1
|
Røgenes H, Finne K, Winge I, Akslen LA, Östman A, Milosevic V. Development of 42 marker panel for in-depth study of cancer associated fibroblast niches in breast cancer using imaging mass cytometry. Front Immunol 2024; 15:1325191. [PMID: 38711512 PMCID: PMC11070582 DOI: 10.3389/fimmu.2024.1325191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Imaging Mass Cytometry (IMC) is a novel, and formidable high multiplexing imaging method emerging as a promising tool for in-depth studying of tissue architecture and intercellular communications. Several studies have reported various IMC antibody panels mainly focused on studying the immunological landscape of the tumor microenvironment (TME). With this paper, we wanted to address cancer associated fibroblasts (CAFs), a component of the TME very often underrepresented and not emphasized enough in present IMC studies. Therefore, we focused on the development of a comprehensive IMC panel that can be used for a thorough description of the CAF composition of breast cancer TME and for an in-depth study of different CAF niches in relation to both immune and breast cancer cell communication. We established and validated a 42 marker panel using a variety of control tissues and rigorous quantification methods. The final panel contained 6 CAF-associated markers (aSMA, FAP, PDGFRa, PDGFRb, YAP1, pSMAD2). Breast cancer tissues (4 cases of luminal, 5 cases of triple negative breast cancer) and a modified CELESTA pipeline were used to demonstrate the utility of our IMC panel for detailed profiling of different CAF, immune and cancer cell phenotypes.
Collapse
Affiliation(s)
- Hanna Røgenes
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kenneth Finne
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ingeborg Winge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Arne Östman
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Vladan Milosevic
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
2
|
De Santis P, Perrone M, Guarini C, Santoro AN, Laface C, Carrozzo D, Oliva GR, Fedele P. Early-stage triple negative breast cancer: the therapeutic role of immunotherapy and the prognostic value of pathological complete response. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:232-250. [PMID: 38464390 PMCID: PMC10918232 DOI: 10.37349/etat.2024.00215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/26/2023] [Indexed: 03/12/2024] Open
Abstract
Triple negative breast cancer (TNBC) represents an aggressive disease associated with a high risk of recurrence after curative treatment and a poor prognosis in the metastatic setting. Chemotherapy was for years the only treatment available in the early and metastatic setting, due to the lack of actionable targets. Clinical practice has changed following the results obtained with the addition of immunotherapy to standard chemotherapy, the development of novel drugs [i.e. antibody-drug conjugates (ADCs)], and the use of targeted treatments for patients carrying germline pathogenic breast cancer susceptibility genes (BRCA) 1 or BRCA 2 variants. The treatment of early-stage disease has had a shift in clinical practice since July 2021, after the Food and Drug Administration (FDA) approval of pembrolizumab in association with chemotherapy as neoadjuvant treatment for TNBC and as a single agent in the subsequent adjuvant setting. This intensive treatment based on the combination of a poly-chemotherapy and an immune checkpoint inhibitor (ICI) led to the improvement of short- and long-term outcomes, but it has highlighted some new unmet clinical needs in the treatment of early-stage TNBC: the selection of the most effective adjuvant therapy and the integration of pembrolizumab with other therapeutic strategies [capecitabine, poly(ADP-ribose) polymerase (PARP) inhibitors] based on the achievement of pathologic complete response (pCR); the identification of predictive biomarkers to select patients who could most benefit from the addition of ICI, to minimize toxicities and to maximize outcomes; the possibility of de-escalating chemotherapy in favor of immune-combo or novel agents, such as ADCs; the role of immunotherapy in estrogen receptor (ER)-low patients. The advent of immunotherapy not only addresses current challenges in TNBC treatment but also holds the promise of a radical transformation in its therapeutic paradigm, enhancing significantly clinical outcomes and offering new perspectives for patients grappling with this aggressive form of breast cancer.
Collapse
Affiliation(s)
- Pierluigi De Santis
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Martina Perrone
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Chiara Guarini
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Anna Natalizia Santoro
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Carmelo Laface
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Daniela Carrozzo
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| | - Gaia Rachele Oliva
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Palma Fedele
- Oncology Unit, Francavilla Fontana Ceglie Messapica Hospital District, 72021 Francavilla Fontana, Italy
| |
Collapse
|
3
|
Nerone M, Rossi L, Condorelli R, Ratti V, Conforti F, Palazzo A, Graffeo R. Beyond PARP Inhibitors in Advanced Breast Cancer Patients with Germline BRCA1/2 Mutations: Focus on CDK4/6-Inhibitors and Data Review on Other Biological Therapies. Cancers (Basel) 2023; 15:3305. [PMID: 37444415 DOI: 10.3390/cancers15133305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
We explored the outcomes of germline BRCA1/2 pathogenic/likely pathogenic variants (PVs/LPVs) in the endocrine-sensitive disease treated with first-line standard of care cyclin-dependent kinase 4/6 (CDK4/6) inhibitors. Three studies retrospectively showed a reduction in the overall survival (OS) and progression-free survival (PFS) in gBRCA1/2m patients compared to both the germinal BRCA1/2 wild type (gBRCA1/2wt) and the untested population. Regarding the efficacy of PI3Kα inhibitors, there are no subgroups or biomarker analyses in which germinal BRCA status was explored. However, the biological interactions between the PIK3CA/AKT/mTOR pathway and BRCA1/2 at a molecular level could help us to understand the activity of these drugs when used to treat BC in BRCA1/2 PVs/LPVs carriers. The efficacy of trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate (ADC) targeting HER2 for HER2-low and HER2-positive (HER2+) BC, has been increasingly described. Unfortunately, data on T-DXd in HER2+ or HER2-low metastatic BC harboring germinal BRCA1/2 PVs/LPVs is lacking. Including germinal BRCA1/2 status in the subgroup analysis of the registration trials of this ADC would be of great interest, especially in the phase III trial DESTINY-breast04. This trial enrolled patients with HER2-negative (HER2-) and both HR+ and HR- metastatic disease, which can now be categorized as HER2-low. The HER2-low subgroup includes tumors that were previously classified as triple negative, so it is highly likely that some women were germline BRCA1/2 PVs/LPVs carriers and this data was not reported. Germline BRCA1/2 status will be available for a higher number of individuals with BC in the near future, and data on the prognostic and predictive role of these PVs/LPVs is needed in order to choose the best treatment options.
Collapse
Affiliation(s)
- Marta Nerone
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Lorenzo Rossi
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Rosaria Condorelli
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Vilma Ratti
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| | - Fabio Conforti
- Oncology Unit, Humanitas Gavazzeni, 24125 Bergamo, Italy
| | - Antonella Palazzo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Rossella Graffeo
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland
| |
Collapse
|
4
|
Fonseca-Montaño MA, Vázquez-Santillán KI, Hidalgo-Miranda A. The current advances of lncRNAs in breast cancer immunobiology research. Front Immunol 2023; 14:1194300. [PMID: 37342324 PMCID: PMC10277570 DOI: 10.3389/fimmu.2023.1194300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy and the leading cause of cancer-related death in women worldwide. Breast cancer development and progression are mainly associated with tumor-intrinsic alterations in diverse genes and signaling pathways and with tumor-extrinsic dysregulations linked to the tumor immune microenvironment. Significantly, abnormal expression of lncRNAs affects the tumor immune microenvironment characteristics and modulates the behavior of different cancer types, including breast cancer. In this review, we provide the current advances about the role of lncRNAs as tumor-intrinsic and tumor-extrinsic modulators of the antitumoral immune response and the immune microenvironment in breast cancer, as well as lncRNAs which are potential biomarkers of tumor immune microenvironment and clinicopathological characteristics in patients, suggesting that lncRNAs are potential targets for immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Marco Antonio Fonseca-Montaño
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado, Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
5
|
Loizides S, Constantinidou A. Triple negative breast cancer: Immunogenicity, tumor microenvironment, and immunotherapy. Front Genet 2023; 13:1095839. [PMID: 36712858 PMCID: PMC9879323 DOI: 10.3389/fgene.2022.1095839] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a biologically diverse subtype of breast cancer characterized by genomic and transcriptional heterogeneity and exhibiting aggressive clinical behaviour and poor prognosis. In recent years, emphasis has been placed on the identification of mechanisms underlying the complex genomic and biological profile of TNBC, aiming to tailor treatment strategies. High immunogenicity, specific immune activation signatures, higher expression of immunosuppressive genes and higher levels of stromal Tumor Infiltrating Lymphocytes, constitute some of the key elements of the immune driven landscape associated with TNBC. The unprecedented response of TNBC to immunotherapy has undoubtedly changed the standard of care in this disease both in the early and the metastatic setting. However, the extent of interplay between immune infiltration and mutational signatures in TNBC is yet to be fully unravelled. In the present review, we present clinical evidence on the immunogenicity and tumour microenvironment influence on TNBC progression and the current treatment paradigms in TNBC based on immunotherapy.
Collapse
Affiliation(s)
- Sotiris Loizides
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - Anastasia Constantinidou
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Medical School, University of Cyprus, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| |
Collapse
|
6
|
When breaks get hot: inflammatory signaling in BRCA1/2-mutant cancers. Trends Cancer 2022; 8:174-189. [PMID: 35000881 DOI: 10.1016/j.trecan.2021.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
Abstract
Genomic instability and inflammation are intricately connected hallmark features of cancer. DNA repair defects due to BRCA1/2 mutation instigate immune signaling through the cGAS/STING pathway. The subsequent inflammatory signaling provides both tumor-suppressive as well as tumor-promoting traits. To prevent clearance by the immune system, genomically instable cancer cells need to adapt to escape immune surveillance. Currently, it is unclear how genomically unstable cancers, including BRCA1/2-mutant tumors, are rewired to escape immune clearance. Here, we summarize the mechanisms by which genomic instability triggers inflammatory signaling and describe adaptive mechanisms by which cancer cells can 'fly under the radar' of the immune system. Additionally, we discuss how therapeutic activation of the immune system may improve treatment of genomically instable cancers.
Collapse
|
7
|
Kossai M, Radosevic-Robin N, Penault-Llorca F. Refining patient selection for breast cancer immunotherapy: beyond PD-L1. ESMO Open 2021; 6:100257. [PMID: 34487970 PMCID: PMC8426207 DOI: 10.1016/j.esmoop.2021.100257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Therapies that modulate immune response to cancer, such as immune checkpoint inhibitors, began an intense development a few years ago; however, in breast cancer (BC), the results have been relatively disappointing so far. Finding biomarkers for better selection of BC patients for various immunotherapies remains a significant unmet medical need. At present, only tumour tissue programmed death-ligand 1 (PD-L1) and mismatch repair deficiency status are approved as theranostic biomarkers for programmed cell death-1 (PD-1)/PD-L1 inhibitors in BC. However, due to the complexity of tumour microenvironment (TME) and cancer response to immunomodulators, none of them is a perfect selector. Therefore, an intense quest is ongoing for complementary tumour- or host-related predictive biomarkers in breast immuno-oncology. Among the upcoming biomarkers, quantity, immunophenotype and spatial distribution of tumour-infiltrating lymphocytes and other TME cells as well as immune gene signatures emerge as most promising and are being increasingly tested in clinical trials. Biomarkers or strategies allowing dynamic assessment of BC response to immunotherapy, such as circulating/exosomal PD-L1, quantity of white/immune blood cell subpopulations and molecular imaging are particularly suitable for immunotreatment monitoring. Finally, host-related factors, such as microbiome and lifestyle, should also be taken into account when planning integration of immunomodulating therapies into BC management. As none of the biomarkers taken separately is accurate enough, the solution could come from composite biomarkers, which would combine clinical, molecular and immunological features of the disease, possibly powered by artificial intelligence. At present, immune checkpoint inhibitors (ICIs) are the only approved immunotherapy drugs in BC. Tumour PD-L1 and microsatellite status are current companion biomarkers for ICIs in BC; however, these need improvement. Evaluation of tumour immune contexture and the dynamics of circulating immune cell counts are promising novel approaches. Development of noninvasive monitoring and composite biomarkers will facilitate cancer immunotherapy, including in BC.
Collapse
Affiliation(s)
- M Kossai
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France
| | - N Radosevic-Robin
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France.
| | - F Penault-Llorca
- Department of Pathology, University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Clermont-Ferrand, France
| |
Collapse
|
8
|
Thomas R, Al-Khadairi G, Decock J. Immune Checkpoint Inhibitors in Triple Negative Breast Cancer Treatment: Promising Future Prospects. Front Oncol 2021; 10:600573. [PMID: 33718107 PMCID: PMC7947906 DOI: 10.3389/fonc.2020.600573] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has emerged as the fifth pillar of cancer treatment alongside surgery, radiotherapy, chemotherapy, and targeted therapy. Immune checkpoint inhibitors are the current superheroes of immunotherapy, unleashing a patient's own immune cells to kill tumors and revolutionizing cancer treatment in a variety of cancers. Although breast cancer was historically believed to be immunologically silent, treatment with immune checkpoint inhibitors has been shown to induce modest responses in metastatic breast cancer. Given the inherent heterogeneity of breast tumors, this raised the question whether certain breast tumors might benefit more from immune-based interventions and which cancer cell-intrinsic and/or microenvironmental factors define the likelihood of inducing a potent and durable anti-tumor immune response. In this review, we will focus on triple negative breast cancer as immunogenic breast cancer subtype, and specifically discuss the relevance of tumor mutational burden, the plethora and diversity of tumor infiltrating immune cells in addition to the immunoscore, the presence of immune checkpoint expression, and the microbiome in defining immune checkpoint blockade response. We will highlight the current immune checkpoint inhibitor treatment options, either as monotherapy or in combination with standard-of-care treatment modalities such as chemotherapy and targeted therapy. In addition, we will look into the potential of immunotherapy-based combination strategies using immune checkpoint inhibitors to enhance both innate and adaptive immune responses, or to establish a more immune favorable environment for cancer vaccines. Finally, the review will address the need for unambiguous predictive biomarkers as one of the main challenges of immune checkpoint blockade. To conclude, the potential of immune checkpoint blockade for triple negative breast cancer treatment could be enhanced by exploration of aforementioned factors and treatment strategies thereby providing promising future prospects.
Collapse
Affiliation(s)
- Remy Thomas
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ghaneya Al-Khadairi
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
9
|
Radulescu D, Baleanu VD, Padureanu V, Radulescu PM, Bordu S, Patrascu S, Socea B, Bacalbasa N, Surlin MV, Georgescu I, Georgescu EF. Neutrophil/Lymphocyte Ratio as Predictor of Anastomotic Leak after Gastric Cancer Surgery. Diagnostics (Basel) 2020; 10:E799. [PMID: 33050137 PMCID: PMC7601164 DOI: 10.3390/diagnostics10100799] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Neutrophil/lymphocyte ratio (NLR) is known as a prognostic for the outcome of the patients with gastric cancer. As no definite risk marker for anastomotic leakage after gastric resection was identified, we investigated the possible role of NLR. METHODS Peripheral blood count for neutrophils and lymphocytes was done at the patient's admission. We retrospectively evaluated 204 gastric cancer patients, who underwent gastric resection, comparing the values of NLR between the group of patients with anastomotic leakage and those without complications. RESULTS Using the ROC curve, we found the cutoff value of NLR, which permitted the comparison of the group with low NLR, presenting increased NLR. The cutoff value for NLR was 3.54. Between the two groups, we could observe statistically significant differences in developing fistula (p < 0.01) and complications leading to death (p < 0.025). The odds ratio for patients with NLR greater than 3.54 to develop anastomotic leak was 17.62, compared to those with lower NLR. CONCLUSION Peripheral blood NLR proved to be a predictor for anastomotic leakage.
Collapse
Affiliation(s)
- Dumitru Radulescu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Vlad Dumitru Baleanu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Vlad Padureanu
- Internal Medicine Department, County Hospital of Craiova, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Patricia Mihaela Radulescu
- “Victor Babes” Clinical Hospital of Infectious Diseases and Pneumophtisiology Craiova, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Silviu Bordu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Stefan Patrascu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Bogdan Socea
- General Surgery Department, “Carol Davila” University of Medicine and Pharmacy, 021659 Bucharest, Romania
| | - Nicolae Bacalbasa
- “Dr. Ion Cantacuzino” Hospital, Gynecology Department, “Carol Davila” University of Medicine and Pharmacy, 020457 Bucharest, Romania;
| | - Marin Valeriu Surlin
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Ion Georgescu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| | - Eugen Florin Georgescu
- General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.R.); (V.D.B.); (M.V.S.); (I.G.); (E.F.G.)
| |
Collapse
|
10
|
Huang Q, Cao Y, Wang S, Zhu R. Creation of a Novel Inflammation-Based Score for Operable Colorectal Cancer Patients. J Inflamm Res 2020; 13:659-671. [PMID: 33116746 PMCID: PMC7547789 DOI: 10.2147/jir.s271541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022] Open
Abstract
Aim Systemic inflammation has been implicated in the progression of patients with colorectal cancer (CRC). We evaluated the prognostic ability of a comprehensive score based on several inflammatory indexes in operable CRC patients. Patients and Methods Between July 2013 and September 2017, this study retrospectively identified 1279 CRC patients receiving radical surgery in Wuhan Union Hospital and randomly assigned them into training (N=921) and validation (N=358) sets. A novel score, the CRC-specific inflammatory index (CSII), was developed from a series of inflammatory indexes significantly associated with survival in patients with CRC. This novel score was then divided into three categories and compared to the well-known systematic inflammatory index (SII) and TNM stage. Finally, a survival nomogram was generated by combining the CSII and other informative clinical features. Results The CSII-OS was calculated as 1.110×lg ALRI + 1.082×CAR + 0.792×PI, while CSII-DFS was 1.709×lg ALRI + 1.033×CAR based on multivariable Cox regression analysis. Patients with high CSII experienced a worse OS (HR=23.72, 95% CI, 11.30-49.78, P <0.001) and worse DFS (HR=15.62, 95% CI, 6.95-35.08, P <0.001) compared to those in CRC patients with low CSII. Moreover, ROC analyses showed that the CSII possessed excellent performance (AUC=0.859) in predicting OS and DFS. The AUC of the OS nomogram based on CSII, TNM stage, and chemotherapy was 0.897, while that of the DFS nomogram based on CSII, T stage, and TNM stage was 0.873. High-quality calibration curves in both OS and DFS nomograms were observed. Verification in the validation dataset showed results consistent with those in the training dataset. Conclusion The CSII is a CRC-specific prognostic score based on the combination of available inflammatory indexes. High CSII is a strong predictor of worse survival outcomes. The CSII also exhibits better predictive performance compared to SII or TNM stage in operable CRC patients.
Collapse
Affiliation(s)
- Qian Huang
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yinghao Cao
- Department of Colorectal Surgery and Gastroenterology, Wuhan Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shouyi Wang
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Rui Zhu
- Department of Pediatric, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
11
|
Marinelli D, Mazzotta M, Pizzuti L, Krasniqi E, Gamucci T, Natoli C, Grassadonia A, Tinari N, Tomao S, Sperduti I, Sanguineti G, Botticelli A, Fabbri A, Botti C, Ciliberto G, Barba M, Vici P. Neoadjuvant Immune-Checkpoint Blockade in Triple-Negative Breast Cancer: Current Evidence and Literature-Based Meta-Analysis of Randomized Trials. Cancers (Basel) 2020; 12:cancers12092497. [PMID: 32899209 PMCID: PMC7565914 DOI: 10.3390/cancers12092497] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Breast cancer is a heterogeneous disease, which encompasses several subgroups of entities widely varying by clinical-pathological features. Triple negative breast cancer is characterized by a particularly aggressive biological behavior. The administration of chemotherapy has long represented the most efficacious weapon in combating triple negative breast cancer in both its initial and late phase of development. A pivot point has been recently reached throughout the approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer. Results from the registrative trial, IMpassion 130, have increasingly fueled the flourishing of studies of immune-checkpoint inhibitors in the early stage of triple negative breast cancer development. We critically interpret results from the most recent literature in light of relevant issues of methodological nature and also present a quantitative summary of data from the inherent trials. Future directions are also highlighted. Abstract Chemotherapy based on the sequential use of anthracyclines and taxanes has long represented the most efficacious approach in the management of early-stage, triple-negative breast cancer, whose aggressive behavior is widely renowned. This standard chemotherapy backbone was subsequently enriched by the use of carboplatin, based on its association with increased pathologic complete response and efficacy in the metastatic setting. Following the results from the IMpassion130 trial, the recent approval of the immunotherapic agent atezolizumab in combination with chemotherapy as first-line treatment for programmed-death ligand 1-positive, unresectable locally advanced, or metastatic triple-negative breast cancer increasingly fueled the flourishing of trials of immune-checkpoint inhibitors in the early setting. In this work, we review the most recent inherent literature in light of key methodological issues and provide a quantitative summary of the results from phase II–III randomized trials of immunotherapic agents combined with chemotherapy in the setting of interest. Hints regarding future directions are also discussed.
Collapse
Affiliation(s)
- Daniele Marinelli
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy;
| | - Marco Mazzotta
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| | - Teresa Gamucci
- Medical Oncology, Sandro Pertini Hospital, 00157 Rome, Italy;
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Antonino Grassadonia
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D’Annunzio University, 66100 Chieti, Italy; (C.N.); (A.G.); (N.T.)
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, ‘Sapienza’ University of Rome, 00161 Rome, Italy;
| | - Isabella Sperduti
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | | | - Agnese Fabbri
- Medical Oncology Unit, Belcolle Hospital, 01100 Viterbo, Italy;
| | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
- Correspondence: (L.P.); (M.B.); Tel.: +39-06-52665698 (L.P.); +39-06-52665419 (M.B.)
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.M.); (E.K.); (P.V.)
| |
Collapse
|
12
|
Glodzik D, Bosch A, Hartman J, Aine M, Vallon-Christersson J, Reuterswärd C, Karlsson A, Mitra S, Niméus E, Holm K, Häkkinen J, Hegardt C, Saal LH, Larsson C, Malmberg M, Rydén L, Ehinger A, Loman N, Kvist A, Ehrencrona H, Nik-Zainal S, Borg Å, Staaf J. Comprehensive molecular comparison of BRCA1 hypermethylated and BRCA1 mutated triple negative breast cancers. Nat Commun 2020; 11:3747. [PMID: 32719340 PMCID: PMC7385112 DOI: 10.1038/s41467-020-17537-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/02/2020] [Indexed: 02/08/2023] Open
Abstract
Homologous recombination deficiency (HRD) is a defining characteristic in BRCA-deficient breast tumors caused by genetic or epigenetic alterations in key pathway genes. We investigated the frequency of BRCA1 promoter hypermethylation in 237 triple-negative breast cancers (TNBCs) from a population-based study using reported whole genome and RNA sequencing data, complemented with analyses of genetic, epigenetic, transcriptomic and immune infiltration phenotypes. We demonstrate that BRCA1 promoter hypermethylation is twice as frequent as BRCA1 pathogenic variants in early-stage TNBC and that hypermethylated and mutated cases have similarly improved prognosis after adjuvant chemotherapy. BRCA1 hypermethylation confers an HRD, immune cell type, genome-wide DNA methylation, and transcriptional phenotype similar to TNBC tumors with BRCA1-inactivating variants, and it can be observed in matched peripheral blood of patients with tumor hypermethylation. Hypermethylation may be an early event in tumor development that progress along a common pathway with BRCA1-mutated disease, representing a promising DNA-based biomarker for early-stage TNBC.
Collapse
Affiliation(s)
- Dominik Glodzik
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA, Cambridge, UK
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Bosch
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Department of Oncology, Skåne University Hospital, SE-22184, Lund, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institute, SE-17177, Stockholm, Sweden
| | - Mattias Aine
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, SE-22184, Lund, Sweden
| | - Johan Vallon-Christersson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Christel Reuterswärd
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Anna Karlsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Shamik Mitra
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Emma Niméus
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Division of Surgery, Department of Clinical Sciences, Lund University, SE-22184, Lund, Sweden
| | - Karolina Holm
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Jari Häkkinen
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Cecilia Hegardt
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Christer Larsson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Martin Malmberg
- Department of Oncology, Skåne University Hospital, SE-22184, Lund, Sweden
| | - Lisa Rydén
- Division of Surgery, Department of Clinical Sciences, Lund University, SE-22184, Lund, Sweden
| | - Anna Ehinger
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Department of Genetics and Pathology, Laboratory Medicine, Region Skåne, SE-22184, Lund, Sweden
| | - Niklas Loman
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Department of Oncology, Skåne University Hospital, SE-22184, Lund, Sweden
| | - Anders Kvist
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Hans Ehrencrona
- Department of Genetics and Pathology, Laboratory Medicine, Region Skåne, SE-22184, Lund, Sweden
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, SE-22184, Lund, Sweden
| | - Serena Nik-Zainal
- Academic Department of Medical Genetics, The Clinical School University of Cambridge, Cambridge Biomedical Research Campus, CB2 0QQ, Cambridge, UK
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden.
| |
Collapse
|
13
|
Ryan D, Paul BT, Koziol J, ElShamy WM. The pro- and anti-tumor roles of mesenchymal stem cells toward BRCA1-IRIS-overexpressing TNBC cells. Breast Cancer Res 2019; 21:53. [PMID: 31014367 PMCID: PMC6480921 DOI: 10.1186/s13058-019-1131-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background To evaluate the cross-talk between BRCA1-IRIS (IRIS)-overexpressing (IRISOE) TNBC cells and tumor-resident mesenchymal stem cells (MSCs) that triggers the aggressiveness or elimination of IRISOE TNBC tumors. Methods We analyzed the effect of silencing or inactivating IRIS on the bi-directional interaction between IRISOE TNBC cells and MSCs on tumor formation and progression. We analyzed the downstream signaling in MSCs induced by IL-6 secreted from IRISOE TNBC cells. We compared the effect of MSCs on the formation and progression of IRIS-proficient and deficient-TNBC cells/tumors using in vitro and in vivo models. Finally, we analyzed the association between IL-6, PTGER2, and PTGER4 overexpression and breast cancer subtype; hormone receptor status; and distant metastasis-free or overall survival. Results We show high-level IL-6 secreted from IRISOE TNBC cells that enhances expression of its receptor (IL-6R) in MSCs, their proliferation, and migration toward IRISOE, in vitro, and recruitment into IRISOE TNBC tumors, in vivo. In serum-free medium, recombinant IL-6 and the IL-6-rich IRISOE TNBC cell condition media (CM) decreased STAT3Y705 phosphorylation (p-STAT3Y705) in MSCs. Inhibiting IRIS expression or activity prolonged STAT3Y705 phosphorylation in MSCs. The interaction with IRISOE TNBC cells skewed MSC differentiation toward prostaglandin E2 (PGE2)-secreting pro-aggressiveness cancer-associated fibroblasts (CAFs). Accordingly, co-injecting human or mouse MSCs with IRISOE TNBC tumor cells promoted the formation of aggressive mammary tumors, high circulating IL-6 and PGE2 levels, and reduced overall survival. In contrast, IRIS-silenced or inactivated cells showed reduced tumor formation ability, limited MSC recruitment into tumors, reduced circulating IL-6 and PGE2 levels, and prolonged overall survival. A positive correlation between IL-6, PTGER2, and PTGER4 expression and basal phenotype; ER-negativity; distant metastasis-free and overall survival in basal; or BRCAmutant carriers was observed. Finally, the bi-directional interaction with MSCs triggered death rather than growth of IRIS-silenced TNBC cells, in vitro and in vivo. Conclusions The IL-6/PGE2-positive feedback loop between IRISOE TNBC tumor cells and MSCs enhances tumor aggressiveness. Inhibiting IRIS expression limits TNBC tumor growth and progression through an MSC-induced death of IRIS-silenced/inactivated TNBC cells. Electronic supplementary material The online version of this article (10.1186/s13058-019-1131-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Ryan
- Breast Cancer Program, San Diego Biomedical Research Institute, 10865 Road to Cure, Suite 100, San Diego, CA, 92121, USA
| | - Bibbin T Paul
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Jim Koziol
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Wael M ElShamy
- Breast Cancer Program, San Diego Biomedical Research Institute, 10865 Road to Cure, Suite 100, San Diego, CA, 92121, USA.
| |
Collapse
|
14
|
Kraya AA, Maxwell KN, Wubbenhorst B, Wenz BM, Pluta J, Rech AJ, Dorfman LM, Lunceford N, Barrett A, Mitra N, Morrissette JJD, Feldman M, Nayak A, Domchek SM, Vonderheide RH, Nathanson KL. Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer. Clin Cancer Res 2019; 25:4363-4374. [PMID: 30914433 DOI: 10.1158/1078-0432.ccr-18-0468] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 12/08/2018] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Breast cancers with BRCA1/2 alterations have a relatively high mutational load, suggesting that immune checkpoint blockade may be a potential treatment option. However, the degree of immune cell infiltration varies widely, and molecular features contributing to this variability remain unknown. EXPERIMENTAL DESIGN We hypothesized that genomic signatures might predict immunogenicity in BRCA1/2 breast cancers. Using The Cancer Genome Atlas (TCGA) genomic data, we compared breast cancers with (89) and without (770) either germline or somatic BRCA1/2 alterations. We also studied 35 breast cancers with germline BRCA1/2 mutations from Penn using WES and IHC. RESULTS We found that homologous recombination deficiency (HRD) scores were negatively associated with expression-based immune indices [cytolytic index (P = 0.04), immune ESTIMATE (P = 0.002), type II IFN signaling (P = 0.002)] despite being associated with a higher mutational/neoantigen burden, in BRCA1/2 mutant breast cancers. Further, absence of allele-specific loss of heterozygosity (LOH negative; P = 0.01) or subclonality (P = 0.003) of germline and somatic BRCA1/2 mutations, respectively, predicted for heightened cytolytic activity. Gene set analysis found that multiple innate and adaptive immune pathways that converge on NF-κB may contribute to this heightened immunogenicity. IHC of Penn breast cancers demonstrated increased CD45+ (P = 0.039) and CD8+ infiltrates (P = 0.037) and increased PDL1 expression (P = 0.012) in HRD-low or LOH-negative cancers. Triple-negative cancers with low HRD had far greater CD8+ T cells (P = 0.0011) and Perforin 1 expression (P = 0.014) compared with hormone receptor-positive HRD-high cancers. CONCLUSIONS HRD scores and hormone receptor subtype are predictive of immunogenicity in BRCA1/2 breast cancers and may inform the design of optimal immune therapeutic strategies.
Collapse
Affiliation(s)
- Adam A Kraya
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bradley Wubbenhorst
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brandon M Wenz
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Pluta
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew J Rech
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liza M Dorfman
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicole Lunceford
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Amanda Barrett
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Feldman
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anupma Nayak
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Susan M Domchek
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania. .,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Li X, Dai D, Chen B, Tang H, Xie X, Wei W. The value of neutrophil-to-lymphocyte ratio for response and prognostic effect of neoadjuvant chemotherapy in solid tumors: A systematic review and meta-analysis. J Cancer 2018; 9:861-871. [PMID: 29581764 PMCID: PMC5868150 DOI: 10.7150/jca.23367] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/28/2018] [Indexed: 12/13/2022] Open
Abstract
Introduction: The neutrophil-to-lymphocyte ratio (NLR) has been found to be an indicator of poor prognosis in many tumour types. However, little is known about the relationship between the NLR and patients with tumours who receive neoadjuvant chemotherapy (NAC) in terms of response rate and prognostic ability. We thus performed this meta-analysis to further investigate this relationship. Methods: An electronic systematic literature search for articles published before September 2017 was performed to explore the association between the pretreatment NLR and outcome in patients treated with NAC. Data were extracted by the reported odds ratios (ORs) and hazard ratios (HRs) with their 95% confidence intervals (CIs) for the response rate and the survival outcome, respectively. The results were pooled using the random-effect or fixed-effect model. Results: Thirty-three studies were eventually included in our study, and all were published no earlier than 2011. An NLR that was higher than the cut-off was associated with a lower pathological complete response (pCR) rate in patients with cancer (OR = 1.72, 95% CI, 1.26-2.33). A lower NLR was associated with better overall survival (OS) (HR = 1.58, 95% CI, 1.34-1.86), cancer-specific survival (CSS) (HR = 2.22, 95% CI, 1.32-3.74), disease-free survival (DFS) (HR = 1.32, 95% CI, 1.10-1.59) and recurrence-free survival (RFS) (HR = 1.90, 95% CI, 1.50-2.40). Conclusion: Overall, an NLR lower than the cut-off value indicated a greater chance for pCR and may predict good survival outcomes after NAC for patients with solid tumours. The use of the NLR for risk stratification before NAC should be further demonstrated by future large-scale prospective studies.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 East Dong feng Road, Guangzhou, 510060, China
| | - Weidong Wei
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 East Dong feng Road, Guangzhou, 510060, China
| |
Collapse
|
16
|
Ryan D, Sinha A, Bogan D, Davies J, Koziol J, ElShamy WM. A niche that triggers aggressiveness within BRCA1-IRIS overexpressing triple negative tumors is supported by reciprocal interactions with the microenvironment. Oncotarget 2017; 8:103182-103206. [PMID: 29262555 PMCID: PMC5732721 DOI: 10.18632/oncotarget.20892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
Production of metastasis capable precursors begins within the primary tumor. Here, we define the bidirectional interactions with stromal cells involved in promoting these precursors within BRCA1-IRIS (hereafter IRIS) overexpressing (IRISOE) TNBC tumors. We define an aggressiveness niche, functionally defined as the necrotic/hypoxic core of the tumor, in which metabolically stressed, hypoxic, and inflamed IRISOE TNBC cells secrete higher levels of cytokines, chemokines and growth factors. One cytokine; IL-1β attracts mesenchymal stem cells (MSCs) to the niche and activates them to secrete CXCL1 that entrains IRISOE cells to secrete higher levels of CCL2 and VEGF. CCL2 attracts macrophages (TAMs) to the niche and activates them to secrete S100A8, and VEGF attracts endothelial cells (ECs) and activates them to secrete IL-8. In concert, CXCL1, S100A8 and IL-8 entrain aggressiveness in IRISOE TNBC cells within the niche. Indeed, compared to IRISOE cells alone, tumors developed by co-injecting IRISOE cells admixed with MSCs (10:1) in athymic mice were bigger and more aggressive. They contained more TAMs and ECs, expressed higher-levels of basal, epithelial to mesenchymal transition, and stemness biomarkers, quickly progressed to lymph-node or visceral metastases, and were highly sensitive to the IL-1β inhibitor “Anakinra”. Our findings supported by human data show that breast cancer patients with high-levels of IL-1β, CXCL1, CCL2, S100A8, VEGF, and IL-8 would show worse clinical outcomes. Our findings argue that this cytokine set is a diagnostic biomarker for patients who may benefit from an IRIS inhibitor-based therapy, and is a blue print for translation of approaches to combining that therapy with inhibitors of these bidirectional interactions to overcome TNBC metastasis.
Collapse
Affiliation(s)
- Daniel Ryan
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| | | | - Danielle Bogan
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Joanna Davies
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Jim Koziol
- Department of Molecular and Experimental Medicine, Scripps Research Institute, San Diego, CA, USA
| | - Wael M ElShamy
- Breast Cancer Program, San Diego Biomedical Research Institute, San Diego, CA, USA
| |
Collapse
|
17
|
Aksoy A, Odabas H, Kaya S, Bozkurt O, Degirmenci M, Topcu TO, Aytekin A, Arpaci E, Avci N, Pilanci KN, Cinkir HY, Bozkaya Y, Cirak Y, Gumus M. Hormone receptor status and survival of medullary breast cancer patients. A Turkish cohort. Saudi Med J 2017; 38:156-162. [PMID: 28133688 PMCID: PMC5329627 DOI: 10.15537/smj.2017.2.18055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objectives: To analyze the relationship between clinical features, hormonal receptor status, and survival in patients who were diagnosed with medullary breast cancer (MBC). Methods: Demographic characteristics, histopathological features, and survival statuses of 201 patients diagnosed with MBC between 1995 and 2015 were retrospectively recorded. Survival analyses were conducted with uni- and multivariate cox regression analysis. Results: Median follow-up time was 54 (4-272) months. Median patient age at the time of diagnosis was 47 years old (26-90). Of the patients, 91.5% were triple negative. Five-year recurrence free survival time (RFS) rate was 87.4% and overalll survival (OS) rate 95.7%. For RFS, progesterone receptor (PR) negativity, atypical histopathological evaluation, absence of lymphovascular invasion, smaller tumor, lower nodal involvement were found to be favourable prognostic factors by univariate analysis (p<0.05). The PR negativity and smaller tumor were found to be favourable factors by univariate analysis (p<0.05). However, none of these factors were determined as significant independent prognostic factors for OS (p>0.05). Conclusion: Turkish MBC patients exhibited good prognosis, which was comparable with survival outcomes achieved in the literature. The PR negativity was related to a better RFS and OS rates.
Collapse
Affiliation(s)
- Asude Aksoy
- Department of Medical Oncology, Medical Faculty, Firat University Elazig, Turkey. E-mail.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mei Z, Shi L, Wang B, Yang J, Xiao Z, Du P, Wang Q, Yang W. Prognostic role of pretreatment blood neutrophil-to-lymphocyte ratio in advanced cancer survivors: A systematic review and meta-analysis of 66 cohort studies. Cancer Treat Rev 2017; 58:1-13. [PMID: 28602879 DOI: 10.1016/j.ctrv.2017.05.005] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
|
19
|
Nicholas DA, Andrieu G, Strissel KJ, Nikolajczyk BS, Denis GV. BET bromodomain proteins and epigenetic regulation of inflammation: implications for type 2 diabetes and breast cancer. Cell Mol Life Sci 2017; 74:231-243. [PMID: 27491296 PMCID: PMC5222701 DOI: 10.1007/s00018-016-2320-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/16/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022]
Abstract
Chronic inflammation drives pathologies associated with type 2 diabetes (T2D) and breast cancer. Obesity-driven inflammation may explain increased risk and mortality of breast cancer with T2D reported in the epidemiology literature. Therapeutic approaches to target inflammation in both T2D and cancer have so far fallen short of the expected improvements in disease pathogenesis or outcomes. The targeting of epigenetic regulators of cytokine transcription and cytokine signaling offers one promising, untapped approach to treating diseases driven by inflammation. Recent work has deeply implicated the Bromodomain and Extra-Terminal domain (BET) proteins, which are acetylated histone "readers", in epigenetic regulation of inflammation. This review focuses on inflammation associated with T2D and breast cancer, and the possibility of targeting BET proteins as an approach to regulating inflammation in the clinic. Understanding inflammation in the context of BET protein regulation may provide a basis for designing promising therapeutics for T2D and breast cancer.
Collapse
Affiliation(s)
- Dequina A Nicholas
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Guillaume Andrieu
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Katherine J Strissel
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Barbara S Nikolajczyk
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Gerald V Denis
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA.
- Section of Hematology/Oncology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, K520, Boston, MA, 02118, USA.
| |
Collapse
|
20
|
Smid M, Rodríguez-González FG, Sieuwerts AM, Salgado R, Prager-Van der Smissen WJC, Vlugt-Daane MVD, van Galen A, Nik-Zainal S, Staaf J, Brinkman AB, van de Vijver MJ, Richardson AL, Fatima A, Berentsen K, Butler A, Martin S, Davies HR, Debets R, Gelder MEMV, van Deurzen CHM, MacGrogan G, Van den Eynden GGGM, Purdie C, Thompson AM, Caldas C, Span PN, Simpson PT, Lakhani SR, Van Laere S, Desmedt C, Ringnér M, Tommasi S, Eyford J, Broeks A, Vincent-Salomon A, Futreal PA, Knappskog S, King T, Thomas G, Viari A, Langerød A, Børresen-Dale AL, Birney E, Stunnenberg HG, Stratton M, Foekens JA, Martens JWM. Breast cancer genome and transcriptome integration implicates specific mutational signatures with immune cell infiltration. Nat Commun 2016; 7:12910. [PMID: 27666519 PMCID: PMC5052682 DOI: 10.1038/ncomms12910] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022] Open
Abstract
A recent comprehensive whole genome analysis of a large breast cancer cohort was used to link known and novel drivers and substitution signatures to the transcriptome of 266 cases. Here, we validate that subtype-specific aberrations show concordant expression changes for, for example, TP53, PIK3CA, PTEN, CCND1 and CDH1. We find that CCND3 expression levels do not correlate with amplification, while increased GATA3 expression in mutant GATA3 cancers suggests GATA3 is an oncogene. In luminal cases the total number of substitutions, irrespective of type, associates with cell cycle gene expression and adverse outcome, whereas the number of mutations of signatures 3 and 13 associates with immune-response specific gene expression, increased numbers of tumour-infiltrating lymphocytes and better outcome. Thus, while earlier reports imply that the sheer number of somatic aberrations could trigger an immune-response, our data suggests that substitutions of a particular type are more effective in doing so than others.
Collapse
Affiliation(s)
- Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - F. Germán Rodríguez-González
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Anieta M. Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, Bd de Waterloo 121, B-1000 Brussels, Belgium
- Department of Pathology/TCRU GZA, 2610 Antwerp, Belgium
| | - Wendy J. C. Prager-Van der Smissen
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Michelle van der Vlugt-Daane
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Anne van Galen
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Serena Nik-Zainal
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 9NB, UK
| | - Johan Staaf
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, SE-223 81 Lund, Sweden
| | - Arie B. Brinkman
- Faculty of Science, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525GA, Nijmegen, The Netherlands
| | - Marc J. van de Vijver
- Department of Pathology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Andrea L. Richardson
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Aquila Fatima
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Kim Berentsen
- Faculty of Science, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525GA, Nijmegen, The Netherlands
| | - Adam Butler
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK
| | - Sancha Martin
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK
| | - Helen R. Davies
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Marion E. Meijer-Van Gelder
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Carolien H. M. van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - Gaëtan MacGrogan
- Département de Biopathologie,Institut Bergonié, CS 61283 33076 Bordeaux, France
| | - Gert G. G. M. Van den Eynden
- Department of Pathology/TCRU GZA, 2610 Antwerp, Belgium
- Molecular Immunology Unit, Jules Bordet Institute, B-1000 Brussels, Belgium
| | - Colin Purdie
- Department of Pathology, Ninewells Hospital & Medical School, Dundee DD1 9SY, UK
| | - Alastair M. Thompson
- Department of Pathology, Ninewells Hospital & Medical School, Dundee DD1 9SY, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Paul N. Span
- Department of Radiation Oncology, Radboud University Medical Center, 6525GA, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, 6525GA, Nijmegen, The Netherlands
| | - Peter T. Simpson
- The University of Queensland: UQ Centre for Clinical Research and School of Medicine, Brisbane 4029, Australia
| | - Sunil R. Lakhani
- The University of Queensland: UQ Centre for Clinical Research and School of Medicine, Brisbane 4029, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Brisbane 4029, Australia
| | - Steven Van Laere
- Center for Oncological Research, University of Antwerp & GZA Hospitals Sint-Augustinus, 2610 Wilrijk, Belgium
| | - Christine Desmedt
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, Bd de Waterloo 121, B-1000 Brussels, Belgium
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, SE-223 81 Lund, Sweden
| | | | - Jorunn Eyford
- Cancer Research Laboratory, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Annegien Broeks
- The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Anne Vincent-Salomon
- Department of Pathology and INSERM U934, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - P. Andrew Futreal
- Department of Genomic Medicine, UT MD Anderson Cancer Center, Houston, TX, 77230, USA
| | - Stian Knappskog
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Department of Oncology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Tari King
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, New York 10065, USA
| | - Gilles Thomas
- Synergie Lyon Cancer,Centre Léon Bérard, 28 rue Laënnec, Cedex 08 Lyon, France
| | - Alain Viari
- Synergie Lyon Cancer,Centre Léon Bérard, 28 rue Laënnec, Cedex 08 Lyon, France
- Equipe Erable, INRIA Grenoble-Rhône-Alpes, 655, Av. de l'Europe, 38330 Montbonnot-Saint Martin, France
| | - Anita Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radiumhospital, 0310, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, 0310 Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital The Norwegian Radiumhospital, 0310, Oslo, Norway
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, 0310 Oslo, Norway
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus,Hinxton CB10 1SD, Cambridgeshire, UK
| | - Hendrik G. Stunnenberg
- Faculty of Science, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525GA, Nijmegen, The Netherlands
| | - Mike Stratton
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridge, UK
| | - John A. Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands
| |
Collapse
|
21
|
Fan Y, Wang Y, Wang K. Prostaglandin E2 stimulates normal bronchial epithelial cell growth through induction of c-Jun and PDK1, a kinase implicated in oncogenesis. Respir Res 2015; 16:149. [PMID: 26684827 PMCID: PMC4699375 DOI: 10.1186/s12931-015-0309-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 12/04/2015] [Indexed: 02/05/2023] Open
Abstract
Background Cyclooxygenase-2-derived prostaglandin E2 (PGE2), a bioactive eicosanoid, has been implicated in many biological processes including reproduction, inflammation and tumor growth. We previously showed that PGE2 stimulated lung cancer cell growth and progression through PGE2 receptor EP2/EP4-mediated kinase signaling pathways. However, the role of PGE2 in controlling lung airway epithelial cell phenotype remains unknown. We evaluated the effects of c-Jun and 3-phosphoinositede dependent protein kinase-1 (PDK1) in mediating epithelial cell hyperplasia induced by PGE2. Method The bronchial epithelial cell lines BEAS-2B and HBEc14-KT were cultured and then treated with PGE2. PDK1 small interfering RNA (siRNA) and a PDK1 inhibitor, an antagonist of the PGE2 receptor subtype EP4 and EP4 siRNA, c-Jun siRNA, and overexpressions of c-Jun and PDK1 have been used to evaluate the effects on cell proliferation. Results We demonstrated that PGE2 increased normal bronchial epithelial cell proliferation through induction of PDK1, an ankyrin repeat-containing Ser/Thr kinase implicated in the induction of apoptosis and the suppression of tumor growth. PDK1 siRNA and a PDK1 inhibitor blocked the effects of PGE2 on normal cell growth. The PGE2-induced PDK1 expression was blocked by an antagonist of the PGE2 receptor subtype EP4 and by EP4 siRNA. In addition, we showed that induction of PDK1 by PGE2 was associated with induction of the transcription factor, c-Jun protein. Silencing of c-Jun using siRNA and point mutations of c-Jun sites in the PDK1 gene promoter resulted in blockade of PDK1 expression and promoter activity induced by PGE2. In contrast, overexpression of c-Jun induced PDK1 gene promoter activity and expression followed increased cell proliferation. Conclusion PGE2 increases normal bronchial epithelial cell proliferation through increased PDK1 gene expression that is dependent on EP4 and induction of c-Jun. Therewith, our data suggest a new role of c-Jun and PDK1 in mediating epithelial cell hyperplasia induced by PGE2.
Collapse
Affiliation(s)
- Yu Fan
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China. .,Department of Radiotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan Province, 610041, China.
| | - Ye Wang
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Ke Wang
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
22
|
Hugo HJ, Saunders C, Ramsay RG, Thompson EW. New Insights on COX-2 in Chronic Inflammation Driving Breast Cancer Growth and Metastasis. J Mammary Gland Biol Neoplasia 2015; 20:109-19. [PMID: 26193871 DOI: 10.1007/s10911-015-9333-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/29/2015] [Indexed: 12/22/2022] Open
Abstract
The medicinal use of aspirin stretches back to ancient times, before it was manufactured in its pure form in the late 19th century. Its accepted mechanistic target, cyclooxygenase (COX), was discovered in the 1970s and since this landmark discovery, the therapeutic application of aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) has increased dramatically. The most significant benefits of NSAIDs are in conditions involving chronic inflammation (CI). Given the recognized role of CI in cancer development, the use of long-term NSAID treatment in the prevention of cancer is an enticing possibility. COX-2 is a key driver of CI, and here we review COX-2 expression as a predictor of survival in various cancer types, including breast. Obesity and post-partum involution are natural inflammatory states that are associated with increased breast cancer risk. We outline the COX-2 mediated mechanisms contributing to the growth of cancers. We dissect the cellular mechanism of epithelial-mesenchymal transition (EMT) and how COX-2 may induce this to facilitate tumor progression. Finally we examine the potential regulation of COX-2 by c-Myb, and the possible interplay between c-Myb/COX-2 in proliferation, and hypoxia inducible factor-1 alpha (HIF1α)/COX-2 in invasive pathways in breast cancer.
Collapse
Affiliation(s)
- Honor J Hugo
- VBCRC Invasion and Metastasis Unit, St Vincent's Institute, Fitzroy, VIC, Australia.
| | - C Saunders
- School of Surgery, University of Western Australia, Perth, Western Australia, Australia
| | - R G Ramsay
- Differentation and Transcription Laboratory, Peter MacCallum Cancer Centre and the Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - E W Thompson
- VBCRC Invasion and Metastasis Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland Institute of Technology, Brisbane, QLD, Australia
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|