1
|
Romo B, Fuentes Z, Randolph L, Mahajan M, Aller EJ, Ebrahimi B, Santhamma B, Pratap UP, Subbarayalu P, Nagandla H, Thomas C, Nair HB, Vadlamudi RK, Viswanadhapalli S. Targeting the Leukemia Inhibitory Factor/Leukemia Inhibitory Factor Receptor Axis Reduces the Growth of Inflammatory Breast Cancer by Promoting Ferroptosis. Cancers (Basel) 2025; 17:790. [PMID: 40075639 PMCID: PMC11898489 DOI: 10.3390/cancers17050790] [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: 01/01/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Inflammatory breast cancer (IBC) is a rare subtype of breast cancer accounting for 7% of breast cancer-related fatalities. There is an urgent need to develop new targeted treatments for IBC. The progression of IBC has been associated with alterations in growth factor and cytokine signaling; however, the function of the LIF (leukemia inhibitory factor)/LIFR (leukemia inhibitory factor receptor) cytokine pathway in the progression of IBC remains unknown. This study evaluated the role of LIFR signaling and tested the efficacy of the LIFR inhibitor EC359 in treating IBC. Methods: The utility of using LIFR inhibition as a treatment strategy in IBC was tested using cell survival, apoptosis, colony formation, invasion, and pre-clinical KPL4 xenografts. Western blotting, siRNA, RT-qPCR, and lipid peroxidation assays were used to establish the mechanism of EC359 therapy. Results: The reduction in LIFR levels using siRNA markedly decreased growth in colony formation assays and reduced the invasion of IBC cells. Pharmacological inhibition of LIFR with EC359 effectively reduced cell survival and the clonogenic capacity of IBC cells. RT-qPCR assays revealed that EC359 markedly decreased the expression of the LIFR target genes. Western blot analyses confirmed that EC359 treatment suppressed downstream LIF/LIFR signaling pathways and promoted apoptosis. Treatment of cells with the ferroptosis inhibitor Fer-1 negated the capacity of EC359 to induce apoptosis. Mechanistic investigations demonstrated that EC359 predominantly triggered ferroptosis by inhibiting the glutathione antioxidant defense system through the downregulation of Glutathione peroxidase 4 (GPX4) levels. EC359 (5 mg/kg/day) was effective in reducing the growth of the IBC KPL4 xenograft tumors. Conclusion: These findings demonstrates that LIFR inhibition promote ferroptosis-mediated cell death in IBC and that EC359 represent novel therapeutic for IBC treatment.
Collapse
Affiliation(s)
- Bianca Romo
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Zenaida Fuentes
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Lois Randolph
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Megharani Mahajan
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Emily J. Aller
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | - Behnam Ebrahimi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
| | | | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Panneerdoss Subbarayalu
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
- Greehey Children Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Harika Nagandla
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.N.); (C.T.)
| | - Christoforos Thomas
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.N.); (C.T.)
| | - Hareesh B. Nair
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Evestra, Inc., San Antonio, TX 78245, USA;
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (B.R.); (Z.F.); (L.R.); (M.M.); (E.J.A.); (B.E.); (U.P.P.)
- Mays Cancer Canter, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| |
Collapse
|
2
|
Gouveia BA, Ramos FR, Silva IKL, Oliveira TESD, Vasconcelos RDO, Xavier JG, Strefezzi RF. Prognostic Implications of Decorin, E-Cadherin and EGFR Expression in Inflammatory and Non-Inflammatory Canine Mammary Carcinomas. Vet Comp Oncol 2025. [PMID: 39853670 DOI: 10.1111/vco.13042] [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: 11/12/2024] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025]
Abstract
Inflammatory mammary carcinoma (IMC) is the most aggressive variant of invasive mammary tumours in dogs and in women. Decorin is an extracellular matrix molecule whose expression can be reduced or absent in various human cancers, which is associated with a poor prognosis. E-cadherin is a cell adhesion protein whose expression is reduced in several neoplasms. However, it is overexpressed in inflammatory breast cancers of women. EGFR is also associated with cancer development and is commonly overexpressed in aggressive neoplasms. This study aimed to characterise the expressions of Decorin, E-cadherin, and EGFR in canine inflammatory and non-inflammatory mammary carcinomas (IMC and non-IMC) and to evaluate their expression levels as prognostic indicators for survival and occurrence of metastases. Thirty-three IMC and 43 non-IMC cases were analysed retrospectively and submitted to immunohistochemical analysis. The reactions were quantified in five high-power field images from areas of the highest intensity and frequency of immunostaining (hot spots). We found significantly lower expression of Decorin and higher of E-cadherin and EGFR in canine IMCs. Patients with tumours that exhibited Decorin expression in less than 26.35% of epithelial cells had shorter survival (p = 0.0410) and a higher occurrence of distant metastases (p = 0.0115). E-cadherin is overexpressed in canine IMCs (p < 0.0001), similar to what occurs in women, reinforcing that dogs can be used as a study model for human IMC. EGFR overexpression in canine IMCs (p = 0.0322) provides evidence for potential targeted therapy with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Bethânia Almeida Gouveia
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fernanda Ramalho Ramos
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ingrid Kester Lima Silva
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | | | | | - Ricardo Francisco Strefezzi
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Villodre ES, Hu X, Larson R, Finetti P, Gomez K, Balema W, Stecklein SR, Santiago‐Sanchez G, Krishnamurthy S, Song J, Su X, Ueno NT, Tripathy D, Van Laere S, Bertucci F, Vivas‐Mejía P, Woodward WA, Debeb BG. Lipocalin 2 promotes inflammatory breast cancer tumorigenesis and skin invasion. Mol Oncol 2021; 15:2752-2765. [PMID: 34342930 PMCID: PMC8486564 DOI: 10.1002/1878-0261.13074] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive form of primary breast cancer characterized by rapid onset and high risk of metastasis and poor clinical outcomes. The biological basis for the aggressiveness of IBC is still not well understood and no IBC-specific targeted therapies exist. In this study, we report that lipocalin 2 (LCN2), a small secreted glycoprotein belonging to the lipocalin superfamily, is expressed at significantly higher levels in IBC vs non-IBC tumors, independently of molecular subtype. LCN2 levels were also significantly higher in IBC cell lines and in their culture media than in non-IBC cell lines. High expression was associated with poor-prognosis features and shorter overall survival in IBC patients. Depletion of LCN2 in IBC cell lines reduced colony formation, migration, and cancer stem cell populations in vitro and inhibited tumor growth, skin invasion, and brain metastasis in mouse models of IBC. Analysis of our proteomics data showed reduced expression of proteins involved in cell cycle and DNA repair in LCN2-silenced IBC cells. Our findings support that LCN2 promotes IBC tumor aggressiveness and offer a new potential therapeutic target for IBC.
Collapse
Affiliation(s)
- Emilly S. Villodre
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoding Hu
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Richard Larson
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Pascal Finetti
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Kristen Gomez
- Department of Biological SciencesThe University of Texas at BrownsvilleTXUSA
| | - Wintana Balema
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Shane R. Stecklein
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ginette Santiago‐Sanchez
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Savitri Krishnamurthy
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of PathologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Juhee Song
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoping Su
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Naoto T. Ueno
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Debu Tripathy
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Steven Van Laere
- Center for Oncological Research (CORE)Integrated Personalized and Precision Oncology Network (IPPON)University of AntwerpBelgium
| | - François Bertucci
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Pablo Vivas‐Mejía
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Wendy A. Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Bisrat G. Debeb
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
4
|
NDRG1 Expression Is an Independent Prognostic Factor in Inflammatory Breast Cancer. Cancers (Basel) 2020; 12:cancers12123711. [PMID: 33321961 PMCID: PMC7763268 DOI: 10.3390/cancers12123711] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Inflammatory breast cancer (IBC) is a rare and aggressive variant of breast cancer that is responsible for a significant number of breast cancer-related deaths. Herein, we describe how the expression of a specific protein named N-myc downstream-regulated gene 1 (NDRG1), commonly described as a gene that prevents the spread of cancer cells to distant organs, may have a paradoxical role in cancer progression in IBC. We found that the level of expression of NDRG1 in tumor tissues predicts the survival outcome of patients with IBC. We also observed that NDRG1, together with other important prognostic factors such as estrogen receptor status and stage, could be used to further analyze prognostic outcome or treatment response of patients. Abstract NDRG1 is widely described as a metastasis suppressor in breast cancer. However, we found that NDRG1 is critical in promoting tumorigenesis and brain metastasis in mouse models of inflammatory breast cancer (IBC), a rare but highly aggressive form of breast cancer. We hypothesized that NDRG1 is a prognostic marker associated with poor outcome in patients with IBC. NDRG1 levels in tissue microarrays from 64 IBC patients were evaluated by immunohistochemical staining with NDRG1 (32 NDRG1-low (≤median), 32 NDRG1-high (>median)). Overall and disease-free survival (OS and DSS) were analyzed with Kaplan–Meier curves and log-rank test. Univariate analysis showed NDRG1 expression, tumor grade, disease stage, estrogen receptor (ER) status, and receipt of adjuvant radiation to be associated with OS and DSS. NDRG1-high patients had poorer 10-year OS and DSS than NDRG1-low patients (OS, 19% vs. 45%, p = 0.0278; DSS, 22% vs. 52%, p = 0.0139). On multivariable analysis, NDRG1 independently predicted OS (hazard ratio (HR) = 2.034, p = 0.0274) and DSS (HR = 2.287, p = 0.0174). NDRG1-high ER-negative tumors had worse outcomes OS, p = 0.0003; DSS, p = 0.0003; and NDRG1-high tumors that received adjuvant radiation treatment had poor outcomes (OS, p = 0.0088; DSS, p = 0.0093). NDRG1 was a significant independent prognostic factor for OS and DSS in IBC patients. Targeting NDRG1 may represent a novel strategy for improving clinical outcomes for patients with IBC.
Collapse
|
5
|
Influence of Biologic Subtype of Inflammatory Breast Cancer on Response to Neoadjuvant Therapy and Cancer Outcomes. Clin Breast Cancer 2018; 18:e501-e506. [DOI: 10.1016/j.clbc.2017.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/01/2017] [Accepted: 10/02/2017] [Indexed: 01/02/2023]
|
6
|
Geospatial Analysis of Inflammatory Breast Cancer and Associated Community Characteristics in the United States. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14040404. [PMID: 28398259 PMCID: PMC5409605 DOI: 10.3390/ijerph14040404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 11/16/2022]
Abstract
Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer, almost always diagnosed at late stage where mortality outcomes and morbidity burdens are known to be worse. Missed by mammography screening, IBC progresses rapidly and reaches late stage by the time of diagnosis. With an unknown etiology and poor prognosis, it is crucial to evaluate the distribution of the disease in the population as well as identify area social and economic contextual risk factors that may be contributing to the observed patterns of IBC incidence. In this study, we identified spatial clustering of county-based IBC rates among US females and examined the underlying community characteristics associated with the clusters. IBC accounted for ~1.25% of all primary breast cancers diagnoses in 2004-2012 and was defined by the Collaborative Stage (CS) Extension code 710 and 730. Global and local spatial clusters of IBC rates were identified and mapped. The Mann-Whitney U test was used to compare median differences in key contextual variables between areas with high and low spatial clusters of IBC rates. High clusters are counties and their neighbors that all exhibit above average rates, clustered together in a fashion that would be extremely unlikely to be observed by chance, and conversely for low clusters. There was statistically significant evidence of spatial clustering into high and low rate clusters. The average rate in the high rate clusters (n = 46) was approximately 12 times the average rate in low rate clusters (n = 126), and 2.2 times the national average across all counties. Significant differences were found in the medians of the underlying race, poverty, and urbanicity variables when comparing the low cluster counties with the high cluster counties (p < 0.05). Cluster analysis confirms that IBC rates differ geographically and may be influenced by social and economic environmental factors. Particular attention may need to be paid to race, urbanicity and poverty when considering risk factors for IBC and when developing interventions and alternative prevention strategies.
Collapse
|
7
|
Jansen MPHM, Sas L, Sieuwerts AM, Van Cauwenberghe C, Ramirez-Ardila D, Look M, Ruigrok-Ritstier K, Finetti P, Bertucci F, Timmermans MM, van Deurzen CHM, Martens JWM, Simon I, Roepman P, Linn SC, van Dam P, Kok M, Lardon F, Vermeulen PB, Foekens JA, Dirix L, Berns EMJJ, Van Laere S. Decreased expression of ABAT and STC2 hallmarks ER-positive inflammatory breast cancer and endocrine therapy resistance in advanced disease. Mol Oncol 2015; 9:1218-33. [PMID: 25771305 DOI: 10.1016/j.molonc.2015.02.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with Estrogen Receptor α-positive (ER+) Inflammatory Breast Cancer (IBC) are less responsive to endocrine therapy compared with ER+ non-IBC (nIBC) patients. The study of ER+ IBC samples might reveal biomarkers for endocrine resistant breast cancer. MATERIALS & METHODS Gene expression profiles of ER+ samples from 201 patients were explored for genes that discriminated between IBC and nIBC. Classifier genes were applied onto clinically annotated expression data from 947 patients with ER+ breast cancer and validated with RT-qPCR for 231 patients treated with first-line tamoxifen. Relationships with metastasis-free survival (MFS) and progression-free survival (PFS) following adjuvant and first-line endocrine treatment, respectively, were investigated using Cox regression analysis. RESULTS A metagene of six genes including the genes encoding for 4-aminobutyrate aminotransferase (ABAT) and Stanniocalcin-2 (STC2) were identified to distinguish 22 ER+ IBC from 43 ER+ nIBC patients and remained discriminatory in an independent series of 136 patients. The metagene and two genes were not prognostic in 517 (neo)adjuvant untreated lymph node-negative ER+ nIBC breast cancer patients. Only ABAT was related to outcome in 250 patients treated with adjuvant tamoxifen. Three independent series of in total 411 patients with advanced disease showed increased metagene scores and decreased expression of ABAT and STC2 to be correlated with poor first-line endocrine therapy outcome. The biomarkers remained predictive for first-line tamoxifen treatment outcome in multivariate analysis including traditional factors or published signatures. In an exploratory analysis, ABAT and STC2 protein expression levels had no relation with PFS after first-line tamoxifen. CONCLUSIONS This study utilized ER+ IBC to identify a metagene including ABAT and STC2 as predictive biomarkers for endocrine therapy resistance.
Collapse
Affiliation(s)
- Maurice P H M Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands.
| | - Leen Sas
- Translational Cancer Research Unit, GZA Hospitals St-Augustinus, Oosterveldlaan 24, Antwerp B2610, Belgium; Department of Medical Oncology, University Hospital Antwerp, Wilrijkstraat 10, B2650 Antwerp, Belgium
| | - Anieta M Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Caroline Van Cauwenberghe
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Diana Ramirez-Ardila
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Maxime Look
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Kirsten Ruigrok-Ritstier
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Pascal Finetti
- Marseille Cancer Research Center (CRCM), UMR891 Inserm, Institut Paoli-Calmettes (IPC), Department of Molecular Oncology, Marseille, France
| | - François Bertucci
- Marseille Cancer Research Center (CRCM), UMR891 Inserm, Institut Paoli-Calmettes (IPC), Department of Molecular Oncology, Marseille, France
| | - Mieke M Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Carolien H M van Deurzen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Iris Simon
- Research and Development, Agendia BV, Amsterdam, The Netherlands
| | - Paul Roepman
- Research and Development, Agendia BV, Amsterdam, The Netherlands
| | - Sabine C Linn
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter van Dam
- Translational Cancer Research Unit, GZA Hospitals St-Augustinus, Oosterveldlaan 24, Antwerp B2610, Belgium
| | - Marleen Kok
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Filip Lardon
- Department of Medical Oncology, University Hospital Antwerp, Wilrijkstraat 10, B2650 Antwerp, Belgium
| | - Peter B Vermeulen
- Translational Cancer Research Unit, GZA Hospitals St-Augustinus, Oosterveldlaan 24, Antwerp B2610, Belgium
| | - John A Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals St-Augustinus, Oosterveldlaan 24, Antwerp B2610, Belgium
| | - Els M J J Berns
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Wytemaweg 80, 3000 CA Rotterdam, The Netherlands
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals St-Augustinus, Oosterveldlaan 24, Antwerp B2610, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Marsan M, Van den Eynden G, Limame R, Neven P, Hauspy J, Van Dam PA, Vergote I, Dirix LY, Vermeulen PB, Van Laere SJ. A core invasiveness gene signature reflects epithelial-to-mesenchymal transition but not metastatic potential in breast cancer cell lines and tissue samples. PLoS One 2014; 9:e89262. [PMID: 24586640 PMCID: PMC3931724 DOI: 10.1371/journal.pone.0089262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/15/2014] [Indexed: 01/07/2023] Open
Abstract
Introduction Metastases remain the primary cause of cancer-related death. The acquisition of invasive tumour cell behaviour is thought to be a cornerstone of the metastatic cascade. Therefore, gene signatures related to invasiveness could aid in stratifying patients according to their prognostic profile. In the present study we aimed at identifying an invasiveness gene signature and investigated its biological relevance in breast cancer. Methods & Results We collected a set of published gene signatures related to cell motility and invasion. Using this collection, we identified 16 genes that were represented at a higher frequency than observed by coincidence, hereafter named the core invasiveness gene signature. Principal component analysis showed that these overrepresented genes were able to segregate invasive and non-invasive breast cancer cell lines, outperforming sets of 16 randomly selected genes (all P<0.001). When applied onto additional data sets, the expression of the core invasiveness gene signature was significantly elevated in cell lines forced to undergo epithelial-mesenchymal transition. The link between core invasiveness gene expression and epithelial-mesenchymal transition was also confirmed in a dataset consisting of 2420 human breast cancer samples. Univariate and multivariate Cox regression analysis demonstrated that CIG expression is not associated with a shorter distant metastasis free survival interval (HR = 0.956, 95%C.I. = 0.896–1.019, P = 0.186). Discussion These data demonstrate that we have identified a set of core invasiveness genes, the expression of which is associated with epithelial-mesenchymal transition in breast cancer cell lines and in human tissue samples. Despite the connection between epithelial-mesenchymal transition and invasive tumour cell behaviour, we were unable to demonstrate a link between the core invasiveness gene signature and enhanced metastatic potential.
Collapse
Affiliation(s)
- Melike Marsan
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- Department of oncology, KU Leuven, Leuven, Belgium
- * E-mail:
| | - Gert Van den Eynden
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Ridha Limame
- Laboratory for Cancer Research and Clinical Oncology, University of Antwerp, Antwerp, Belgium
| | | | - Jan Hauspy
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | | | | | - Luc Y. Dirix
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Peter B. Vermeulen
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Steven J. Van Laere
- Translational Cancer Research Unit, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
- Department of oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Sas L, Vermeulen PB, van Dam P, Dirix LY, Lardon F, Van Laere SJ. Contribution of ER and NF-κB to endocrine resistance in inflammatory breast cancer. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.13.72] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
SUMMARY Inflammatory breast cancer (IBC) is a very aggressive form of breast cancer with a high mortality rate. Most patients have lymph node metastasis at the time of diagnosis and 30% of patients already have metastases in distant organs. IBC is normally treated with multimodality therapy. Endocrine therapy is administered in cases of ER-positive tumors. Nevertheless, IBC has a high HOXB13:Il17RB ratio, predicting a poor response to tamoxifen treatment. These data suggest a possible role for IBC as a model for endocrine resistance. Previous studies have shown that NF-κB, a transcription factor regulating different cellular processes, is more highly activated in IBC than in non-IBC, while ER is often downregulated in this tumor type. This article summarizes the activity of ER and NF-κB in IBC and their possible contribution to endocrine resistance in this breast cancer subtype.
Collapse
Affiliation(s)
- Leen Sas
- Department of Oncology, University of Antwerp, Antwerp, Belgium
- Translational Cancer Research Unit Antwerp, Laboratory of Pathology GZA, Hospitals Sint Augustinus, Antwerp, Belgium.
| | - Peter B Vermeulen
- Translational Cancer Research Unit Antwerp, Laboratory of Pathology GZA, Hospitals Sint Augustinus, Antwerp, Belgium
| | - Peter van Dam
- Translational Cancer Research Unit Antwerp, Laboratory of Pathology GZA, Hospitals Sint Augustinus, Antwerp, Belgium
- Department of Oncology, University of Antwerp, Antwerp, Belgium
| | - Luc Y Dirix
- Translational Cancer Research Unit Antwerp, Laboratory of Pathology GZA, Hospitals Sint Augustinus, Antwerp, Belgium
| | - Filip Lardon
- Department of Oncology, University of Antwerp, Antwerp, Belgium
| | - Steven J Van Laere
- Translational Cancer Research Unit Antwerp, Laboratory of Pathology GZA, Hospitals Sint Augustinus, Antwerp, Belgium
- Division of Gyneacological Oncology, Department of Oncology, University Hospital Leuven, Catholic University Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Van Laere SJ, Ueno NT, Finetti P, Vermeulen P, Lucci A, Robertson FM, Marsan M, Iwamoto T, Krishnamurthy S, van Dam P, Woodward WA, Viens P, Cristofanilli M, Birnbaum D, Dirix L, Reuben JM, Bertucci F. Uncovering the molecular secrets of inflammatory breast cancer biology: an integrated analysis of three distinct affymetrix gene expression datasets. Clin Cancer Res 2013; 19:4685-96. [PMID: 23396049 PMCID: PMC6156084 DOI: 10.1158/1078-0432.ccr-12-2549] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Inflammatory breast cancer (IBC) is a poorly characterized form of breast cancer. So far, the results of expression profiling in IBC are inconclusive due to various reasons including limited sample size. Here, we present the integration of three Affymetrix expression datasets collected through the World IBC Consortium allowing us to interrogate the molecular profile of IBC using the largest series of IBC samples ever reported. EXPERIMENTAL DESIGN Affymetrix profiles (HGU133-series) from 137 patients with IBC and 252 patients with non-IBC (nIBC) were analyzed using unsupervised and supervised techniques. Samples were classified according to the molecular subtypes using the PAM50-algorithm. Regression models were used to delineate IBC-specific and molecular subtype-independent changes in gene expression, pathway, and transcription factor activation. RESULTS Four robust IBC-sample clusters were identified, associated with the different molecular subtypes (P<0.001), all of which were identified in IBC with a similar prevalence as in nIBC, except for the luminal A subtype (19% vs. 42%; P<0.001) and the HER2-enriched subtype (22% vs. 9%; P<0.001). Supervised analysis identified and validated an IBC-specific, molecular subtype-independent 79-gene signature, which held independent prognostic value in a series of 871 nIBCs. Functional analysis revealed attenuated TGF-β signaling in IBC. CONCLUSION We show that IBC is transcriptionally heterogeneous and that all molecular subtypes described in nIBC are detectable in IBC, albeit with a different frequency. The molecular profile of IBC, bearing molecular traits of aggressive breast tumor biology, shows attenuation of TGF-β signaling, potentially explaining the metastatic potential of IBC tumor cells in an unexpected manner.
Collapse
Affiliation(s)
- Steven J. Van Laere
- Translational Cancer Research Unit Antwerp, Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
- Department Oncology, KU Leuven, Leuven, Belgium
| | - Naoto T. Ueno
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pascal Finetti
- Département d’Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Peter Vermeulen
- Translational Cancer Research Unit Antwerp, Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Anthony Lucci
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fredika M. Robertson
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melike Marsan
- Translational Cancer Research Unit Antwerp, Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
- Department Oncology, KU Leuven, Leuven, Belgium
| | - Takayuki Iwamoto
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter van Dam
- Translational Cancer Research Unit Antwerp, Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - Wendy A. Woodward
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrice Viens
- Département d’Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Massimo Cristofanilli
- Department of Medical Oncology,G. Morris Dorrance Jr. Endowed Chair in Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel Birnbaum
- Département d’Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Luc Dirix
- Translational Cancer Research Unit Antwerp, Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | - James M. Reuben
- Morgan Welch Inflammatory Breast Cancer Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - François Bertucci
- Département d’Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes (IPC), Marseille, France
| |
Collapse
|
11
|
Nokes BT, Cunliffe HE, Lafleur B, Mount DW, Livingston RB, Futscher BW, Lang JE. In Vitro Assessment of the Inflammatory Breast Cancer Cell Line SUM 149: Discovery of 2 Single Nucleotide Polymorphisms in the RNase L Gene. J Cancer 2013; 4:104-16. [PMID: 23386909 PMCID: PMC3563072 DOI: 10.7150/jca.5002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 11/12/2012] [Indexed: 01/04/2023] Open
Abstract
Background: Inflammatory breast cancer (IBC) is a rare, highly aggressive form of breast cancer. The mechanism of IBC carcinogenesis remains unknown. We sought to evaluate potential genetic risk factors for IBC and whether or not the IBC cell lines SUM149 and SUM190 demonstrated evidence of viral infection. Methods: We performed single nucleotide polymorphism (SNP) genotyping for 2 variants of the ribonuclease (RNase) L gene that have been correlated with the risk of prostate cancer due to a possible viral etiology. We evaluated dose-response to treatment with interferon-alpha (IFN-α); and assayed for evidence of the putative human mammary tumor virus (HMTV, which has been implicated in IBC) in SUM149 cells. A bioinformatic analysis was performed to evaluate expression of RNase L in IBC and non-IBC. Results: 2 of 2 IBC cell lines were homozygous for RNase L common missense variants 462 and 541; whereas 2 of 10 non-IBC cell lines were homozygous positive for the 462 variant (p= 0.09) and 0 of 10 non-IBC cell lines were homozygous positive for the 541 variant (p = 0.015). Our real-time polymerase chain reaction (RT-PCR) and Southern blot analysis for sequences of HMTV revealed no evidence of the putative viral genome. Conclusion: We discovered 2 SNPs in the RNase L gene that were homozygously present in IBC cell lines. The 462 variant was absent in non-IBC lines. Our discovery of these SNPs present in IBC cell lines suggests a possible biomarker for risk of IBC. We found no evidence of HMTV in SUM149 cells. A query of a panel of human IBC and non-IBC samples showed no difference in RNase L expression. Further studies of the RNase L 462 and 541 variants in IBC tissues are warranted to validate our in vitro findings.
Collapse
|
12
|
A randomized phase II study of lapatinib + pazopanib versus lapatinib in patients with HER2+ inflammatory breast cancer. Breast Cancer Res Treat 2012; 137:471-82. [PMID: 23239151 PMCID: PMC3539065 DOI: 10.1007/s10549-012-2369-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 11/30/2012] [Indexed: 01/04/2023]
Abstract
This multi-center Phase II study evaluated lapatinib, pazopanib, and the combination in patients with relapsed HER2+ inflammatory breast cancer. In Cohort 1, 76 patients were randomized 1:1 to receive lapatinib 1,500 mg + placebo or lapatinib 1,500 mg + pazopanib 800 mg (double-blind) once daily until disease progression, unacceptable toxicity, or death. Due to high-grade diarrhea observed with this dose combination in another study (VEG20007), Cohort 1 was closed. The protocol was amended such that an additional 88 patients (Cohort 2) were randomized in a 5:5:2 ratio to receive daily monotherapy lapatinib 1,500 mg, lapatinib 1,000 mg + pazopanib 400 mg, or monotherapy pazopanib 800 mg, respectively. The primary endpoint was overall response rate (ORR). Secondary endpoints included duration of response, progression-free survival (PFS), overall survival, and safety. In Cohort 1, ORR for the lapatinib (n = 38) and combination (n = 38) arms was 29 and 45 %, respectively; median PFS was 16.1 and 14.3 weeks, respectively. Grade ≥3 adverse events (AEs) were more frequent in the combination arm (71 %) than in the lapatinib arm (24 %). Dose reductions and interruptions due to AEs were also more frequent in the combination arm (45 and 53 %, respectively) than in the lapatinib monotherapy arm (0 and 11 %, respectively). In Cohort 2, ORR for patients treated with lapatinib (n = 36), lapatinib + pazopanib (n = 38), and pazopanib (n = 13) was 47, 58, and 31 %, respectively; median PFS was 16.0, 16.0, and 11.4 weeks, respectively. In the lapatinib, combination, and pazopanib therapy arms, grade ≥3 AEs were reported for 17, 50, and 46 % of patients, respectively, and the incidence of discontinuations due to AEs was 0, 24, and 23 %, respectively. The lapatinib-pazopanib combination was associated with a numerically higher ORR but no increase in PFS compared to lapatinib alone. The combination also had increased toxicity resulting in more dose reductions, modifications, and treatment delays. Activity with single-agent lapatinib was confirmed in this population.
Collapse
|
13
|
Carkaci S, Adrada BE, Rohren E, Wei W, Quraishi MA, Mawlawi O, Buchholz TA, Yang W. Semiquantitative analysis of maximum standardized uptake values of regional lymph nodes in inflammatory breast cancer: is there a reliable threshold for differentiating benign from malignant? Acad Radiol 2012; 19:535-41. [PMID: 22300741 PMCID: PMC4128395 DOI: 10.1016/j.acra.2012.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/03/2012] [Accepted: 01/03/2012] [Indexed: 10/14/2022]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to determine an optimum standardized uptake value (SUV) threshold for identifying regional nodal metastasis on 18F-fluorodeoxyglucose (FDG) positron emission tomographic (PET)/computed tomographic (CT) studies of patients with inflammatory breast cancer. MATERIALS AND METHODS A database search was performed of patients newly diagnosed with inflammatory breast cancer who underwent 18F-FDG PET/CT imaging at the time of diagnosis at a single institution between January 1, 2001, and September 30, 2009. Three radiologists blinded to the histopathology of the regional lymph nodes retrospectively analyzed all 18F-FDG PET/CT images by measuring the maximum SUV (SUVmax) in visually abnormal nodes. The accuracy of 18F-FDG PET/CT image interpretation was correlated with histopathology when available. Receiver-operating characteristic curve analysis was performed to assess the diagnostic performance of PET/CT imaging. Sensitivity, specificity, positive predictive value, and negative predictive value were calculated using three different SUV cutoff values (2.0, 2.5, and 3.0). RESULTS A total of 888 regional nodal basins, including bilateral axillary, infraclavicular, internal mammary, and supraclavicular lymph nodes, were evaluated in 111 patients (mean age, 56 years). Of the 888 nodal basins, 625 (70%) were negative and 263 (30%) were positive for metastasis. Malignant lymph nodes had significantly higher SUVmax than benign lymph nodes (P < .0001). An SUVmax of 2.0 showed the highest overall sensitivity (89%) and specificity (99%) for the diagnosis of malignant disease. CONCLUSIONS SUVmax of regional lymph nodes on 18F-FDG PET/CT imaging may help differentiate benign and malignant lymph nodes in patients with inflammatory breast cancer. An SUV cutoff of 2 provided the best accuracy in identifying regional nodal metastasis in this patient population.
Collapse
Affiliation(s)
- Selin Carkaci
- Department of Diagnostic Radiology, Unit 1350, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
A critical evaluation of in vitro cell culture models for high-throughput drug screening and toxicity. Pharmacol Ther 2012; 134:82-106. [DOI: 10.1016/j.pharmthera.2012.01.001] [Citation(s) in RCA: 276] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 12/22/2011] [Indexed: 01/10/2023]
|
15
|
|
16
|
Ohshiro K, Schwartz AM, Levine PH, Kumar R. Alternate estrogen receptors promote invasion of inflammatory breast cancer cells via non-genomic signaling. PLoS One 2012; 7:e30725. [PMID: 22295107 PMCID: PMC3266301 DOI: 10.1371/journal.pone.0030725] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 12/27/2011] [Indexed: 11/18/2022] Open
Abstract
Although Inflammatory Breast Cancer (IBC) is a rare and an aggressive type of locally advanced breast cancer with a generally worst prognosis, little work has been done in identifying the status of non-genomic signaling in the invasiveness of IBC. The present study was performed to explore the status of non-genomic signaling as affected by various estrogenic and anti-estrogenic agents in IBC cell lines SUM149 and SUM190. We have identified the presence of estrogen receptor α (ERα) variant, ERα36 in SUM149 and SUM190 cells. This variant as well as ERβ was present in a substantial concentration in IBC cells. The treatment with estradiol (E2), anti-estrogenic agents 4-hydroxytamoxifen and ICI 182780, ERβ specific ligand DPN and GPR30 agonist G1 led to a rapid activation of p-ERK1/2, suggesting the involvement of ERα36, ERβ and GPR30 in the non-genomic signaling pathway in these cells. We also found a substantial increase in the cell migration and invasiveness of SUM149 cells upon the treatment with these ligands. Both basal and ligand-induced migration and invasiveness of SUM149 cells were drastically reduced in the presence of MEK inhibitor U0126, implicating that the phosphorylation of ERK1/2 by MEK is involved in the observed motility and invasiveness of IBC cells. We also provide evidence for the upregulation of p-ERK1/2 through immunostaining in IBC patient samples. These findings suggest a role of non-genomic signaling through the activation of p-ERK1/2 in the hormonal dependence of IBC by a combination of estrogen receptors. These findings only explain the failure of traditional anti-estrogen therapies in ER-positive IBC which induces the non-genomic signaling, but also opens newer avenues for design of modified therapies targeting these estrogen receptors.
Collapse
Affiliation(s)
- Kazufumi Ohshiro
- Department of Biochemistry and Molecular Biology, The George Washington University Medical Center, Washington, D.C., United States of America
| | - Arnold M. Schwartz
- Department of Pathology, The George Washington University Medical Center, Washington, D.C., United States of America
| | - Paul H. Levine
- Department of Epidemiology and Biostatistics, The George Washington University Medical Center, Washington, D.C., United States of America
| | - Rakesh Kumar
- Department of Biochemistry and Molecular Biology, The George Washington University Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
17
|
Ladoux B, Anon E, Lambert M, Rabodzey A, Hersen P, Buguin A, Silberzan P, Mège RM. Strength dependence of cadherin-mediated adhesions. Biophys J 2010; 98:534-42. [PMID: 20159149 DOI: 10.1016/j.bpj.2009.10.044] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 10/09/2009] [Accepted: 10/29/2009] [Indexed: 01/16/2023] Open
Abstract
Traction forces between adhesive cells play an important role in a number of collective cell processes. Intercellular contacts, in particular cadherin-based intercellular junctions, are the major means of transmitting force within tissues. We investigated the effect of cellular tension on the formation of cadherin-cadherin contacts by spreading cells on substrates with tunable stiffness coated with N-cadherin homophilic ligands. On the most rigid substrates, cells appear well-spread and present cadherin adhesions and cytoskeletal organization similar to those classically observed on cadherin-coated glass substrates. However, when cells are cultured on softer substrates, a change in morphology is observed: the cells are less spread, with a more disorganized actin network. A quantitative analysis of the cells adhering on the cadherin-coated surfaces shows that forces are correlated with the formation of cadherin adhesions. The stiffer the substrates, the larger are the average traction forces and the more developed are the cadherin adhesions. When cells are treated with blebbistatin to inhibit myosin II, the forces decrease and the cadherin adhesions disappear. Together, these findings are consistent with a mechanosensitive regulation of cadherin-mediated intercellular junctions through the cellular contractile machinery.
Collapse
Affiliation(s)
- Benoit Ladoux
- Laboratoire Matière et Systèmes Complexes, Centre National de la Recherche Scientifique, Unite Mixte de Recherche 7057, Université Paris Diderot, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Thapaliya P, Karlin NJ. An update on inflammatory breast cancer. Oncol Rev 2009. [DOI: 10.1007/s12156-009-0010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
19
|
Van Laere S, Beissbarth T, Van der Auwera I, Van den Eynden G, Trinh XB, Elst H, Van Hummelen P, van Dam P, Van Marck E, Vermeulen P, Dirix L. Relapse-free survival in breast cancer patients is associated with a gene expression signature characteristic for inflammatory breast cancer. Clin Cancer Res 2009; 14:7452-60. [PMID: 19010862 DOI: 10.1158/1078-0432.ccr-08-1077] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We hypothesize that a gene expression profile characteristic for inflammatory breast cancer (IBC), an aggressive form of breast cancer associated with rapid cancer dissemination and poor survival, might be related to tumor aggressiveness in non-IBC (nIBC). EXPERIMENTAL DESIGN RNA from 17 IBC samples and 40 nIBC samples was hybridized onto Affymetrix chips. A gene signature predictive of IBC was identified and applied onto 1,157 nIBC samples with survival data of 881 nIBC samples. Samples were classified as IBC-like or nIBC-like. The IBC signature classification was compared with the classifications according to other prognostically relevant gene signatures and clinicopathologic variables. In addition, relapse-free survival (RFS) was compared by the Kaplan-Meyer method. RESULTS Classification according to the IBC signature is significantly (P < 0.05) associated with the cell-of-origin subtypes, the wound healing response, the invasive gene signature, the genomic grade index, the fibroblastic neoplasm signature, and the 70-gene prognostic signature. Significant associations (P < 0.01) were found between the IBC signature and tumor grade, estrogen receptor status, ErbB2 status, and patient age at diagnosis. Patients with an IBC-like phenotype show a significantly shorter RFS interval (P < 0.05). Oncomine analysis identified cell motility as an important concept linked with the IBC signature. CONCLUSIONS We show that nIBC carcinomas having an IBC-like phenotype have a reduced RFS interval. This suggests that IBC and nIBC show comparable phenotypic traits, for example augmented cell motility, with respect to aggressive tumor cell behavior. This observation lends credit to the use of IBC to study aggressive tumor cell behavior.
Collapse
Affiliation(s)
- Steven Van Laere
- Translational Cancer Research Group, Laboratory of Pathology, University of Antwerp and Oncology Center, General Hospital Sint-Augustinus, Wilrijk, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Aberrant methylation of the Adenomatous Polyposis Coli (APC) gene promoter is associated with the inflammatory breast cancer phenotype. Br J Cancer 2008; 99:1735-42. [PMID: 18841156 PMCID: PMC2584952 DOI: 10.1038/sj.bjc.6604705] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aberrant methylation of the adenomatous polyposis coli (APC) gene promoter occurs in about 40% of breast tumours and has been correlated with reduced APC protein levels. To what extent epigenetic alterations of the APC gene may differ according to specific breast cancer phenotypes, remains to be elucidated. Our aim was to explore the role of APC methylation in the inflammatory breast cancer (IBC) phenotype. The status of APC gene promoter hypermethylation was investigated in DNA from normal breast tissues, IBC and non-IBC by both conventional and real-time quantitative methylation-specific PCR (MSP). APC methylation levels were compared with APC mRNA and protein levels. Hypermethylation of the APC gene promoter was present in 71% of IBC samples (n=21) and 43% of non-IBC samples (n=30) by conventional MSP (P=0.047). The APC gene also showed an increased frequency of high methylation levels in IBC (in 74% of cases, n=19) vs non-IBC (in 46% of cases, n=35) using a qMSP assay (P=0.048). We observed no significant association between APC methylation levels by qMSP and APC mRNA or protein expression levels. In conclusion, for the first time, we report the association of aberrant methylation of the APC gene promoter with the IBC phenotype, which might be of biological and clinical importance.
Collapse
|
21
|
Abstract
Imaging plays several key roles in the diagnosis and assessment of inflammatory breast cancer (IBC). These include characterization of the known tumor, delineation of locoregional disease in the ipsilateral and contralateral breast and regional lymph node basins, diagnosis of distant metastases, and evaluation of treatment response. We review the role of conventional imaging modalities, including mammography and sonography. We also discuss the potential of using evolving imaging modalities such as magnetic resonance imaging (MRI), positron emission tomography with computed tomography (PET/CT), and more advanced or emerging imaging techniques in the assessment of IBC.
Collapse
Affiliation(s)
- Carisa H Le-Petross
- Department of Diagnostic Radiology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | |
Collapse
|
22
|
Renz DM, Baltzer PAT, Böttcher J, Thaher F, Gajda M, Camara O, Runnebaum IB, Kaiser WA. Inflammatory breast carcinoma in magnetic resonance imaging: a comparison with locally advanced breast cancer. Acad Radiol 2008; 15:209-21. [PMID: 18206620 DOI: 10.1016/j.acra.2007.09.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 09/08/2007] [Accepted: 09/10/2007] [Indexed: 11/18/2022]
Abstract
RATIONALE AND OBJECTIVES Although inflammatory breast carcinoma (IBC) accounts for 1%-4% of all breast cancer cases, the appearance of this highly malignant tumor in magnetic resonance imaging (MRI) is still not well characterized. The aim of this study was to identify typical imaging features of IBC in comparison with noninflammatory locally advanced breast carcinoma (LABC). MATERIALS AND METHODS MRIs of 48 patients with IBC were compared with an equivalent cohort of 52 subjects with LABC. Age and histopathologic subtype were equivalent between the two groups. To delineate characteristic features, a multitude of dynamic and morphologic parameters were evaluated using T1- and T2-weighted sequences. RESULTS No significant differences of prevalences could be found for the following criteria: dynamic tumor signal characteristics, prominent vessels, perifocal edema, axillary lymph node involvement, morphology of focal masses, and morphologic pattern of non-mass like enhancement. Otherwise, the quantity of focal masses and the spatial distribution of the tumoral infiltration significantly differed between the two cancer groups. The following parameters occurred more frequently in the IBC cases: edema (cutaneous/subcutaneous 81.3%, perimamillar 70.8%, diffuse 89.6%, prepectoral 72.9%, intramuscular pectoral 41.7%), thickening (75.0%) and pathologic enhancement (60.4%) of Cooper's ligaments, skin thickening (83.3%), punched-out sign (initially strong, focal increase of some dermal or subcutaneous parts followed by slow-continuous enhancement of the surrounding skin; 56.3%). CONCLUSIONS Inflammatory breast carcinoma seems to represent a specific biological entity resulting in typical MRI characteristics. Some of the parameters are supposed to visualize the characteristic extensive lymphovascular infiltration and therefore may improve the diagnosis of IBC.
Collapse
Affiliation(s)
- Diane M Renz
- Institute of Diagnostic and Interventional Radiology, Friedrich-Schiller-University Jena, Erlanger Allee 101, Jena, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Güth U, Jane Huang D, Holzgreve W, Wight E, Singer G. T4 breast cancer under closer inspection: A case for revision of the TNM classification. Breast 2007; 16:625-36. [PMID: 17604172 DOI: 10.1016/j.breast.2007.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 05/13/2007] [Indexed: 01/27/2023] Open
Abstract
The presence of skin involvement in breast cancer results in the classification of the tumor into the highest tumor category, and accordingly into the highest non-metastatic disease stage (current TNM classification: T4/stage III). This traditional view is no longer justifiable, as tumors that show non-inflammatory skin involvement (T4b) make up a considerably heterogeneous group with a high percentage of small-sized tumors. Classifying all lesions demonstrating this feature together results in the combination of tumors with widely differing prognostic and therapeutic implications into a single group. This violates the basic principle of the TNM concept in that only tumors exhibiting similar extension and prognosis should be grouped into one category/stage. Furthermore, the currently valid definitions of non-inflammatory skin involvement are misconceived for the substantial group of small tumors which often have ambiguous morphologic findings: the clinical classification depends on the subjective perception of the individual observer, and the pathologic staging considers histologic criteria that are not justifiable from a functional-morphological point of view. For these reasons, we strongly feel that there is a need to revise the current T4 category. We recommend that breast carcinomas currently classified as T4a-c should be eliminated from the T4 category and classified simply according to their tumor size (T1-3). The prognostically very unfavorable inflammatory carcinoma (T4d) should be maintained as the only clinicopathologic entity in the T4 category. This proposal, which will also lead to a revision of the stage III group, adheres more closely to the goals and principles of the TNM classification than do the current classification guidelines. Through the revision of the T4 category, the definitions and guidelines of inflammatory breast carcinoma should be adapted to the internationally accepted nomenclature.
Collapse
Affiliation(s)
- Uwe Güth
- University Hospital Basel, Department of Gynecology and Obstetrics, Spitalstrasse 21, CH-4031 Basel, Switzerland.
| | | | | | | | | |
Collapse
|
24
|
Van Laere S, Van der Auwera I, Van den Eynden G, Van Hummelen P, van Dam P, Van Marck E, Vermeulen PB, Dirix L. Distinct molecular phenotype of inflammatory breast cancer compared to non-inflammatory breast cancer using Affymetrix-based genome-wide gene-expression analysis. Br J Cancer 2007; 97:1165-74. [PMID: 17848951 PMCID: PMC2360452 DOI: 10.1038/sj.bjc.6603967] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The present study aims at a platform-independent confirmation of previously obtained cDNA microarray results on inflammatory breast cancer (IBC) using Affymetrix chips. Gene-expression data of 19 IBC and 40 non-IBC specimens were subjected to clustering and principal component analysis. The performance of a previously identified IBC signature was tested using clustering and gene set enrichment analysis. The presence of different cell-of-origin subtypes in IBC was investigated and confirmed using immunohistochemistry on a TMA. Differential gene expression was analysed using SAM and topGO was used to identify the fingerprints of a pro-metastatic-signalling pathway. IBC and non-IBC have distinct gene-expression profiles. The differences in gene expression between IBC and non-IBC are captured within an IBC signature, identified in a platform-independent manner. Part of the gene-expression differences between IBC and non-IBC are attributable to the differential presence of the cell-of-origin subtypes, since IBC primarily segregated into the basal-like or ErbB2-overexpressing group. Strikingly, IBC tumour samples more closely resemble the gene-expression profile of T1/T2 tumours than the gene-expression profile or T3/T4 tumours. We identified the insulin-like growth factor-signalling pathway, potentially contributing to the biology of IBC. Our previous results have been validated in a platform-independent manner. The distinct biological behaviour of IBC is reflected in a distinct gene-expression profile. The fact that IBC tumours are quickly arising tumours might explain the close resemblance of the IBC gene-expression profile to the expression profile of T1/T2 tumours.
Collapse
Affiliation(s)
- S Van Laere
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| | - I Van der Auwera
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| | - G Van den Eynden
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| | - P Van Hummelen
- VIB Microarray Facility, UZ-Gasthuisberg, O&N, Leuven 3000, Belgium
| | - P van Dam
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| | - E Van Marck
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| | - P B Vermeulen
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
- Department Pathology – AZ Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium. E-mail:
| | - L Dirix
- Translational Cancer Research Group (Lab Pathology), University of Antwerp, Universiteitsplein 1, Wilrijk B2610, Belgium
- Oncology Center, General Hospital Sint-Augustinus, Oosterveldlaan 24, Wilrijk B2610, Belgium
| |
Collapse
|
25
|
Yang WT, Le-Petross HT, Macapinlac H, Carkaci S, Gonzalez-Angulo AM, Dawood S, Resetkova E, Hortobagyi GN, Cristofanilli M. Inflammatory breast cancer: PET/CT, MRI, mammography, and sonography findings. Breast Cancer Res Treat 2007; 109:417-26. [PMID: 17653852 DOI: 10.1007/s10549-007-9671-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Accepted: 06/26/2007] [Indexed: 11/27/2022]
Abstract
PURPOSE To describe the role of Positron Emission Tomography/Computed Tomography (PET/CT), Magnetic Resonance Imaging (MRI), sonography, and mammography in patients with inflammatory breast cancer (IBC). MATERIALS AND METHODS Patients who had been newly diagnosed with IBC and who had undergone mammography, sonography, MRI, PET/CT, or a combination of these were included in this study. The visibility of breast parenchymal lesion (BPLs), skin abnormalities, regional (axillary, supraclavicular, or internal mammary) nodal disease, and distant metastatic disease was documented with the imaging techniques. RESULTS Eighty patients (median age, 51 years, [range, 25-78 years]) were included in this study: 75 (94%) had undergone mammography, 76 (95%) sonography, 33 (41%) MRI, and 24 (30%) PET/CT. A primary BPL was found in 60 patients (80%) on mammography (mass or calcifications), 72 (95%) on sonography (mass or architectural distortion), 23 (96%) on PET/CT (hypermetabolic BPL), and 33 (100%) on MRI (enhancing BPL). Regional axillary nodal disease was found in 74 patients (93%) by histologic or cytologic examination, in 71 patients (93%) on sonography, in 21 (88%) on PET/CT, in 29 (88%) on MRI, and in 34 (45%) on mammography. Distant metastases in the bone, liver, and contralateral lymph nodes were diagnosed in nine patients (38%) on PET/CT. CONCLUSION MRI was the most accurate imaging technique in detecting a primary BPL in IBC patients. Sonography can be useful in diagnosing regional nodal disease. PET/CT provides additional information on distant metastasis, and it should be considered in the initial staging of IBC.
Collapse
Affiliation(s)
- Wei T Yang
- The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|