutts
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Sarah Hrebien
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Fabrice André
- Department of Medical Oncology, Institut Gustave Roussy, Université Paris Sud, Villejuif, France
| | - Sibylle Loibl
- German Breast Group, Martin Behaim-Strasse 12, Neu-Isenburg, Germany
| | - Sherene Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Australia
| | - Isaac Garcia-Murillas
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Massimo Cristofanilli
- Robert H Lurie Comprehensive Cancer Centre, Feinberg School of Medicine, Chicago, IL, USA
| | | | - Nicholas C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, Royal Marsden Hospital, London, UK
| |
Collapse
|
17
|
Shahdoust M, Mahjub H, Pezeshk H, Sadeghi M. A Network-Based Comparison Between Molecular Apocrine Breast Cancer Tumor and Basal and Luminal Tumors by Joint Graphical Lasso. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1555-1562. [PMID: 30990436 DOI: 10.1109/tcbb.2019.2911074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Joint graphical lasso (JGL) approach is a Gaussian graphical model to estimate multiple graphical models corresponding to distinct but related groups. Molecular apocrine (MA) breast cancer tumor has similar characteristics to luminal and basal subtypes. Due to the relationship between MA tumor and two other subtypes, this paper investigates the similarities and differences between the MA genes association network and the ones corresponding to other tumors by taking advantageous of JGL properties. Two distinct JGL graphical models are applied to two sub-datasets including the gene expression information of the MA and the luminal tumors and also the MA and the basal tumors. Then, topological comparisons between the networks such as finding the shared edges are applied. In addition, several support vector machine (SVM) classification models are performed to assess the discriminating power of some critical nodes in the networks, like hub nodes, to discriminate the tumors sample. Applying the JGL approach prepares an appropriate tool to observe the networks of the MA tumor and other subtypes in one map. The results obtained by comparing the networks could be helpful to generate new insight about MA tumor for future studies.
Collapse
|
18
|
Pathak M, Deo SVS, Dwivedi SN, Thakur B, Sreenivas V, Rath GK. Regimens of neo-adjuvant chemotherapy in the treatment of breast cancer: A systematic review & network meta-analysis with PRISMA-NMA compliance. Crit Rev Oncol Hematol 2020; 153:103015. [DOI: 10.1016/j.critrevonc.2020.103015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 01/16/2023] Open
|
19
|
Li K, Liao N, Chen B, Zhang G, Wang Y, Guo L, Wei G, Jia M, Wen L, Ren C, Cao L, Mok H, Li C, Lin J, Chen X, Zhang Z, Hou T, Li M, Liu J, Balch CM, Liao N. Genetic mutation profile of Chinese HER2-positive breast cancers and genetic predictors of responses to Neoadjuvant anti-HER2 therapy. Breast Cancer Res Treat 2020; 183:321-332. [PMID: 32638235 PMCID: PMC7383038 DOI: 10.1007/s10549-020-05778-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Despite the therapeutic success of existing HER2-targeted therapies, tumors respond quite differently to them. This study aimed at figuring out genetic mutation profile of Chinese HER2-positive patients and investigating predictive factors of neoadjuvant anti-HER2 responses. METHODS We employed two cohorts. The first cohort was comprised of 181 HER2-positive patients treated at Guangdong Provincial People's Hospital from 2012 to 2018. The second cohort included 40 patients from the first cohort who underwent HER2-targeted neoadjuvant chemotherapy. Genetic mutations were characterized using next-generation sequencing. We employed the most commonly used definition of pathological complete response (pCR)-eradication of tumor from both breast and lymph nodes (ypT0/is ypN0). RESULTS In Chinese HER2-positive breast cancer patients, TP53 (74.6%), CDK12 (64.6%) and PIK3CA (46.4%) have the highest mutation frequencies. In cohort 2, significant differences were found between pCR and non-pCR groups in terms of the initial Ki67 status, TP53 missense mutations, TP53 LOF mutations, PIK3CA mutations and ROS1 mutations (p = 0.028, 0.019, 0.005, 0.013, 0.049, respectively). Furthermore, TP53 LOF mutations and initial Ki67 status (OR 7.086, 95% CI 1.366-36.749, p = 0.020 and OR 6.007, 95% CI 1.120-32.210, p = 0.036, respectively) were found to be predictive of pCR status. CONCLUSION TP53 LOF mutations and initial Ki67 status in HER2-positive breast cancer are predictive of pCR status after HER2-targeted NACT.
Collapse
Affiliation(s)
- Kai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Bo Chen
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Guochun Zhang
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yulei Wang
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Liping Guo
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guangnan Wei
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Minghan Jia
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Lingzhu Wen
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Chongyang Ren
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Li Cao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Hsiaopei Mok
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Cheukfai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Jiali Lin
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoqing Chen
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | | | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | - Min Li
- Burning Rock Biotech, Guangzhou, China
| | - Jing Liu
- Burning Rock Biotech, Guangzhou, China
| | - Charles M Balch
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Huang M, O’Shaughnessy J, Zhao J, Haiderali A, Cortes J, Ramsey S, Briggs A, Karantza V, Aktan G, Qi CZ, Gu C, Xie J, Yuan M, Cook J, Untch M, Schmid P, Fasching PA. Evaluation of Pathologic Complete Response as a Surrogate for Long-Term Survival Outcomes in Triple-Negative Breast Cancer. J Natl Compr Canc Netw 2020; 18:1096-1104. [DOI: 10.6004/jnccn.2020.7550] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 11/17/2022]
Abstract
Background: Pathologic complete response (pCR) is a common efficacy endpoint in neoadjuvant therapy trials for triple-negative breast cancer (TNBC). Previous studies have shown that pCR is strongly associated with improved long-term survival outcomes, including event-free survival (EFS) and overall survival (OS). However, the trial-level associations between treatment effect on pCR and long-term survival outcomes are not well established. This study sought to evaluate these associations by incorporating more recent clinical trials in TNBC. Methods: A literature review identified published randomized controlled trials (RCTs) of neoadjuvant therapy for TNBC that reported results for both pCR and EFS/OS. Meta-regression models were performed to evaluate the association of treatment effect on pCR and EFS/OS. Sensitivity analyses were conducted to assess the impact of divergent study designs. Results: Ten comparisons from 8 RCTs (N=2,478 patients) were identified from the literature review. The log (odds ratio) of pCR was a significant predictor of the log (hazard ratio) of EFS (P=.003), with a coefficient of determination of 0.68 (95% CI, 0.41–0.95). There was a weaker association between pCR and OS (P=.18), with a coefficient of determination of 0.24 (95% CI, 0.01–0.77). Consistent results were found in the exploratory analysis and sensitivity analyses. Conclusions: This is the first study that has shown a trial-level association between pCR and survival outcomes in TNBC. By incorporating the most up-to-date RCTs, this study showed a significant trial-level association between pCR and EFS. A positive association between pCR and OS was also recorded.
Collapse
Affiliation(s)
- Min Huang
- 1Merck & Co., Inc., Kenilworth, New Jersey
| | - Joyce O’Shaughnessy
- 2Baylor University Medical Center, Texas Oncology, and U.S. Oncology, Dallas, Texas
| | - Jing Zhao
- 1Merck & Co., Inc., Kenilworth, New Jersey
| | | | - Javier Cortes
- 3IOB Institute of Oncology, Quironsalud Group, Madrid and Barcelona, Spain
- 4Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | - Scott Ramsey
- 5Fred Hutchinson Cancer Research Center, and University of Washington, Seattle, Washington
| | - Andrew Briggs
- 6London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | | | - Chenyang Gu
- 8Analysis Group, Inc., Los Angeles, California
| | - Jipan Xie
- 8Analysis Group, Inc., Los Angeles, California
| | - Muhan Yuan
- 7Analysis Group, Inc., Boston, Massachusetts
| | - John Cook
- 9Complete HEOR Solutions, North Wales, Pennsylvania
| | - Michael Untch
- 10Department of Gynecology, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Peter Schmid
- 11Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Peter A. Fasching
- 12Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
21
|
Spring LM, Fell G, Arfe A, Sharma C, Greenup R, Reynolds KL, Smith BL, Alexander B, Moy B, Isakoff SJ, Parmigiani G, Trippa L, Bardia A. Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis. Clin Cancer Res 2020; 26:2838-2848. [PMID: 32046998 PMCID: PMC7299787 DOI: 10.1158/1078-0432.ccr-19-3492] [Citation(s) in RCA: 418] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/16/2019] [Accepted: 02/04/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE While various studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy (NAT), the impact of additional adjuvant therapy after pCR is not known. EXPERIMENTAL DESIGN PubMed was searched for studies with NAT for breast cancer and individual patient-level data was extracted for analysis using plot digitizer software. HRs, with 95% probability intervals (PI), measuring the association between pCR and overall survival (OS) or event-free survival (EFS), were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models including pCR as predictor. RESULTS Overall, 52 of 3,209 publications met inclusion criteria, totaling 27,895 patients. Patients with a pCR after NAT had significantly better EFS (HR = 0.31; 95% PI, 0.24-0.39), particularly for triple-negative (HR = 0.18; 95% PI, 0.10-0.31) and HER2+ (HR = 0.32; 95% PI, 0.21-0.47) disease. Similarly, pCR after NAT was also associated with improved survival (HR = 0.22; 95% PI, 0.15-0.30). The association of pCR with improved EFS was similar among patients who received subsequent adjuvant chemotherapy (HR = 0.36; 95% PI, 0.19-0.67) and those without adjuvant chemotherapy (HR = 0.36; 95% PI, 0.27-0.54), with no significant difference between the two groups (P = 0.60). CONCLUSIONS Achieving pCR following NAT is associated with significantly better EFS and OS, particularly for triple-negative and HER2+ breast cancer. The similar outcomes with or without adjuvant chemotherapy in patients who attain pCR likely reflects tumor biology and systemic clearance of micrometastatic disease, highlighting the potential of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response.See related commentary by Esserman, p. 2771.
Collapse
Affiliation(s)
- Laura M Spring
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | | | - Chandni Sharma
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - Kerry L Reynolds
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Barbara L Smith
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Brian Alexander
- Harvard Medical School, Boston, Massachusetts
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Giovanni Parmigiani
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard TH Chan School of Public Health, Boston, Massachusetts
| | - Lorenzo Trippa
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard TH Chan School of Public Health, Boston, Massachusetts
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Yi Z, Ma F, Rong G, Guan Y, Li C, Xu B. Clinical spectrum and prognostic value of TP53 mutations in circulating tumor DNA from breast cancer patients in China. Cancer Commun (Lond) 2020; 40:260-269. [PMID: 32436611 PMCID: PMC7307233 DOI: 10.1002/cac2.12032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/09/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background TP53 mutations are common in breast cancer. There is currently no large‐scale cohort study to investigate the TP53 landscape in breast cancer patients from China. The predictive value of TP53 mutations for the efficacy of human epidermal growth factor receptor 2 (HER2)‐targeted therapy in breast cancer remains controversial. In the present study, we aimed to analyze the clinical spectrum and prognostic value of TP53 mutations in circulating tumor DNA (ctDNA) from breast cancer patients in China. Methods We retrospectively analyzed the clinical data and TP53 mutation features in ctDNA samples from 804 patients with metastatic breast cancer. TP53 mutations were detected by target region capture‐based next‐generation sequencing. The relationship between TP53 mutation status and disease‐free survival (DFS) was analyzed in 444 patients with metastatic breast cancer. Moreover, the relationship between TP53 mutation status and progression‐free survival (PFS) was analyzed in 55 HER2‐positive patients treated with first‐line trastuzumab‐based therapy. Kaplan‐Meier analysis was performed to estimate the survival curves of the different subgroups, and the log‐rank test was used to compare the curves. A Cox regression model was used to estimate multivariable‐adjusted hazard ratios and their 95% confidence intervals (CIs) associated with the DFS and PFS. Results Among the 804 investigated patients, 431 (53.6%) patients harbored TP53 mutations. TP53 mutations were differentially distributed among different molecular subtypes of breast cancer (P < 0.05). Patients with TP53 mutations had a shorter DFS than those with wild‐type TP53 (hazard ratio = 1.32, 95% CI = 1.09‐1.61, P = 0.005). TP53 mutations in exons 5‐8 were associated with worse outcome (hazard ratio = 1.50, 95% CI = 1.11‐2.03, P = 0.009). However, TP53 mutation status was not significantly associated with PFS in HER2‐positive patients who received first‐line trastuzumab‐based therapy (P = 0.966). Interestingly, in the taxane combination group, patients with TP53 mutations exhibited longer PFS than those without TP53 mutations (hazard ratio = 0.08, 95% CI = 0.02‐0.30, P < 0.001). However, in the non‐taxane combination group, patients with TP53 mutations displayed shorter PFS than those with wild‐type TP53 (hazard ratio = 4.84, 95% CI = 1.60‐14.66, P = 0.005). Conclusions TP53 mutations in exons 5‐8 may be an independent prognostic marker for short DFS in patients with metastatic breast cancer. TP53 mutations had opposite effects on trastuzumab‐treated patients treated with and without taxanes.
Collapse
Affiliation(s)
- Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Guohua Rong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Yanfang Guan
- Geneplus-Beijing Institute, Beijing, 100021, P. R. China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| |
Collapse
|
23
|
BAD sensitizes breast cancer cells to docetaxel with increased mitotic arrest and necroptosis. Sci Rep 2020; 10:355. [PMID: 31942016 PMCID: PMC6962214 DOI: 10.1038/s41598-019-57282-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer patients are commonly treated with taxane (e.g. docetaxel) chemotherapy, despite poor outcomes and eventual disease relapse. We previously identified the Bcl-2-associated death promoter (BAD) as a prognostic indicator of good outcome in taxane-treated breast cancer patients. We also demonstrated that BAD expression in human breast carcinoma cells generated larger tumors in mouse xenograft models. These paradoxical results suggest that BAD-expressing tumors are differentially sensitive to taxane treatment. We validated this here and show that docetaxel therapy preferentially reduced growth of BAD-expressing xenograft tumors. We next explored the cellular mechanism whereby BAD sensitizes cells to docetaxel. Taxanes are microtubule inhibiting agents that cause cell cycle arrest in mitosis whereupon the cells either die in mitosis or aberrantly exit (mitotic slippage) and survive as polyploid cells. In response to docetaxel, BAD-expressing cells had lengthened mitotic arrest with a higher proportion of cells undergoing death in mitosis with decreased mitotic slippage. Death in mitosis was non-apoptotic and not dependent on Bcl-XL interaction or caspase activation. Instead, cell death was necroptotic, and dependent on ROS. These results suggest that BAD is prognostic for favourable outcome in response to taxane chemotherapy by enhancing necroptotic cell death and inhibiting the production of potentially chemoresistant polyploid cells.
Collapse
|
24
|
Alfirevic A, Pirmohamed M, Marinovic B, Harcourt‐Smith L, Jorgensen AL, Cooper TE. Genetic testing for prevention of severe drug-induced skin rash. Cochrane Database Syst Rev 2019; 7:CD010891. [PMID: 31314143 PMCID: PMC6636675 DOI: 10.1002/14651858.cd010891.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Drug-induced skin reactions present with a range of clinical symptoms, from mild maculopapular skin rashes to potentially fatal blistering skin rashes - such as Stevens-Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN) - which may result in death. Milder reactions may be troublesome and lead to low drug compliance. The pathogenesis of these drug reactions is not yet fully understood; however, there is evidence that pretreatment genetic testing may help to predict and prevent these reactions in some cases. OBJECTIVES To assess the effects of prospective pharmacogenetic screening to reduce drug-associated skin reactions in a patient population. SEARCH METHODS We searched the following databases up to July 2018: the Cochrane Skin Specialised Register, CENTRAL, MEDLINE, Embase and LILACS. We also searched five trials registers, and checked the reference lists of included studies and relevant reviews for further references to relevant randomised controlled trials (RCTs). SELECTION CRITERIA We included RCTs of participants who had prospective pharmacogenetic screening to determine genetic variants associated with hypersensitivity reactions, compared with those who did not have prospective pharmacogenetic screening. We included participants in any setting, who were of any age, gender, and ethnicity, who had been prescribed drugs known to cause delayed type hypersensitivity reactions. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. To assess studies for inclusion, two review authors independently screened all of the titles and abstracts of publications identified by the searches. Because there was only one included study, many of the planned data analyses were not applicable to the review. We used GRADE to assess the quality of the included study.The review's primary outcomes were the incidence of severe skin rashes with systemic symptoms (such as fever and multiple organ involvement), and long-term effects (such as scarring of eyelids or lung tissue). Secondary outcomes were hospitalisation for drug-induced skin reactions, blistering skin reactions (such as SJS, hypersensitivity (HSS) syndrome), and death. MAIN RESULTS One study, which was a randomised, double-blind, controlled, multicentre trial, fulfilled our inclusion criteria. The trial included 1956 adult participants (74% men, with a mean age of 42 years) across 265 centres (medical centres, hospitals, outpatient clinics) in 19 countries around the world who were infected with HIV-type 1 and who had not received abacavir previously. The participants, who had a clinical need for treatment with an antiretroviral-drug regimen containing abacavir, were randomly assigned to undergo prospective human leukocyte antigen (HLA) Class I, locus B, allele 57:01 (HLA-B*57:01) screening (prospective-screening group) before this treatment, or to undergo a standard-care approach of abacavir use without prospective HLA-B*57:01 screening (control group). Participants who tested positive for HLA-B*57:01 were not given abacavir; instead, they received antiretroviral therapy that did not include abacavir. The control group did have retrospective HLA-B*57:01 pharmacogenetic testing. The trial duration was six months. Each participant was observed for six weeks. Assessments were performed at the time of study entry, at baseline (day one of abacavir treatment), and at weeks one, two and six. This study was funded by the manufacturer of abacavir, GlaxoSmithKline.The study did not assess any of our primary outcomes, and it measured none of our secondary outcomes in isolation. However, it did assess an outcome of (characteristically severe) hypersensitivity reaction which included (but was not limited to) our secondary outcomes of HSS and SJS/TEN.The study demonstrated that prospective HLA-B*57:01 screening probably reduces the incidence of hypersensitivity reaction to abacavir. The incidence of clinically diagnosed HSS reaction to abacavir was lower in the screening arm (risk ratio (RR) 0.43, 95% confidence interval (CI) 0.28 to 0.67; 1650 participants; moderate-quality evidence), as was immunologically confirmed HSS reaction (RR 0.02, 95% 0.00 to 0.37; 1644 participants; moderate-quality evidence). A positive result from an epicutaneous patch test performed six to ten weeks after clinical diagnosis provided immunological confirmation.Overall, the study demonstrates a low risk of bias across five out of seven domains. There was a high risk of detection bias because hypersensitivity reactions were diagnosed by the principal investigator at the recruitment site without the use of predefined clinical criteria. Although there was also high risk of attrition bias due to excluding participants with incomplete follow-up from analyses, the authors did undertake a series of sensitivity analyses based on the intention-to-treat population, which demonstrated consistent results with the primary analysis. We rated the study quality as moderate-quality using GRADE criteria. AUTHORS' CONCLUSIONS Prospective screening for HLA-B*57:01 probably reduces severe hypersensitivity skin reactions to abacavir in patients positive for HIV-type 1. However, these results are only based on one study, which was at high risk of attrition and detection bias.Our primary outcomes (incidence of severe skin rashes with systemic symptoms, and long-term effects) were not assessed by the trial, and only one of the review's secondary outcomes was measured (hypersensitivity reaction); thus, we found no evidence relating to hospitalisation, death, or long-term conditions resulting from drug injury.We found no eligible evidence on genetic testing for severe drug-induced skin rash in relation to different drugs and classes of drugs. Further clinical trials based on other drugs, and in different patient populations, would be useful for advising policy changes for improving the prevention of adverse skin reactions to drug treatments.
Collapse
Affiliation(s)
- Ana Alfirevic
- Institute of Translational Medicine, University of LiverpoolDepartment of Molecular and Clinical PharmacologyCentre for Personalised Medicine, Block A: Waterhouse Building1‐5 Brownlow StreetLiverpoolUKL69 3GE
| | - Munir Pirmohamed
- Institute of Translational Medicine, University of LiverpoolDepartment of Molecular and Clinical PharmacologyCentre for Personalised Medicine, Block A: Waterhouse Building1‐5 Brownlow StreetLiverpoolUKL69 3GE
| | - Branka Marinovic
- University Hospital Centre Zagreb, School of Medicine, University of ZagrebDepartment of Dermatology and VenereologySalata 4ZagrebCroatia10000
| | - Linda Harcourt‐Smith
- The University of Nottinghamc/o Cochrane Skin GroupA103, King's Meadow CampusLenton LaneNottinghamUKNG7 2NR
| | - Andrea L Jorgensen
- University of LiverpoolCentre for Medical Statistics and Health EvaluationShelley's CottageBrownlow StreetLiverpoolUKL69 3 GS
| | - Tess E Cooper
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchWestmeadNSWAustralia2145
| | | |
Collapse
|
25
|
Pathak M, Dwivedi SN, Deo SVS, Thakur B, Sreenivas V, Rath GK. Effectiveness of taxanes over anthracyclines in neoadjuvant setting: A systematic-review and meta-analysis. World J Meta-Anal 2019; 7:170-183. [DOI: 10.13105/wjma.v7.i4.170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anthracyclines and taxanes are more active group of chemotherapy regimen. Randomized controlled trials (RCTs) reported variable evidences regarding efficacy of taxanes over anthracyclines for tumor response and survival outcomes. The present study compares the relative efficacy of taxanes over anthracyclines using pathological complete response (pCR), clinical responses, breast-conserving surgeries and survival outcomes in female breast cancer patients by systematic review and meta-analysis of available RCTs.
AIM To assess the effectiveness of taxanes over anthracyclines in neoadjuvant setting in terms of tumor response and survival outcomes.
METHODS All RCTs assessing efficacy of taxanes over anthracyclines in neoadjuvant setting for management of breast cancer searched through PubMed and Cochrane register of controlled trials on 28 April 2017 and published in English language were considered. Following PRISMA guideline, retrieved records were screened and data were extracted by two independent reviewers. Meta-analysis was performed using fixed effect or random effect method depending on heterogeneity assessed using I2 statistic. Subgroup meta-analyses on the basis of taxane alone or taxane along with anthracycline in comparison to anthracycline alone were also performed for each considered outcomes.
RESULTS A total of 16 RCTs involving 6752 breast cancer patients were found eligible. Taxanes based chemotherapy significantly improved pCR (n = 7, RR = 1.48, 95%CI: 1.04-2.12), disease free survival [n = 6, RR = 0.89 (0.80-0.99)] and loco-regional recurrence free survival [n = 4, RR = 0.74 (0.59-0.94)]. Interestingly in subgroup analysis, addition of taxane to anthracyclines showed better effectiveness regarding these survivals over anthracyclines than taxane alone over anthracycline.
CONCLUSION Addition of taxanes to anthracyclines based chemotherapy significantly improves pCR, disease free survival and loco-regional recurrence free survival but with no significant impact on breast conservation rates.
Collapse
Affiliation(s)
- Mona Pathak
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi 110029, India
- Division of Biostatistics, Kalinga Institute of Medical Sciences, Bhubaneswar 751024, India
| | - Sada Nand Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - SVS Deo
- Department of Surgical Oncology, BRAIRCH, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bhaskar Thakur
- Division of Biostatistics, Kalinga Institute of Medical Sciences, Bhubaneswar 751024, India
| | - Vishnubhatla Sreenivas
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Goura Kishor Rath
- Department of Radiotherapy, BRAIRCH, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
26
|
Mullan PB, Bingham V, Haddock P, Irwin GW, Kay E, McQuaid S, Buckley NE. NUP98 - a novel predictor of response to anthracycline-based chemotherapy in triple negative breast cancer. BMC Cancer 2019; 19:236. [PMID: 30935371 PMCID: PMC6444590 DOI: 10.1186/s12885-019-5407-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
Background Triple Negative breast cancer (TNBC) is a poor outcome subgroup of breast cancer defined based on the absence of expression of ERα and PR and HER2 amplification. These hard to treat cancers lack targeted treatment options and are therefore treated with a standard of care (SoC) generic cocktail of DNA damaging chemotherapy, with a wide range of clinical responses. While a subset of TNBC patients respond very well to this treatment, others receive no clinical benefit and die from their disease within a short time period. We currently lack biomarkers to prospectively identify patients likely to relapse and we lack alternate treatment options. Methods NUP98 protein expression was investigated in patient samples using two independent tissue microarrays (TMAs), as well as a normal breast TMA. Correlation with pathological response to various chemotherapy regimens was investigated. Results We have shown that high NUP98 is significantly associated with poor outcome in TNBC patient samples both by gene expression and IHC-based protein analysis. While trends linking NUP98 expression with poorer outcomes were observed in breast cancer overall (and more specifically in the LuminalB Her2- subgroup), significant correlations were observed in TNBC. This appeared to be specific to anthracycline based regimens as the association between NUP98 and response was not observed in patients treated with taxane-based chemotherapy. Conclusions We have identified a novel biomarker, NUP98, that can predict response to anthracycline based chemotherapy in TNBC. The ability to prospectively identify patients who are less likely to respond to SoC chemotherapy is a vital step in improving the overall survival of these patients. Electronic supplementary material The online version of this article (10.1186/s12885-019-5407-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paul B Mullan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland
| | - Victoria Bingham
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland
| | - Paula Haddock
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland
| | - Gareth W Irwin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland.,Nightingale Breast Centre, Wythenshawe Hospital, Manchester University Foundation Trust, Manchester, UK
| | - Elaine Kay
- Department of Surgery, Beaumont Hospital and Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephen McQuaid
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland
| | - Niamh E Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland.
| |
Collapse
|
27
|
Bonnefoi H, MacGrogan G, Poncet C, Iggo R, Pommeret F, Grellety T, Larsimont D, Bécette V, Kerdraon O, Bibeau F, Ghnassia JP, Picquenot JM, Thomas J, Tille JC, Slaets L, Bodmer A, Bergh J, Cameron D. Molecular apocrine tumours in EORTC 10994/BIG 1-00 phase III study: pathological response after neoadjuvant chemotherapy and clinical outcomes. Br J Cancer 2019; 120:913-921. [PMID: 30899086 PMCID: PMC6734658 DOI: 10.1038/s41416-019-0420-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 02/09/2019] [Accepted: 02/20/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We explored, within the EORTC10994 study, the outcomes for patients with molecular apocrine (MA) breast cancer, and defined immunohistochemistry (IHC) as androgen-receptor (AR) positive, oestrogen (ER) and progesterone (PR) negative. We also assessed the concordance between IHC and gene expression arrays (GEA) in the identification of MA cancers. METHODS Centrally assessed biopsies for AR, ER, PR, HER2 and Ki67 by IHC were classified into six subtypes: MA, triple-negative (TN) basal-like, luminal A, luminal B HER2 negative, luminal B HER2 positive and "other". The two main objectives were the pCR rates and survival outcomes in the overall MA subtype (and further divided by HER2 status) and the remaining five subtypes. RESULTS IHC subtyping was obtained in 846 eligible patients. Ninety-three (11%) tumours were classified as the MA subtype. Both IHC and GEA data were available for 64 patients. In this subset, IHC concordance was 88.3% in identifying MA tumours compared with GEA. Within the MA subtype, pCR was observed in 33.3% of the patients (95% CI: 29.4-43.9) and the 5-year recurrence-free interval was 59.2% (95% CI: 48.2-68.6). Patients with MA and TN basal-like tumours have lower survival outcomes. CONCLUSIONS Irrespective of their HER2 status, the prognosis for MA tumours remains poor and adjuvant trials evaluating anti-androgens should be considered.
Collapse
Affiliation(s)
- Hervé Bonnefoi
- Department of Medical Oncology, Institut Bergonié Unicancer, University of Bordeaux, INSERM U1218, INSERM CIC1401, Bordeaux, France.
| | - Gaetan MacGrogan
- Department of BioPathology, Institut Bergonié Unicancer, INSERM U1218, Bordeaux, France
| | - Coralie Poncet
- European Organisation for Research and Treatment of Cancer EORTC) Headquarters, Brussels, Belgium
| | - Richard Iggo
- Institut Bergonié Unicancer, INSERM U1218, Bordeaux, France
| | - Fanny Pommeret
- Department of Medical Oncology, Institut Bergonié Unicancer, University of Bordeaux, INSERM U1218, INSERM CIC1401, Bordeaux, France
| | - Thomas Grellety
- Department of Medical Oncology, Institut Bergonié Unicancer, University of Bordeaux, INSERM U1218, INSERM CIC1401, Bordeaux, France
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | - Véronique Bécette
- Department of Pathology, Institut Curie-Hôpital René Huguenin, Saint-Cloud, France
| | - Olivier Kerdraon
- Department of Pathology, Centre René Gauducheau, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Frédéric Bibeau
- Department of Pathology, Institut de Cancérologie de Montpellier (ICM), Montpellier, France
| | | | | | - Jeremy Thomas
- Department of Pathology, Edinburgh Cancer Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Jean-Christophe Tille
- Department of Pathologie, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland
| | - Leen Slaets
- European Organisation for Research and Treatment of Cancer EORTC) Headquarters, Brussels, Belgium
| | - Alexandre Bodmer
- Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland
| | - Jonas Bergh
- Swedish Breast Cancer Group (SweBCG), Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - David Cameron
- Anglo-Celtic Cooperative Oncology Group (ACCOG), Edinburgh, United Kingdom
- Department of Medical Oncology, Edinburgh Cancer Centre, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Stoll G, Kremer M, Bloy N, Joseph A, Castedo M, Meurice G, Klein C, Galluzzi L, Michels J, Kroemer G. Metabolic enzymes expressed by cancer cells impact the immune infiltrate. Oncoimmunology 2019; 8:e1571389. [PMID: 31069148 DOI: 10.1080/2162402x.2019.1571389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
The expression of two metabolic enzymes, i.e., aldehyde dehydrogenase 7 family, member A1 (ALDH7A1) and lipase C, hepatic type (LIPC) by malignant cells, has been measured by immunohistochemical methods in non-small cell lung carcinoma (NSCLC) biopsies, and has been attributed negative and positive prognostic value, respectively. Here, we demonstrate that the protein levels of ALDH7A1 and LIPC correlate with the levels of the corresponding mRNAs. Bioinformatic analyses of gene expression data from 4921 cancer patients revealed that the expression of LIPC positively correlates with abundant tumor infiltration by myeloid and lymphoid cells in NSCLC, breast carcinoma, colorectal cancer and melanoma samples. In contrast, high levels of ALDH7A1 were associated with a paucity of immune effectors within the tumor bed. These data reinforce the notion that the metabolism of cancer cells has a major impact on immune and inflammatory processes in the tumor microenvironment, pointing to hitherto unsuspected intersections between oncometabolism and immunometabolism.
Collapse
Affiliation(s)
- Gautier Stoll
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Margerie Kremer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Normal Bloy
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Adrien Joseph
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Castedo
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guillaume Meurice
- Bioinformatics Core Facility, Gustave Roussy Cancer Campus, Villejuif, France
| | - Christophe Klein
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Centre d'Histologie, Imagerie cellulaire et Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Judith Michels
- Department of Medical Oncology, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Centre de Recherche des Cordeliers, Equipe 11 labellisée Ligue Nationale contre le Cancer, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
29
|
Janiaud P, Serghiou S, Ioannidis JP. New clinical trial designs in the era of precision medicine: An overview of definitions, strengths, weaknesses, and current use in oncology. Cancer Treat Rev 2019; 73:20-30. [DOI: 10.1016/j.ctrv.2018.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
|
30
|
Hashmi AA, Naz S, Hashmi SK, Hussain ZF, Irfan M, Khan EY, Faridi N, Khan A, Edhi MM. Prognostic significance of p16 & p53 immunohistochemical expression in triple negative breast cancer. BMC Clin Pathol 2018; 18:9. [PMID: 30305801 PMCID: PMC6171321 DOI: 10.1186/s12907-018-0077-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022] Open
Abstract
Background p16 and p53 genes are frequently mutated in triple negative breast cancer & prognostic value of these mutations have been shown; however, their role as immunohistochemical overexpression has not been fully validated. Therefore we aimed to evaluate the association of p16 and p53 overexpression in triple negative breast cancer with various prognostic parameters. Methods Total 150 cases of triple negative breast cancers were selected from records of pathology department archives that underwent surgeries at Liaquat National hospital, Karachi from January 2008 till December 2013. ER, PR and Her2neu immunohistochemistry were re-performed to confirm triple negative status. p16 & p53 immunohistochemistry was performed on all cases and association with various clinicopathologic parameters was determined. Results Mean age of the patients involved in the study was 48.9 years. Most of the patients presented at stage T2 with a high mean ki67 index i.e. 46.9%. 42.7% of cases had nodal metastasis. Although 84% cases were of invasive ductal carcinoma; however a significant proportion of cases were of metaplastic histology (9.3%). Fifty-one percent (76 cases) of cases showed positive p53 expression while 49% (74 cases) were negative. Higher percentage of p53 expression was found to correlate with higher T stage, high ki67 index and higher nodal stage. On the other hand, strong intensity of p53 expression was positively correlated with higher tumor grade and ki67 index. Seventy-one percent (98 cases) of cases showed positive p16 expression, whereas 24.8% (34 cases) were negative and 3.6% (5 cases) showed focal positive p16 expression. However, no significant association was found between p16 expression and various clinical and pathologic parameters. Similarly, no significant association of either p16 or p53 over-expression was noted with recurrence status of patients. Conclusion On the basis of significant association of p53 over-expression with worse prognostic factors in triple negative breast cancer, therefore we suggest that more large scale studies are needed to validate this finding in loco-regional population. Moreover, high expression of p16 in triple negative breast cancer suggests a potential role of this biomarker in triple negative breast cancer pathogenesis which should be investigated with molecular based research in our population.
Collapse
Affiliation(s)
- Atif Ali Hashmi
- 1Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Samreen Naz
- 1Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | | | - Muhammad Irfan
- 1Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Erum Yousuf Khan
- 1Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Naveen Faridi
- 1Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Amir Khan
- 3Kandahar University, North, Kandahar 3802 Afghanistan
| | | |
Collapse
|
31
|
Bae SY, Jung SP, Lee SK, Yu J, Lee JE, Kim SW, Nam SJ. Prognostic value of immunohistochemically detected p53 in adjuvant chemotherapy-treated triple negative breast cancer. Kaohsiung J Med Sci 2018; 34:663-672. [PMID: 30527200 DOI: 10.1016/j.kjms.2018.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/09/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
Mutations of the p53 gene are the most common genomic alterations associated with triple-negative breast cancer (TNBC) and are reported in 60-88% cases. Despite the high incidence of such mutations, there is no consensus about the clinical application of p53 detection in breast cancer management. This study investigates the prognostic value of immunohistochemically detected p53 in TNBC patients who received adjuvant chemotherapy. We reviewed the clinicopathologic features of 1088 TNBC patients who received curative surgery and adjuvant chemotherapy. Immunohistochemically, nuclear staining of >10% was defined as p53 "positive." Of the total 1088 TNBC patients, 709 (65.2%) had no lymph node metastasis (N0). Among the N0 patients, 408 (57.5%) were p53- positive (p53+), and 301 (42.5%) were p53- negative (p53-). p53 + tumors showed a tendency for better breast cancer-specific survival (BCSS, p = 0.052) and overall survival (OS, p = 0.079) compared to p53- tumors. In multivariate analysis, p53 + tumors showed significantly better BCSS (p53 + vs. p53-; HR 2.8, 95% confidence interval: 1.1-7.3, p = 0.034); however, in TNBC patients with lymph node metastasis, there was no correlation between p53 status, clinicopathologic characteristics, and survival. Consequently, in TNBC patients who received adjuvant chemotherapy, immunohistochemical p53 expression was associated with better BCSS in N0 patients.
Collapse
Affiliation(s)
- Soo Youn Bae
- Department of Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Seung Pil Jung
- Department of Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Se Kyung Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jonghan Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seok Jin Nam
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Stoll G, Pol J, Soumelis V, Zitvogel L, Kroemer G. Impact of chemotactic factors and receptors on the cancer immune infiltrate: a bioinformatics study revealing homogeneity and heterogeneity among patient cohorts. Oncoimmunology 2018; 7:e1484980. [PMID: 30288345 PMCID: PMC6169589 DOI: 10.1080/2162402x.2018.1484980] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 01/19/2023] Open
Abstract
Multiple soluble factors including proteins (in particular chemokines), non-proteinaceous factors released by dead cells, as well as receptors for such factors (in particular chemokine receptors, formyl peptide receptors and purinergic receptors), influence the recruitment of distinct cell subsets into the tumor microenvironment. We performed an extensive bioinformatic analysis on tumor specimens from 5953 cancer patients to correlate the mRNA expression levels of chemotactic factors/receptors with the density of immune cell types infiltrating the malignant lesions. This meta-analysis, which included specimens from breast, colorectal, lung, ovary and head and neck carcinomas as well as melanomas, revealed that a subset of chemotactic factors/receptors exhibited a positive and reproducible correlation with several infiltrating cell types across various solid cancers, revealing a universal pattern of association. Hence, this meta-analysis distinguishes between homogeneous associations that occur across different cancer types and heterogeneous correlations, that are specific of one organ. Importantly, in four out of five breast cancer cohorts for which clinical data were available, the levels of expression of chemotactic factors/receptors that exhibited universal (rather than organ-specific) positive correlations with the immune infiltrate had a positive impact on the response to neoadjuvant chemotherapy. These results support the notion that general (rather than organ-specific) rules governing the recruitment of immune cells into the tumor bed are particularly important in determining local immunosurveillance and response to therapy.
Collapse
Affiliation(s)
- Gautier Stoll
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jonathan Pol
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Vassili Soumelis
- pôle de biopathologie, Institut Curie, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U932, Paris, France.,CIC IGR-Curie 1428, Paris, France.,PSL, Paris, France
| | - Laurence Zitvogel
- Equipe labellisée Ligue Nationale Contre le Cancer, Institut National de la Santé et de la Recherche Médicale, U1015, Villejuif, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
33
|
Pathak M, Dwivedi SN, Deo SVS, Thakur B, Sreenivas V, Rath GK. Neoadjuvant chemotherapy regimens in treatment of breast cancer: a systematic review and network meta-analysis protocol. Syst Rev 2018; 7:89. [PMID: 29945652 PMCID: PMC6020442 DOI: 10.1186/s13643-018-0754-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 06/06/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT), a standard of care for locally advanced breast cancer patients, is widely used for early breast cancer patients also. The varying role of regimens used as NACT needs to be investigated. Despite availability of some randomized controlled trials (RCTs), it is unclear which treatment regimen suits best. Further, there is no study comparing all the three regimens. Accordingly, present study will compare the efficacy of anthracyclines, taxanes, and targeted therapy administered in neoadjuvant setting on the basis of oncological outcomes and functional outcomes. METHOD/DESIGN Online databases PubMed and Cochrane Register of Controlled Trials will be searched to acquire eligible studies. Further, content of relevant journals, references of relevant articles, and proceedings of major related conference will also be searched. The RCTs comparing any of abovementioned regimen as NACT on breast cancer patients will be eligible. Two reviewers independently and in duplicate will screen the records on the basis of title and abstract and complete full-text review to determine eligibility. Similarly, data extraction and risk of bias assessment will be done by two independent reviewers. The pair-wise meta-analysis as well as network meta-analysis will be conducted to assess the relative efficacy of anthracyclines, taxanes, and targeted therapy regimens. DISCUSSION The present systematic review will improve the understanding of the relative efficacies of the three treatment regimens and possibly guide the clinical practices by providing the current best evidence on the efficacy of various regimens of NACT in the management of breast cancer patients. SYSTEMATIC REVIEW REGISTRATION PROSPERO ( CRD42016027236 ).
Collapse
Affiliation(s)
- Mona Pathak
- Department of Biostatistics, All India Institute of Medical Sciences, Room No.5, New Delhi, 110029, India
| | - Sada Nand Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences, Room No.5, New Delhi, 110029, India.
| | - S V S Deo
- Department of Surgical Oncology, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Bhaskar Thakur
- Department of Biostatistics, All India Institute of Medical Sciences, Room No.5, New Delhi, 110029, India
| | - Vishnubhatla Sreenivas
- Department of Biostatistics, All India Institute of Medical Sciences, Room No.5, New Delhi, 110029, India
| | - G K Rath
- Department of Radiotherapy, Dr. BRA IRCH, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
34
|
Fan H, Li C, Xiang Q, Xu L, Zhang Z, Liu Q, Zhang T, Zhou Y, Zhao X, Cui Y. PIK3CA mutations and their response to neoadjuvant treatment in early breast cancer: A systematic review and meta-analysis. Thorac Cancer 2018; 9:571-579. [PMID: 29575819 PMCID: PMC5928352 DOI: 10.1111/1759-7714.12618] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 02/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND PIK3CA mutations frequently occur in breast cancer patients. This study was conducted to evaluate the relationship between PIK3CA mutations and neoadjuvant treatment response and to analyze the clinical implications. METHODS PubMed, Embase, and the Cochrane database were searched for relevant studies in September 2017. The pooled risk ratio (RR) was estimated using fixed effects or random effects models according to heterogeneity among studies. RESULTS This meta-analysis included 20 studies with 4392 patients. The pooled RR showed that PIK3CA mutation is correlated to lower pathological complete response (pCR) in unselected HER2+ patients (RR = 0.73; 95% confidence interval [CI] 0.66-0.81), thus the predictive value of PIK3CA status may be stronger in HER2+/HR+ patients (RR = 0.50; 95% CI 0.27-0.93) and those administered dual-targeting treatment (RR = 0.55; 95% CI 0.39-0.78). In contrast with wild type, either exon 9 (RR = 0.55; 95% CI 0.39-0.78) or exon 20 (RR = 0.71; 95% CI 0.58-0.89) mutations were significantly associated with lower pCR. The predictive value of exon 9 mutations was not significantly greater than exon 20 mutations (RR = 0.76; 95% CI 0.51-1.13). CONCLUSION In early breast cancer, PIK3CA mutations seem to identify HER2+ patients who are less likely to reach pCR. The clinical implications of PIK3CA mutations tend to vary between exon 9 and exon 20. This mechanism should be explored in further studies.
Collapse
Affiliation(s)
- Hongwei Fan
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Chao Li
- Department of PharmacyPeking University First HospitalBeijingChina
- National Cancer Center/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Qian Xiang
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Ling Xu
- Breast Disease CenterPeking University First HospitalBeijingChina
| | - Zhuo Zhang
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Qianxin Liu
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Tonttong Zhang
- National Cancer Center/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Ying Zhou
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Xia Zhao
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Yimin Cui
- Department of PharmacyPeking University First HospitalBeijingChina
| |
Collapse
|
35
|
Sande CM, Chang B, Monga V, Bossler AD, Ma D. Biallelic TP53 gain of function mutations in rapidly progressing solid tumors. Cancer Genet 2018; 222-223:20-24. [PMID: 29666004 DOI: 10.1016/j.cancergen.2018.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/31/2018] [Accepted: 02/19/2018] [Indexed: 01/04/2023]
Abstract
Recent studies are discovering TP53 mutations with gain of function (GOF) properties that promote tumorigenesis via a variety of mechanisms. To our knowledge, all reported compound mutations are allelic. We identified two patients with biallelic GOF TP53 mutations in their tumors and a third with allelic compound variants. The correlation with p53 expression was also examined. Genomic DNA was extracted from formalin-fixed, paraffin-embedded tissue and mutational analysis was performed using Ion AmpliSeq™Cancer HotSpot Panel V2. Biallelic GOF mutations (p.R273H and p.R273C) were identified in a 19-year-old male with glioblastoma (allele frequencies 94% and 48%) and a 54-year-old with pT3 penile squamous cell carcinoma (allele frequencies 19% and 27%). Immunohistochemistry showed nuclear accumulation of p53. The third patient, a 62-year-old female with metastatic lung adenocarcinoma, had allelic p.P278S (GOF) and p.R283L (non-GOF) variants at frequencies of 61% but with null staining for p53. Germline testing for Patient 1 confirmed wildtype TP53. No other variants were discovered among the genes tested in these cases. All patients succumbed within two years of diagnosis despite aggressive treatment. In conclusion, implementation of TP53 mutation analysis in clinical practice may predict patient outcome, and inhibition of GOF p53 could represent an attractive target for therapy.
Collapse
Affiliation(s)
- Christopher M Sande
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City 52241, IA, USA
| | - Brian Chang
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City 52241, IA, USA
| | - Varun Monga
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City 52241, IA, USA
| | - Aaron D Bossler
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City 52241, IA, USA
| | - Deqin Ma
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City 52241, IA, USA .
| |
Collapse
|
36
|
Duffy MJ, Synnott NC, Crown J. Mutant p53 in breast cancer: potential as a therapeutic target and biomarker. Breast Cancer Res Treat 2018; 170:213-219. [PMID: 29564741 DOI: 10.1007/s10549-018-4753-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/13/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The aim of this article is to discuss mutant p53 as a possible therapeutic target and biomarker for breast cancer. RESULTS TP53 (p53) is the most frequently mutated gene in invasive breast cancer. Although mutated in 30-35% of all cases, p53 is mutated in approximately 80% of triple-negative (TN) tumors (i.e., tumors negative for ER, PR, and HER2). Because of this high prevalence, mutated p53 is both a potential biomarker and therapeutic target for patients with breast cancer, especially for those with the TN subtype. Although several retrospective studies have investigated a potential prognostic and therapy predictive role for mutant p53 in breast cancer, the results to date are mixed. Thus, at present, mutant p53 cannot be recommended as a prognostic or therapy predictive biomarker in breast cancer. In contrast to the multiple reports on a potential biomarker role, few studies had until recently, investigated mutant p53 as a potential target for breast cancer treatment. In the last decade, however, several compounds have become available which can reactivate mutant p53 protein and convert it to a conformation with wild-type properties. Some of these compounds, especially PRIMA-1, APR-246 PK11007, and COTI-2, have been found to exhibit anticancer activity in preclinical models of breast cancer. CONCLUSION Since p53 is mutated in the vast majority of TN breast cancers, compounds such as APR-246, PK11007, and COTI-2 are potential treatments for patients with this subform of the disease. Further research is necessary to identify a potential biomarker role for mutant p53 in breast cancer.
Collapse
Affiliation(s)
- Michael J Duffy
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin 4, Ireland. .,UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| | - Naoise C Synnott
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John Crown
- Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
37
|
Aalders KC, Touati N, Tryfonidis K, Annonay M, Litiere S, Bergh J, Bodmer A, Cameron DA, Bonnefoi HR. Do patients whose tumor achieved a pathological response relapse at specific sites? A substudy of the EORTC 10994/BIG-1-00 trial. Breast Cancer Res Treat 2018; 169:497-505. [DOI: 10.1007/s10549-018-4698-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 01/31/2023]
|
38
|
Darb-Esfahani S, Denkert C, Stenzinger A, Salat C, Sinn B, Schem C, Endris V, Klare P, Schmitt W, Blohmer JU, Weichert W, Möbs M, Tesch H, Kümmel S, Sinn P, Jackisch C, Dietel M, Reimer T, Loi S, Untch M, von Minckwitz G, Nekljudova V, Loibl S. Role of TP53 mutations in triple negative and HER2-positive breast cancer treated with neoadjuvant anthracycline/taxane-based chemotherapy. Oncotarget 2018; 7:67686-67698. [PMID: 27611952 PMCID: PMC5356512 DOI: 10.18632/oncotarget.11891] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/24/2016] [Indexed: 11/27/2022] Open
Abstract
Background TP53 mutations are frequent in breast cancer, however their clinical relevance in terms of response to chemotherapy is controversial. Methods 450 pre-therapeutic, formalin-fixed, paraffin-embedded core biopsies from the phase II neoadjuvant GeparSixto trial that included HER2-positive and triple negative breast cancer (TNBC) were subjected to Sanger sequencing of exons 5-8 of the TP53 gene. TP53 status was correlated to response to neoadjuvant anthracycline/taxane-based chemotherapy with or without carboplatin and trastuzumab/lapatinib in HER2-positive and bevacizumab in TNBC. p53 protein expression was evaluated by immunohistochemistry in the TNBC subgroup. Results Of 450 breast cancer samples 297 (66.0%) were TP53 mutant. Mutations were significantly more frequent in TNBC (74.8%) compared to HER2-positive cancers (55.4%, P < 0.0001). Neither mutations nor different mutation types and effects were associated with pCR neither in the whole study group nor in molecular subtypes (P > 0.05 each). Missense mutations tended to be associated with a better survival compared to all other types of mutations in TNBC (P = 0.093) and in HER2-positive cancers (P = 0.071). In TNBC, missense mutations were also linked to higher numbers of tumor-infiltrating lymphocytes (TILs, P = 0.028). p53 protein overexpression was also linked with imporved survival (P = 0.019). Conclusions Our study confirms high TP53 mutation rates in TNBC and HER2-positive breast cancer. Mutations did not predict the response to an intense neoadjuvant chemotherapy in these two molecular breast cancer subtypes.
Collapse
Affiliation(s)
| | - Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, (DKTK), Berlin, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Pathology, Center for Integrated Diagnostics (CID), Massachusetts General Hospital, Boston, MA, USA
| | | | - Bruno Sinn
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Schem
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Hostein, Kiel, Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Klare
- Praxisklinik Krebsheilkunde für Frauen/Brustzentrum, Berlin, Germany
| | - Wolfgang Schmitt
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jens-Uwe Blohmer
- Department of Gynecology and Obstetrics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wilko Weichert
- German Cancer Consortium, (DKTK), Berlin, Germany.,Institute of Pathology, Technical University Munich, Munich, Germany
| | - Markus Möbs
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Hans Tesch
- Center for Hematology and Oncology Bethanien, Frankfurt/Main, Germany
| | | | - Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Jackisch
- Department of Gynecology and Obstetrics, Sana Klinikum Offenbach, Offenbach, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Toralf Reimer
- Department of Gynecology, Klinikum Südstadt Rostock, Rostock, Germany
| | - Sherene Loi
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Michael Untch
- Department of Gynecology and Obstetrics, Helios Klinikum Berlin-Buch, Berlin, Germany
| | | | | | - Sibylle Loibl
- German Breast Group c/o (GBG Forschungs GmbH), Neu-Isenburg, Germany
| |
Collapse
|
39
|
Matikas A, Margolin S, Hellström M, Johansson H, Bengtsson NO, Karlsson L, Edlund P, Karlsson P, Lidbrink E, Linderholm B, Lindman H, Malmstrom P, Villman K, Foukakis T, Bergh J. Long-term safety and survival outcomes from the Scandinavian Breast Group 2004-1 randomized phase II trial of tailored dose-dense adjuvant chemotherapy for early breast cancer. Breast Cancer Res Treat 2017; 168:349-355. [PMID: 29190004 PMCID: PMC5838137 DOI: 10.1007/s10549-017-4599-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Although adjuvant polychemotherapy improves outcomes for early breast cancer, the significant variability in terms of pharmacokinetics results in differences in efficacy and both short and long-term toxicities. Retrospective studies support the use of dose tailoring according to the hematologic nadirs. METHODS The SBG 2004-1 trial was a randomized feasibility phase II study which assessed tailored dose-dense epirubicin and cyclophosphamide (EC) followed by docetaxel (T) (group A), the same regimen with fixed doses (group B) and the TAC regimen (group C). Women aged 18-65 years, ECOG PS 0-1 with at least one positive axillary lymph node were randomized 1:1:1. The primary endpoint of the study was the safety and feasibility of the treatment. Toxicity was graded according to CTC-AE version 3.0. The design and short-term toxicity have been previously published. Here, we report safety and efficacy data after 10 years of follow-up. RESULTS A total of 124 patients were included in the study. After a median follow-up of 10.3 years, the probability for 10-year survival was 78.5, 75.1, and 63.4% and for relapse free survival 64.1, 71.0, and 59.5% for groups A, B, and C, respectively. There were no cases of clinically diagnosed cardiotoxicity or hematologic malignancies. No patient was lost to follow-up. CONCLUSIONS In this randomized phase II trial, tailored dose adjuvant chemotherapy was feasible, without an increased risk for long-term adverse events after a median follow-up of 10 years.
Collapse
Affiliation(s)
- Alexios Matikas
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden.
| | - Sara Margolin
- Department of Oncology, Stockholm South General Hospital, Stockholm, Sweden
| | - Mats Hellström
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Hemming Johansson
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Lidbrink
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Barbro Linderholm
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Lindman
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Per Malmstrom
- Department of Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | - Theodoros Foukakis
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology, Radiumhemmet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Tian Q, Du P, Li S, Bai Z, Yang Y, Zeng J. Effect of antitumor treatments on triple-negative breast cancer patients: A PRISMA-compliant network meta-analysis of randomized controlled trials. Medicine (Baltimore) 2017; 96:e8389. [PMID: 29137021 PMCID: PMC5690714 DOI: 10.1097/md.0000000000008389] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) lacks the expression of the estrogen receptor, progesterone receptor, and receptor tyrosine-protein kinase erbB-2 (HER2/neu), which renders hormone-related endocrine and targeted therapy essentially futile. OBJECTIVE We performed a meta-analysis to assess the effects of antitumor regimens in the treatment of TNBC patients. METHODS We searched electronic databases, including PubMed, Embase, and the Cochrane Library, through January 2017 using the following keywords: "triple negative breast cancer," "TNBC," and "random*" without language restrictions. The major outcome in the present analysis was the overall response rate (ORR), and the secondary outcomes were progression-free survival (PFS) and overall survival (OS). A network meta-analysis and multilevel mixed-effects logistic regression were used to compare antitumor regimens. RESULTS We included 35 articles assessing a total of 8476 TNBC patients in our systematic review. The regimen of Bevacizumab, Carboplatin, and Paclitaxel (78.2%) was the most likely to improve the ORR in TNBC patients, followed by EndoTAG-1 and Paclitaxel (69.7%), Carboplatin and Paclitaxel (65.0%), and Bevacizumab and Paclitaxel (61.8%). In the patients without metastasis, the regimen of Bevacizumab, Carboplatin, and Paclitaxel (74.9%) remained the most likely to improve the ORR. We could not analyze the results for patients with metastasis or outcomes of PFS and OS because no >4 regimens formed a network. In the regression analysis, Bevacizumab (odds ratio [OR], 1.71; 95% confidence interval [CI], 1.43-2.05; P < .001) and Carboplatin (OR, 2.07; 95% CI, 1.62-2.64; P < .001) correlated with superior ORR outcome, and Iniparib (OR, 1.51; 95% CI, 1.11-2.07; P = .009) correlated with superior OS outcome. CONCLUSION The regimen including Bevacizumab, Carboplatin, and Paclitaxel was the most likely to improve the ORR in TNBC patients and in advanced metastatic TNBC patients. The administration of Bevacizumab and Carboplatin provided greater benefit toward improved patient ORR.
Collapse
Affiliation(s)
| | - Peng Du
- Department of Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sen Li
- Department of Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | | | | | - Jinsheng Zeng
- Department of Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
41
|
Gillon P, Touati N, Breton-Callu C, Slaets L, Cameron D, Bonnefoi H. Factors predictive of locoregional recurrence following neoadjuvant chemotherapy in patients with large operable or locally advanced breast cancer: An analysis of the EORTC 10994/BIG 1-00 study. Eur J Cancer 2017; 79:226-234. [PMID: 28527420 DOI: 10.1016/j.ejca.2017.04.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Identification of clinicopathological factors predicting for a locoregional recurrence (LRR) after neoadjuvant chemotherapy (NAC) could help to decide on the optimal locoregional radiotherapy. The objective of this trial is to identify those factors in the context of a phase III trial (European Organisation for Research and Treatment of Cancer 10994). METHODS Patients received NAC followed by surgery with or without radiotherapy. Radiotherapy was administered according to pre-specified guidelines. Patients with hormone receptor positive tumours received adjuvant hormonal therapy. A proportion of patients with human epidermal growth factor receptor 2 (HER2) positive cancer received adjuvant trastuzumab. The predictive factors for LRR were identified by multivariate analysis with time to LRR as first event as the primary end-point. RESULTS The median follow-up was 4.4 years. In 1553 eligible patients, there were 76 LRRs with a 5-year cumulative incidence of 4.9% (95% confidence interval, CI [3.76-6.04]). In multivariate analysis, breast cancer subtype was a significant predictor of LRR (p < 0.0001): hazard ratio (HR) 6.44 (95% CI [2.83-14.69]) for triple negative, 6.26 (95% CI [2.81-13.93]) for HER2+ without trastuzumab (T) and 3.37 (95% CI [1.10-10.34]) for HER2+ with T cancers, all compared to luminal A patients. Lack of pathological response was also associated with significantly higher LRR risk in case of ≥4 pathologically positive nodes, HR 2.43 (95% CI [1.34-4.40], p < 0.0001). CONCLUSION Breast cancer subtype and lack of pathological response are predictive factors for high LRR after NAC.
Collapse
Affiliation(s)
- Pauline Gillon
- Department of Radiotherapy, Institut Bergonié Unicancer, Bordeaux, France
| | - Nathan Touati
- EORTC, Statistics Department, Avenue Emmanuel Mounier 83b11, 1200 Brussels, Belgium
| | | | - Leen Slaets
- EORTC, Statistics Department, Avenue Emmanuel Mounier 83b11, 1200 Brussels, Belgium
| | - David Cameron
- Western General Hospital, Edinburgh Cancer Centre, Crewe Road South, GB Edinburgh EH4 2XU, United Kingdom
| | - Hervé Bonnefoi
- Department of Medical Oncology, Institut Bergonié Unicancer, Univ. Bordeaux, INSERM U1218, INSERM CIC1401 Bordeaux, France.
| |
Collapse
|
42
|
Resistance to Taxanes in Triple-Negative Breast Cancer Associates with the Dynamics of a CD49f+ Tumor-Initiating Population. Stem Cell Reports 2017; 8:1392-1407. [PMID: 28457887 PMCID: PMC5425727 DOI: 10.1016/j.stemcr.2017.03.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/04/2022] Open
Abstract
Taxanes are a mainstay of treatment for breast cancer, but resistance often develops followed by metastatic disease and mortality. Aiming to reveal the mechanisms underlying taxane resistance, we used breast cancer patient-derived orthoxenografts (PDX). Mimicking clinical behavior, triple-negative breast tumors (TNBCs) from PDX models were more sensitive to docetaxel than luminal tumors, but they progressively acquired resistance upon continuous drug administration. Mechanistically, we found that a CD49f+ chemoresistant population with tumor-initiating ability is present in sensitive tumors and expands during the acquisition of drug resistance. In the absence of the drug, the resistant CD49f+ population shrinks and taxane sensitivity is restored. We describe a transcriptional signature of resistance, predictive of recurrent disease after chemotherapy in TNBC. Together, these findings identify a CD49f+ population enriched in tumor-initiating ability and chemoresistance properties and evidence a drug holiday effect on the acquired resistance to docetaxel in triple-negative breast cancer. PDX models mimic the clinical response to docetaxel in breast cancer patients Sensitivity to docetaxel can be regained in metastatic resistant TNBC A tumor-initiating CD49f chemoresistant population is present in TNBC Docetaxel resistance associates with the expansion of a CD49f+ population in TNBC
Collapse
|
43
|
Vandenberghe S, Duchateau L, Slaets L, Bogaerts J, Vansteelandt S. Surrogate marker analysis in cancer clinical trials through time-to-event mediation techniques. Stat Methods Med Res 2017; 27:3367-3385. [DOI: 10.1177/0962280217702179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The meta-analytic approach is the gold standard for validation of surrogate markers, but has the drawback of requiring data from several trials. We refine modern mediation analysis techniques for time-to-event endpoints and apply them to investigate whether pathological complete response can be used as a surrogate marker for disease-free survival in the EORTC 10994/BIG 1-00 randomised phase 3 trial in which locally advanced breast cancer patients were randomised to either taxane or anthracycline based neoadjuvant chemotherapy. In the mediation analysis, the treatment effect is decomposed into an indirect effect via pathological complete response and the remaining direct effect. It shows that only 4.2% of the treatment effect on disease-free survival after five years is mediated by the treatment effect on pathological complete response. There is thus no evidence from our analysis that pathological complete response is a valuable surrogate marker to evaluate the effect of taxane versus anthracycline based chemotherapies on progression free survival of locally advanced breast cancer patients. The proposed analysis strategy is broadly applicable to mediation analyses of time-to-event endpoints, is easy to apply and outperforms existing strategies in terms of precision as well as robustness against model misspecification.
Collapse
Affiliation(s)
- Sjouke Vandenberghe
- Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, Belgium
| | - Luc Duchateau
- Department of Comparative Physiology and Biometrics, Ghent University, Belgium
| | - Leen Slaets
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Stijn Vansteelandt
- Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, Belgium
| |
Collapse
|
44
|
P53 and SOX2 Protein Expression Predicts Esophageal Adenocarcinoma in Response to Neoadjuvant Chemoradiotherapy. Ann Surg 2017; 265:347-355. [DOI: 10.1097/sla.0000000000001625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
45
|
Touati N, Tryfonidis K, Caramia F, Bonnefoi H, Cameron D, Slaets L, Parker BS, Loi S. Correlation between severe infection and breast cancer metastases in the EORTC 10994/BIG 1-00 trial: Investigating innate immunity as a tumour suppressor in breast cancer. Eur J Cancer 2016; 72:95-102. [PMID: 28027521 DOI: 10.1016/j.ejca.2016.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Breast cancer cells which express an innate immune signature regulated by interferon regulatory factor 7 (IRF7) have reduced metastatic potential. Infections can induce interferon signalling and may activate an anti-tumour immune response. We investigated whether 'severe infection' can be a clinical surrogate of this phenomenon and/or the presence of high levels of the IRF7 signature at diagnosis before neo-adjuvant chemotherapy (NACT) is associated with a reduced distant relapse risk, specifically in bones. METHODS Clinical data of the European Organisation for Research and Treatment of Cancer 10994/BIG 1-00 phase III trial which randomised 1856 patients treated with NACT between 2001 and 2006, were used. Severe infection was febrile neutropenia or any other grade III-IV infective adverse event during NACT. The IRF7 signature was calculated from gene expression data available for 160 patients on a pre-NACT biopsy. Cox models for distant relapse-free interval (DRFI) investigated the effect of the severe infection and IRF7. Fine and Gray models studied the occurrence of bone metastases as first distant relapse. RESULTS Median follow-up was 4.8 years. No association between severe infection and DFRI was observed in the entire population (n = 1615 eligible patients) hazard ratio [(HR] = 0.99, 90% CI, confidence interval [CI] = 0.81-1.20). For IRF7 (N = 160), a trend towards an association with DRFI was observed (HR = 0.89 for a 50 unit increase, 90% CI = 0.78-1.02, p = 0.081). Higher levels of the IRF7 signature were significantly associated with a decreased bone metastases risk: (HR = 0.76 for a 50 unit increase, 95% CI, 0.62-0.94, p = 0.012). CONCLUSIONS In this study it was shown that severe infection during NACT was not associated with decreased DRFI while high expression of the IRF7 gene signature was significantly associated with reduced bone relapse. This result may be useful for future adjuvant bisphosphonate/denosumab use.
Collapse
Affiliation(s)
- Nathan Touati
- EORTC, Statistics Department, Avenue Emmanuel Mounier 83b11, 1200 Brussels, Belgium.
| | | | - Franco Caramia
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
| | - Hervé Bonnefoi
- Department of Medical Oncology, Institut Bergonié Unicancer, INSERM CIC1401, Bordeaux, France.
| | - David Cameron
- Western General Hospital, Edinburgh Cancer Centre, Crewe Road South, GB Edinburgh EH4 2XU, UK.
| | - Leen Slaets
- EORTC, Statistics Department, Avenue Emmanuel Mounier 83b11, 1200 Brussels, Belgium.
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia.
| | - Sherene Loi
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
46
|
Rudewicz J, Soueidan H, Uricaru R, Bonnefoi H, Iggo R, Bergh J, Nikolski M. MICADo - Looking for Mutations in Targeted PacBio Cancer Data: An Alignment-Free Method. Front Genet 2016; 7:214. [PMID: 28008336 PMCID: PMC5143680 DOI: 10.3389/fgene.2016.00214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022] Open
Abstract
Targeted sequencing is commonly used in clinical application of NGS technology since it enables generation of sufficient sequencing depth in the targeted genes of interest and thus ensures the best possible downstream analysis. This notwithstanding, the accurate discovery and annotation of disease causing mutations remains a challenging problem even in such favorable context. The difficulty is particularly salient in the case of third generation sequencing technology, such as PacBio. We present MICADo, a de Bruijn graph based method, implemented in python, that makes possible to distinguish between patient specific mutations and other alterations for targeted sequencing of a cohort of patients. MICADo analyses NGS reads for each sample within the context of the data of the whole cohort in order to capture the differences between specificities of the sample with respect to the cohort. MICADo is particularly suitable for sequencing data from highly heterogeneous samples, especially when it involves high rates of non-uniform sequencing errors. It was validated on PacBio sequencing datasets from several cohorts of patients. The comparison with two widely used available tools, namely VarScan and GATK, shows that MICADo is more accurate, especially when true mutations have frequencies close to backgound noise. The source code is available at http://github.com/cbib/MICADo.
Collapse
Affiliation(s)
- Justine Rudewicz
- Centre de BioInformatique de Bordeaux, University of BordeauxBordeaux, France; Laboratoire Bordelais de Recherche en Informatique, Centre National de la Recherche Scientifique, University of BordeauxBordeaux, France; Bergonié Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1218, University of BordeauxBordeaux, France
| | - Hayssam Soueidan
- Centre de BioInformatique de Bordeaux, University of BordeauxBordeaux, France; Laboratoire Bordelais de Recherche en Informatique, Centre National de la Recherche Scientifique, University of BordeauxBordeaux, France
| | - Raluca Uricaru
- Centre de BioInformatique de Bordeaux, University of BordeauxBordeaux, France; Laboratoire Bordelais de Recherche en Informatique, Centre National de la Recherche Scientifique, University of BordeauxBordeaux, France
| | - Hervé Bonnefoi
- Bergonié Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1218, University of Bordeaux Bordeaux, France
| | - Richard Iggo
- Bergonié Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1218, University of Bordeaux Bordeaux, France
| | - Jonas Bergh
- Karolinska Institute and University Hospital Stockholm, Sweden
| | - Macha Nikolski
- Centre de BioInformatique de Bordeaux, University of BordeauxBordeaux, France; Laboratoire Bordelais de Recherche en Informatique, Centre National de la Recherche Scientifique, University of BordeauxBordeaux, France
| |
Collapse
|
47
|
Curigliano G, Criscitiello C, Esposito A, Pruneri G. Over-using chemotherapy in the adjuvant setting. Breast 2016; 31:303-308. [PMID: 27866835 DOI: 10.1016/j.breast.2016.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 02/07/2023] Open
Abstract
Avoidance of unnecessary or ineffective treatment should be one of the main goals in adjuvant breast oncology today. Unfortunately, both patients and doctors hunt for tiny statistical differences in survival curves. This search could not only lead to an oncological approach of unlimited addition that we will not be able to afford, but would also end inevitably in indeterminate overtreatment with substantial risks of unexpected toxic effects eating away whatever progress we might make. "Do not harm" remains the main principle in medicine. To be able to follow this rule, we need to better understand the biology of breast cancer. The mistake of "one treatment fits all" can only be changed when we critically review trial designs of adjuvant breast oncology. The risk of overtreatment is there and selection of precisely defined cohorts for phase 3 trials is necessary, despite pressure of scientific ambition, pragmatism, and demands of industry. The "add on" clinical trial design model accepts the inability to confirm that standard therapy is still necessary if a positive result from the addition of the new therapy is obtained. The same model can be applied to "extended" adjuvant treatments in breast cancer subtypes. Addition of "miraculin" to the standard of care should generate a new standard. Such trials that show a modest benefit on average at a population level take us a step away from refining care for the individual, and might support the use of multiple and costly interventions with potential short and long term side effects. It is essential to escalate treatment when necessary and to de-escalate when un-necessary.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- Istituto Europeo di Oncologia, Division of Early Drug Development for Innovative Therapies, Via Ripamonti 435, 20133 Milano, Italy.
| | - Carmen Criscitiello
- Istituto Europeo di Oncologia, Division of Early Drug Development for Innovative Therapies, Via Ripamonti 435, 20133 Milano, Italy
| | - Angela Esposito
- Istituto Europeo di Oncologia, Division of Early Drug Development for Innovative Therapies, Via Ripamonti 435, 20133 Milano, Italy
| | - Giancarlo Pruneri
- Division of Pathology, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy; University of Milan, Breast Cancer Program, Via Ripamonti 435, 20133 Milano, Italy
| |
Collapse
|
48
|
Bozovic-Spasojevic I, Zardavas D, Brohée S, Ameye L, Fumagalli D, Ades F, de Azambuja E, Bareche Y, Piccart M, Paesmans M, Sotiriou C. The Prognostic Role of Androgen Receptor in Patients with Early-Stage Breast Cancer: A Meta-analysis of Clinical and Gene Expression Data. Clin Cancer Res 2016; 23:2702-2712. [PMID: 28151718 DOI: 10.1158/1078-0432.ccr-16-0979] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 10/03/2016] [Accepted: 10/27/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Androgen receptor (AR) expression has been observed in about 70% of patients with breast cancer, but its prognostic role remains uncertain.Experimental Design: To assess the prognostic role of AR expression in early-stage breast cancer, we performed a meta-analysis of studies that evaluated the impact of AR at the protein and gene expression level on disease-free survival (DFS) and/or overall survival (OS). Eligible studies were identified by systematic review of electronic databases using the MeSH-terms "breast neoplasm" and "androgen receptor" and were selected after a qualitative assessment based on the REMARK criteria. A pooled gene expression analysis of 35 publicly available microarray data sets was also performed from patients with early-stage breast cancer with available gene expression and clinical outcome data.Results: Twenty-two of 33 eligible studies for the clinical meta-analysis, including 10,004 patients, were considered as evaluable for the current study after the qualitative assessment. AR positivity defined by IHC was associated with improved DFS in all patients with breast cancer [multivariate (M) analysis, HR 0.46; 95% confidence interval (CI) 0.37-0.58, P < 0.001] and better OS [M-HR 0.53; 95% CI, 0.38-0.73, P < 0.001]. Thirty-five datasets including 7,220 patients were eligible for the pooled gene expression analysis. High AR mRNA levels were found to confer positive prognosis overall in terms of DFS (HR 0.82; 95% CI 0.72-0.92;P = 0.0007) and OS (HR 0.84; 95% CI, 0.75-0.94; P = 0.02) only in univariate analysis.Conclusions: Our analysis, conducted among more than 17,000 women with early-stage breast cancer included in clinical and gene expression analysis, demonstrates that AR positivity is associated with favorable clinical outcome. Clin Cancer Res; 23(11); 2702-12. ©2016 AACR.
Collapse
Affiliation(s)
- Ivana Bozovic-Spasojevic
- Breast Data Centre, Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium.,Institute for Oncology and Radiology of Serbia, National Cancer Research Centre, Belgrade, Republic of Serbia
| | | | - Sylvain Brohée
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium
| | - Lieveke Ameye
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Felipe Ades
- Breast Data Centre, Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium.,Hospital Albert Einstein, São Paulo, Brazil
| | - Evandro de Azambuja
- Breast Data Centre, Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium
| | - Yacine Bareche
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium
| | - Martine Piccart
- Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Marianne Paesmans
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brusells, Belgium.
| |
Collapse
|
49
|
Hainaut P, Pfeifer GP. Somatic TP53 Mutations in the Era of Genome Sequencing. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026179. [PMID: 27503997 DOI: 10.1101/cshperspect.a026179] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amid the complexity of genetic alterations in human cancer, TP53 mutation appears as an almost invariant component, representing by far the most frequent genetic alteration overall. Compared with previous targeted sequencing studies, recent integrated genomics studies offer a less biased view of TP53 mutation patterns, revealing that >20% of mutations occur outside the DNA-binding domain. Among the 12 mutations representing each at least 1% of all mutations, five occur at residues directly involved in specific DNA binding, four affect the tertiary fold of the DNA-binding domain, and three are nonsense mutations, two of them in the carboxyl terminus. Significant mutations also occur in introns, affecting alternative splicing events or generating rearrangements (e.g., in intron 1 in sporadic osteosarcoma). In aggressive cancers, mutation is so common that it may not have prognostic value (all these cancers have impaired p53 function caused by mutation or by other mechanisms). In several other cancers, however, mutation makes a clear difference for prognostication, as, for example, in HER2-enriched breast cancers and in lung adenocarcinoma with EGFR mutations. Thus, the clinical significance of TP53 mutation is dependent on tumor subtype and context. Understanding the clinical impact of mutation will require integrating mutation-specific information (type, frequency, and predicted impact) with data on haplotypes and on loss of heterozygosity.
Collapse
Affiliation(s)
- Pierre Hainaut
- University Grenoble Alpes, Institut Albert Bonniot, Institut National de la Santé et de la Recherche Médicale (INSERM), 823 Grenoble, France
| | - Gerd P Pfeifer
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan 49503
| |
Collapse
|
50
|
Santarpia L, Bottai G, Kelly CM, Győrffy B, Székely B, Pusztai L. Deciphering and Targeting Oncogenic Mutations and Pathways in Breast Cancer. Oncologist 2016; 21:1063-78. [PMID: 27384237 PMCID: PMC5016060 DOI: 10.1634/theoncologist.2015-0369] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/16/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED : Advances in DNA and RNA sequencing revealed substantially greater genomic complexity in breast cancer than simple models of a few driver mutations would suggest. Only very few, recurrent mutations or copy-number variations in cancer-causing genes have been identified. The two most common alterations in breast cancer are TP53 (affecting the majority of triple-negative breast cancers) and PIK3CA (affecting almost half of estrogen receptor-positive cancers) mutations, followed by a long tail of individually rare mutations affecting <1%-20% of cases. Each cancer harbors from a few dozen to a few hundred potentially high-functional impact somatic variants, along with a much larger number of potentially high-functional impact germline variants. It is likely that it is the combined effect of all genomic variations that drives the clinical behavior of a given cancer. Furthermore, entirely new classes of oncogenic events are being discovered in the noncoding areas of the genome and in noncoding RNA species driven by errors in RNA editing. In light of this complexity, it is not unexpected that, with the exception of HER2 amplification, no robust molecular predictors of benefit from targeted therapies have been identified. In this review, we summarize the current genomic portrait of breast cancer, focusing on genetic aberrations that are actively being targeted with investigational drugs. IMPLICATIONS FOR PRACTICE Next-generation sequencing is now widely available in the clinic, but interpretation of the results is challenging, and its impact on treatment selection is often limited. This work provides an overview of frequently encountered molecular abnormalities in breast cancer and discusses their potential therapeutic implications. This review emphasizes the importance of administering investigational targeted therapies, or off-label use of approved targeted drugs, in the context of a formal clinical trial or registry programs to facilitate learning about the clinical utility of tumor target profiling.
Collapse
Affiliation(s)
- Libero Santarpia
- Oncology Experimental Therapeutics, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Institute, Milan, Italy
| | - Giulia Bottai
- Oncology Experimental Therapeutics, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Clinical and Research Institute, Milan, Italy
| | | | - Balázs Győrffy
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Borbala Székely
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Lajos Pusztai
- Yale Cancer Center, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|