1
|
Yousafzai NA, El Khalki L, Wang W, Szpendyk J, Sossey-Alaoui K. Kindlin-2 regulates the oncogenic activities of integrins and TGF-β in triple-negative breast cancer progression and metastasis. Oncogene 2024; 43:3291-3305. [PMID: 39300257 PMCID: PMC11534691 DOI: 10.1038/s41388-024-03166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Kindlin-2, an adapter protein, is dysregulated in various human cancers, including triple-negative breast cancer (TNBC), where it drives tumor progression and metastasis by influencing several cancer hallmarks. One well-established role of Kindlin-2 involves the regulation of integrin signaling, achieved by directly binding to the cytoplasmic tail of the integrin β subunit. In this study, we present novel insights into Kindlin-2's involvement in stabilizing the β1-Integrin:TGF-β type 1 receptor (TβRI) complexes, acting as a physical bridge that links β1-Integrin to TβRI. Loss of Kindlin-2 results in the degradation of this protein complex, leading to the inhibition of downstream oncogenic pathways. We used a diverse range of in vitro assays, including CRISPR/Cas9 gene editing, cell migration, 3D-tumorsphere formation and invasion, solid binding, co-immunoprecipitation, cell adhesion and spreading assays, as well as western blot and flow cytometry analyses, utilizing MDA-MB-231 and 4T1 TNBC cell lines. Additionally, preclinical in vivo mouse models of TNBC tumor progression and metastasis were employed to substantiate our findings. Our studies established the direct interaction between Kindlin-2 and β1-Integrin and between Kindlin-2 and TβRI. Disruption of these interactions, via CRISPR/Cas9-mediated knockout of Kindlin-2, led to the degradation of β1-Integrin and TβRI, resulting in the inhibition of oncogenic pathways downstream of both proteins, subsequently hindering tumor growth and metastasis. Treatment of Kindlin-2-deficient cells with the proteasome inhibitor MG-132 restored the expression of both β1-Integrin and TβRI. Furthermore, the rescue of Kindlin-2 expression reinstated their oncogenic activities in vitro and in vivo, while Kindlin-2 lacking domains involved in the interaction of Kindlin-2 with β1-Integrin or TβRI did not. This study identifies a novel function of Kindlin-2 in stabilizing the β1-Integrin:TβRI complexes and regulating their downstream oncogenic signaling. The translational implications of these findings are substantial, potentially unveiling new therapeutically targeted pathways crucial for the treatment of TNBC tumors.
Collapse
Affiliation(s)
- Neelum Aziz Yousafzai
- MetroHealth System, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44109, USA
| | - Lamyae El Khalki
- MetroHealth System, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44109, USA
| | - Wei Wang
- MetroHealth System, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44109, USA
| | | | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, OH, USA.
- Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44109, USA.
| |
Collapse
|
2
|
Yousafzai NA, El Khalki L, Wang W, Szpendyk J, Sossey-Alaoui K. Kindlin-2 Regulates the Oncogenic Activities of Integrins and TGF-β In Triple Negative Breast Cancer Progression and Metastasis. RESEARCH SQUARE 2024:rs.3.rs-3914650. [PMID: 38405979 PMCID: PMC10889066 DOI: 10.21203/rs.3.rs-3914650/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background Kindlin-2, an adaptor protein, is dysregulated in various human cancers, including triple negative breast cancer (TNBC), where it drives tumor progression and metastasis by influencing several cancer hallmarks. One well-established role of Kindlin-2 involves the regulation of integrin signaling, achieved by directly binding to the cytoplasmic tail of the integrin β subunit. In this study, we present novel insights into Kindlin-2's involvement in stabilizing the β1-Integrin:TGF-β type 1 receptor (TβRI) complexes, acting as a physical bridge that links β1-Integrin to TβRI. The loss of Kindlin-2 results in the degradation of this protein complex, leading to the inhibition of downstream oncogenic pathways. Methods Our methodology encompassed a diverse range of in vitro assays, including CRISPR/Cas9 gene editing, cell migration, 3D tumorsphere formation and invasion, solid binding, co-immunoprecipitation, cell adhesion and spreading assays, as well as western blot and flow cytometry analyses, utilizing MDA-MB-231 and 4T1 TNBC cell lines. Additionally, preclinical in vivo mouse models of TNBC tumor progression and metastasis were employed to substantiate our findings. Results The investigation revealed that the direct interaction between Kindlin-2 and β1-Integrin is mediated through the C-terminal F3 domain of Kindlin-2, while the interaction between Kindlin-2 and TβRI is facilitated through the F2 domain of Kindlin-2. Disruption of this bridge, achieved via CRISPR/Cas9-mediated knockout of Kindlin-2, led to the degradation of β1-Integrin and TβRI, resulting in the inhibition of oncogenic pathways downstream of both proteins, subsequently hindering tumor growth and metastasis. Treatment of Kindlin-2-deficient cells with the proteasome inhibitor MG-132 restored the expression of both β1-Integrin and TβRI. Furthermore, the rescue of Kindlin-2 expression reinstated their oncogenic activities both in vitro and in vivo. Conclusions This study identifies a novel function of Kindlin-2 in stabilizing the β1-Integrin:TβR1 complexes and regulating their downstream oncogenic signaling. The translational implications of these findings are substantial, potentially unveiling new therapeutically targeted pathways crucial for the treatment of TNBC tumors.
Collapse
|
3
|
Eroğlu Güneş C, Güçlü E, Vural H, Kurar E. Knockdown of lncRNA ZEB2NAT suppresses epithelial mesenchymal transition, metastasis and proliferation in breast cancer cells. Gene 2021; 805:145904. [PMID: 34418470 DOI: 10.1016/j.gene.2021.145904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/08/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer is the second most common cause of cancer-related mortality in women. Breast cancer metastasis which usually is observed at the last stage is the major cause of breast cancer-related death. Long non-coding RNAs (lncRNAs) are member of the non-coding RNA family. It is known that lncRNAs have important functions in the genes regulation of different processes and pathways such as EMT (Epithelial mesenchymal transition), metastasis and apoptosis. Therefore, it is inevitable that lncRNAs have potential contribution for the understanding of cancer pathogenesis. lncRNA-ZEB2NAT is the natural antisense transcript of ZEB2. Herein, we investigated the effects of lncRNA-ZEB2NAT on process of EMT, metastasis and apoptosis in MCF7 and MDA-MB-231 breast cancer cells. The effect of ZEB2NAT on the expression of important genes in EMT, metastasis and apoptosis, and some protein levels was determined by qRT-PCR and western blot analysis, respectively. The effects of ZEB2NAT on cell proliferation, apoptosis, invasion and colony formation were evaluated using XTT, annexin V, invasion and colony assays, respectively. The ZEB2NAT knockdown caused anti-metastatic and apoptotic effects. The ZEB2NAT knockdown resulted in a decrease in ZEB2 and N-cadherin but an increase in E-cadherin protein levels. In addition, it was determined that ZEB2NAT knockdown significantly decreased cell proliferation and stimulated apoptosis in both cells. It was found that ZEB2NAT knockdown significantly decreased invasion and colony formation in both cells. ZEB2NAT knockdown showed anti-metastatic and apoptotic effect by affecting the important genes in both cells. These results have suggested that ZEB2NAT has an important role in EMT, metastasis and apoptosis in breast cancer and ZEB2NAT knockdown caused significant anti-cancer activities.
Collapse
Affiliation(s)
- Canan Eroğlu Güneş
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey.
| | - Ebru Güçlü
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hasibe Vural
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ercan Kurar
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
4
|
YB1 Is a Major Contributor to Health Disparities in Triple Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13246262. [PMID: 34944882 PMCID: PMC8699660 DOI: 10.3390/cancers13246262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Triple negative breast cancer (TNBC) is a devastating disease that affects many women, due to the lack of FDA-approved targeted therapy. In the absence of cell surface receptors ER, PR, and Her2 that can be targeted with hormonal and antibody treatments, cytotoxic chemotherapy remains the major course of treatment, with a dismal response and rapid recurrence due to the acquisition of resistance. TNBC is also twice as more prevalent in African American (AA) when compared to Caucasian American (CA) women. This study investigated the role of the YB1 gene in the disparities in TNBC between AA and CA women. We found that YB1 is highly expressed in TNBC tumors of AA origin when compared to CAs. Increased expression levels and activity of YB1 correlates with poor disease outcomes, resistance to chemotherapy, and the activation of the cancer stem cell (CSC) phenotype, with higher levels in AA than in CA TNBC tumors. More importantly, we found that the targeted inhibition of the expression and activity of YB1 significantly inhibited the oncogenic behavior of AA tumors through sensitization to chemotherapy and inhibition of CSCs. Our study is the first to show that YB1 activity may be a major biological contributor to the health disparities in TNBC, and that development of therapies that specifically target YB1 could reduce these disparities. Abstract Triple negative breast cancer (TNBC) is the most aggressive amongst all breast cancer (BC) subtypes. While TNBC tumors represent less than 20% of all BC subtypes, they are responsible for the most BC-related deaths. More significantly, when considering TNBC incidence across all racial/ethnic groups, TNBC accounts for less than 20% of all BCs. However, in non-Hispanic black women, the incidence rate of TNBC is more than 40%, which may be a contributing factor to the higher BC-related death rate in this population. These disparities remain strong even after accounting for differences in socioeconomic status, healthcare access, and lifestyle factors. Increased evidence now points to biological mechanisms that are intrinsic to the tumor that contribute to disparate TNBC disease burdens. Here, we show that YB1, a multifunction gene, plays a major role in the TNBC disparities between African American (AA) and Caucasian American (CA) women. We show in three independent TNBC tumors cohorts, that YB1 is significantly highly expressed in AA TNBC tumors when compared to CAs, and that increased levels of YB1 correlate with poor survival of AA patients with TNBC. We used a combination of genetic manipulation of YB1 and chemotherapy treatment, both in vitro and in animal models of TNBC to show that YB1 oncogenic activity is more enhanced in TNBC cell lines of AA origin, by increasing their tumorigenic and aggressive behaviors, trough the activation of cancer stem cell phenotype and resistance to chemotherapeutic treatments.
Collapse
|
5
|
Kansakar U, Wang W, Markovic V, Sossey-Alaoui K. Elucidating the molecular signaling pathways of WAVE3. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:900. [PMID: 32793744 DOI: 10.21037/atm.2020.02.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer metastasis is a complex, multistep process that requires tumor cells to evade from the original site and form new tumors at a distant site or a different organ, often via bloodstream or the lymphatic system. Metastasis is responsible for more than 90% of cancer-related deaths. WAVE3 belongs to the Wiskott-Aldrich syndrome protein (WASP) family, which regulate actin cytoskeleton remodeling as well as several aspects of cell migration, invasion, and metastasis. In fact, WAVE3 has been established as a driver of tumor progression and metastasis in cancers from several origins, including triple negative breast cancers (TNBCs), which are classified as the most lethal subtype of breast cancer, due to their resistance to standard of care therapy and highly metastatic behavior. In this review, we will attempt to summarize the recent advances that have been made to understand how WAVE3 contributes to the molecular mechanisms that control cancer progression and metastasis. We will also review the signaling pathways that are involved in the regulation of WAVE3 expression and function to identify potential therapeutic options targeted against WAVE3 for the treatment of patients with metastatic tumors.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Wei Wang
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Vesna Markovic
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| | - Khalid Sossey-Alaoui
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Rammelkamp Center for Research, MetroHealth, Cleveland, OH, USA
| |
Collapse
|
6
|
Jiang L, Xiao Y, Ding Y, Tang J, Guo F. Discovering Cancer Subtypes via an Accurate Fusion Strategy on Multiple Profile Data. Front Genet 2019; 10:20. [PMID: 30804977 PMCID: PMC6370730 DOI: 10.3389/fgene.2019.00020] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/15/2019] [Indexed: 01/03/2023] Open
Abstract
Discovering cancer subtypes is useful for guiding clinical treatment of multiple cancers. Progressive profile technologies for tissue have accumulated diverse types of data. Based on these types of expression data, various computational methods have been proposed to predict cancer subtypes. It is crucial to study how to better integrate these multiple profiles of data. In this paper, we collect multiple profiles of data for five cancers on The Cancer Genome Atlas (TCGA). Then, we construct three similarity kernels for all patients of the same cancer by gene expression, miRNA expression and isoform expression data. We also propose a novel unsupervised multiple kernel fusion method, Similarity Kernel Fusion (SKF), in order to integrate three similarity kernels into one combined kernel. Finally, we make use of spectral clustering on the integrated kernel to predict cancer subtypes. In the experimental results, the P-values from the Cox regression model and survival curve analysis can be used to evaluate the performance of predicted subtypes on three datasets. Our kernel fusion method, SKF, has outstanding performance compared with single kernel and other multiple kernel fusion strategies. It demonstrates that our method can accurately identify more accurate subtypes on various kinds of cancers. Our cancer subtype prediction method can identify essential genes and biomarkers for disease diagnosis and prognosis, and we also discuss the possible side effects of therapies and treatment.
Collapse
Affiliation(s)
- Limin Jiang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Yongkang Xiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Yijie Ding
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Jijun Tang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
- Department of Computer Science and Engineering, University of South Carolina, Columbia, SC, United States
| | - Fei Guo
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| |
Collapse
|
7
|
Sossey-Alaoui K, Pluskota E, Szpak D, Schiemann WP, Plow EF. The Kindlin-2 regulation of epithelial-to-mesenchymal transition in breast cancer metastasis is mediated through miR-200b. Sci Rep 2018; 8:7360. [PMID: 29743493 PMCID: PMC5943603 DOI: 10.1038/s41598-018-25373-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/20/2018] [Indexed: 12/20/2022] Open
Abstract
Metastasis is the main cause of death in cancer patients, including breast cancer (BC). Despite recent progress in understanding the biological and molecular determinants of BC metastasis, effective therapeutic treatments are yet to be developed. Among the multitude of molecular mechanisms that regulate cancer metastasis, the epithelial-to-mesenchymal transition (EMT) program plays a key role in the activation of the biological steps leading to the metastatic phenotype. Kindlin-2 has been associated with the pathogenesis of several types of cancers, including BC. The role of Kindlin-2 in the regulation of BC metastasis, and to a lesser extent in EMT is not well understood. In this study, we show that Kindlin-2 is closely associated with the development of the metastatic phenotype in BC. We report that knockout of Kindlin-2 in either human or mouse BC cells, significantly inhibits metastasis in both human and mouse models of BC metastasis. We also report that the Kindlin-2-mediated inhibition of metastasis is the result of inhibition of expression of key molecular markers of the EMT program. Mechanistically, we show that miR-200b, a master regulator of EMT, directly targets and inhibits the expression of Kindlin-2, leading to the subsequent inhibition of EMT and metastasis. Together, our data support the targeting of Kindlin-2 as a therapeutic strategy against BC metastasis.
Collapse
Affiliation(s)
- Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Elzbieta Pluskota
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Dorota Szpak
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Edward F Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
8
|
Bledzka K, Schiemann B, Schiemann WP, Fox P, Plow EF, Sossey-Alaoui K. The WAVE3-YB1 interaction regulates cancer stem cells activity in breast cancer. Oncotarget 2017; 8:104072-104089. [PMID: 29262622 PMCID: PMC5732788 DOI: 10.18632/oncotarget.22009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Resistance to therapy is the main cause of tumor recurrence and metastasis and cancer stem cells (CSCs) play a crucial role in this process, especially in triple-negative breast cancers (TNBCs). Unfortunately, no FDA-approved treatment is currently available for this subtype of BC, which explains the high rate of mortality in patients with TNBC tumors. WAVE3, a member of the WASP/WAVE actin-cytoskeleton remodeling family of protein, has been established as a major driver of tumor progression and metastasis of several solid tumors, including those originating in the breast. Our recently published studies found WAVE3 to mediate the process of chemoresistance in TNBCs. The molecular mechanisms whereby WAVE3 regulates chemoresistance in TNBC tumors remains largely unknown, as does the role of WAVE3 in CSC maintenance. Here we show that WAVE3 promotes CSC self-renewal and regulates transcription of CSC-specific genes, which, in part, provides a mechanistic explanation for the function of WAVE3 in chemoresistance in TNBCs. Our data show that WAVE3 is enriched in the CSC-subpopulation of TNBC cell lines. Knockout of WAVE3 via CRISPR/Cas9 significantly attenuates the CSC-subpopulation and inhibits transcription of CSC transcription factors. Mechanistically, we established a link between WAVE3 and the Y-box-binding protein-1 (YB1), a transcription factor and CSC-maintenance gene. Indeed, the interaction of WAVE3 with YB1 is required for YB1 translocation to the nucleus of cancer cells, and activation of transcription of CSC-specific genes. Our findings identify a new WAVE3/YB1 signaling axis that regulates the CSC-mediated resistance to therapy and opens a new therapeutic window for TNBCs treatment.
Collapse
Affiliation(s)
- Kamila Bledzka
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Paul Fox
- Department of Cellular and Molecular Medicine, Cleveland, Ohio, USA
| | - Edward F Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Wilkinson JB, Shah C, Amin M, Nadeau L, Shaitelman SF, Chen PY, Grills IS, Martinez AA, Mitchell CK, Wallace MF, Vicini FA. Outcomes According to Breast Cancer Subtype in Patients Treated With Accelerated Partial Breast Irradiation. Clin Breast Cancer 2016; 17:55-60. [PMID: 27666436 DOI: 10.1016/j.clbc.2016.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/03/2016] [Accepted: 07/20/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND The purpose of the study was to determine outcomes for patients treated with accelerated partial breast irradiation (APBI) on the basis of breast cancer subtype (BCST). PATIENTS AND METHODS Our single-institution, institutional review board-approved APBI database was queried for patients who had complete testing results for the estrogen (ER), progesterone (PR), and HER2/neu receptors to determine outcomes for each BCST. Women were assigned as luminal A (LA), luminal B (LB), HER2, and basal BCST using their ER, PR, and HER2/neu receptor status. Degree of ER expression supplemented the receptor-based luminal BCST assignment. Two hundred seventy-eight patients had results for all 3 receptors (LA = 164 [59%], LB = 81 [29%], HER2 = 5 [2%], basal = 28 [10%]), which were submitted for analysis (ipsilateral breast tumor recurrence [IBTR], regional nodal failure, distant metastasis [DM], disease-free survival [DFS], cause-specific survival [CSS], and overall survival [OS]). RESULTS Median follow-up was 5.4 years (range, 0.1-12.4 years). Basal and HER2 subtype patients had higher histologic grades (Grade 3 = 75% vs. 10% LA/LB; P < .001), larger tumors (13.0 mm basal vs. 10.7 mm LA/LB; P = .059), and were more likely to receive chemotherapy (68% vs. 15% LA/LB; P < .001). Margin and nodal status were similar among BCSTs. At 5 years, IBTR rates were similar (1.8%, 2.9%, 0%, and 4.8%) for LA, LB, HER2, and basal subtypes, respectively (P = .62). DM was only seen in LA (2.9%) and LB (1.3%) (P = .83). DFS (95%-100%), CSS (97%-100%), and OS (80%-100%) were not statistically different (P = .97, .87, .46, respectively). CONCLUSION Five-year local control rates after breast-conserving surgery, APBI, and appropriate systemic therapy are excellent for luminal, HER2, and basal phenotypes of early-stage breast cancer; however, further study of receptor subtype effect on risk stratification in early-stage breast cancer is needed.
Collapse
Affiliation(s)
- J Ben Wilkinson
- Department of Radiation Oncology, Willis-Knighton Health System, Louisiana State University Health Sciences Center, Shreveport, LA.
| | - Chirag Shah
- Department of Radiation Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Mitual Amin
- Department of Pathology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Laura Nadeau
- Department of Medical Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Simona F Shaitelman
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Peter Y Chen
- Department of Radiation Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Inga S Grills
- Department of Radiation Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Alvaro A Martinez
- Department of Radiation Oncology, Michigan Healthcare Professionals, 21st Century Oncology, Farmington Hills, MI
| | - Christina K Mitchell
- Department of Radiation Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Michelle F Wallace
- Department of Radiation Oncology, Beaumont Cancer Institute, Oakland University William Beaumont School of Medicine, Royal Oak, MI
| | - Frank A Vicini
- Department of Radiation Oncology, Michigan Healthcare Professionals, 21st Century Oncology, Farmington Hills, MI
| |
Collapse
|
10
|
Wu Q, Guo L, Jiang F, Li L, Li Z, Chen F. Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER- breast cancer cell lines. J Cell Mol Med 2015; 19:2874-87. [PMID: 26416600 PMCID: PMC4687702 DOI: 10.1111/jcmm.12681] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 08/14/2015] [Indexed: 12/22/2022] Open
Abstract
Recently, rapid advances in bioinformatics analysis have expanded our understanding of the transcriptome to a genome‐wide level. miRNA–mRNA–lncRNA interactions have been shown to play critical regulatory role in cancer biology. In this study, we discussed the use of an integrated systematic approach to explore new facets of the oestrogen receptor (ER)‐regulated transcriptome. The identification of RNAs that are related to the expression status of the ER may be useful in clinical therapy and prognosis. We used a network modelling strategy. First, microarray expression profiling of mRNA, lncRNA and miRNA was performed in MCF‐7 (ER‐positive) and MDA‐MB‐231 cells (ER‐ negative). A co‐expression network was then built using co‐expression relationships of the differentially expressed mRNAs and lncRNAs. Finally, the selected miRNA–mRNA network was added to the network. The key miRNA–mRNA–lncRNA interaction can be inferred from the network. The mRNA and non‐coding RNA expression profiles of the cells with different ER phenotypes were distinct. Among the aberrantly expressed miRNAs, the expression levels of miR‐19a‐3p, miR‐19b‐3p and miR‐130a‐3p were much lower in the MCF‐7 cells, whereas that of miR‐148b‐3p was much higher. In a cluster of miR‐17‐92, the expression levels of six of seven miRNAs were lower in the MCF‐7 cells, in addition to miR‐20b in the miR‐106a‐363 cluster. However, the levels of all the miRNAs in the miR‐106a‐25 cluster were higher in the MCF‐7 cells. In the co‐expression networking, CD74 and FMNL2 gene which is involved in the immune response and metastasis, respectively, had a stronger correlation with ER. Among the aberrantly expressed lncRNAs, lncRNA‐DLEU1 was highly expressed in the MCF‐7 cells. A statistical analysis revealed that there was a co‐expression relationship between ESR1 and lncRNA‐DLEU1. In addition, miR‐19a and lncRNA‐DLEU1 are both located on the human chromosome 13q. We speculate that miR‐19a might be co‐expressed with lncRNA‐DLEU1 to co‐regulate the expression of ESR1, which influences the occurrence and development of breast cancer cells with different levels of ER expression. Our findings reveal that the status of ER is mainly due to the differences in the mRNA and ncRNA profile between the breast cancer cell lines, and highlight the importance of studying the miRNA–mRNA–lncRNA interactions to completely illustrate the intricate transcriptome.
Collapse
Affiliation(s)
- Qian Wu
- The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Enviromental Health Sciences, Bloomburg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Li Guo
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Fei Jiang
- Department of Nutrition and Food Hygiene and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Li
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhong Li
- Department of Nutrition and Food Hygiene and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Putluri N, Maity S, Kommagani R, Kommangani R, Creighton CJ, Putluri V, Chen F, Nanda S, Bhowmik SK, Terunuma A, Dorsey T, Nardone A, Fu X, Shaw C, Sarkar TR, Schiff R, Lydon JP, O'Malley BW, Ambs S, Das GM, Michailidis G, Sreekumar A. Pathway-centric integrative analysis identifies RRM2 as a prognostic marker in breast cancer associated with poor survival and tamoxifen resistance. Neoplasia 2015; 16:390-402. [PMID: 25016594 PMCID: PMC4198742 DOI: 10.1016/j.neo.2014.05.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/15/2014] [Accepted: 05/19/2014] [Indexed: 01/14/2023] Open
Abstract
Breast cancer (BCa) molecular subtypes include luminal A, luminal B, normal-like, HER-2-enriched, and basal-like tumors, among which luminal B and basal-like cancers are highly aggressive. Biochemical pathways associated with patient survival or treatment response in these more aggressive subtypes are not well understood. With the limited availability of pathologically verified clinical specimens, cell line models are routinely used for pathway-centric studies. We measured the metabolome of luminal and basal-like BCa cell lines using mass spectrometry, linked metabolites to biochemical pathways using Gene Set Analysis, and developed a novel rank-based method to select pathways on the basis of their enrichment in patient-derived omics data sets and prognostic relevance. Key mediators of the pathway were then characterized for their role in disease progression. Pyrimidine metabolism was altered in luminal versus basal BCa, whereas the combined expression of its associated genes or expression of one key gene, ribonucleotide reductase subunit M2 (RRM2) alone, associated significantly with decreased survival across all BCa subtypes, as well as in luminal patients resistant to tamoxifen. Increased RRM2 expression in tamoxifen-resistant patients was verified using tissue microarrays, whereas the metabolic products of RRM2 were higher in tamoxifen-resistant cells and in xenograft tumors. Both genetic and pharmacological inhibition of this key enzyme in tamoxifen-resistant cells significantly decreased proliferation, reduced expression of cell cycle genes, and sensitized the cells to tamoxifen treatment. Our study suggests for evaluating RRM2-associated metabolites as noninvasive markers for tamoxifen resistance and its pharmacological inhibition as a novel approach to overcome tamoxifen resistance in BCa.
Collapse
Affiliation(s)
- Nagireddy Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Suman Maity
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Ramakrishna Kommagani
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Vasanta Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Fengju Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sarmishta Nanda
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Salil Kumar Bhowmik
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Atsushi Terunuma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Agostina Nardone
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoyong Fu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Chad Shaw
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Tapasree Roy Sarkar
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rachel Schiff
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA; Verna and Marrs McLean Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Davuluri G, Schiemann WP, Plow EF, Sossey-Alaoui K. Loss of WAVE3 sensitizes triple-negative breast cancers to chemotherapeutics by inhibiting the STAT-HIF-1α-mediated angiogenesis. JAKSTAT 2015; 3:e1009276. [PMID: 26413422 DOI: 10.1080/21623996.2015.1009276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 01/10/2023] Open
Abstract
Chemoresistance allows for disease to recur and ultimately causes the death of most breast cancer patients. This scenario is particularly relevant in patients harboring triple-negative breast cancer (TNBC) tumors for which there are no effective FDA-approved drugs. However, a recent study determined that TNBCs can be segregated into 6 genetically distinct subtypes that do in fact exhibit differential rates of pathological complete response (pCR) to standard-of-care chemotherapies. Of these, the mesenchymal and mesenchymal stem-like subtypes of TNBCs exhibit the lowest rates of pCR when treated with standard-of-care chemotherapies. WAVE3 is an actin-cytoskeleton remodeling protein, and recent studies have highlighted a potential role for WAVE3 in promoting tumor progression and metastasis in TNBC. However, whether WAVE3 activity is involved in the development of chemoresistance in TNBCs remains unclear. Here we show that loss of WAVE3 expression resensitizes human TNBC cells to doxorubicin and docetaxel, as measured by increased apoptosis and cell death. We also show that WAVE3 knockdown in the chemotherapy-treated TNBC cells results in inhibition of STAT1 phosphorylation, as well as a significant decrease in expression levels of its downstream effector HIF-1α. Since HIF-1α is a major activator of VEGF-A production, and therefore a stimulator of tumor angiogenesis, loss of HIF-1α in the WAVE3-knockdown cells resulted in the inhibition the chemotherapy-mediated VEGF-A secretion and the downstream activation of angiogenesis, a phenomenon that often accompanies chemoresistance. Our data identify a critical role of WAVE3 in sensitizing TNBC to chemotherapy by inhibiting the STAT1→HIF-1α→VEGF-A signaling axis, and support the possibility that WAVE3 inhibition may be a promising target for TNBC cancer therapy.
Collapse
Affiliation(s)
- Gangarao Davuluri
- Department of Molecular Cardiology; Cleveland Clinic Lerner Institute ; Cleveland, OH USA
| | - William P Schiemann
- Case Comprehensive Cancer Center; Case Western Reserve University ; Cleveland, OH USA
| | - Edward F Plow
- Department of Molecular Cardiology; Cleveland Clinic Lerner Institute ; Cleveland, OH USA
| | - Khalid Sossey-Alaoui
- Department of Molecular Cardiology; Cleveland Clinic Lerner Institute ; Cleveland, OH USA
| |
Collapse
|
13
|
Sossey-Alaoui K. Surfing the big WAVE: Insights into the role of WAVE3 as a driving force in cancer progression and metastasis. Semin Cell Dev Biol 2013; 24:287-97. [PMID: 23116924 PMCID: PMC4207066 DOI: 10.1016/j.semcdb.2012.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/16/2012] [Accepted: 10/17/2012] [Indexed: 02/06/2023]
Abstract
WAVE3 belongs to the WASP/WAVE family of actin cytoskeleton remodeling proteins. These proteins are known to be involved in several biological functions ranging from controlling cell shape and movement, to being closely associated with pathological conditions such as cancer progression and metastasis. Last decade has seen an explosion in the literature reporting significant scientific advances on the molecular mechanisms whereby the WASP/WAVE proteins are regulated both in normal physiological as well as pathological conditions. The purpose of this review is to present the major findings pertaining to how WAVE3 has become a critical player in the regulation of signaling pathways involved in cancer progression and metastasis. The review will conclude with suggesting options for the potential use of WAVE3 as a therapeutic target to prevent the progression of cancer to the lethal stage that is the metastatic disease.
Collapse
Affiliation(s)
- Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Cleveland Clinic Lerner Research Institute, 9500 Euclid Ave., NB-50, Cleveland, OH 44195, USA.
| |
Collapse
|
14
|
Eastman A, Tammaro Y, Moldrem A, Andrews V, Huth J, Euhus D, Leitch M, Rao R. Outcomes of delays in time to treatment in triple negative breast cancer. Ann Surg Oncol 2013; 20:1880-5. [PMID: 23292484 DOI: 10.1245/s10434-012-2835-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND Compared with other breast cancer subtypes, triple negative breast cancers (TNBC) are associated with higher recurrence rates and worse survival. Because of the aggressive nature of TNBC, outcomes may be more sensitive to delays in time to treatment. This study evaluates whether delays from diagnosis to initial treatment in TNBC impacts survival or locoregional recurrence (LRR). METHODS Retrospective review of TNBC patients treated between January 2004 and January 2011 at an academic center was performed. Data collected included demographics, pathology, treatment, recurrence, and survival. Interval to treatment was defined as days from pathologic diagnosis to first local or systemic treatment. The t test, Cox regression, and Kaplan-Meier analyses were used to evaluate impact of time to treatment on overall survival and LRR. RESULTS Median follow-up was 40 months for 301 TNBC patients. Mean interval to treatment was 46 ± 2 days. Higher initial stage yielded worse survival (p < .0001). Interval to treatment did not impact overall survival (p = .24), although there was a trend toward worse survival with delays of >90 days (p = .06). LRR was seen in 20 patients (7 %). Median time to recurrence was 15 months. Time to treatment was 38 ± 6 days for patients with LRR versus 44 ± 2 days without a recurrence (p = .37). Short delay in time to treatment did not impact LRR (p = .54). CONCLUSIONS In TNBC, a short delay from pathologic diagnosis to initial treatment does not appear to adversely affect survival or LRR. Appropriate time to perform evaluations such as genetic testing, imaging, or additional consultation can be taken to guide optimal treatment options.
Collapse
Affiliation(s)
- Amy Eastman
- Department of Surgery, Division of Surgical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Kulkarni S, Augoff K, Rivera L, McCue B, Khoury T, Groman A, Zhang L, Tian L, Sossey-Alaoui K. Increased expression levels of WAVE3 are associated with the progression and metastasis of triple negative breast cancer. PLoS One 2012; 7:e42895. [PMID: 22952619 PMCID: PMC3428347 DOI: 10.1371/journal.pone.0042895] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/13/2012] [Indexed: 12/15/2022] Open
Abstract
Background Breast Cancer (BC) is a heterogeneous disease comprised of at least five genetically distinct subtypes, which together form the second leading cause of cancer death in women in the United States. Within BC subtypes, those classified as Triple Negative BCs (TNBCs) exhibit dismal survival rates due to their propensity to develop distant metastases. We have identified the WAVE3 protein, which is a critical regulator of actin cytoskeleton dynamics that are required for the motility and invasion of cancer cells through its activation of the Arp2/3 complex, as a key regulator of the different steps of the invasion-metastasis cascade in BC, especially in the more aggressive TNBCs. Our published studies have also shown that elevated expression levels of WAVE3 in the TNBC cell lines directly contribute to their increased invasion and metastasis potentials both in vitro and in vivo in murine models of BC metastasis. Methodology/Principal Findings Herein, we utilized both immunohistochemistry (IHC) of primary human BC tumors as well as quantitative real-time RT-PCR of WAVE3 in the peripheral blood of BC patients to clearly establish that WAVE3 is a predictive marker of overall BC patients’ survival. High levels of WAVE3 were predictive for reduced distant recurrence-free survival as well as for decreased disease-specific mortality. Our analysis of WAVE3 expression levels in the peripheral blood of BC patients showed that WAVE3 is highly expressed in the blood of patients who developed metastatic breast cancer compared to those who did not. WAVE3 expression was also highly upregulated in the blood of BC patients with the more aggressive TNBC subtype. Conclusions Together, these findings establish WAVE3 as a novel marker for increased risk of breast-cancer-specific mortality and for the metastatic potential of the TNBCs, and also identify WAVE3 as an attractive therapeutic target for the treatment of metastatic BC.
Collapse
Affiliation(s)
- Swati Kulkarni
- Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- * E-mail: (SK); (KSA)
| | - Katarzyna Augoff
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Louis Rivera
- Department of Surgery, Naval Medical Center San Diego, San Diego, California, United States of America
| | - Brian McCue
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Thaer Khoury
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Adrienne Groman
- Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Li Zhang
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lili Tian
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail: (SK); (KSA)
| |
Collapse
|
16
|
Lavasani MA, Moinfar F. Molecular classification of breast carcinomas with particular emphasis on "basal-like" carcinoma: a critical review. JOURNAL OF BIOPHOTONICS 2012; 5:345-366. [PMID: 22232077 DOI: 10.1002/jbio.201100097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 12/14/2011] [Accepted: 12/16/2011] [Indexed: 05/31/2023]
Abstract
During the last 11 years, 5 molecular subtypes of breast carcinoma (luminal A, luminal B, Her2-positive, basal-like, and normal breast-like) have been characterized and intensively studied. As genomic research evolves, further subtypes of breast cancers into new "molecular entities" are expected to occur. For example, a new and rare breast cancer subtype, known as claudin-low, has been recently found in human carcinomas and in breast cancer cell lines. There is no doubt that global gene expression analyses using high-throughput biotechnologies have drastically improved our understanding of breast cancer as a heterogeneous disease. The main question is, however, whether new molecular techniques such as gene expression profiling (or signature) should be regarded as the gold standard for identifying breast cancer subtypes. A critical review of the literature clearly shows major problems with current molecular techniques and classification including poor definitions, lack of reproducibility, and lack of quality control. Therefore, the current molecular approaches cannot be incorporated into routine clinical practice and treatment decision making as they are immature or even can be misleading. This review particularly focuses on the "basal-like" breast cancer subtype that represents one of the most popular breast cancer "entities". It critically shows major problems and misconceptions with and about this subtype and challenges the common claim that it represents a "distinct entity". It concludes that the term "basal-like" is misleading and states that there is no evidence that expression of basal-type cytokeratins in a given breast cancer, regardless of other established prognostic factors, does have any impact on clinical outcome.
Collapse
Affiliation(s)
- Mohammad Ali Lavasani
- Unit of Breast and Gynecologic Pathology, Department of Pathology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
17
|
Eswaran J, Cyanam D, Mudvari P, Reddy SDN, Pakala SB, Nair SS, Florea L, Fuqua SAW, Godbole S, Kumar R. Transcriptomic landscape of breast cancers through mRNA sequencing. Sci Rep 2012; 2:264. [PMID: 22355776 PMCID: PMC3278922 DOI: 10.1038/srep00264] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 01/17/2012] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a heterogeneous disease with a poorly defined genetic landscape, which poses a major challenge in diagnosis and treatment. By massively parallel mRNA sequencing, we obtained 1.2 billion reads from 17 individual human tissues belonging to TNBC, Non-TNBC, and HER2-positive breast cancers and defined their comprehensive digital transcriptome for the first time. Surprisingly, we identified a high number of novel and unannotated transcripts, revealing the global breast cancer transcriptomic adaptations. Comparative transcriptomic analyses elucidated differentially expressed transcripts between the three breast cancer groups, identifying several new modulators of breast cancer. Our study also identified common transcriptional regulatory elements, such as highly abundant primary transcripts, including osteonectin, RACK1, calnexin, calreticulin, FTL, and B2M, and "genomic hotspots" enriched in primary transcripts between the three groups. Thus, our study opens previously unexplored niches that could enable a better understanding of the disease and the development of potential intervention strategies.
Collapse
Affiliation(s)
- Jeyanthy Eswaran
- McCormick Genomic and Proteomics Center, The George Washington University, Washington, DC 20037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Augoff K, McCue B, Plow EF, Sossey-Alaoui K. miR-31 and its host gene lncRNA LOC554202 are regulated by promoter hypermethylation in triple-negative breast cancer. Mol Cancer 2012; 11:5. [PMID: 22289355 PMCID: PMC3298503 DOI: 10.1186/1476-4598-11-5] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/30/2012] [Indexed: 12/19/2022] Open
Abstract
Background microRNAs have been established as powerful regulators of gene expression in normal physiological as well as in pathological conditions, including cancer progression and metastasis. Recent studies have demonstrated a key role of miR-31 in the progression and metastasis of breast cancer. Downregulation of miR-31 enhances several steps of the invasion-metastasis cascade in breast cancer, i.e., local invasion, extravasation and survival in the circulation system, and metastatic colonization of distant sites. miR-31 exerts its metastasis-suppressor activity by targeting a cohort of pro-metastatic genes, including RhoA and WAVE3. The molecular mechanisms that lead to the loss of miR-31 and the activation of its pro-metastatic target genes during these specific steps of the invasion-metastasis cascade are however unknown. Results In the present report, we identify promoter hypermethylation as one of the major mechanisms for silencing miR-31 in breast cancer, and in the triple-negative breast cancer (TNBC) cell lines of basal subtype, in particular. miR-31 maps to the intronic sequence of a novel long non-coding (lnc)RNA, LOC554202 and the regulation of its transcriptional activity is under control of LOC554202. Both miR-31 and the host gene LOC554202 are down-regulated in the TNBC cell lines of basal subtype and over-expressed in the luminal counterparts. Treatment of the TNBC cell lines with either a de-methylating agent alone or in combination with a de-acetylating agent resulted in a significant increase of both miR-31 and its host gene, suggesting an epigenetic mechanism for the silencing of these two genes by promoter hypermethylation. Finally, both methylation-specific PCR and sequencing of bisulfite-converted DNA demonstrated that the LOC554202 promoter-associated CpG island is heavily methylated in the TNBC cell lines and hypomethylated in the luminal subtypes. Conclusion Loss of miR-31 expression in TNBC cell lines is attributed to hypermethylation of its promoter-associated CpG island. Together, our results provide the initial evidence for a mechanism by which miR-31, an important determinant of the invasion metastasis cascade, is regulated in breast cancer.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | |
Collapse
|
19
|
Expression Profiling-Based Subtyping Identifies Novel Non-small Cell Lung Cancer Subgroups and Implicates Putative Resistance to Pemetrexed Therapy. J Thorac Oncol 2012; 7:105-14. [DOI: 10.1097/jto.0b013e3182352a45] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Higa GM. The microtubule as a breast cancer target. Breast Cancer 2010; 18:103-19. [PMID: 20862571 DOI: 10.1007/s12282-010-0224-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 08/24/2010] [Indexed: 01/19/2023]
Abstract
Manifestations of non-equilibrium polarity, random transgressions, and catastrophes are not conditions usually associated with a sense of normalcy. Yet these disquieting features distinguish a utilitarian behavior known as dynamic instability, the signature characteristic of the microtubule. Long known to be a tumor target, disruption of this fragile attribute is associated with some of the most effective agents used to treat breast cancer today. Although the biology of the microtubule is under intense investigation much still remains unknown. As such, our understanding of regulatory molecules and resistance mechanisms are still rudimentary, further compromising our ability to develop novel therapeutic strategies to improve microtubule inhibitors. This review focuses on several classes of anti-microtubule agents and their effects on the functional dynamics of the targeted polymer. The primary objective is to critically examine the molecular mechanisms that contribute to tumor cell death, tumor-resistance, and incident neurotoxicity.
Collapse
Affiliation(s)
- Gerald M Higa
- Mary Babb Randolph Cancer Center, West Virginia University Schools of Pharmacy and Medicine, Morgantown, WV, USA.
| |
Collapse
|
21
|
Sandhu R, Parker JS, Jones WD, Livasy CA, Coleman WB. Microarray-Based Gene Expression Profiling for Molecular Classification of Breast Cancer and Identification of New Targets for Therapy. Lab Med 2010. [DOI: 10.1309/lmlik0vie3cjk0wd] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
22
|
Perez EA, Patel T, Moreno-Aspitia A. Efficacy of ixabepilone in ER/PR/HER2-negative (triple-negative) breast cancer. Breast Cancer Res Treat 2010; 121:261-71. [DOI: 10.1007/s10549-010-0824-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 02/25/2010] [Indexed: 12/11/2022]
|
23
|
Sambade MJ, Camp JT, Kimple RJ, Sartor CI, Shields JM. Mechanism of lapatinib-mediated radiosensitization of breast cancer cells is primarily by inhibition of the Raf>MEK>ERK mitogen-activated protein kinase cascade and radiosensitization of lapatinib-resistant cells restored by direct inhibition of MEK. Radiother Oncol 2009; 93:639-44. [PMID: 19853943 DOI: 10.1016/j.radonc.2009.09.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 08/25/2009] [Accepted: 09/15/2009] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE We recently showed that lapatinib, an EGFR/HER2 inhibitor, radiosensitized breast cancer cells of the basal and HER2+ subtypes. The purpose of this study was to identify the downstream signaling pathways responsible for lapatinib-mediated radiosensitization in breast cancer. MATERIALS AND METHODS Response of EGFR downstream signaling pathways was assessed by Western blot and clonogenic cell survival assays in breast tumor cells after irradiation (5Gy), lapatinib, CI-1040, or combined treatment. RESULTS In SUM102 cells, an EGFR+ basal breast cancer cell line, exposure to ionizing radiation elicited strong activation of ERK1/2 and JNK, which was blocked by lapatinib, and weak/no activation of p38, AKT or STAT3. Direct inhibition of MEK1 with CI-1040 resulted in 95% inhibition of surviving colonies when combined with radiation while inhibition of JNK with SP600125 had no effect. Lapatinib-mediated radiosensitization of SUM102 cells was completely abrogated with expression of constitutively active Raf. Treatment of lapatinib-resistant SUM185 cells with CI-1040 restored radiosensitization with 45% fewer surviving colonies when combined with radiation. CONCLUSIONS These data suggest that radiosensitization by lapatinib is mediated largely through inhibition of MEK/ERK and that direct inhibition of this pathway may provide an additional avenue of radiosensitization in EGFR+ or HER2+ breast cancers.
Collapse
Affiliation(s)
- Maria J Sambade
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
24
|
ErbB family immunohistochemical expression in colorectal cancer patients with higher risk of recurrence after radical surgery. Int J Colorectal Dis 2009; 24:1059-68. [PMID: 19390858 DOI: 10.1007/s00384-009-0702-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/01/2009] [Indexed: 02/04/2023]
Abstract
PURPOSE Investigate ErbB family expression in colorectal cancer patients with higher risk of recurrence after surgical treatment. METHODS We studied 109 individuals with high risk stage II and stage III patients submitted to radical surgery. ErbB expression was assessed by tissue microarray technique. RESULTS The immunohistochemical expression was considered positive for EGFR, ErbB2, ErbB3, ErbB4 membrane, and ErbB4 cytoplasmic in respectively 57.8%, 8.3%, 69.7%, 11%, and 19.3% of patients. ErbB3 negative expression was associated with lymphovascular invasion. EGFR, ErbB2, and cytoplasmic ErbB4 expression was not associated with prognosis. Membranous positive ErbB4 expression was an independent prognostic factor for recurrence. ErbB3 negative expression was an independent prognostic factor for recurrence and survival in the multivariate analysis. CONCLUSIONS The immunohistochemical expression of ErbB3 and ErbB4 may identify a subgroup with stage II and III colorectal cancer at higher risk of recurrence.
Collapse
|
25
|
O'Brien C, Cavet G, Pandita A, Hu X, Haydu L, Mohan S, Toy K, Rivers CS, Modrusan Z, Amler LC, Lackner MR. Functional genomics identifies ABCC3 as a mediator of taxane resistance in HER2-amplified breast cancer. Cancer Res 2008; 68:5380-9. [PMID: 18593940 DOI: 10.1158/0008-5472.can-08-0234] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Breast cancer is a heterogeneous disease with distinct molecular subtypes characterized by differential response to targeted and chemotherapeutic agents. Enhanced understanding of the genetic alterations characteristic of different subtypes is needed to pave the way for more personalized administration of therapeutic agents. We have taken a functional genomics approach using a well-characterized panel of breast cancer cell lines to identify putative biomarkers of resistance to antimitotic agents such as paclitaxel and monomethyl-auristatin-E (MMAE). In vitro studies revealed a striking difference in sensitivity to these agents between cell lines from different subtypes, with basal-like cell lines being significantly more sensitive to both agents than luminal or HER2-amplified cell lines. Genome-wide association studies using copy number data from Affymetrix single nucleotide polymorphism arrays identified amplification of the chromosome 17q21 region as being highly associated with resistance to both paclitaxel and MMAE. An unbiased approach consisting of RNA interference and high content analysis was used to show that amplification and concomitant overexpression of the gene encoding the ABCC3 drug transporter is responsible for conferring in vitro resistance to paclitaxel and MMAE. We also show that amplification of ABCC3 is present in primary breast tumors and that it occurs predominantly in HER2-amplified and luminal tumors, and we report on development of a specific fluorescence in situ hybridization assay that may have utility as a predictive biomarker of taxane resistance in breast cancer.
Collapse
Affiliation(s)
- Carol O'Brien
- Department of Oncology, Genentech, Inc, South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Higher Incidence of Aggressive Breast Cancers in African-American Women: A Review. J Natl Med Assoc 2008; 100:698-702. [DOI: 10.1016/s0027-9684(15)31344-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
Nishimura R, Arima N. Is triple negative a prognostic factor in breast cancer? Breast Cancer 2008; 15:303-8. [PMID: 18369692 DOI: 10.1007/s12282-008-0042-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 01/08/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Breast cancer is characterized by hormone dependency, and endocrine therapy is a key treatment in breast cancer. Recently, targeted therapies such as Trastuzumab treatment for HER2-positive breast cancer has been important. Triple-negative (TN) breast cancer is characterized by lack of expression of estrogen receptor (ER) and progesterone receptor (PgR), and the absence of HER2 protein overexpression, and so there is no targeted therapy for this subtype. In this study, we examined the biological and prognostic characteristics in TN breast cancer. PATIENTS AND METHODS Between January 1998 and September 2006, 1,552 patients with primary breast cancer were investigated retrospectively in this study and ER, PgR and HER2 status were evaluated in all cases. Furthermore, p53 overexpression and Ki67 values were examined immunohistochemically. RESULTS Patient distribution according to ER, PgR or HER2 status was as follows: ER and PgR positive: 57.9%, and ER and PgR negative: 25.1%. With regards to the HER2 status, HER2 positive was 23.3%, and triple negative (TN) was 14.0%. TN breast cancer has a high proliferation rate, high nuclear grade and frequent p53 overexpression. Patients with TN tumors had a significantly poorer disease-free survival (DFS) than those with non-TN tumors. After recurrence the overall survival (OS) rate in TN cases was significantly lower than that of the non-TN cases. Multivariate analysis revealed that TN was a significant factor for DFS and OS after recurrence. CONCLUSION TN breast cancer is a rare subtype with a high proliferation rate and a high nuclear grade, p53 overexpression, and lower DFS/OS. To improve the prognosis of TN breast cancer, a new effective strategy needs to be developed.
Collapse
Affiliation(s)
- Reiki Nishimura
- Department of Breast and Endocrine Surgery, Kumamoto City Hospital, Kumamoto 862-8505, Japan.
| | | |
Collapse
|