1
|
Blondeaux E, Arecco L, Punie K, Graffeo R, Toss A, De Angelis C, Trevisan L, Buzzatti G, Linn SC, Dubsky P, Cruellas M, Partridge AH, Balmaña J, Paluch-Shimon S, Lambertini M. Germline TP53 pathogenic variants and breast cancer: A narrative review. Cancer Treat Rev 2023; 114:102522. [PMID: 36739824 DOI: 10.1016/j.ctrv.2023.102522] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/02/2023]
Abstract
Approximately 10% of breast cancers are associated with the inheritance of a pathogenic variant (PV) in one of the breast cancer susceptibility genes. Multiple breast cancer predisposing genes, including TP53, are responsible for the increased breast cancer risk. Tumor protein-53 (TP53) germline PVs are associated with Li-Fraumeni syndrome, a rare autosomal dominant inherited cancer predisposition syndrome associated with early-onset pediatric and multiple primary cancers such as soft tissue and bone sarcomas, breast cancer, brain tumors, adrenocortical carcinomas and leukemias. Women harboring a TP53 PV carry a lifetime risk of developing breast cancer of 80-90%. The aim of the present narrative review is to provide a comprehensive overview of the criteria for offering TP53 testing, prevalence of TP53 carriers among patients with breast cancer, and what is known about its prognostic and therapeutic implications. A summary of the current indications of secondary cancer surveillance and survivorship issues are also provided. Finally, the spectrum of TP53 alteration and testing is discussed. The optimal strategies for the treatment of breast cancer in patients harboring TP53 PVs poses certain challenges. Current guidelines favor the option of performing mastectomy rather than lumpectomy to avoid adjuvant radiotherapy and subsequent risk of radiation-induced second primary malignancies, with careful consideration of radiation when indicated post-mastectomy. Some studies suggest that patients with breast cancer and germline TP53 PV might have worse survival outcomes compared to patients with breast cancer and wild type germline TP53 status. Annual breast magnetic resonance imaging (MRI) and whole-body MRI are recommended as secondary prevention.
Collapse
Affiliation(s)
- Eva Blondeaux
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Kevin Punie
- Department of General Medical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Rossella Graffeo
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Angela Toss
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Lucia Trevisan
- Hereditary Cancer Unit, Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giulia Buzzatti
- Hereditary Cancer Unit, Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sabine C Linn
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter Dubsky
- Breast Centre, Hirslanden Klinik St Anna, Luzern, Switzerland
| | - Mara Cruellas
- Department of Medical Oncology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Judith Balmaña
- Department of Medical Oncology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Shani Paluch-Shimon
- Breast Cancer Unit, Sharett Institute of Oncology, Hadassah Medical Center & Faculty of Medicine, Hebrew University, 91120 Jerusalem, Israel
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
2
|
Yao J, Chen Q, Zhu J, Cai R. Targeted gene next‐generation sequencing reveals genomic profile in a cohort of 46 Chinese patients with breast cancer. J Gene Med 2022; 24:e3420. [PMID: 35470535 DOI: 10.1002/jgm.3420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jie Yao
- Department of Medical Oncology, Fu Xing Hospital Capital Medical University Beijing China
| | | | | | - Rui‐gang Cai
- 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 China
| |
Collapse
|
3
|
Significant impact of circulating tumour DNA mutations on survival in metastatic breast cancer patients. Sci Rep 2021; 11:6761. [PMID: 33762647 PMCID: PMC7990915 DOI: 10.1038/s41598-021-86238-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Mutational analysis of circulating tumour (ct) DNA holds promise as an effective tool to predict the course of metastatic breast cancer (MBC). In the present study we used targeted next generation sequencing of ctDNA to evaluate the impact of cancer driven mutations on the prognosis of MBC. The study included 59 oestrogen receptor-positive (ER+), HER2-negative MBC patients. Sequencing analysis was performed in ESR1, PIK3CA, ERBB2, PTEN, TP53, KRAS, HRAS, NRAS, and AR. At baseline, patients started receiving either chemotherapy (34%; n = 20) or cyclin-dependent kinase 4/6 inhibitor therapy in combination with endocrine therapy (CDK4/6i+ET; 66%; n = 39). Overall, 64.4% (n = 38) of the patients carried at least one pathogenic or likely-pathogenic mutation. Number of ctDNA mutations was significantly linked with worse progression free survival (PFS; p = 0.003) and overall survival (OS; p = 0.007). Furthermore, ctDNA load, defined by the number of mutant ctDNA molecules per mL plasma, significantly correlated with PFS (p < 0.001) and OS (p = 0.001). Furthermore, mutational status of ESR1 and TP53 significantly predicted PFS (p = 0.024 and p = 0.035, respectively) and OS (p < 0.001 and p = 0.035, respectively). These results emphasizes the clinical value of ctDNA mutational analysis in the management of advanced breast cancer.
Collapse
|
4
|
Shahbandi A, Nguyen HD, Jackson JG. TP53 Mutations and Outcomes in Breast Cancer: Reading beyond the Headlines. Trends Cancer 2020; 6:98-110. [PMID: 32061310 PMCID: PMC7931175 DOI: 10.1016/j.trecan.2020.01.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/11/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022]
Abstract
TP53 is the most frequently mutated gene in breast cancer, but its role in survival is confounded by different studies concluding that TP53 mutations are associated with negative, neutral, or positive outcomes. Closer examination showed that many studies were limited by factors such as imprecise methods to detect TP53 mutations and small cohorts that combined patients treated with drugs having very different mechanisms of action. When only studies of patients receiving the same treatment(s) were compared, they tended to agree. These analyses reveal a role for TP53 in response to different treatments as complex as its different biological activities. We discuss studies that have assessed the role of TP53 mutations in breast cancer treatment and limitations in interpreting reported results.
Collapse
Affiliation(s)
- Ashkan Shahbandi
- Tulane School of Medicine, Department of Biochemistry and Molecular Biology, 1430 Tulane Avenue #8543, New Orleans, LA 70112, USA
| | - Hoang D Nguyen
- Tulane School of Medicine, Department of Biochemistry and Molecular Biology, 1430 Tulane Avenue #8543, New Orleans, LA 70112, USA
| | - James G Jackson
- Tulane School of Medicine, Department of Biochemistry and Molecular Biology, 1430 Tulane Avenue #8543, New Orleans, LA 70112, USA.
| |
Collapse
|
5
|
Alvarado-Ortiz E, de la Cruz-López KG, Becerril-Rico J, Sarabia-Sánchez MA, Ortiz-Sánchez E, García-Carrancá A. Mutant p53 Gain-of-Function: Role in Cancer Development, Progression, and Therapeutic Approaches. Front Cell Dev Biol 2020; 8:607670. [PMID: 33644030 PMCID: PMC7905058 DOI: 10.3389/fcell.2020.607670] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 02/05/2023] Open
Abstract
Frequent p53 mutations (mutp53) not only abolish tumor suppressor capacities but confer various gain-of-function (GOF) activities that impacts molecules and pathways now regarded as central for tumor development and progression. Although the complete impact of GOF is still far from being fully understood, the effects on proliferation, migration, metabolic reprogramming, and immune evasion, among others, certainly constitute major driving forces for human tumors harboring them. In this review we discuss major molecular mechanisms driven by mutp53 GOF. We present novel mechanistic insights on their effects over key functional molecules and processes involved in cancer. We analyze new mechanistic insights impacting processes such as immune system evasion, metabolic reprogramming, and stemness. In particular, the increased lipogenic activity through the mevalonate pathway (MVA) and the alteration of metabolic homeostasis due to interactions between mutp53 and AMP-activated protein kinase (AMPK) and Sterol regulatory element-binding protein 1 (SREBP1) that impact anabolic pathways and favor metabolic reprograming. We address, in detail, the impact of mutp53 over metabolic reprogramming and the Warburg effect observed in cancer cells as a consequence, not only of loss-of-function of p53, but rather as an effect of GOF that is crucial for the imbalance between glycolysis and oxidative phosphorylation. Additionally, transcriptional activation of new targets, resulting from interaction of mutp53 with NF-kB, HIF-1α, or SREBP1, are presented and discussed. Finally, we discuss perspectives for targeting molecules and pathways involved in chemo-resistance of tumor cells resulting from mutp53 GOF. We discuss and stress the fact that the status of p53 currently constitutes one of the most relevant criteria to understand the role of autophagy as a survival mechanism in cancer, and propose new therapeutic approaches that could promote the reduction of GOF effects exercised by mutp53 in cancer.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Karen Griselda de la Cruz-López
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jared Becerril-Rico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sánchez
- Programa de Posgrado en Ciencias Bioquímicas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Alejandro García-Carrancá
- Laboratorio de Virus and Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- *Correspondence: Alejandro García-Carrancá
| |
Collapse
|
6
|
Li JP, Zhang XM, Zhang Z, Zheng LH, Jindal S, Liu YJ. Association of p53 expression with poor prognosis in patients with triple-negative breast invasive ductal carcinoma. Medicine (Baltimore) 2019; 98:e15449. [PMID: 31045815 PMCID: PMC6504250 DOI: 10.1097/md.0000000000015449] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/24/2019] [Accepted: 04/05/2019] [Indexed: 01/06/2023] Open
Abstract
TP53 gene is mutated in approximately 80% of triple-negative breast cancer (TNBC). However, the prognostic significance of immunohistochemical (IHC)-detected p53 protein expression remains controversial in TNBC. In this study, we retrospectively analyzed the association between IHC-detected p53 expression and the prognosis in a cohort of 278 patients with stage I-III triple-negative breast invasive ductal carcinoma (IDC), who received surgery at the department of breast surgery in the Fourth Hospital of Hebei Medical University from 2010-01 to 2012-12. We found a positive expression ratio of IHC-detected p53 in triple-negative breast IDC of 58.6% (163/278). Furthermore, levels of expression were significantly associated with vessel tumor emboli and higher histologic grade (P = .038, P = .043, respectively), with the highest expression level observed in G3 breast cancer (64.7%). Additionally, Kaplan-Meier analysis showed that p53 expression indicated worse overall survival (OS) in the whole cohort (79.6% vs 89.6%, Log-rank test P = .025) as well as in stratified prognostic stage II patients (90.8% vs 100%, Log-rank test P = .027). The mortality risk of p53 expression patients was 2.22 times higher than that of p53 negative patients (HR: 2.222; 95%CI: 1.147-4.308). In addition, p53 expression was also associated with poor disease-free survival (DFS) (76.7% vs 86.8%, P = .020). Cox proportional hazard ratio model showed p53 expression was an independent risk factor for OS (P = .018) and DFS (P = .018) after controlling for tumor size, lymph node status, and vessel tumor emboli. Altogether, our data showed that IHC-detected p53 expression is a promising prognostic candidate for poor survival in triple-negative breast IDC patients. However, more studies are needed to determine if p53 may be applied to clinical practice as a biomarker and/or novel therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jing-ping Li
- Department of Breast Surgery, the Fourth Hospital of Hebei Medical University
| | - Xiang-mei Zhang
- Research Center, the Fourth Hospital of Hebei Medical University
| | - Zhenzhen Zhang
- Division of Hematology & Medical Oncology, Oregon Health & Science University
| | - Li-hua Zheng
- Department of Vascular Surgery, the First Hospital of Hebei Medical University
| | - Sonali Jindal
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Shijiazhuang, Hebei, China
| | - Yun-jiang Liu
- Department of Breast Surgery, the Fourth Hospital of Hebei Medical University
| |
Collapse
|
7
|
An IRAK1-PIN1 signalling axis drives intrinsic tumour resistance to radiation therapy. Nat Cell Biol 2019; 21:203-213. [PMID: 30664786 PMCID: PMC6428421 DOI: 10.1038/s41556-018-0260-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/07/2018] [Indexed: 11/30/2022]
Abstract
Drug-based strategies to overcome tumour resistance to radiotherapy (R-RT) remain limited by the single-agent toxicity of traditional radiosensitizers (e.g., platinums) and a lack of targeted alternatives. In a screen for compounds that restore radiosensitivity in p53 mutant zebrafish while tolerated in non-irradiated wild-type animals, we identified the benzimidazole anthelmintic, oxfendazole. Surprisingly, oxfendazole acts via inhibition of IRAK1, a kinase otherwise involved in Interleukin-1 and Toll-like receptor (IL-1R/TLR) immune responses. IRAK1 drives R-RT in a pathway involving IRAK4 and TRAF6 but not the IL-1R/TLR—IRAK adaptor MyD88. Rather than stimulating NF-κB, radiation-activated IRAK1 acts to prevent apoptosis mediated by the PIDDosome complex (PIDD/RAIDD/caspase-2). Countering this pathway with IRAK1 inhibitors suppresses R-RT in tumour models derived from cancers in which TP53 mutations predict R-RT. Lastly, IRAK1 inhibitors synergize with inhibitors of PIN1, a prolyl isomerase essential for IRAK1 activation in response to pathogens and, as shown here, ionizing radiation. These data identify an IRAK1 radiation-response pathway as a rational chemo-RT target.
Collapse
|
8
|
Hu ZY, Xie N, Tian C, Yang X, Liu L, Li J, Xiao H, Wu H, Lu J, Gao J, Hu X, Cao M, Shui Z, Xiao M, Tang Y, He Q, Chang L, Xia X, Yi X, Liao Q, Ouyang Q. Identifying Circulating Tumor DNA Mutation Profiles in Metastatic Breast Cancer Patients with Multiline Resistance. EBioMedicine 2018; 32:111-118. [PMID: 29807833 PMCID: PMC6020712 DOI: 10.1016/j.ebiom.2018.05.015] [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: 03/07/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 12/27/2022] Open
Abstract
Purpose In cancer patients, tumor gene mutations contribute to drug resistance and treatment failure. In patients with metastatic breast cancer (MBC), these mutations increase after multiline treatment, thereby decreasing treatment efficiency. The aim of this study was to evaluate gene mutation patterns in MBC patients to predict drug resistance and disease progression. Method A total of 68 MBC patients who had received multiline treatment were recruited. Circulating tumor DNA (ctDNA) mutations were evaluated and compared among hormone receptor (HR)/human epidermal growth factor receptor 2 (HER2) subgroups. Results The baseline gene mutation pattern (at the time of recruitment) varied among HR/HER2 subtypes. BRCA1 and MED12 were frequently mutated in triple negative breast cancer (TNBC) patients, PIK3CA and FAT1 mutations were frequent in HR+ patients, and PIK3CA and ERBB2 mutations were frequent in HER2+ patients. Gene mutation patterns also varied in patients who progressed within either 3 months or 3–6 months of chemotherapy treatment. For example, in HR+ patients who progressed within 3 months of treatment, the frequency of TERT mutations significantly increased. Other related mutations included FAT1 and NOTCH4. In HR+ patients who progressed within 3–6 months, PIK3CA, TP53, MLL3, ERBB2, NOTCH2, and ERS1 were the candidate mutations. This suggests that different mechanisms underlie disease progression at different times after treatment initiation. In the COX model, the ctDNA TP53 + PIK3CA gene mutation pattern successfully predicted progression within 6 months. Conclusion ctDNA gene mutation profiles differed among HR/HER2 subtypes of MBC patients. By identifying mutations associated with treatment resistance, we hope to improve therapy selection for MBC patients who received multiline treatment. Doctors felt difficult to design effective regimen for MBC patients after multi-line treatment. ctDNA testing provide potential treatment targets and reflect treatment response of tumors. ctDNA gene mutation pattern varies among four HR/HER2 subgroups. The gene mutation patterns also varied between resistant patients and sensitive patients. In COX model, ctDNA gene mutation pattern could successfully predict progression within 6 months.
In this study, we clarified the baseline ctDNA mutation pattern for metastatic breast cancer patients. We also selected out treatment-resistance related mutations by ctDNA testing. Here, we showed that PIK3CA were significantly related to HR+. Moreover, in this study, we also showed a plenty of other rare mutations, including DDR2, CDK12, etc. For different HR/HER2 subtypes, MED12 was frequent in TNBC samples, FAT1 was frequent in HR+ samples, and DDR2 was frequent in HER2+ samples. These findings need further intensive investigations in larger samples.
Collapse
Affiliation(s)
- Zhe-Yu Hu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Central Laboratory, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Ning Xie
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Can Tian
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Xiaohong Yang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Liping Liu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Jing Li
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Huawu Xiao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Hui Wu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Jun Lu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Jianxiang Gao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Xuming Hu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Min Cao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Zhengrong Shui
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Mengjia Xiao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Yu Tang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Qiongzhi He
- Geneplus-Beijing Institute, Beijing 102206, China
| | | | - Xuefeng Xia
- Geneplus-Beijing Institute, Beijing 102206, China
| | - Xin Yi
- Geneplus-Beijing Institute, Beijing 102206, China
| | - Qianjin Liao
- Geneplus-Beijing Institute, Beijing 102206, China
| | - Quchang Ouyang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha 410013, China; Department of Breast Cancer Medical Oncology, The Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China.
| |
Collapse
|
9
|
Meric-Bernstam F, Zheng X, Shariati M, Damodaran S, Wathoo C, Brusco L, Demirhan ME, Tapia C, Eterovic AK, Basho RK, Ueno NT, Janku F, Sahin A, Rodon J, Broaddus R, Kim TB, Mendelsohn J, Mills Shaw KR, Tripathy D, Mills GB, Chen K. Survival Outcomes by TP53 Mutation Status in Metastatic Breast Cancer. JCO Precis Oncol 2018; 2018. [PMID: 30035249 DOI: 10.1200/po.17.00245] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose We sought to determine the significant genomic alterations in patients with metastatic breast cancer (MBC), and survival outcomes in common genotypes. Patients and Methods High-depth next generation sequencing was performed for 202 genes in tumor and normal DNA from 257 patients with MBC, including 165 patients with ER/PR+ HER2- (hormone receptor positive, HR+ positive), 32 patients with HER2+ and 60 patients with triple negative (ER/PR/HER2-) cancer. Kaplan Meier survival analysis was performed in our discovery set, in breast cancer patients analyzed in The Cancer Genome Atlas, and in a separate cohort of 98 patients with MBC who underwent clinical genomic testing. Results Significantly mutated genes (SMGs) varied by histology and tumor subtype, but TP53 was a SMG in all three subtypes. The most SMGs in HR+ patients included PIK3CA (32%), TP53 (29%), GATA3 (15%), CDH1 (8%), MAP3K1 (8%), PTEN (5%), TGFBR2 (4%), AKT1 (4%), and MAP2K4 (4%). TP53 mutations were associated with shorter recurrence-free survival (P=0.004), progression-free survival (P=0.00057) and overall survival (P=0.003). Further, TP53 status was prognostic among HR+ patients with PIK3CA mutations. TP53 mutations were also associated with poorer overall survival in the 442 HR+ breast cancer patients in the TCGA (P=0.042) and in an independent set of 96 HR+ MBC who underwent clinical sequencing (P=0.0004). Conclusions SMGs differ by tumor subtype but TP53 is significantly mutated in all three breast cancer subtypes. TP53 mutations are associated with poor prognosis in HR+ breast cancer. TP53 mutations should be considered in the design and interpretation of precision oncology trials.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xiaofeng Zheng
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Maryam Shariati
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Senthil Damodaran
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Chetna Wathoo
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Lauren Brusco
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Mehmet Esat Demirhan
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Coya Tapia
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Agda Karina Eterovic
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Reva K Basho
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX 77030.,current address: Cedars-Sinai, Los Angeles, CA 90048
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Aysegul Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Jordi Rodon
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Russell Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Tae-Beom Kim
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - John Mendelsohn
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Kenna R Mills Shaw
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Gordon B Mills
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Ken Chen
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
10
|
Fujii T, Matsuda N, Kono M, Harano K, Chen H, Luthra R, Roy-Chowdhuri S, Sahin AA, Wathoo C, Joon AY, Tripathy D, Meric-Bernstam F, Ueno NT. Prior systemic treatment increased the incidence of somatic mutations in metastatic breast cancer. Eur J Cancer 2017; 89:64-71. [PMID: 29232568 DOI: 10.1016/j.ejca.2017.11.015] [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: 07/30/2017] [Revised: 11/01/2017] [Accepted: 11/10/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Understanding the biology of breast cancer is important for guiding treatment strategies and revealing resistance mechanisms. Our objectives were to investigate the relationship between previous systemic therapy exposure and mutational spectrum in metastatic breast cancer and to identify clinicopathological factors associated with identified frequent somatic mutations. METHODS Archival tissues of patients with metastatic breast cancer were subjected to hotspot molecular testing by next-generation sequencing. The variables that significantly differed (P < 0.05) in univariate analysis were selected to fit multivariate models. Logistic models were fit to estimate the association between mutation status and clinical variables of interest. Five-fold cross-validation was performed to estimate the prediction error of each model. RESULTS A total of 922 patients were included in the analysis. In multivariate analysis, previous systemic treatment before molecular testing (N = 186) was associated with a significantly higher rate of TP53 and PIK3CA mutations compared with the lack of systemic treatment (P < 0.001 for both). CONCLUSION Systemic treatment exposure is an independent risk factor for high rates of TP53 and PIK3CA mutation, which suggests the importance of testing samples after systemic therapy to accurately assess mutations. It is worth testing the gene profile when tumours become resistant to systemic treatments.
Collapse
Affiliation(s)
- Takeo Fujii
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Naoko Matsuda
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Miho Kono
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Kenichi Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rajyalakshmi Luthra
- Molecular Diagnostic Laboratory, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Chetna Wathoo
- Sheikh Khalifa Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Aron Y Joon
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Funda Meric-Bernstam
- Sheikh Khalifa Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Department of Investigational Cancer Therapeutics (Phase I Trials Department), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Yndestad S, Austreid E, Knappskog S, Chrisanthar R, Lilleng PK, Lønning PE, Eikesdal HP. High PTEN gene expression is a negative prognostic marker in human primary breast cancers with preserved p53 function. Breast Cancer Res Treat 2017; 163:177-190. [PMID: 28213783 PMCID: PMC5387035 DOI: 10.1007/s10549-017-4160-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 02/13/2017] [Indexed: 12/22/2022]
Abstract
Purpose PTEN is an important tumor suppressor in breast cancer. Here, we examined the prognostic and predictive value of PTEN and PTEN pseudogene (PTENP1) gene expression in patients with locally advanced breast cancer given neoadjuvant chemotherapy. Methods The association between pretreatment PTEN and PTENP1 gene expression, response to neoadjuvant chemotherapy, and recurrence-free and disease-specific survival was assessed in 364 patients with locally advanced breast cancer given doxorubicin, 5-fluorouracil/mitomycin, or epirubicin versus paclitaxel in three phase II prospective studies. Further, protein expression of PTEN or phosphorylated Akt, S6 kinase, and 4EBP1 was assessed in a subgroup of 187 tumors. Results Neither PTEN nor PTENP1 gene expression level predicted response to any of the chemotherapy regimens tested (n = 317). Among patients without distant metastases (n = 282), a high pretreatment PTEN mRNA level was associated with inferior relapse-free (RFS; p = 0.001) and disease-specific survival (DSS; p = 0.003). Notably, this association was limited to patients harboring TP53 wild-type tumors (RFS; p = 0.003, DSS; p = 0.009). PTEN mRNA correlated significantly with PTENP1 mRNA levels (rs = 0.456, p < 0.0001) and PTEN protein staining (rs = 0.163, p = 0.036). However, no correlation between PTEN, phosphorylated Akt, S6 kinase or 4EBP1 protein staining, and survival was recorded. Similarly, no correlation between PTENP1 gene expression and survival outcome was observed. Conclusion High intratumoral PTEN gene expression was associated with poor prognosis in patients with locally advanced breast cancers harboring wild-type TP53. Electronic supplementary material The online version of this article (doi:10.1007/s10549-017-4160-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Synnøve Yndestad
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Eilin Austreid
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Ranjan Chrisanthar
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Section of Molecular Pathology, Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Peer Kåre Lilleng
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Hans Petter Eikesdal
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway. .,Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
12
|
Kim J, de Sampaio PC, Lundy DM, Peng Q, Evans KW, Sugimoto H, Gagea M, Kienast Y, Amaral NSD, Rocha RM, Eikesdal HP, Lønning PE, Meric-Bernstam F, LeBleu VS. Heterogeneous perivascular cell coverage affects breast cancer metastasis and response to chemotherapy. JCI Insight 2016; 1:e90733. [PMID: 28018977 PMCID: PMC5161212 DOI: 10.1172/jci.insight.90733] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Angiogenesis and co-optive vascular remodeling are prerequisites of solid tumor growth. Vascular heterogeneity, notably perivascular composition, may play a critical role in determining the rate of cancer progression. The contribution of vascular pericyte heterogeneity to cancer progression and therapy response is unknown. Here, we show that angiopoietin-2 (Ang2) orchestrates pericyte heterogeneity in breast cancer with an effect on metastatic disease and response to chemotherapy. Using multispectral imaging of human breast tumor specimens, we report that perivascular composition, as defined by the ratio of PDGFRβ- and desmin+ pericytes, provides information about the response to epirubicin but not paclitaxel. Using 17 distinct patient-derived breast cancer xenografts, we demonstrate a cancer cell-derived influence on stromal Ang2 production and a cancer cell-defined control over tumor vasculature and perivascular heterogeneity. The aggressive features of tumors and their distinct response to therapies may thus emerge by the cancer cell-defined engagement of distinct and heterogeneous angiogenic programs.
Collapse
Affiliation(s)
| | | | | | | | - Kurt W Evans
- Department of Investigational Cancer Therapeutics, and
| | | | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yvonne Kienast
- Discovery Oncology, Roche Pharmaceutical Research and Early Development, (pRED), Roche Innovation Center, Munich, Germany
| | | | - Rafael Malagoli Rocha
- Molecular Gynecology Laboratory, Gynecology Department, Federal University of São Paulo, Brazil
| | - Hans Petter Eikesdal
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, and.,Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
13
|
Kim JY, Park K, Jung HH, Lee E, Cho EY, Lee KH, Bae SY, Lee SK, Kim SW, Lee JE, Nam SJ, Ahn JS, Im YH, Park YH. Association between Mutation and Expression of TP53 as a Potential Prognostic Marker of Triple-Negative Breast Cancer. Cancer Res Treat 2016; 48:1338-1350. [PMID: 26910472 PMCID: PMC5080805 DOI: 10.4143/crt.2015.430] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/28/2016] [Indexed: 12/21/2022] Open
Abstract
Purpose TP53, the most frequently mutated gene in breast cancer, is more frequently altered in HER2-enriched and basal-like breast cancer. However, no studies have clarified the role of TP53 status as a prognostic and predictive marker of triple-negative breast cancer (TNBC). Materials and Methods We performed p53 immunohistochemistry (IHC), nCounter mRNA expression assay, and DNA sequencing to determine the relationship between TP53 alteration and clinical outcomes of TNBC patients. Results Seventy-seven of 174 TNBC patients were found to harbor a TP53 mutation. Patients with missense mutations showed high protein expression in contrast to patients with deletion mutations (positivity of IHC: wild type vs. missense vs. deletion mutation, 53.6% vs. 89.8% vs. 25.0%, respectively; p < 0.001). TP53 mRNA expression was influenced by mutation status (mRNA expression [median]: wild type vs. missense vs. deletion mutation, 207.36± 132.73 vs. 339.61±143.21 vs. 99.53±99.57, respectively; p < 0.001). According to survival analysis, neither class of mutation nor protein or mRNA expression status had any impact on patient prognosis. In subgroup analysis, low mRNA expression was associated with poor prognosis in patients with a TP53 missense mutation (5-year distant recurrence-free survival [5Y DRFS]: low vs. high, 50.0% vs. 87.8%; p=0.009), while high mRNA expression with a TP53 deletion mutation indicated poor prognosis (5Y DRFS: low vs. high, 91.7% vs. 75.0%; p=0.316). Conclusion Association between TP53 mutation and expression indicates a potential prognostic marker of TNBC; hence both DNA sequencing and mRNA expression analysis may be required to predict the prognosis of TNBC patients.
Collapse
Affiliation(s)
- Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyunghee Park
- Samsung Genome Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae Hyun Jung
- Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eunjin Lee
- Samsung Genome Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Yoon Cho
- Cancer of Companion Diagnostics, Innovative Cancer Medicine Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kwang Hee Lee
- Life Science Solutions Group, Thermo Fisher Scientific Corporation, Seoul, Korea
| | - Soo Youn Bae
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Kyung Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Nam
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Hyuck Im
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea.,Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea.,Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Wang Y, Xu Y, Chen J, Ouyang T, Li J, Wang T, Fan Z, Fan T, Lin B, Xie Y. TP53 mutations are associated with higher rates of pathologic complete response to anthracycline/cyclophosphamide-based neoadjuvant chemotherapy in operable primary breast cancer. Int J Cancer 2015; 138:489-96. [PMID: 26238069 DOI: 10.1002/ijc.29715] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 07/01/2015] [Accepted: 07/21/2015] [Indexed: 12/20/2022]
Abstract
The role of TP53 mutations in predicting response to neoadjuvant chemotherapy in breast cancer remains controversial. The aims of this study were to investigate whether TP53 mutations were associated with response and survival in breast cancer patients who received neoadjuvant chemotherapy. Therefore, we identified TP53 mutations in the core-needle biopsy tumor samples obtained before the neoadjuvant chemotherapy from 351 operable primary breast cancer patients who either received anthracycline/cyclophosphamide-based (n = 252) or paclitaxel (n = 99) neoadjuvant chemotherapy. We found that 41.0% (144 of 351) of patients harbored TP53 mutations, and 14.8% of patients achieved a pCR (pathologic complete response) after neoadjuvant chemotherapy. Among patients treated with anthracycline/cyclophosphamide (n = 252), patients with TP53 mutations had a significantly higher pCR rate than those with wild-type (28.6 vs.7.1%; p < 0.001), and TP53 mutation was an independent favorable predictor of pCR [odds ratio (OR) = 3.41; 95% confidence interval (CI) 1.50-7.77; p = 0.003] in this group; moreover, patients with TP53 mutation had a better distant recurrence-free survival (DRFS) than those with wild-type [unadjusted hazard ratio (HR) = 0.43; 95% CI 0.20-0.94; p = 0.030] in this group. Among patients treated with paclitaxel (n = 99), no significant difference in pCR rates was observed between patients with or without TP53 mutations (15.2 vs. 11.3%; p = 0.57). Our results suggested that patients with TP53 mutations are more likely to respond to anthracycline/ cyclophosphamide-based neoadjuvant chemotherapy and have a favorable survival.
Collapse
Affiliation(s)
- Yuxia Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ye Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jiuan Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Tao Ouyang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jinfeng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Tianfeng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhaoqing Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Tie Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Benyao Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yuntao Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|