1
|
Finkelman BS, Zhang H, Hicks DG, Turner BM. The Evolution of Ki-67 and Breast Carcinoma: Past Observations, Present Directions, and Future Considerations. Cancers (Basel) 2023; 15:808. [PMID: 36765765 PMCID: PMC9913317 DOI: 10.3390/cancers15030808] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The 1983 discovery of a mouse monoclonal antibody-the Ki-67 antibody-that recognized a nuclear antigen present only in proliferating cells represented a seminal discovery for the pathologic assessment of cellular proliferation in breast cancer and other solid tumors. Cellular proliferation is a central determinant of prognosis and response to cytotoxic chemotherapy in patients with breast cancer, and since the discovery of the Ki-67 antibody, Ki-67 has evolved as an important biomarker with both prognostic and predictive potential in breast cancer. Although there is universal recognition among the international guideline recommendations of the value of Ki-67 in breast cancer, recommendations for the actual use of Ki-67 assays in the prognostic and predictive evaluation of breast cancer remain mixed, primarily due to the lack of assay standardization and inconsistent inter-observer and inter-laboratory reproducibility. The treatment of high-risk ER-positive/human epidermal growth factor receptor-2 (HER2) negative breast cancer with the recently FDA-approved drug abemaciclib relies on a quantitative assessment of Ki-67 expression in the treatment decision algorithm. This further reinforces the urgent need for standardization of Ki-67 antibody selection and staining interpretation, which will hopefully lead to multidisciplinary consensus on the use of Ki-67 as a prognostic and predictive marker in breast cancer. The goals of this review are to highlight the historical evolution of Ki-67 in breast cancer, summarize the present literature on Ki-67 in breast cancer, and discuss the evolving literature on the use of Ki-67 as a companion diagnostic biomarker in breast cancer, with consideration for the necessary changes required across pathology practices to help increase the reliability and widespread adoption of Ki-67 as a prognostic and predictive marker for breast cancer in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Bradley M. Turner
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14620, USA
| |
Collapse
|
2
|
Kreipe H, Harbeck N, Christgen M. Clinical validity and clinical utility of Ki67 in early breast cancer. Ther Adv Med Oncol 2022; 14:17588359221122725. [PMID: 36105888 PMCID: PMC9465566 DOI: 10.1177/17588359221122725] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Ki67 represents an immunohistochemical nuclear localized marker that is widely
used in surgical pathology. Nuclear immunoreactivity for Ki67 indicates that
cells are cycling and are in G1- to S-phase. The percentage of Ki67-positive
tumor cells (Ki67 index) therefore provides an estimate of the growth fraction
in tumor specimens. In breast cancer (BC), tumor cell proliferation rate is one
of the most relevant prognostic markers and Ki67 is consequently helpful in
prognostication similar to histological grading and mRNA profiling-based BC risk
stratification. In BCs treated with short-term preoperative endocrine therapy,
Ki67 dynamics enable distinguishing between endocrine sensitive and resistant
tumors. Despite its nearly universal use in pathology laboratories worldwide, no
internationally accepted consensus has yet been achieved for some methodological
details related to Ki67 immunohistochemistry (IHC). Controversial issues refer
to choice of IHC antibody clones, scoring methods, inter-laboratory
reproducibility, and the potential value of computer-assisted imaging analysis
and/or artificial intelligence for Ki67 assessment. Prospective clinical trials
focusing on BC treatment have proven that Ki67, as determined by standardized
central pathology assessment, is of clinical validity. Clinical utility has been
demonstrated in huge observational studies.
Collapse
Affiliation(s)
- Hans Kreipe
- Institute of Pathology, Hannover Medical School, Carl-Neubergstraße 1, Hannover 30625, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München (LMU) Frauenklinik Maistrasse-Innenstadt und Klinikum Großhadern, Germany
| | | |
Collapse
|
3
|
Kreipe HH, Sinn P. [Relevant mutations in predictive breast cancer pathology]. DER PATHOLOGE 2021; 42:399-404. [PMID: 33822253 DOI: 10.1007/s00292-021-00929-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/24/2021] [Indexed: 11/29/2022]
Abstract
Whereas predictive immunohistochemistry has represented a core element of breast cancer classification for decades, predictive molecular pathology, with the exception of in situ hybridization for assessment of HER2 amplification, has only recently gained importance because novel drugs have been approved for treatment of metastatic disease. For the use of PARP inhibitors, proof of BRCA1 or BRCA2 mutation is mandatory. When mutation of the catalytic subunit α of the phosphatidylinositol‑4.5‑bisphosphate 3‑kinase gene (PIK3CA) is present, which can be encountered in up to 40% of luminal breast cancers, the option for treatment with the specific inhibitor alpelisib arises. The HER2 -encoded growth factor receptor contributes to neoplastic transformation not only by amplification and overexpression but also by activating the mutation of the kinase domain, which is responsive to tyrosine kinase inhibitors of the tucatinib/neratinib type. Up to 30% of metastatic and endocrine treated luminal breast cancers acquire an activating mutation of the estrogen receptor gene ESR1, resulting in an autocrine and ligand-independent growth stimulation resistant to aromatase inhibitors. Larotrectinib-sensitive mutation of tropomyosinreceptor kinase is present in up to 50% of secretory breast cancers, whereas the other histologic subtypes display an incidence of below 1%. In conclusion, predictive molecular pathology has gained importance in metastatic breast cancer.
Collapse
Affiliation(s)
- Hans H Kreipe
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland.
| | - P Sinn
- Institut für Pathologie, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| |
Collapse
|
4
|
Tendl-Schulz KA, Rössler F, Wimmer P, Heber UM, Mittlböck M, Kozakowski N, Pinker K, Bartsch R, Dubsky P, Fitzal F, Filipits M, Eckel FC, Langthaler EM, Steger G, Gnant M, Singer CF, Helbich TH, Bago-Horvath Z. Factors influencing agreement of breast cancer luminal molecular subtype by Ki67 labeling index between core needle biopsy and surgical resection specimens. Virchows Arch 2020; 477:545-555. [PMID: 32383007 PMCID: PMC7508960 DOI: 10.1007/s00428-020-02818-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/20/2020] [Accepted: 04/16/2020] [Indexed: 11/09/2022]
Abstract
Reliable determination of Ki67 labeling index (Ki67-LI) on core needle biopsy (CNB) is essential for determining breast cancer molecular subtype for therapy planning. However, studies on agreement between molecular subtype and Ki67-LI between CNB and surgical resection (SR) specimens are conflicting. The present study analyzed the influence of clinicopathological and sampling-associated factors on agreement. Molecular subtype was determined visually by Ki67-LI in 484 pairs of CNB and SR specimens of invasive estrogen receptor (ER)-positive, human epidermal growth factor (HER2)-negative breast cancer. Luminal B disease was defined by Ki67-LI > 20% in SR. Correlation of molecular subtype agreement with age, menopausal status, CNB method, Breast Imaging Reporting and Data System imaging category, time between biopsies, type of surgery, and pathological tumor parameters was analyzed. Recurrence-free survival (RFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. CNB had a sensitivity of 77.95% and a specificity of 80.97% for identifying luminal B tumors in CNB, compared with the final molecular subtype determination after surgery. The correlation of Ki67-LI between CNB and SR was moderate (ROC-AUC 0.8333). Specificity and sensitivity for CNB to correctly define molecular subtype of tumors according to SR were significantly associated with tumor grade, immunohistochemical progesterone receptor (PR) and p53 expression (p < 0.05). Agreement of molecular subtype did not significantly impact RFS and OS (p = 0.22 for both). The identified factors likely mirror intratumoral heterogeneity that might compromise obtaining a representative CNB. Our results challenge the robustness of a single CNB-driven measurement of Ki67-LI to identify luminal B breast cancer of low (G1) or intermediate (G2) grade.
Collapse
Affiliation(s)
- Kristina A Tendl-Schulz
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Fabian Rössler
- Department of Surgery and Transplantation, University Hospital and University of Zurich, Zurich, Switzerland
| | - Philipp Wimmer
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Ulrike M Heber
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Martina Mittlböck
- Center for Medical Statistics, Informatics, and Intelligent Systems, Section for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | - Nicolas Kozakowski
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Katja Pinker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rupert Bartsch
- Department for Medicine I/Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Peter Dubsky
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Hirslanden Klinik St. Anna Brustzentrum, Lucerne, Switzerland
| | - Florian Fitzal
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Martin Filipits
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Fanny Carolina Eckel
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Eva-Maria Langthaler
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Günther Steger
- Department for Medicine I/Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian F Singer
- Department of Obstetrics and Gynaecology and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria.
| |
Collapse
|
5
|
Danzinger S, Tan YY, Rudas M, Kastner MT, Weingartshofer S, Muhr D, Singer CF. Differential Claudin 3 and EGFR Expression Predicts BRCA1 Mutation in Triple-Negative Breast Cancer. Cancer Invest 2018; 36:378-388. [DOI: 10.1080/07357907.2018.1499934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Sabine Danzinger
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Yen Yen Tan
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Marie-Theres Kastner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sigrid Weingartshofer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daniela Muhr
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian F. Singer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
6
|
Prediction of BRCA1 germline mutation status in women with ovarian cancer using morphology-based criteria: identification of a BRCA1 ovarian cancer phenotype. Am J Surg Pathol 2012; 36:1170-7. [PMID: 22790858 DOI: 10.1097/pas.0b013e31825d9b8d] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Specific morphologic features that may predict BRCA1 germline mutation in ovarian cancer have neither been well described nor independently tested. We identified 5 morphologic features associated with BRCA1 mutation status in a series of 20 ovarian cancers from BRCA1 mutation carriers: (1) modified Nottingham grade 3; (2) serous/undifferentiated histology; (3) prominent intraepithelial lymphocytes; (4) marked nuclear atypia with giant/bizarre forms; and (5) abundant mitotic figures. These morphologic features were then tested on 325 ovarian tumors drawn from a population-based Greater Bay Area Cancer Registry and classified into 3 categories independent of the BRCA1 status: "Compatible with BRCA1," "Possibly compatible with BRCA1," and "Not compatible with BRCA1." All "Compatible with BRCA1" tumors were additionally investigated for presence of dominant adnexal mass, fallopian tube mucosal involvement, and uterine cornu involvement. The positive and negative predictive values for "Compatible with BRCA1" were 11/42 (26.2%) and 267/283 (94.3%), respectively, whereas combining the "Compatible with BRCA1" and "Possibly compatible with BRCA1" had positive and negative predictive values of 18/85 (21.2%) and 231/240 (96.3%), respectively. Although dominant adnexal mass and uterine cornu involvement did not add further predictive value, the likelihood of BRCA1 positivity increased to 42.9% when a tumor with "Compatible with BRCA1" histology was also associated with fallopian tube mucosal involvement. The combination of modified Nottingham grade 3 serous or undifferentiated histology, prominent intraepithelial lymphocytes, marked nuclear atypia with giant/bizarre nuclei, and high mitotic index should help to identify women for BRCA1 mutational analysis in the appropriate clinical setting. Ovarian tumors lacking this specific phenotype are unlikely to be associated with BRCA1 and should not undergo mutational analysis in the absence of other indications.
Collapse
|
7
|
Focken T, Steinemann D, Skawran B, Hofmann W, Ahrens P, Arnold N, Kroll P, Kreipe H, Schlegelberger B, Gadzicki D. Human BRCA1-associated breast cancer: no increase in numerical chromosomal instability compared to sporadic tumors. Cytogenet Genome Res 2011; 135:84-92. [PMID: 22024613 DOI: 10.1159/000332005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2011] [Indexed: 12/28/2022] Open
Abstract
BRCA1 is a major gatekeeper of genomic stability. Acting in multiple central processes like double-strand break repair, centrosome replication, and checkpoint control, BRCA1 participates in maintaining genomic integrity and protects the cell against genomic instability. Chromosomal instability (CIN) as part of genomic instability is an inherent characteristic of most solid tumors and is also involved in breast cancer development. In this study, we determined the extent of CIN in 32 breast cancer tumors of women with a BRCA1 germline mutation compared to 62 unselected breast cancers. We applied fluorescence in situ hybridization (FISH) with centromere-specific probes for the chromosomes 1, 7, 8, 10, 17, and X and locus-specific probes for 3q27 (BCL6), 5p15.2 (D5S23), 5q31 (EGR1), 10q23.3 (PTEN), and 14q32 (IGH@) on formalin-fixed paraffin-embedded tissue microarray sections. Our hypothesis of an increased level of CIN in BRCA1-associated breast cancer could not be confirmed by this approach. Surprisingly, we detected no significant difference in the extent of CIN in BRCA1-mutated versus sporadic tumors. The only exception was the CIN value for chromosome 1. Here, the extent of CIN was slightly higher in the group of sporadic tumors.
Collapse
Affiliation(s)
- T Focken
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
[A new scoring system for the diagnosis of BRCA1/2 associated breast-ovarian cancer predisposition]. Bull Cancer 2011; 98:779-95. [PMID: 21708517 DOI: 10.1684/bdc.2011.1397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Criteria have been proposed for genetic testing of breast and ovarian cancer susceptibility genes BRCA1 and BRCA2. Using simulations, this study evaluates the efficiency (sensitivity, positive predictive value [PPV] and specificity) of the various criteria used in France. The efficiency of the criteria published in 1998, which are largely used, is not optimal. We show that some extensions of these criteria provide an increase in sensitivity with a low decrease in specificity and PPV. The study shows that scoring systems (Manchester, Eisinger) have similar efficiency that may be improved. In this aim, we propose a new scoring system that takes into account unaffected individuals and kinship coefficients between family members. This system increases sensitivity without affecting PPV and specificity. Finally, we propose a two-step procedure with a large screening by the physician for recommending genetic counselling, followed by a more stringent selection by the geneticist for prescribing genetic testing. This procedure would result in an increase of genetic counselling activity but would allow the identification of almost 80% of mutation carriers among affected individuals, with a mutation detection rate of 15% and a specificity of 88%.
Collapse
|
9
|
Romero Q, Bendahl PO, Klintman M, Loman N, Ingvar C, Rydén L, Rose C, Grabau D, Borgquist S. Ki67 proliferation in core biopsies versus surgical samples - a model for neo-adjuvant breast cancer studies. BMC Cancer 2011; 11:341. [PMID: 21819622 PMCID: PMC3163632 DOI: 10.1186/1471-2407-11-341] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/07/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An increasing number of neo-adjuvant breast cancer studies are being conducted and a novel model for tumor biological studies, the "window-of-opportunity" model, has revealed several advantages. Change in tumor cell proliferation, estimated by Ki67-expression in pre-therapeutic core biopsies versus post-therapeutic surgical samples is often the primary end-point. The aim of the present study was to investigate potential differences in proliferation scores between core biopsies and surgical samples when patients have not received any intervening anti-cancer treatment. Also, a lack of consensus concerning Ki67 assessment may raise problems in the comparison of neo-adjuvant studies. Thus, the secondary aim was to present a novel model for Ki67 assessment. METHODS Fifty consecutive breast cancer cases with both a core biopsy and a surgical sample available, without intervening neo-adjuvant therapy, were collected and tumor proliferation (Ki67, MIB1 antibody) was assessed immunohistochemically. A theoretical model for the assessment of Ki67 was constructed based on sequential testing of the null hypothesis 20% Ki67-positive cells versus the two-sided alternative more or less than 20% positive cells.. RESULTS Assessment of Ki67 in 200 tumor cells showed an absolute average proliferation difference of 3.9% between core biopsies and surgical samples (p = 0.046, paired t-test) with the core biopsies being the more proliferative sample type. A corresponding analysis on the log-scale showed the average relative decrease from the biopsy to the surgical specimen to be 19% (p = 0.063, paired t-test on the log-scale). The difference was significant when using the more robust Wilcoxon matched-pairs signed-ranks test (p = 0.029). After dichotomization at 20%, 12 of the 50 sample pairs had discrepant proliferation status, 10 showed high Ki67 in the core biopsy compared to two in the surgical specimen (p = 0.039, McNemar's test). None of the corresponding results for 1000 tumor cells were significant - average absolute difference 2.2% and geometric mean of the ratios 0.85 (p = 0.19 and p = 0.18, respectively, paired t-tests, p = 0.057, Wilcoxon's test) and an equal number of discordant cases after dichotomization. Comparing proliferation values for the initial 200 versus the final 800 cancer cells showed significant absolute differences for both core biopsies and surgical samples 5.3% and 3.2%, respectively (p < 0.0001, paired t-test). CONCLUSIONS A significant difference between core biopsy and surgical sample proliferation values was observed despite no intervening therapy. Future neo-adjuvant breast cancer studies may have to take this into consideration.
Collapse
Affiliation(s)
- Quinci Romero
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gadzicki D, Evans DG, Harris H, Julian-Reynier C, Nippert I, Schmidtke J, Tibben A, van Asperen CJ, Schlegelberger B. Genetic testing for familial/hereditary breast cancer-comparison of guidelines and recommendations from the UK, France, the Netherlands and Germany. J Community Genet 2011; 2:53-69. [PMID: 22109790 PMCID: PMC3186026 DOI: 10.1007/s12687-011-0042-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 02/09/2011] [Indexed: 12/19/2022] Open
Abstract
In this review, the national guidelines and recommendations for genetic testing for familial/hereditary breast cancer from the UK, France, the Netherlands and Germany were evaluated as to the inclusion criteria for genetic testing. In all four countries, access to genetic testing relies basically on the family history of breast and ovarian cancer. Similarities are obvious for most selection criteria. All four guidelines recommend embedding genetic testing within a framework of genetic counselling, and all agree to perform genetic testing first in an affected person. However, there are differences regarding the thresholds based on certain familial constellations, detailed description of selection criteria, the degree of relatedness between affected individuals and the counsellee, the age of diagnosis, the individual history of early onset breast cancer, bilateral breast cancer, the tumour morphology or the access to intensified surveillance. These differences and open questions not covered by the guidelines, e.g. on how to deal with phenocopies, unclassified variants, genetic variants in newly identified breast cancer susceptibility genes or with family constellations not fitting the criteria, are discussed. New evidence is usually slowly integrated into the guidelines. An exchange process towards the harmonization of the guidelines will ensure high quality health care across Europe.
Collapse
Affiliation(s)
- Dorothea Gadzicki
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Liedtke C, Kiesel L. Current Issues of Targeted Therapy in Metastatic Triple-Negative Breast Cancer. ACTA ACUST UNITED AC 2011; 6:234-239. [PMID: 21779231 DOI: 10.1159/000328964] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Patients with triple-negative breast cancer are characterized by a poor prognosis compared with patients with other breast cancer subtypes. The angiogenesis inhibitor bevacizumab is effective in the palliative treatment of patients with triple-negative breast cancer as well as in other breast cancer subtypes. PARP inhibitors represent the first group of targeted agents to be developed under the particular aspect of treating patients with hereditary and triple-negative breast cancer. In addition, an increasing number of studies have demonstrated a significant and clinically relevant change in phenotype between primary tumor and metastasis. Consequently, it should be an essential component of the design of modern clinical trials of targeted agents in metastatic breast cancer to determine the relevant tumor phenotype and, depending on the clinical situation, confirm the presence of the therapeutic target in metastatic lesions.
Collapse
Affiliation(s)
- Cornelia Liedtke
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Universitätsklinikum Münster, Germany
| | | |
Collapse
|
12
|
Vargas AC, Reis-Filho JS, Lakhani SR. Phenotype-genotype correlation in familial breast cancer. J Mammary Gland Biol Neoplasia 2011; 16:27-40. [PMID: 21400086 DOI: 10.1007/s10911-011-9204-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/01/2011] [Indexed: 12/25/2022] Open
Abstract
Familial breast cancer accounts for a small but significant proportion of breast cancer cases worldwide. Identification of the candidate genes is always challenging specifically in patients with little or no family history. Therefore, a multidisciplinary team is required for the proper detection and further management of these patients. Pathologists have played a pivotal role in the cataloguing of genotypic-phenotypic correlations in families with hereditary cancer syndromes. These efforts have led to the identification of histological and phenotypic characteristics that can help predict the presence or absence of germline mutations of specific cancer predisposition genes. However, the panoply of cancer phenotypes associated with mutations of genes other than in BRCA1 is yet to be fully characterised; in fact, many cancer syndromes, germline mutations and gene sequence variants are under investigation for their possible morphological associations. Here we review the current understanding of phenotype-genotype correlation in familial breast cancer.
Collapse
Affiliation(s)
- Ana Cristina Vargas
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | | | | |
Collapse
|
13
|
Vargas AC, Da Silva L, Lakhani SR. The contribution of breast cancer pathology to statistical models to predict mutation risk in BRCA carriers. Fam Cancer 2011; 9:545-53. [PMID: 20577821 DOI: 10.1007/s10689-010-9362-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BRCA1 and BRCA2 associated breast cancer comprises a small but important group of hereditary breast cancer. Testing for BRCA1 and BRCA2 has significant clinical and personal implications for the patients in terms of therapy and follow-up of individual family members. The sequencing of the genes is expensive and since the information derived may have a profound effect on the individual and family members, it is important that testing is done only when the risk of carrying a mutation is thought to be high. Over the last decade, researchers have developed a number of statistical models for predicting risk for harboring mutations in these genes and the risk of subsequently developing breast and ovarian cancer. These models usually take into account the type of tumor and age at occurrence as well as family history. Data from pathological analysis show that although breast tumours are heterogeneous, there are histological characteristics that are seen more frequently in carriers of BRCA1 germ line mutations compared to BRCA2 and sporadic breast cancers. A number of authors have suggested that the addition of pathological data to risk algorithms may improve the predictive power of these models and provide a more accurate way of identifying individuals who may benefit from testing. Here we review the pathology of familial breast cancer and assess the evidence to justify the use of pathology in refining risk assessment models.
Collapse
Affiliation(s)
- Ana Cristina Vargas
- Molecular and Cellular Pathology, The University of Queensland Centre for Clinical Research, The Royal Brisbane and Women's Hospital, Building 71/918, Brisbane, QLD 4029, Australia
| | | | | |
Collapse
|
14
|
Mulligan AM, Pinnaduwage D, Bane AL, Bull SB, O'Malley FP, Andrulis IL. CK8/18 expression, the basal phenotype, and family history in identifying BRCA1-associated breast cancer in the Ontario site of the breast cancer family registry. Cancer 2010; 117:1350-9. [PMID: 21425134 DOI: 10.1002/cncr.25642] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 07/06/2009] [Accepted: 08/09/2009] [Indexed: 01/07/2023]
Abstract
BACKGROUND BRCA1-associated breast cancer had been shown to be morphologically and genetically distinct from sporadic cancers. The aim of this study was to determine the association of CK8/18 with BRCA1-associated tumors and if, by using CK8/18 and basal biomarkers in conjunction with morphologic features and family history characteristics, the specificity of the BRCA1-associated tumor profile in a pathologically well-characterized cohort would be improved. METHODS Fifty-eight patients with known BRCA1 germline mutations and 221 control (familial non-BRCA) patients were selected from the Ontario Familial Breast Cancer Registry. From this database, information on family history and morphologic features was abstracted. Tissue microarrays were constructed and immunohistochemistry to determine expression of several biomarkers was performed. After a logistic regression fit, a best-subsets variable-selection procedure using model performance and predictive ability measures was applied to find a best predictor to distinguish BRCA1-associated tumors from non-BRCA associated tumors. RESULTS BRCA1-associated tumors differed significantly from control tumors in terms of morphology, family history, and biomarker profile. CK8/18 was highly significantly associated with BRCA1 tumors. Consistently, BRCA1 cancers showed low levels of CK8/18 compared to non-BRCA tumors, whether they were basal-like or not. A combination of 7 factors, including CK8/18 and family history, best predicted the BRCA1-associated cancers. CONCLUSIONS CK8/18 expression was independently associated with BRCA1-associated breast cancers. Reduced CK8/18 expression in conjunction with the basal-like phenotype and family history may have improved the ability to identify which tumors were likely to be associated with a BRCA1 germline mutation and thereby help streamline genetic testing.
Collapse
Affiliation(s)
- Anna Marie Mulligan
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital and the Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Ontario, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|