1
|
Boudin L, De Nonneville A, Finetti P, Guittard G, Nunes JA, Birnbaum D, Mamessier E, Bertucci F. CISH Expression Is Associated with Metastasis-Free Interval in Triple-Negative Breast Cancer and Refines the Prognostic Value of PDL1 Expression. Cancers (Basel) 2022; 14:3356. [PMID: 35884417 PMCID: PMC9316839 DOI: 10.3390/cancers14143356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Strategies are being explored to increase the efficiency of immune checkpoint inhibitors (ICIs) targeting PD1/PDL1 in triple-negative breast cancer (TNBC), including combination with therapies inhibiting intracellular immune checkpoints such as CISH (Cytokine-induced SH2 protein). Correlation between CISH expression and TNBC features is unknown. We retrospectively analyzed CISH expression in 1936 clinical TNBC samples and searched for correlations with clinical variables, including metastasis-free interval (MFI). Among TNBCs, 44% were identified as "CISH-up" and 56% "CISH-down". High expression was associated with pathological axillary lymph node involvement, more adjuvant chemotherapy, and Lehmann's immunomodulatory and luminal AR subtypes. The "CISH-up" class showed longer 5-year MFI (72%) than the "CISH-down" class (60%; p = 2.8 × 10-2). CISH upregulation was associated with activation of IFNα and IFNγ pathways, antitumor cytotoxic immune response, and signatures predictive for ICI response. When CISH and PDL1 were upregulated together, the 5-year MFI was 81% versus 52% when not upregulated (p = 6.21 × 10-6). The two-gene model provided more prognostic information than each gene alone and maintained its prognostic value in multivariate analysis. CISH expression is associated with longer MFI in TNBC and refines the prognostic value of PDL1 expression. Such observation might reinforce the therapeutic relevance of combining CISH inhibition with an anti-PD1/PDL1 ICI.
Collapse
Affiliation(s)
- Laurys Boudin
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
- Department of Medical Oncology, Hôpital d’Instruction des Armées Sainte-Anne, 83000 Toulon, France
| | - Alexandre De Nonneville
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
| | - Geoffrey Guittard
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (G.G.); (J.A.N.)
| | - Jacques A. Nunes
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (G.G.); (J.A.N.)
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
| | - François Bertucci
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, Inserm UMR1068, CNRS UMR725, 13009 Marseille, France; (L.B.); (A.D.N.); (P.F.); (D.B.); (E.M.)
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France
| |
Collapse
|
2
|
Bertucci F, Boudin L, Finetti P, Van Berckelaer C, Van Dam P, Dirix L, Viens P, Gonçalves A, Ueno NT, Van Laere S, Birnbaum D, Mamessier E. Immune landscape of inflammatory breast cancer suggests vulnerability to immune checkpoint inhibitors. Oncoimmunology 2021; 10:1929724. [PMID: 34104544 PMCID: PMC8158040 DOI: 10.1080/2162402x.2021.1929724] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background. Anti-PD1/PDL1 immune checkpoint inhibitors (ICIs) showed promising results in breast cancer, and exploration of additional actionable immune checkpoints is ongoing. Inflammatory breast cancer (IBC) is an aggressive form of disease, the immune tumor microenvironment (TME) of which is poorly known. We aimed at providing the first comprehensive immune portrait of IBCs. Methods. From the gene expression profiles of 137 IBC and 252 non-IBC clinical samples, we measured the fractions of 22 immune cell types, expression of signatures associated with tertiary lymphoid structures (TLS) and with the response to ICIs (T cell-inflamed signature: TIS) and of 18 genes coding for major actionable immune checkpoints. The IBC/non-IBC comparison was adjusted upon the clinicopathological variables. Results. The immune profiles of IBCs were heterogeneous. CIBERSORT analysis showed profiles rich in macrophages, CD8+ and CD4 + T-cells, with remarkable similarity with melanoma TME. The comparison with non-IBCs showed significant enrichment in M1 macrophages, γδ T-cells, and memory B-cells. IBCs showed higher expression of TLS and TIS signatures. The TIS signature displayed values in IBCs close to those observed in other cancers sensitive to ICIs. Two-thirds of actionable immune genes (HAVCR2/TIM3, CD27, CD70, CTLA4, ICOS, IDO1, LAG3, PDCD1, TNFRSF9, PVRIG, CD274/PDL1, and TIGIT) were overexpressed in IBCs as compared to normal breast and two-thirds were overexpressed in IBCs versus non-IBCs, with very frequent co-overexpression. For most of them, the overexpression was associated with better pathological response to chemotherapy. Conclusion. Our results suggest the potential higher vulnerability of IBC to ICIs. Clinical trials.
Collapse
Affiliation(s)
- François Bertucci
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Laurys Boudin
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Christophe Van Berckelaer
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Peter Van Dam
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium.,Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Patrice Viens
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Anthony Gonçalves
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France.,Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Naoto T Ueno
- Breast Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp Belgium.,Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| | - Emilie Mamessier
- Predictive Oncology Laboratory, "Equipe Labellisée Ligue Contre Le Cancer", Centre De Recherche En Cancérologie De Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille Université, Marseille, France
| |
Collapse
|
3
|
Elimimian EB, Samuel TA, Liang H, Elson L, Bilani N, Nahleh ZA. Clinical and Demographic Factors, Treatment Patterns, and Overall Survival Associated With Rare Triple-Negative Breast Carcinomas in the US. JAMA Netw Open 2021; 4:e214123. [PMID: 33844001 PMCID: PMC8042532 DOI: 10.1001/jamanetworkopen.2021.4123] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPORTANCE Triple-negative breast cancers are known collectively to demonstrate a more aggressive clinical course and earlier recurrence than cancers of other histological subtypes. However, the literature on rare triple-negative breast cancers and the association of histological type with survival and risk of metastasis is sparse. OBJECTIVE To present the clinical and demographic characteristics, treatment patterns, and overall survival (OS) for histologically rare (<10% of breast cancers) triple-negative breast cancer types: medullary carcinoma, adenoid cystic carcinoma, and metaplastic breast carcinoma. DESIGN, SETTING, AND PARTICIPANTS This cohort study was performed in the US using data reported by the National Cancer Database between 2010 and 2016. Confirmed cases of medullary carcinoma, adenoid cystic carcinoma, and metaplastic breast cancer were analyzed. Univariable analyses and multivariable Cox regression models were performed. Data analysis was performed from April to May 2020. MAIN OUTCOMES AND MEASURES The primary outcome was 5-year OS. Secondary outcomes included site of metastasis, effect of immunohistochemistry, management, and 2-year mortality. RESULTS A total of 8479 patients with breast cancer (mean [SD] age; 62.6 [14.3] years; 8435 women [99.48%]) were analyzed. Metaplastic carcinoma was the most commonly diagnosed histological type in this cohort, with 6867 patients (81%), followed by 1357 (16%) with adenoid cystic carcinoma and only 255 (3%) with medullary carcinoma. Medullary carcinoma presented earlier in life, at a median (interquartile range) age of 53 (45-62) years, compared with 62 (53-72) years for patients with adenoid cystic carcinoma and 63 (52-74) years for patients with metaplastic carcinoma. The proportion of tumors with triple-negative immunohistochemistry varied by histological type for medullary carcinoma (57 patients [22.4%]), adenoid cystic carcinoma (653 patients [48.1%]), and metaplastic carcinoma (3637 patients [53.0%]). Patients with adenoid cystic carcinoma were less likely to receive radiotherapy (711 patients [52.4%]) and chemotherapy (175 patients [12.9%]) compared with patients with medullary carcinoma (radiotherapy, 156 patients [61.2%]; chemotherapy, 190 patients [74.5%]) and metaplastic carcinoma (radiotherapy, 3416 patients [49.7%]; chemotherapy, 4709 patients [68.6%]). The 5-year OS rate was superior for patients with medullary (91.7%) and adenoid cystic carcinoma (88.4%) compared with patients with metaplastic carcinoma (63.1%). The 5-year mortality rate for adenoid cystic carcinoma was 8.33% vs 36.91% for metaplastic carcinoma. CONCLUSIONS AND RELEVANCE Nationally, over the course of 7 years, medullary carcinoma was most common and metaplastic carcinoma had the worst 5-year OS among the rare histological breast cancer subtypes analyzed. Factors associated with a poor prognosis for metaplastic carcinoma included advanced stage, lung metastasis, older age, and not receiving chemotherapy or radiation therapy. Future research focusing on rare subtypes of breast cancer is desirable and could inform the optimal management of these relatively understudied carcinomas.
Collapse
Affiliation(s)
- Elizabeth B. Elimimian
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| | - Thomas A. Samuel
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| | - Hong Liang
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| | - Leah Elson
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| | - Nadeem Bilani
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| | - Zeina A. Nahleh
- Department of Hematology-Oncology, Maroone Cancer Center, Cleveland Clinic Florida, Weston, Florida
| |
Collapse
|
4
|
Rocha D, García IA, González Montoro A, Llera A, Prato L, Girotti MR, Soria G, Fernández EA. Pan-Cancer Molecular Patterns and Biological Implications Associated with a Tumor-Specific Molecular Signature. Cells 2020; 10:E45. [PMID: 33396205 PMCID: PMC7823585 DOI: 10.3390/cells10010045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Studying tissue-independent components of cancer and defining pan-cancer subtypes could be addressed using tissue-specific molecular signatures if classification errors are controlled. Since PAM50 is a well-known, United States Food and Drug Administration (FDA)-approved and commercially available breast cancer signature, we applied it with uncertainty assessment to classify tumor samples from over 33 cancer types, discarded unassigned samples, and studied the emerging tumor-agnostic molecular patterns. The percentage of unassigned samples ranged between 55.5% and 86.9% in non-breast tissues, and gene set analysis suggested that the remaining samples could be grouped into two classes (named C1 and C2) regardless of the tissue. The C2 class was more dedifferentiated, more proliferative, with higher centrosome amplification, and potentially more TP53 and RB1 mutations. We identified 28 gene sets and 95 genes mainly associated with cell-cycle progression, cell-cycle checkpoints, and DNA damage that were consistently exacerbated in the C2 class. In some cancer types, the C1/C2 classification was associated with survival and drug sensitivity, and modulated the prognostic meaning of the immune infiltrate. Our results suggest that PAM50 could be repurposed for a pan-cancer context when paired with uncertainty assessment, resulting in two classes with molecular, biological, and clinical implications.
Collapse
Affiliation(s)
- Darío Rocha
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
| | - Iris A. García
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina;
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina;
| | - Aldana González Montoro
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
- Facultad de Matemática, Astronomía y Física, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Andrea Llera
- Laboratorio de Terapia Molecular y Celular—Genocan, Fundación Instituto Leloir, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1405BWE, Argentina;
| | - Laura Prato
- Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María, Villa María, Córdoba X5900, Argentina;
| | - María R. Girotti
- Laboratorio de Inmuno Oncología Traslacional, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1428ADN, Argentina;
| | - Gastón Soria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina;
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba X5000HUA, Argentina
| | - Elmer A. Fernández
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (D.R.); (A.G.M.)
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina;
| |
Collapse
|
5
|
Taurin S, Alkhalifa H. Breast cancers, mammary stem cells, and cancer stem cells, characteristics, and hypotheses. Neoplasia 2020; 22:663-678. [PMID: 33142233 PMCID: PMC7586061 DOI: 10.1016/j.neo.2020.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022]
Abstract
The cellular heterogeneity of breast cancers still represents a major therapeutic challenge. The latest genomic studies have classified breast cancers in distinct clusters to inform the therapeutic approaches and predict clinical outcomes. The mammary epithelium is composed of luminal and basal cells, and this seemingly hierarchical organization is dependent on various stem cells and progenitors populating the mammary gland. Some cancer cells are conceptually similar to the stem cells as they can self-renew and generate bulk populations of nontumorigenic cells. Two models have been proposed to explain the cell of origin of breast cancer and involve either the reprogramming of differentiated mammary cells or the dysregulation of mammary stem cells or progenitors. Both hypotheses are not exclusive and imply the accumulation of independent mutational events. Cancer stem cells have been isolated from breast tumors and implicated in the development, metastasis, and recurrence of breast cancers. Recent advances in single-cell sequencing help deciphering the clonal evolution within each breast tumor. Still, few clinical trials have been focused on these specific cancer cell populations.
Collapse
Affiliation(s)
- Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain.
| | - Haifa Alkhalifa
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
6
|
Shahdoust M, Mahjub H, Pezeshk H, Sadeghi M. A Network-Based Comparison Between Molecular Apocrine Breast Cancer Tumor and Basal and Luminal Tumors by Joint Graphical Lasso. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1555-1562. [PMID: 30990436 DOI: 10.1109/tcbb.2019.2911074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Joint graphical lasso (JGL) approach is a Gaussian graphical model to estimate multiple graphical models corresponding to distinct but related groups. Molecular apocrine (MA) breast cancer tumor has similar characteristics to luminal and basal subtypes. Due to the relationship between MA tumor and two other subtypes, this paper investigates the similarities and differences between the MA genes association network and the ones corresponding to other tumors by taking advantageous of JGL properties. Two distinct JGL graphical models are applied to two sub-datasets including the gene expression information of the MA and the luminal tumors and also the MA and the basal tumors. Then, topological comparisons between the networks such as finding the shared edges are applied. In addition, several support vector machine (SVM) classification models are performed to assess the discriminating power of some critical nodes in the networks, like hub nodes, to discriminate the tumors sample. Applying the JGL approach prepares an appropriate tool to observe the networks of the MA tumor and other subtypes in one map. The results obtained by comparing the networks could be helpful to generate new insight about MA tumor for future studies.
Collapse
|
7
|
Atukorale PU, Raghunathan SP, Raguveer V, Moon TJ, Zheng C, Bielecki PA, Wiese ML, Goldberg AL, Covarrubias G, Hoimes CJ, Karathanasis E. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNβ-Driven Antitumor Immunity. Cancer Res 2019; 79:5394-5406. [PMID: 31431457 DOI: 10.1158/0008-5472.can-19-0381] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/14/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNβ. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.
Collapse
Affiliation(s)
- Prabhani U Atukorale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Shruti P Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Vanitha Raguveer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Carolyn Zheng
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Peter A Bielecki
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Michelle L Wiese
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Amy L Goldberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Christopher J Hoimes
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Radiology, Case Western Reserve University, Cleveland, Ohio.,Case Center for Imaging Research, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
8
|
Savage SR, Shi Z, Liao Y, Zhang B. Graph Algorithms for Condensing and Consolidating Gene Set Analysis Results. Mol Cell Proteomics 2019; 18:S141-S152. [PMID: 31142576 PMCID: PMC6692773 DOI: 10.1074/mcp.tir118.001263] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/22/2019] [Indexed: 01/04/2023] Open
Abstract
Gene set analysis plays a critical role in the functional interpretation of omics data. Although this is typically done for one omics experiment at a time, there is an increasing need to combine gene set analysis results from multiple experiments performed on the same or different omics platforms, such as in multi-omics studies. Integrating results from multiple experiments is challenging, and annotation redundancy between gene sets further obscures clear conclusions. We propose to use a weighted set cover algorithm to reduce redundancy of gene sets identified in a single experiment. Next, we use affinity propagation to consolidate similar gene sets identified from multiple experiments into clusters and to automatically determine the most representative gene set for each cluster. Using three examples from over representation analysis and gene set enrichment analysis, we showed that weighted set cover outperformed a previously published set cover method and reduced the number of gene sets by 52-77%. Focusing on overlapping genes between the list of input genes and the enriched gene sets in over-representation analysis and leading-edge genes in gene set enrichment analysis further reduced the number of gene sets. A use case combining enrichment analysis results from RNA-Seq and proteomics data comparing basal and luminal A breast cancer samples highlighted the known difference in proliferation and DNA damage response. Finally, we used these algorithms for a pan-cancer survival analysis. Our analysis clearly revealed prognosis-related pathways common to multiple cancer types or specific to individual cancer types, as well as pathways associated with prognosis in different directions in different cancer types. We implemented these two algorithms in an R package, Sumer, which generates tables and static and interactive plots for exploration and publication. Sumer is publicly available at https://github.com/bzhanglab/sumer.
Collapse
Affiliation(s)
- Sara R Savage
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Yuxing Liao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
9
|
Association between mammographic density and tumor marker-defined breast cancer subtypes: a case-control study. Eur J Cancer Prev 2019; 27:239-247. [PMID: 28957821 DOI: 10.1097/cej.0000000000000353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High mammographic density (MD) is the most important risk factor for breast cancer. This study aimed to clarify the relationship between MD and breast cancer subtypes defined by tumor markers. We enrolled 642 women with breast cancer (69% premenopausal) and 1241 controls matched for age and menopausal status. Absolute mammographic dense area (ADA), percent mammographic dense area (PDA), and nondense area were assessed using a computer-assisted thresholding technique. We classified breast cancer cases into four subtypes using information on tumor marker expression such as estrogen receptor (ER), progesterone receptor (PR), and Cerb2 receptor (HER2); luminal A (ER+ and/or PR+, HER2-), luminal B (ER+ and/or PR+, HER2+), HER2-overexpressing (ER-, PR-, and HER2+), and triple-negative (ER-, PR-, and HER2-). Analysis was carried out using a conditional logistic regression model with adjustment for covariates. ADA and PDA were associated positively with the risk of breast cancer overall. Both ADA and PDA tended to have a positive association with breast cancer with any ER, any PR, or HER2-, but not for HER2+. The risk of luminal A breast cancer increased significantly 1.11 times (95% confidence interval: 1.01-1.23) for ADA and 1.12 times (95% confidence interval: 1.01-1.24) for PDA, estimated per 1 SD of the age and BMI-adjusted MD. However, the risk of breast cancer with luminal B, HER2-overexpressing, and triple-negative subtypes did not differ (P>0.10). Differential associations between MD measures and breast cancer by tumor marker status or tumor marker-defined subtypes were not detected. These findings suggested that the association between MD and breast cancer subtype may be because of other causal pathways.
Collapse
|
10
|
Yang K, Gao J, Luo M. Identification of key pathways and hub genes in basal-like breast cancer using bioinformatics analysis. Onco Targets Ther 2019; 12:1319-1331. [PMID: 30863098 PMCID: PMC6388944 DOI: 10.2147/ott.s158619] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Basal-like breast cancer (BLBC) is the most aggressive subtype of breast cancer (BC) and links to poor outcomes. As the molecular mechanism of BLBC has not yet been completely discovered, identification of key pathways and hub genes of this disease is an important way for providing new insights into exploring the mechanisms of BLBC initiation and progression. OBJECTIVE The aim of this study was to identify potential gene signatures of the development and progression of the BLBC via bioinformatics analysis. METHODS AND RESULTS The differential expressed genes (DEGs) including 40 up-regulated and 21 down-regulated DEGs were identified between GSE25066 and GSE21422 microarrays, and these DEGs were significantly enriched in the terms related to oncogenic or suppressive roles in BLBC progression. In addition, KEGG pathway and GSEA (Gene Set Enrichment Analysis) enrichment analyses were performed for DEGs between the basal type and non-basal-type breast cancer from GSE25066 microarray. These DEGs were enriched in pathways such as cell cycle, cytokine-cytokine receptor interaction, chemokine signaling pathway, central carbon metabolism signaling and TNF signaling pathway. Moreover, the protein-protein interaction (PPI) network was constructed with those 61 DEGs using the Cytoscape software, and the biological significance of putative modules was established using MCODE. The module 1 was found to be closely related with a term of mitosis regulation and enriched in cell cycle pathway, and thus confirmed the pathological characteristic of BLBC with a high mitotic index. Furthermore, prediction values of the top 10 hub genes such as CCNB2, BUB1, NDC80, CENPE, KIF2C, TOP2A, MELK, TPX2, CKS2 and KIF20A were validated using Oncomine and Kaplan-Meier plotter. CONCLUSION Our results suggest the intriguing possibility that the hub genes and modules in the PPI network contributed to in-depth knowledge about the molecular mechanism of BLBC, paving a way for more accurate discovery of potential treatment targets for BLBC patients.
Collapse
Affiliation(s)
- Kaidi Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China,
- Key Laboratory of Tumor Immunology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jian Gao
- Department of Life Sciences and Technology, Yangtze Normal University, Chongqing, China
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China,
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China,
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China,
| |
Collapse
|
11
|
Hua H, Zhang H, Kong Q, Jiang Y. Mechanisms for estrogen receptor expression in human cancer. Exp Hematol Oncol 2018; 7:24. [PMID: 30250760 PMCID: PMC6148803 DOI: 10.1186/s40164-018-0116-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023] Open
Abstract
Estrogen is a steroid hormone that has critical roles in reproductive development, bone homeostasis, cardiovascular remodeling and brain functions. However, estrogen also promotes mammary, ovarian and endometrial tumorigenesis. Estrogen antagonists and drugs that reduce estrogen biosynthesis have become highly successful therapeutic agents for breast cancer patients. The effects of estrogen are largely mediated by estrogen receptor (ER) α and ERβ, which are members of the nuclear receptor superfamily of transcription factors. The mechanisms underlying the aberrant expression of ER in breast cancer and other types of human tumors are complex, involving considerable alternative splicing of ERα and ERβ, transcription factors, epigenetic and post-transcriptional regulation of ER expression. Elucidation of mechanisms for ER expression may not only help understand cancer progression and evolution, but also shed light on overcoming endocrine therapy resistance. Herein, we review the complex mechanisms for regulating ER expression in human cancer.
Collapse
Affiliation(s)
- Hui Hua
- 1Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- 2Laboratory of Oncogene, West China Hospital, Sichuan University, Chengdu, China
| | - Qingbin Kong
- 2Laboratory of Oncogene, West China Hospital, Sichuan University, Chengdu, China
| | - Yangfu Jiang
- 2Laboratory of Oncogene, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Li X, Huang J, Luo X, Yang D, Yin X, Peng W, Bi C, Ren G, Xiang T. Paired box 5 is a novel marker of breast cancers that is frequently downregulated by methylation. Int J Biol Sci 2018; 14:1686-1695. [PMID: 30416383 PMCID: PMC6216036 DOI: 10.7150/ijbs.27599] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/15/2018] [Indexed: 12/20/2022] Open
Abstract
Identifying markers for breast cancer is important for both diagnosis and the design of treatment strategies. Recent studies have implicated Paired box 5 (PAX5) as a suppressor in various cancer types, where it is silenced by hypermethylation. However, determining the role of PAX5 in breast cancer requires further study, and the relationship between PAX5 methylation and breast cancer remains unclear. In this study, we found that PAX5 expression was frequently silenced or reduced by methylation in breast cancer cell lines as well as in breast cancer tissues. Restoring expression of PAX5 in breast cancer cells led to tumor suppression through inhibited proliferation and invasion, which resulted from modulation of the cell cycle and altered vascular endothelial growth factor (VEGF) expression. Most importantly, we found that PAX5 methylation status in breast cancer tissues was significantly correlated with patients' age, estrogen receptor (ER) status, progesterone receptor (PR) status, indicating that PAX5 could serve as a marker for breast cancer diagnosis and treatment strategy design.
Collapse
Affiliation(s)
- Xia Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianbo Huang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinrong Luo
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dejuan Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuedong Yin
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Can Bi
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrine and Breast Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Peiris PM, He F, Covarrubias G, Raghunathan S, Turan O, Lorkowski M, Gnanasambandam B, Wu C, Schiemann WP, Karathanasis E. Precise targeting of cancer metastasis using multi-ligand nanoparticles incorporating four different ligands. NANOSCALE 2018; 10:6861-6871. [PMID: 29620124 PMCID: PMC5908762 DOI: 10.1039/c8nr02513d] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Metastasis displays a highly heterogeneous cellular population with cancer cells continuously evolving. As a result, a single-ligand nanoparticle cannot account for the continuously changing expression of targetable biomarkers over time and space. To effectively direct nanoparticles to metastasis, we developed a multi-ligand nanoparticle by using four different types of ligands on the same nanoparticle that target biomarkers on the endothelium associated with metastatic disease. These vascular targets included αvβ3 integrin, P-selectin, EGFR and fibronectin. Using terminal and in vivo imaging studies, the targeting performance of the multi-ligand nanoparticles was compared to the single-ligand nanoparticle variants. All four single-ligand nanoparticle variants achieved significant targeting of lung metastasis in the 4T1 mouse model of breast cancer metastasis with about 2.5% of the injected dose being deposited into metastasis. A dual-ligand nanoparticle resulted in a nearly 2-fold higher deposition into lung metastases than its single-ligand counterparts. The multi-ligand nanoparticle significantly outperformed its targeting nanoparticle counterparts achieving a deposition of ∼7% of its injected nanoparticles into lung metastases. Using the high sensitivity of radionuclide imaging, PET imaging showed that a multi-ligand nanoparticle labeled with [18F]fluoride was able to precisely target metastatic disease at its very early stage of development in three different animal models of metastatic breast cancer.
Collapse
Affiliation(s)
- P M Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
M-Rabet M, Cabaud O, Josselin E, Finetti P, Castellano R, Farina A, Agavnian-Couquiaud E, Saviane G, Collette Y, Viens P, Gonçalves A, Ginestier C, Charafe-Jauffret E, Birnbaum D, Olive D, Bertucci F, Lopez M. Nectin-4: a new prognostic biomarker for efficient therapeutic targeting of primary and metastatic triple-negative breast cancer. Ann Oncol 2017; 28:769-776. [PMID: 27998973 DOI: 10.1093/annonc/mdw678] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Indexed: 12/17/2022] Open
Abstract
Background Triple-negative breast cancers (TNBCs) are associated with a poor prognosis. In contrast to other molecular subtypes, they have no identified specific target and chemotherapy remains the only available systemic treatment. The adhesion molecule nectin-4 represents a new potential therapeutic target in different cancer models. Here, we have tested the prognostic value of nectin-4 expression and assessed the therapeutic efficiency of an anti-nectin 4 antibody drug conjugate (ADC) on localised and metastatic TNBC in vitro and in vivo. Materials and methods We analysed nectin-4/PVRL4 mRNA expression in 5673 invasive breast cancers and searched for correlations with clinicopathological features including metastasis-free survival (MFS). Immunohistochemistry was carried out in 61 TNBCs and in samples of primary TNBC Patient-Derived Xenografts (PDXs). An anti-nectin-4 antibody eligible for ADC was produced and tested in vitro and in vivo in localised and metastatic TNBC PDXs. Results High nectin-4/PVRL4 mRNA expression was associated with poor-prognosis features including the TN and basal subtypes. High PVRL4 mRNA expression showed independent negative prognostic value for MFS in multivariate analysis in TNBCs. Nectin-4 protein expression was not detected in adult healthy tissues including mammary tissue. Membranous protein expression was found in 62% of TNBCs, with strong correlation with mRNA expression. We developed an ADC (N41mab-vcMMAE) comprising a human anti-nectin-4 monoclonal antibody conjugated to monomethyl auristatin-E (MMAE). In vitro, this ADC bound to nectin-4 with high affinity and specificity and induced its internalisation as well as dose-dependent cytotoxicity on nectin-4-expressing breast cancer cell lines. In vivo, this ADC induced rapid, complete and durable responses on nectin-4-positive xenograft TNBC samples including primary tumours, metastatic lesions, and local relapses; efficiency was dependent on both the dose and the nectin-4 tumour expression level. Conclusion Nectin-4 is both a new promising prognostic biomarker and specific therapeutic target for ADC in the very limited armamentarium against TNBC.
Collapse
Affiliation(s)
- M M-Rabet
- Centre de Cancérologie de Marseille, INSERM U1068, Equipe Immunité et Cancer, Institut Paoli-Calmettes, Aix-Marseille Université, CNRS, UMR7258, Marseille, France
| | - O Cabaud
- Molecular Oncology "Equipe labellisée Ligue Contre le Cancer," Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille, France
| | - E Josselin
- CRCM, INSERM, U1068; Institut Paoli-Calmettes; Aix-Marseille Université; CNRS, UMR 7258, Marseille, France
| | - P Finetti
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| | - R Castellano
- TrGET Platform, Inserm, U1068, Marseille F-13009, France
| | - A Farina
- ICEP Platform, Inserm, U1068, CRCM, Institut Paoli Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258
| | - E Agavnian-Couquiaud
- ICEP Platform, Inserm, U1068, CRCM, Institut Paoli Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258
| | - G Saviane
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| | - Y Collette
- TrGET Platform, Inserm, U1068, Marseille F-13009, France
| | - P Viens
- Département d'Oncologie Médicale, Institut Paoli Calmettes, Aix-Marseille Université, UM 105, Marseille, France
| | - A Gonçalves
- Département d'Oncologie Médicale, Institut Paoli Calmettes, Aix-Marseille Université, UM 105, Marseille, France
| | - C Ginestier
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| | - E Charafe-Jauffret
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| | - D Birnbaum
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| | - D Olive
- Centre de Cancérologie de Marseille, INSERM U1068, Equipe Immunité et Cancer, Institut Paoli-Calmettes, Aix-Marseille Université, CNRS, UMR7258, Marseille, France
| | - F Bertucci
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
- Département d'Oncologie Médicale, Institut Paoli Calmettes, Aix-Marseille Université, UM 105, Marseille, France
| | - M Lopez
- Aix Marseille Université, CNRS, INSERM , Institut Paoli-Calmettes, CRCM, Equipe Oncologie Moléculaire labellisée 'Ligue contre le cancer' , Marseille , France
| |
Collapse
|
15
|
Mumcuoglu M, Gurkan-Alp AS, Buyukbingol E, Cetin-Atalay R. Retinoid N-(1H-benzo[d]imidazol-2-yl)-5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalene-2-carboxamide induces p21-dependent senescence in breast cancer cells. Steroids 2016; 108:31-8. [PMID: 26898539 DOI: 10.1016/j.steroids.2016.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 01/20/2016] [Accepted: 02/11/2016] [Indexed: 12/27/2022]
Abstract
Retinoids have been implicated as pharmacological agents for the prevention and treatment of various types of cancers, including breast cancers. We analyzed 27 newly synthesized retinoids for their bioactivity on breast, liver, and colon cancer cells. Majority of the retinoids demonstrated selective bioactivity on breast cancer cells. Retinoid 17 had a significant inhibitory activity (IC50 3.5 μM) only on breast cancer cells while no growth inhibition observed with liver and colon cancer cells. The breast cancer selective growth inhibitory action by retinoid 17 was defined as p21-dependent cell death, reminiscent of senescence, which is an indicator of targeted receptor mediated bioactivity. A comparative analysis of retinoid receptor gene expression levels in different breast cancer cells and IC50 values of 17 indicated the involvement of Retinoid X receptors in the cytotoxic bioactivity of retinoid 17 in the senescence associated cell death. Furthermore, siRNA knockdown studies with RXRγ induced decrease in cell proliferation. Therefore, we suggest that retinoid derivatives that target RXRγ, can be considered for breast cancer therapies.
Collapse
Affiliation(s)
- Mine Mumcuoglu
- LOSEV the Foundation for Children with Leukemia, Cancer Genetics Research Laboratory, Ankara, Turkey; Department of Molecular Biology and Genetics, Bilkent University, Bilkent, 06800 Ankara, Turkey
| | - A Selen Gurkan-Alp
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Turkey
| | - Erdem Buyukbingol
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Turkey
| | - Rengul Cetin-Atalay
- LOSEV the Foundation for Children with Leukemia, Cancer Genetics Research Laboratory, Ankara, Turkey; Graduate School of Informatics, Cancer Systems Biology Laboratory, METU, 06800 Ankara, Turkey.
| |
Collapse
|
16
|
Perez-Janices N, Blanco-Luquin I, Torrea N, Liechtenstein T, Escors D, Cordoba A, Vicente-Garcia F, Jauregui I, De La Cruz S, Illarramendi JJ, Coca V, Berdasco M, Kochan G, Ibañez B, Lera JM, Guerrero-Setas D. Differential involvement of RASSF2 hypermethylation in breast cancer subtypes and their prognosis. Oncotarget 2015; 6:23944-58. [PMID: 26284587 PMCID: PMC4695163 DOI: 10.18632/oncotarget.4062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a heterogeneous disease that can be subdivided into clinical, histopathological and molecular subtypes (luminal A-like, luminal B-like/HER2-negative, luminal B-like/HER2-positive, HER2-positive, and triple-negative). The study of new molecular factors is essential to obtain further insights into the mechanisms involved in the tumorigenesis of each tumor subtype. RASSF2 is a gene that is hypermethylated in breast cancer and whose clinical value has not been previously studied. The hypermethylation of RASSF1 and RASSF2 genes was analyzed in 198 breast tumors of different subtypes. The effect of the demethylating agent 5-aza-2'-deoxycytidine in the re-expression of these genes was examined in triple-negative (BT-549), HER2 (SK-BR-3), and luminal cells (T-47D). Different patterns of RASSF2 expression for distinct tumor subtypes were detected by immunohistochemistry. RASSF2 hypermethylation was much more frequent in luminal subtypes than in non-luminal tumors (p = 0.001). The re-expression of this gene by lentiviral transduction contributed to the differential cell proliferation and response to antineoplastic drugs observed in luminal compared with triple-negative cell lines. RASSF2 hypermethylation is associated with better prognosis in multivariate statistical analysis (P = 0.039). In conclusion, RASSF2 gene is differently methylated in luminal and non-luminal tumors and is a promising suppressor gene with clinical involvement in breast cancer.
Collapse
Affiliation(s)
- Noemi Perez-Janices
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet (FMS), Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Navarra, Spain
- Division of Infection and Immunity, Rayne Institute, University College London (UCL), London, United Kingdom
| | - Idoia Blanco-Luquin
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet (FMS), Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Navarra, Spain
- Division of Infection and Immunity, Rayne Institute, University College London (UCL), London, United Kingdom
- Cancer Immunomodulation Group, Navarrabiomed-Fundacion Miguel Servet, IdiSNA, Navarra, Spain
| | - Natalia Torrea
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Therese Liechtenstein
- Division of Infection and Immunity, Rayne Institute, University College London (UCL), London, United Kingdom
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - David Escors
- Division of Infection and Immunity, Rayne Institute, University College London (UCL), London, United Kingdom
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alicia Cordoba
- Department of Pathology, Complejo Hospitalario de Navarra, Navarra Health Service, Pamplona, Navarra, Spain
| | | | - Isabel Jauregui
- Department of Pathology, Complejo Hospitalario de Navarra, Navarra Health Service, Pamplona, Navarra, Spain
| | - Susana De La Cruz
- Department of Medical Oncology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - José Juan Illarramendi
- Department of Medical Oncology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - Valle Coca
- Biobank Unit, Navarrabiomed-Fundacion Miguel Servet, IdiSNA, Navarra, Spain
| | - Maria Berdasco
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Programme (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Grazyna Kochan
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Berta Ibañez
- Red de Evaluación en Servicios Sanitarios y Enfermedades Cronicas (REDISSEC), Navarrabiomed-Fundación Miguel Servet, IdiSNA, Navarra, Spain
| | - José Miguel Lera
- Department of Surgery, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - David Guerrero-Setas
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet (FMS), Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Navarra, Spain
| |
Collapse
|
17
|
Panis C, Pizzatti L, Herrera AC, Corrêa S, Binato R, Abdelhay E. Label-Free Proteomic Analysis of Breast Cancer Molecular Subtypes. J Proteome Res 2014; 13:4752-72. [DOI: 10.1021/pr500676x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Carolina Panis
- Laboratório
de Células Tronco, Instituto Nacional do Câncer, INCA, Rio de
Janeiro, Brazil
- Laboratório
de Mediadores Inflamatórios, Universidade Estadual do Oeste do Paraná, UNIOESTE, Campus Francisco Beltrão, Paraná, Brazil
| | - Luciana Pizzatti
- Laboratório
de Células Tronco, Instituto Nacional do Câncer, INCA, Rio de
Janeiro, Brazil
- Departamento
de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Ana Cristina Herrera
- Pontifícia Universidade Católica do Paraná, PUC−PR, Campus Londrina, Londrina, Paraná, Brazil
| | - Stephany Corrêa
- Laboratório
de Células Tronco, Instituto Nacional do Câncer, INCA, Rio de
Janeiro, Brazil
| | - Renata Binato
- Laboratório
de Células Tronco, Instituto Nacional do Câncer, INCA, Rio de
Janeiro, Brazil
| | - Eliana Abdelhay
- Laboratório
de Células Tronco, Instituto Nacional do Câncer, INCA, Rio de
Janeiro, Brazil
| |
Collapse
|
18
|
Razzaghi H, Troester MA, Gierach GL, Olshan AF, Yankaskas BC, Millikan RC. Association between mammographic density and basal-like and luminal A breast cancer subtypes. Breast Cancer Res 2014; 15:R76. [PMID: 24008056 PMCID: PMC3978452 DOI: 10.1186/bcr3470] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 07/18/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction Mammographic density is a strong risk factor for breast cancer overall, but few studies have examined the association between mammographic density and specific subtypes of breast cancer, especially aggressive basal-like breast cancers. Because basal-like breast cancers are less frequently screen-detected, it is important to understand how mammographic density relates to risk of basal-like breast cancer. Methods We estimated associations between mammographic density and breast cancer risk according to breast cancer subtype. Cases and controls were participants in the Carolina Breast Cancer Study (CBCS) who also had mammograms recorded in the Carolina Mammography Registry (CMR). A total of 491 cases had mammograms within five years prior to and one year after diagnosis and 528 controls had screening or diagnostic mammograms close to the dates of selection into CBCS. Mammographic density was reported to the CMR using Breast Imaging Reporting and Data System categories. The expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 1 and 2 (HER1 and HER2), and cytokeratin 5/6 (CK5/6) were assessed by immunohistochemistry and dichotomized as positive or negative, with ER+ and/or PR+, and HER2- tumors classified as luminal A and ER-, PR-, HER2-, HER1+ and/or CK5/6+ tumors classified as basal-like breast cancer. Triple negative tumors were defined as negative for ER, PR and HER2. Of the 491 cases 175 were missing information on subtypes; the remaining cases included 181 luminal A, 17 luminal B, 48 basal-like, 29 ER-/PR-/HER2+, and 41 unclassified subtypes. Odds ratios comparing each subtype to all controls and case-case odds ratios comparing mammographic density distributions in basal-like to luminal A breast cancers were estimated using logistic regression. Results Mammographic density was associated with increased risk of both luminal A and basal-like breast cancers, although estimates were imprecise. The magnitude of the odds ratio associated with mammographic density was not substantially different between basal-like and luminal A cancers in case–control analyses and case-case analyses (case-case OR = 1.08 (95% confidence interval: 0.30, 3.84)). Conclusions These results suggest that risk estimates associated with mammographic density are not distinct for separate breast cancer subtypes (basal-like/triple negative vs. luminal A breast cancers). Studies with a larger number of basal-like breast cancers are needed to confirm our findings.
Collapse
|
19
|
Peiris PM, Toy R, Abramowski A, Vicente P, Tucci S, Bauer L, Mayer A, Tam M, Doolittle E, Pansky J, Tran E, Lin D, Schiemann WP, Ghaghada KB, Griswold MA, Karathanasis E. Treatment of cancer micrometastasis using a multicomponent chain-like nanoparticle. J Control Release 2013; 173:51-8. [PMID: 24188960 DOI: 10.1016/j.jconrel.2013.10.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/17/2013] [Accepted: 10/25/2013] [Indexed: 12/30/2022]
Abstract
While potent cytotoxic agents are available to oncologists, the clinical utility of these agents is limited due to their non-specific distribution in the body and toxicity to normal tissues leading to use of suboptimal doses for eradication of metastatic disease. Furthermore, treatment of micrometastases is impeded by several biobarriers, including their small size and high dispersion to organs, making them nearly inaccessible to drugs. To circumvent these limitations in treating metastatic disease, we developed a multicomponent, flexible chain-like nanoparticle (termed nanochain) that possesses a unique ability to gain access to and be deposited at micrometastatic sites. Moreover, coupling nanochain particles to radiofrequency (RF)-triggered cargo delivery facilitated widespread delivery of drug into hard-to-reach cancer cells. Collectively, these features synergistically facilitate effective treatment and ultimately eradication of micrometastatic disease using a low dose of a cytotoxic drug.
Collapse
Affiliation(s)
- Pubudu M Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Randall Toy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Aaron Abramowski
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Pete Vicente
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Samantha Tucci
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lisa Bauer
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Physics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Aaron Mayer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Morgan Tam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Elizabeth Doolittle
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jenna Pansky
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Emily Tran
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dishen Lin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ketan B Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX 77030, USA; Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark A Griswold
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
20
|
Myc is required for β-catenin-mediated mammary stem cell amplification and tumorigenesis. Mol Cancer 2013; 12:132. [PMID: 24171719 PMCID: PMC4176121 DOI: 10.1186/1476-4598-12-132] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/23/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Basal-like breast cancer is a heterogeneous disease characterized by the expression of basal cell markers, no estrogen or progesterone receptor expression and a lack of HER2 overexpression. Recent studies have linked activation of the Wnt/β-catenin pathway, and its downstream target, Myc, to basal-like breast cancer. Transgenic mice K5ΔNβcat previously generated by our team present a constitutive activation of Wnt/β-catenin signaling in the basal myoepithelial cell layer, resulting in focal mammary hyperplasias that progress to invasive carcinomas. Mammary lesions developed by K5ΔNβcat mice consist essentially of basal epithelial cells that, in contrast to mammary myoepithelium, do not express smooth muscle markers. METHODS Microarray analysis was used to compare K5ΔNβcat mouse tumors to human breast tumors, mammary cancer cell lines and the tumors developed in other mouse models. Cre-Lox approach was employed to delete Myc from the mammary basal cell layer of K5ΔNβcat mice. Stem cell amplification in K5ΔNβcat mouse mammary epithelium was assessed with 3D-culture and transplantation assays. RESULTS Histological and microarray analyses of the mammary lesions of K5ΔNβcat females revealed their high similarity to a subset of basal-like human breast tumors with squamous differentiation. As in human basal-like carcinomas, the Myc pathway appeared to be activated in the mammary lesions of K5ΔNβcat mice. We found that a basal cell population with stem/progenitor characteristics was amplified in K5ΔNβcat mouse preneoplastic glands. Finally, the deletion of Myc from the mammary basal layer of K5ΔNβcat mice not only abolished the regenerative capacity of basal epithelial cells, but, in addition, completely prevented the tumorigenesis. CONCLUSIONS These results strongly indicate that β-catenin-induced stem cell amplification and tumorigenesis rely ultimately on the Myc pathway activation and reinforce the hypothesis that basal stem/progenitor cells may be at the origin of a subset of basal-like breast tumors.
Collapse
|
21
|
Charafe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski J, Finetti P, Josselin E, Adelaide J, Nguyen TT, Monville F, Jacquemier J, Thomassin-Piana J, Pinna G, Jalaguier A, Lambaudie E, Houvenaeghel G, Xerri L, Harel-Bellan A, Chaffanet M, Viens P, Birnbaum D. ALDH1-positive cancer stem cells predict engraftment of primary breast tumors and are governed by a common stem cell program. Cancer Res 2013; 73:7290-300. [PMID: 24142344 DOI: 10.1158/0008-5472.can-12-4704] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer stem-like cells (CSC) have been widely studied, but their clinical relevance has yet to be established in breast cancer. Here, we report the establishment of primary breast tumor-derived xenografts (PDX) that encompass the main diversity of human breast cancer and retain the major clinicopathologic features of primary tumors. Successful engraftment was correlated with the presence of ALDH1-positive CSCs, which predicted prognosis in patients. The xenografts we developed showed a hierarchical cell organization of breast cancer with the ALDH1-positive CSCs constituting the tumorigenic cell population. Analysis of gene expression from functionally validated CSCs yielded a breast CSC signature and identified a core transcriptional program of 19 genes shared with murine embryonic, hematopoietic, and neural stem cells. This generalized stem cell program allowed the identification of potential CSC regulators, which were related mainly to metabolic processes. Using an siRNA genetic screen designed to target the 19 genes, we validated the functional role of this stem cell program in the regulation of breast CSC biology. Our work offers a proof of the functional importance of CSCs in breast cancer, and it establishes the reliability of PDXs for use in developing personalized CSC therapies for patients with breast cancer.
Collapse
Affiliation(s)
- Emmanuelle Charafe-Jauffret
- Authors' Affiliations: INSERM, U1068, CRCM, Molecular Oncology, Institut Paoli-Calmettes, Biopathology, Aix-Marseille Univ, UM 105, F-13284, Départements d'Oncologie Médicale, Chirugie oncologique, and Radiologie, Institut Paoli-Calmettes, Marseille; and Plateforme ARN interference PArI, CEA SACLAY, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Peiris PM, Tam M, Vicente P, Abramowski A, Toy R, Bauer L, Mayer A, Pansky J, Doolittle E, Tucci S, Schmidt E, Shoup C, Rao S, Murray K, Gopalakrishnan R, Keri RA, Basilion JP, Griswold MA, Karathanasis E. On-command drug release from nanochains inhibits growth of breast tumors. Pharm Res 2013; 31:1460-8. [PMID: 23934254 DOI: 10.1007/s11095-013-1102-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE To evaluate the ability of radiofrequency (RF)-triggered drug release from a multicomponent chain-shaped nanoparticle to inhibit the growth of an aggressive breast tumor. METHODS A two-step solid phase chemistry was employed to synthesize doxorubicin-loaded nanochains, which were composed of three iron oxide nanospheres and one doxorubicin-loaded liposome assembled in a 100-nm-long linear nanochain. The nanochains were tested in the 4T1-LUC-GFP orthotopic mouse model, which is a highly aggressive breast cancer model. The 4T1-LUC-GFP cell line stably expresses firefly luciferase, which allowed the non-invasive in vivo imaging of tumor response to the treatment using bioluminescence imaging (BLI). RESULTS Longitudinal BLI imaging showed that a single nanochain treatment followed by application of RF resulted in an at least 100-fold lower BLI signal compared to the groups treated with nanochains (without RF) or free doxorubicin followed by RF. A statistically significant increase in survival time of the nanochain-treated animals followed by RF (64.3 days) was observed when compared to the nanochain-treated group without RF (35.7 days), free doxorubicin-treated group followed by RF (38.5 days), and the untreated group (30.5 days; n=5 animals per group). CONCLUSIONS These studies showed that the combination of RF and nanochains has the potential to effectively treat highly aggressive cancers and prolong survival.
Collapse
Affiliation(s)
- Pubudu M Peiris
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bertucci F, Bouvier-Labit C, Finetti P, Metellus P, Adelaide J, Mokhtari K, Figarella-Branger D, Decouvelaere AV, Miquel C, Coindre JM, Birnbaum D. Gene expression profiling of solitary fibrous tumors. PLoS One 2013; 8:e64497. [PMID: 23734203 PMCID: PMC3667191 DOI: 10.1371/journal.pone.0064497] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/15/2013] [Indexed: 12/12/2022] Open
Abstract
Background Solitary fibrous tumors (SFTs) are rare spindle-cell tumors. Their cell-of-origin and molecular basis are poorly known. They raise several clinical problems. Differential diagnosis may be difficult, prognosis is poorly apprehended by histoclinical features, and no effective therapy exists for advanced stages. Methods We profiled 16 SFT samples using whole-genome DNA microarrays and analyzed their expression profiles with publicly available profiles of 36 additional SFTs and 212 soft tissue sarcomas (STSs). Immunohistochemistry was applied to validate the expression of some discriminating genes. Results SFTs displayed whole-genome expression profiles more homogeneous and different from STSs, but closer to genetically-simple than genetically-complex STSs. The SFTs/STSs comparison identified a high percentage (∼30%) of genes as differentially expressed, most of them without any DNA copy number alteration. One of the genes most overexpressed in SFTs encoded the ALDH1 stem cell marker. Several upregulated genes and associated ontologies were also related to progenitor/stem cells. SFTs also overexpressed genes encoding therapeutic targets such as kinases (EGFR, ERBB2, FGFR1, JAK2), histone deacetylases, or retinoic acid receptors. Their overexpression was found in all SFTs, regardless the anatomical location. Finally, we identified a 31-gene signature associated with the mitotic count, containing many genes related to cell cycle/mitosis, including AURKA. Conclusion We established a robust repertoire of genes differentially expressed in SFTs. Certain overexpressed genes could provide new diagnostic (ALDH1A1), prognostic (AURKA) and/or therapeutic targets.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm; Marseille, France
- Département d'Oncologie Médicale, IPC, CRCM, UMR1068 Inserm, Marseille, France
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
- * E-mail:
| | - Corinne Bouvier-Labit
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
- Département d’Anatomopathologie, Hôpital de la Timone, Marseille, France
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm; Marseille, France
| | - Philippe Metellus
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
- Département de Neurochirurgie, Hôpital de la Timone, Marseille, France
| | - José Adelaide
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm; Marseille, France
| | - Karima Mokhtari
- Département de Neuropathologie, Hôpital Pitié Salpétrière, Paris, France
| | - Dominique Figarella-Branger
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
- Département d’Anatomopathologie, Hôpital de la Timone, Marseille, France
| | | | - Catherine Miquel
- Département de Neuropathologie, Hôpital Sainte Anne, Paris, France
| | | | - Daniel Birnbaum
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm; Marseille, France
| |
Collapse
|
24
|
Abou-Bakr AA, Eldweny HI. p16 expression correlates with basal-like triple-negative breast carcinoma. Ecancermedicalscience 2013; 7:317. [PMID: 23717338 PMCID: PMC3660155 DOI: 10.3332/ecancer.2013.317] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Indexed: 12/31/2022] Open
Abstract
Background: Basal-like breast carcinoma (BLBC) has attracted considerable attention over the past few years. It has been suggested that tumours expressing basal markers have a more aggressive clinical behaviour. However, a molecular basis for this disease remains unclear, and it lacks currently used therapeutic targets. Therefore developing a novel treatment strategy is crucial for improving the prognosis. The aim of this study was to characterise the immunohistochemical (IHC) expression of p16 in patients with BLBC compared with non-BLBC. Materials and methods: Eighty-five cases of grade-3 invasive ductal carcinomas not otherwise specified (IDC-NOS) were analyzed. Immunohistochemical stains for oestrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor type 2 (HER2), cytokeratin (CK) 5/6, epidermal growth factor receptor (EGFR) and p16 were performed. BLBC was defined as ER-, PR-, Her2- and CK5/6+, and/or EGFR+. Results: Twenty cases were categorised as BLBC versus 65 as non-basal. High mitotic count and presence of necrosis were associated with basal-like phenotype. Distant metastasis developed in 40% of cases of BLBC with frequent spread to brain and lung. p16 had significantly higher expression in the basal subgroup (80% versus 50.8%, P = 0.04). Patients with BLBCs were found to have a lower disease-free survival (DFS) rate (60% versus 70.8%, P = 0.03). Conclusion: BLBC typically demonstrates a unique profile. p16 is frequently expressed in breast cancers with basal-like phenotype; this suggests that p16 may play a role in the poor prognosis of this tumour, and it may be used in the development of a targeted therapy that will result in improved patient prognostication and outcome.
Collapse
Affiliation(s)
- Amany A Abou-Bakr
- Department of Pathology, National Cancer Institute, Cairo University, Egypt
| | | |
Collapse
|
25
|
Shields BJ, Wiede F, Gurzov EN, Wee K, Hauser C, Zhu HJ, Molloy TJ, O'Toole SA, Daly RJ, Sutherland RL, Mitchell CA, McLean CA, Tiganis T. TCPTP regulates SFK and STAT3 signaling and is lost in triple-negative breast cancers. Mol Cell Biol 2013; 33:557-70. [PMID: 23166300 PMCID: PMC3554209 DOI: 10.1128/mcb.01016-12] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 11/12/2012] [Indexed: 01/08/2023] Open
Abstract
Tyrosine phosphorylation-dependent signaling, as mediated by members of the epidermal growth factor receptor (EGFR) family (ErbB1 to -4) of protein tyrosine kinases (PTKs), Src family PTKs (SFKs), and cytokines such as interleukin-6 (IL-6) that signal via signal transducer and activator of transcription 3 (STAT3), is critical to the development and progression of many human breast cancers. EGFR, SFKs, and STAT3 can serve as substrates for the protein tyrosine phosphatase TCPTP (PTPN2). Here we report that TCPTP protein levels are decreased in a subset of breast cancer cell lines in vitro and that TCPTP protein is absent in a large proportion of "triple-negative" primary human breast cancers. Homozygous TCPTP deficiency in murine mammary fat pads in vivo is associated with elevated SFK and STAT3 signaling, whereas TCPTP deficiency in human breast cancer cell lines enhances SFK and STAT3 signaling. On the other hand, TCPTP reconstitution in human breast cancer cell lines severely impaired cell proliferation and suppressed anchorage-independent growth in vitro and xenograft growth in vivo. These studies establish TCPTP's potential to serve as a tumor suppressor in human breast cancer.
Collapse
Affiliation(s)
- Benjamin J. Shields
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Esteban N. Gurzov
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kenneth Wee
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Christine Hauser
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Timothy J. Molloy
- The Kinghorn Cancer Centre & Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Sandra A. O'Toole
- The Kinghorn Cancer Centre & Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Central Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Roger J. Daly
- The Kinghorn Cancer Centre & Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Kensington, New South Wales, Australia
| | - Robert L. Sutherland
- The Kinghorn Cancer Centre & Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Kensington, New South Wales, Australia
| | - Christina A. Mitchell
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Catriona A. McLean
- Department of Anatomical Pathology, Alfred Hospital, Prahran, Victoria, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
26
|
Ginestier C, Monville F, Wicinski J, Cabaud O, Cervera N, Josselin E, Finetti P, Guille A, Larderet G, Viens P, Sebti S, Bertucci F, Birnbaum D, Charafe-Jauffret E. Mevalonate metabolism regulates Basal breast cancer stem cells and is a potential therapeutic target. Stem Cells 2012; 30:1327-37. [PMID: 22605458 DOI: 10.1002/stem.1122] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is increasing evidence that breast tumors are organized in a hierarchy, with a subpopulation of tumorigenic cancer cells, the cancer stem cells (CSCs), which sustain tumor growth. The characterization of protein networks that govern CSC behavior is paramount to design new therapeutic strategies targeting this subpopulation of cells. We have sought to identify specific molecular pathways of CSCs isolated from 13 different breast cancer cell lines of luminal or basal/mesenchymal subtypes. We compared the gene expression profiling of cancer cells grown in adherent conditions to those of matched tumorsphere cultures. No specific pathway was identified to be commonly regulated in luminal tumorspheres, resulting from a minor CSC enrichment in tumorsphere passages from luminal cell lines. However, in basal/mesenchymal tumorspheres, the enzymes of the mevalonate metabolic pathway were overexpressed compared to those in cognate adherent cells. Inhibition of this pathway with hydroxy-3-methylglutaryl CoA reductase blockers resulted in a reduction of breast CSC independent of inhibition of cholesterol biosynthesis and of protein farnesylation. Further modulation of this metabolic pathway demonstrated that protein geranylgeranylation (GG) is critical to breast CSC maintenance. A small molecule inhibitor of the geranylgeranyl transferase I (GGTI) enzyme reduced the breast CSC subpopulation both in vitro and in primary breast cancer xenografts. We found that the GGTI effect on the CSC subpopulation is mediated by inactivation of Ras homolog family member A (RHOA) and increased accumulation of P27(kip1) in the nucleus. The identification of protein GG as a major contributor to CSC maintenance opens promising perspectives for CSC targeted therapy in basal breast cancer.
Collapse
Affiliation(s)
- Christophe Ginestier
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, INSERM, Institut Paoli-Calmettes, 27 Boulevard Leï Roure, Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ossovskaya V, Wang Y, Budoff A, Xu Q, Lituev A, Potapova O, Vansant G, Monforte J, Daraselia N. Exploring molecular pathways of triple-negative breast cancer. Genes Cancer 2012; 2:870-9. [PMID: 22593799 DOI: 10.1177/1947601911432496] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a high rate of proliferation and metastasis, as well as poor prognosis for advanced-stage disease. Although TNBC was previously classified together with basal-like and BRCA1/2-related breast cancers, genomic profiling now shows that there is incomplete overlap, with important distinctions associated with each subtype. The biology of TNBC is still poorly understood; therefore, to define the relative contributions of major cellular pathways in TNBC, we have studied its molecular signature based on analysis of gene expression. Comparisons were then made with normal breast tissue. Our results suggest the existence of molecular networks in TNBC, characterized by explicit alterations in the cell cycle, DNA repair, nucleotide synthesis, metabolic pathways, NF-κB signaling, inflammatory response, and angiogenesis. Moreover, we also characterized TNBC as a cancer of mixed phenotypes, suggesting that TNBC extends beyond the basal-like molecular signature and may constitute an independent subtype of breast cancer. The data provide a new insight into the biology of TNBC.
Collapse
Affiliation(s)
- Valeria Ossovskaya
- BiPar Sciences, Inc. (subsidiary of Sanofi), South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Carracedo A, Weiss D, Leliaert AK, Bhasin M, de Boer VCJ, Laurent G, Adams AC, Sundvall M, Song SJ, Ito K, Finley LS, Egia A, Libermann T, Gerhart-Hines Z, Puigserver P, Haigis MC, Maratos-Flier E, Richardson AL, Schafer ZT, Pandolfi PP. A metabolic prosurvival role for PML in breast cancer. J Clin Invest 2012; 122:3088-100. [PMID: 22886304 DOI: 10.1172/jci62129] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 07/05/2012] [Indexed: 12/26/2022] Open
Abstract
Cancer cells exhibit an aberrant metabolism that facilitates more efficient production of biomass and hence tumor growth and progression. However, the genetic cues modulating this metabolic switch remain largely undetermined. We identified a metabolic function for the promyelocytic leukemia (PML) gene, uncovering an unexpected role for this bona fide tumor suppressor in breast cancer cell survival. We found that PML acted as both a negative regulator of PPARγ coactivator 1A (PGC1A) acetylation and a potent activator of PPAR signaling and fatty acid oxidation. We further showed that PML promoted ATP production and inhibited anoikis. Importantly, PML expression allowed luminal filling in 3D basement membrane breast culture models, an effect that was reverted by the pharmacological inhibition of fatty acid oxidation. Additionally, immunohistochemical analysis of breast cancer biopsies revealed that PML was overexpressed in a subset of breast cancers and enriched in triple-negative cases. Indeed, PML expression in breast cancer correlated strikingly with reduced time to recurrence, a gene signature of poor prognosis, and activated PPAR signaling. These findings have important therapeutic implications, as PML and its key role in fatty acid oxidation metabolism are amenable to pharmacological suppression, a potential future mode of cancer prevention and treatment.
Collapse
Affiliation(s)
- Arkaitz Carracedo
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gamble C, McIntosh K, Scott R, Ho KH, Plevin R, Paul A. Inhibitory kappa B Kinases as targets for pharmacological regulation. Br J Pharmacol 2012; 165:802-19. [PMID: 21797846 PMCID: PMC3312479 DOI: 10.1111/j.1476-5381.2011.01608.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 06/20/2011] [Accepted: 07/02/2011] [Indexed: 01/04/2023] Open
Abstract
The inhibitory kappa B kinases (IKKs) are well recognized as key regulators of the nuclear factor kappa B (NF-κB) cascade and as such represent a point of convergence for many extracellular agents that activate this pathway. The IKKs generally serve to transduce pro-inflammatory and growth stimulating signals that contribute to major cellular processes but also play a key role in the pathogenesis of a number of human diseases. Therefore, the catalytic IKKs represent attractive targets for intervention with small molecule kinase inhibitors. IKK isoforms are assembled as variable multi-subunit IKK complexes that regulate not only NF-κB dimers, but also protein substrates out-with this cascade. Consequently, close consideration of how these individual complexes transduce extracellular signals and more importantly what impact small molecule inhibitors of the IKKs have on functional outcomes are demanded. A number of adenosine triphosphate (ATP)-competitive IKKβ-selective inhibitors have been developed but have demonstrated a lack of activity against IKKα. A number of these chemicals have also exhibited detrimental outcomes such as cellular toxicity and immuno-suppression. The impact of small molecule inhibitors of IKK catalytic activity will therefore be reappraised, examining the advantages and potential disadvantages to this type of intervention strategy in the treatment of diseases such as arthritis, intestinal inflammation and cancer. Furthermore, we will outline some emerging strategies, particularly the disruption of protein-protein interactions within the IKK complex, as an alternative route towards the development of novel pharmacological agents. Whether these alternatives may negate the limitations of ATP-competitive molecules and potentially avoid the issues of toxicity will be discussed.
Collapse
Affiliation(s)
- Carly Gamble
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | | | | | | | | | | |
Collapse
|
30
|
Bertucci F, Finetti P, Birnbaum D. Basal breast cancer: a complex and deadly molecular subtype. Curr Mol Med 2012; 12:96-110. [PMID: 22082486 PMCID: PMC3343384 DOI: 10.2174/156652412798376134] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/24/2011] [Accepted: 11/02/2011] [Indexed: 12/15/2022]
Abstract
During the last decade, gene expression profiling of breast cancer has revealed the existence of five molecular subtypes and allowed the establishment of a new classification. The basal subtype, which represents 15-25% of cases, is characterized by an expression profile similar to that of myoepithelial normal mammary cells. Basal tumors are frequently assimilated to triple-negative (TN) breast cancers. They display epidemiological and clinico-pathological features distinct from other subtypes. Their pattern of relapse is characterized by frequent and early relapses and visceral locations. Despite a relative sensitivity to chemotherapy, the prognosis is poor. Recent characterization of their molecular features, such as the dysfunction of the BRCA1 pathway or the frequent expression of EGFR, provides opportunities for optimizing the systemic treatment. Several clinical trials dedicated to basal or TN tumors are testing cytotoxic agents and/or molecularly targeted therapies. This review summarizes the current state of knowledge of this aggressive and hard-to-treat subtype of breast cancer.
Collapse
Affiliation(s)
- F Bertucci
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Marseille, France.
| | | | | |
Collapse
|
31
|
Ali AM, Ueno T, Tanaka S, Takada M, Ishiguro H, Abdellah AZ, Toi M. Determining circulating endothelial cells using CellSearch system during preoperative systemic chemotherapy in breast cancer patients. Eur J Cancer 2011; 47:2265-72. [DOI: 10.1016/j.ejca.2011.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 06/07/2011] [Accepted: 06/07/2011] [Indexed: 12/12/2022]
|
32
|
Sabatier R, Finetti P, Mamessier E, Raynaud S, Cervera N, Lambaudie E, Jacquemier J, Viens P, Birnbaum D, Bertucci F. Kinome expression profiling and prognosis of basal breast cancers. Mol Cancer 2011; 10:86. [PMID: 21777462 PMCID: PMC3156788 DOI: 10.1186/1476-4598-10-86] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 07/21/2011] [Indexed: 01/05/2023] Open
Abstract
Background Basal breast cancers (BCs) represent ~15% of BCs. Although overall poor, prognosis is heterogeneous. Identification of good- versus poor-prognosis patients is difficult or impossible using the standard histoclinical features and the recently defined prognostic gene expression signatures (GES). Kinases are often activated or overexpressed in cancers, and constitute targets for successful therapies. We sought to define a prognostic model of basal BCs based on kinome expression profiling. Methods DNA microarray-based gene expression and histoclinical data of 2515 early BCs from thirteen datasets were collected. We searched for a kinome-based GES associated with disease-free survival (DFS) in basal BCs of the learning set using a metagene-based approach. The signature was then tested in basal tumors of the independent validation set. Results A total of 591 samples were basal. We identified a 28-kinase metagene associated with DFS in the learning set (N = 73). This metagene was associated with immune response and particularly cytotoxic T-cell response. On multivariate analysis, a metagene-based predictor outperformed the classical prognostic factors, both in the learning and the validation (N = 518) sets, independently of the lymphocyte infiltrate. In the validation set, patients whose tumors overexpressed the metagene had a 78% 5-year DFS versus 54% for other patients (p = 1.62E-4, log-rank test). Conclusions Based on kinome expression, we identified a predictor that separated basal BCs into two subgroups of different prognosis. Tumors associated with higher activation of cytotoxic tumor-infiltrative lymphocytes harbored a better prognosis. Such classification should help tailor the treatment and develop new therapies based on immune response manipulation.
Collapse
Affiliation(s)
- Renaud Sabatier
- Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Institut Paoli-Calmettes, 27 bd Leï Roure, 13009 Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Prolonged mammosphere culture of MCF-7 cells induces an EMT and repression of the estrogen receptor by microRNAs. Breast Cancer Res Treat 2011; 132:75-85. [PMID: 21553120 DOI: 10.1007/s10549-011-1534-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 04/16/2011] [Indexed: 12/21/2022]
Abstract
Mammosphere culture has been used widely for the enrichment of mammary epithelial stem cells and breast cancer stem cells (CSCs). Epithelial-to-mesenchymal transition (EMT) also induces stem cell features in normal and transformed mammary cells. We examined whether mammosphere culture conditions per se induced EMT in the epithelial MCF-7 breast cancer cell line. MCF-7 cells were cultured as mammospheres for 5 weeks, with dispersal and reseeding at the end of each week. This mammosphere culture induced a complete EMT by 3 weeks. Return of the cells to standard adherent culture conditions in serum-supplemented media generated a cell population (called MCF-7(M) cells), which displays a stable mesenchymal and CSC-like CD(44+)/CD(24-/low) phenotype. EMT was accompanied by a stable, marked increase in EMT-associated transcription factors and mesenchymal markers, and a decrease in epithelial markers and estrogen receptor α (ERα). MCF-7(M) cells showed increased motility, proliferation and chemoresistance in vitro, and produced larger tumors in immunodeficient mice with or without estrogen supplementation. MicroRNA analysis showed suppression of miR-200c, miR-203, and miR-205; and increases in miR-222 and miR-221. Antisense hairpin RNA inhibitor targeting miR-221 resulted in re-expression of ERα in MCF-7(M) cells. This study provides the first example of mammosphere culture conditions inducing EMT and of EMT regulating microRNAs that target ERα.
Collapse
|
34
|
D'Anello L, Sansone P, Storci G, Mitrugno V, D'Uva G, Chieco P, Bonafé M. Epigenetic control of the basal-like gene expression profile via Interleukin-6 in breast cancer cells. Mol Cancer 2010; 9:300. [PMID: 21092249 PMCID: PMC3002335 DOI: 10.1186/1476-4598-9-300] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 11/23/2010] [Indexed: 12/26/2022] Open
Abstract
Background Basal-like carcinoma are aggressive breast cancers that frequently carry p53 inactivating mutations, lack estrogen receptor-α (ERα) and express the cancer stem cell markers CD133 and CD44. These tumors also over-express Interleukin 6 (IL-6), a pro-inflammatory cytokine that stimulates the growth of breast cancer stem/progenitor cells. Results Here we show that p53 deficiency in breast cancer cells induces a loss of methylation at IL-6 proximal promoter region, which is maintained by an IL-6 autocrine loop. IL-6 also elicits the loss of methylation at the CD133 promoter region 1 and of CD44 proximal promoter, enhancing CD133 and CD44 gene transcription. In parallel, IL-6 induces the methylation of estrogen receptor (ERα) promoter and the loss of ERα mRNA expression. Finally, IL-6 induces the methylation of IL-6 distal promoter and of CD133 promoter region 2, which harbour putative repressor regions. Conclusion We conclude that IL-6, whose methylation-dependent autocrine loop is triggered by the inactivation of p53, induces an epigenetic reprogramming that drives breast carcinoma cells towards a basal-like/stem cell-like gene expression profile.
Collapse
Affiliation(s)
- Laura D'Anello
- Center for Applied Biomedical Research (CRBA), St, Orsola-Malpighi University Hospital, via Massarenti 9, 40138 Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Storci G, Sansone P, Mari S, D'Uva G, Tavolari S, Guarnieri T, Taffurelli M, Ceccarelli C, Santini D, Chieco P, Marcu KB, Bonafè M. TNFalpha up-regulates SLUG via the NF-kappaB/HIF1alpha axis, which imparts breast cancer cells with a stem cell-like phenotype. J Cell Physiol 2010; 225:682-91. [PMID: 20509143 DOI: 10.1002/jcp.22264] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular and intracellular mediators of inflammation, such as tumor necrosis factor alpha (TNFα) and NF-kappaB (NF-κB), play major roles in breast cancer pathogenesis, progression and relapse. SLUG, a mediator of the epithelial-mesenchymal transition process, is over-expressed in CD44(+)/CD24(-) tumor initiating breast cancer cells and in basal-like carcinoma, a subtype of aggressive breast cancer endowed with a stem cell-like gene expression profile. Cancer stem cells also over-express members of the pro-inflammatory NF-κB network, but their functional relationship with SLUG expression in breast cancer cells remains unclear. Here, we show that TNFα treatment of human breast cancer cells up-regulates SLUG with a dependency on canonical NF-κB/HIF1α signaling, which is strongly enhanced by p53 inactivation. Moreover, SLUG up-regulation engenders breast cancer cells with stem cell-like properties including enhanced expression of CD44 and Jagged-1 in conjunction with estrogen receptor alpha down-regulation, growth as mammospheres, and extracellular matrix invasiveness. Our results reveal a molecular mechanism whereby TNFα, a major pro-inflammatory cytokine, imparts breast cancer cells with stem cell-like features, which are connected to increased tumor aggressiveness.
Collapse
Affiliation(s)
- Gianluca Storci
- Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dias Pereira P, Lopes C, Matos A, Cortez P, Gärtner F, Medeiros R, Lopes C. Caveolin-1 in Diagnosis and Prognosis of Canine Mammary Tumours: Comparison of Evaluation Systems. J Comp Pathol 2010; 143:87-93. [DOI: 10.1016/j.jcpa.2010.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 11/30/2009] [Accepted: 01/04/2010] [Indexed: 11/28/2022]
|
37
|
Fleming IN, Gilbert FJ, Miles KA, Cameron D. Opportunities for PET to deliver clinical benefit in cancer: breast cancer as a paradigm. Cancer Imaging 2010; 10:144-52. [PMID: 20605761 PMCID: PMC2904028 DOI: 10.1102/1470-7330.2010.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The glucose analogue fluorodeoxyglucose (FDG) has demonstrated enhanced uptake in the majority of tumours as a result of increased uptake and fixation by phosphorylation. It is the most widely used radiotracer in positron emission tomography (PET), being used in >90% of scans, and is useful for diagnosis, staging and detection of residual/recurrent cancer. However, there are limits to the utility of FDG, particularly in certain tumour types. The development of new radiotracers to study molecular processes such as proliferation, apoptosis, angiogenesis and hypoxia will complement FDG by providing additional information on the cell biology of tumours. The aim of this paper is to consider how the availability of new tracers, or new applications for existing PET/CT technologies, could deliver clinical benefit in cancer, using breast cancer as a paradigm.
Collapse
Affiliation(s)
- Ian N Fleming
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | | | | | | |
Collapse
|
38
|
A straightforward but not piecewise relationship between age and lymph node status in Chinese breast cancer patients. PLoS One 2010; 5:e11035. [PMID: 20543953 PMCID: PMC2882951 DOI: 10.1371/journal.pone.0011035] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 05/17/2010] [Indexed: 12/01/2022] Open
Abstract
Purpose To investigate the relationship between age and axillary lymph node (LN) involvement in Chinese breast cancer patients, and to replicate a recently identified piecewise relationship between age and LN involvement. Methods A dataset, consisting of 3,715 patients (with complete information on study variables) with operable breast cancer consecutively surgically treated between 1996 and 2006, was derived from the database of Shanghai Cancer Hospital. Univariate and multivariate logistic regression were employed to analyze the relationship between age and LN. We subsequently performed a similar analysis on another dataset including 1,832 consecutive patients treated between 2007 and 2008 to replicate our findings in the first dataset. Results A U-shaped relationship (previously observed in two European populations) between age and LN status failed to be replicated in our dataset of Chinese patients. Instead, we observed a linear rather than piecewise relationship. After multivariate adjustment, the linear relationship was still present. Moreover, the interaction between age and LN involvement was not modified by tumor size. The odds of LN involvement decreased by 1.5% for each year increase in age (OR 0.985, 95% CI 0.979–0.991, P<0.001). Breast cancer subtypes were also associated with LN status. Proportions of basal-like and ERBB2+ subtypes decreased with increasing age. The observations in the first dataset were successfully replicated in a second independent dataset. Conclusion We confirmed a straightforward but not piecewise relationship between age and LN status in Chinese patients. The different pattern between Chinese and European elderly patients should be considered when making clinical decisions.
Collapse
|
39
|
Choo JR, Nielsen TO. Biomarkers for Basal-like Breast Cancer. Cancers (Basel) 2010; 2:1040-65. [PMID: 24281106 PMCID: PMC3835118 DOI: 10.3390/cancers2021040] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/11/2010] [Accepted: 05/19/2010] [Indexed: 12/24/2022] Open
Abstract
Initially recognized through microarray-based gene expression profiling, basal-like breast cancer, for which we lack effective targeted therapies, is an aggressive form of carcinoma with a predilection for younger women. With some success, immunohistochemical studies have attempted to reproduce the expression profile classification of breast cancer through identification of subtype-specific biomarkers. This review aims to present an in depth summary and analysis of the current status of basal-like breast cancer biomarker research. While a number of biomarkers show promise for future clinical application, the next logical step is a comprehensive investigation of all biomarkers against a gene expression profile gold standard for breast cancer subtype assignment.
Collapse
Affiliation(s)
- Jennifer R Choo
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.
| | | |
Collapse
|
40
|
A gene expression signature identifies two prognostic subgroups of basal breast cancer. Breast Cancer Res Treat 2010; 126:407-20. [PMID: 20490655 DOI: 10.1007/s10549-010-0897-9] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Accepted: 04/12/2010] [Indexed: 01/05/2023]
Abstract
Prognosis of basal breast cancers is poor but heterogeneous. Medullary breast cancers (MBC) display a basal profile, but a favorable prognosis. We hypothesized that a previously published 368-gene expression signature associated with MBC might serve to define a prognostic classifier in basal cancers. We collected public gene expression and histoclinical data of 2145 invasive early breast adenocarcinomas. We developed a Support Vector Machine (SVM) classifier based on this 368-gene list in a learning set, and tested its predictive performances in an independent validation set. Then, we assessed its prognostic value and that of six prognostic signatures for disease-free survival (DFS) in the remaining 2034 samples. The SVM model accurately classified all MBC samples in the learning and validation sets. A total of 466 cases were basal across other sets. The SVM classifier separated them into two subgroups, subgroup 1 (resembling MBC) and subgroup 2 (not resembling MBC). Subgroup 1 exhibited 71% 5-year DFS, whereas subgroup 2 exhibited 50% (P = 9.93E-05). The classifier outperformed the classical prognostic variables in multivariate analysis, conferring lesser risk for relapse in subgroup 1 (HR = 0.52, P = 3.9E-04). This prognostic value was specific to the basal subtype, in which none of the other prognostic signatures was informative. Ontology analysis revealed effective immune response (IR), enhanced tumor cell apoptosis, elevated levels of metastasis-inhibiting factors and low levels of metastasis-promoting factors in the good-prognosis subgroup, and a more developed cell migration system in the poor-prognosis subgroup. In conclusion, based on this 368-gene SVM model derived from an MBC signature, basal breast cancers were classified in two prognostic subgroups, suggesting that MBC and basal breast cancers share similar molecular alterations associated with aggressiveness. This signature could help define the prognosis, adapt the systemic treatment, and identify new therapeutic targets.
Collapse
|
41
|
Germline mutations and polymorphisms in the origins of cancers in women. JOURNAL OF ONCOLOGY 2010; 2010:297671. [PMID: 20111735 PMCID: PMC2810468 DOI: 10.1155/2010/297671] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 11/14/2009] [Indexed: 12/18/2022]
Abstract
Several female malignancies including breast, ovarian, and endometrial cancers can be characterized based on known somatic and germline mutations. Initiation and propagation of tumors reflect underlying genomic alterations such as mutations, polymorphisms, and copy number variations found in genes of multiple cellular pathways. The contributions of any single genetic variation or mutation in a population depend on its frequency and penetrance as well as tissue-specific functionality. Genome wide association studies, fluorescence in situ hybridization, comparative genomic hybridization, and candidate gene studies have enumerated genetic contributors to cancers in women. These include p53, BRCA1, BRCA2, STK11, PTEN, CHEK2, ATM, BRIP1, PALB2, FGFR2, TGFB1, MDM2, MDM4 as well as several other chromosomal loci. Based on the heterogeneity within a specific tumor type, a combination of genomic alterations defines the cancer subtype, biologic behavior, and in some cases, response to therapeutics. Consideration of tumor heterogeneity is therefore important in the critical analysis of gene associations in cancer.
Collapse
|
42
|
Abstract
Cancer remains an outstanding cause of global morbidity and mortality, despite intensive research and unprecedented insights into the basic mechanisms of cancer development. A plethora of clinical and experimental evidence suggests that cancers from individual patients are likely to be molecularly heterogeneous in their use of distinct oncogenic pathways and biological programs. Efforts to significantly impact cancer patient outcomes will almost certainly require the development of robust strategies to subdivide such heterogeneous panels of cancers into biologically and clinically homogenous subgroups, for the purposes of personalizing treatment protocols and identifying optimal drug targets. In this review, I describe recent progress in the development of both targeted and genome-wide approaches for the molecular stratification of cancers, drawing examples from both the haematopoietic and solid tumor malignancies.
Collapse
Affiliation(s)
- Patrick Tan
- Duke-NUS Graduate Medical School, Genome Institute of Singapore, Singapore; Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
43
|
Abstract
Metastasis is the major cause of death in breast cancer patients. Gene-expression studies have shown that the likelihood of metastasis can be predicted from analysis of primary tumors. Two recent papers in BMC Medicine and BMC Cancer have established new operational expression signatures containing a limited number of genes involved in angiogenesis or cell proliferation.
Collapse
|
44
|
Birnbaum D, Sircoulomb F, Imbert J. A reason why the ERBB2 gene is amplified and not mutated in breast cancer. Cancer Cell Int 2009; 9:5. [PMID: 19226453 PMCID: PMC2649042 DOI: 10.1186/1475-2867-9-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 02/18/2009] [Indexed: 12/15/2022] Open
Abstract
Alterations of receptor-type tyrosine kinases (RTK) are frequent in human cancers. They can result from translocation, mutation or amplification. The ERBB2 RTK is encoded by a gene that is amplified in about 20% breast cancers. The question is: why is this RTK specifically subjected to this type of alteration? We propose that ERBB2 gene amplification is used to overcome repression of its expression by sequence-specific transcription factors.
Collapse
Affiliation(s)
- Daniel Birnbaum
- Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm and Institut Paoli-Calmettes, Marseille, F-13009, France.
| | | | | |
Collapse
|