2651
|
Lin H, Wu Y, Liang G, Chen L. Establishing a predicted model to evaluate prognosis for initially diagnosed metastatic Her2-positive breast cancer patients and exploring the benefit from local surgery. PLoS One 2020; 15:e0242155. [PMID: 33170907 PMCID: PMC7654787 DOI: 10.1371/journal.pone.0242155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/27/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND For patients initially diagnosed with metastatic Her2-positive breast cancer (MHBC), we intended to construct a nomogram with risk stratification to predict prognosis and to explore the role of local surgery. METHODS We retrieved data from the Surveillance, Epidemiology, and End Results (SEER) database. Kaplan-Meier (KM) method and log-rank test were used for the selection of significant variables. Cox regression analysis and Fine-Gray test were utilized to confirm independent prognostic factors of overall survival (OS) and breast cancer-specific survival (BCSS). A nomogram predicting 1-year, 3-year, and 5-year OS was developed and validated. Patients were stratified based on the optimal cut-off values of total personal score. KM method and log-rank test were used to estimate OS prognosis and benefit from local surgery and chemotherapy. RESULTS There were 1680 and 717 patients in the training and validation cohort. Age, race, marriage, T stage, estrogen receptor (ER) status, visceral metastasis (bone, brain, liver and lung) were identified as independent prognostic factors for OS and BCSS, while histology was also corelated with OS. C-indexes in the training and validation cohort were 0.70 and 0.68, respectively. Calibration plots indicated precise predictive ability. The total population was divided into low- (<141 points), intermediate- (142-208 points), and high-risk (>208 points) prognostic groups. Local surgery and chemotherapy brought various degrees of survival benefit for patients with diverse-risk prognosis. CONCLUSIONS We constructed a model with accurate prediction and discrimination. It would provide a reference for clinicians' decision-making. Surgery on the primary lesion was recommended for patients with good physical performance status, while further study on optimal surgical opportunity was needed.
Collapse
Affiliation(s)
- Hong Lin
- Department of Oncology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Yanxuan Wu
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Guoxi Liang
- Department of Oncology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Liming Chen
- Department of Oncology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
2652
|
Du C, Wang Y, Zhang Y, Zhang J, Zhang L, Li J. LncRNA DLX6-AS1 Contributes to Epithelial-Mesenchymal Transition and Cisplatin Resistance in Triple-negative Breast Cancer via Modulating Mir-199b-5p/Paxillin Axis. Cell Transplant 2020; 29:963689720929983. [PMID: 32686982 PMCID: PMC7563824 DOI: 10.1177/0963689720929983] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive cancer types with high recurrence, metastasis, and drug resistance. Recent studies report that long noncoding RNAs (lncRNAs)-mediated competing endogenous RNAs (ceRNA) play an important role in tumorigenesis and drug resistance of TNBC. Although elevated lncRNA DLX6 antisense RNA 1 (DLX6-AS1) has been observed to promote carcinogenesis in various cancers, the role in TNBC remained unclear. In this study, expression levels of DLX6-AS1 were increased in TNBC tissues and cell lines when compared with normal tissues or breast fibroblast cells which were determined by quantitative real-time PCR (RT-qPCR). Then, CCK-8 assay, cell colony formation assay and western blot were performed in CAL-51 cells transfected with siRNAs of DLX6-AS1 or MDA-MB-231 cells transfected with DLX6-AS1 over expression plasmids. Knock down of DLX6-AS1 inhibited cell proliferation, epithelial-mesenchymal transition (EMT), decreased expression levels of BCL2 apoptosis regulator (Bcl-2), Snail family transcriptional repressor 1 (Snail) as well as N-cadherin and decreased expression levels of cleaved caspase-3, γ-catenin as well as E-cadherin, while up regulation of DLX6-AS1 had the opposite effect. Besides, knockdown of DLX6-AS1 in CAL-51 cells or up regulation of DLX6-AS1 in MDA-MB-231 cells also decreased or increased cisplatin resistance of those cells analyzed by MTT assay. Moreover, by using dual luciferase reporter assay, RNA immunoprecipitation and RNA pull down assay, a ceRNA which was consisted by lncRNA DLX6-AS1, microRNA-199b-5p (miR-199b-5p) and paxillin (PXN) was identified. And DLX6-AS1 function through miR-199b-5p/PXN in TNBC cells. Finally, results of xenograft experiments using nude mice showed that DLX6-AS1 regulated cell proliferation, EMT and cisplatin resistance by miR-199b-5p/PXN axis in vivo. In brief, DLX6-AS1 promoted cell proliferation, EMT, and cisplatin resistance through miR-199b-5p/PXN signaling in TNBC in vitro and in vivo.
Collapse
Affiliation(s)
- Chuang Du
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Yan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Yingying Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Jianhua Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Linfeng Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Jingruo Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, China
| |
Collapse
|
2653
|
Pattarawat P, Wallace S, Pfisterer B, Odoi A, Wang HCR. Formulation of a triple combination gemcitabine plus romidepsin + cisplatin regimen to efficaciously and safely control triple-negative breast cancer tumor development. Cancer Chemother Pharmacol 2019; 85:141-152. [PMID: 31865420 DOI: 10.1007/s00280-019-04013-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive, lethal, and heterogeneous subtype of breast cancers, tending to have lower 5-year survival rates than other BC subtypes in response to conventional chemotherapies. This study's aim was to identify advanced regimens to effectively control TNBC tumor development. METHODS We investigated the combination of the DNA synthesis inhibitor gemcitabine, the DNA-damaging agent cisplatin, and the histone deacetylase inhibitor romidepsin to control a variety of breast cells in vitro. We studied the toxicity of drug doses and administration schedules to determine tolerable combination regimens in immune-deficient nude and -competent BALB/c mice. We then studied the efficacy of tolerable regimens in controlling TNBC cell-derived xenograft development in nude mice. By reducing clinically equivalent doses of each agent in combination, we formulated tolerable regimens in animals. We verified that the tolerable triple combination gemcitabine plus romidepsin + cisplatin regimen more efficacious than double combination regimens in controlling xenograft tumor development in nude mice. RESULTS A triple combination of gemcitabine + romidepsin + cisplatin synergistically induced death of the TNBC M.D. Anderson-Metastatic Breast cancer (MDA-MB) 231 and MDA-MB468, as well as Michigan Cancer Foundation (MCF) 7, MCF10A, and MCF10A-Ras cells. Cell death induced by gemcitabine + romidepsin + cisplatin was in a reactive oxygen species-dependent manner. CONCLUSION Considering the high costs for developing a new anticancer agent, we used the FDA-approved drugs gemcitabine, romidepsin (is approved for T-cell lymphoma and is under clinical trial for TNBC), and cisplatin to economically formulate an efficacious and safe combination regimen. The highly efficacious gemcitabine plus romidepsin + cisplatin regimen should be poised for efficient translation into clinical trials, ultimately contributing to reduced mortality and improved quality of life for TNBC patients.
Collapse
Affiliation(s)
- Pawat Pattarawat
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA.,UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Shelby Wallace
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Bianca Pfisterer
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Agricola Odoi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
2654
|
Xiao W, Zheng S, Zou Y, Yang A, Xie X, Tang H, Xie X. CircAHNAK1 inhibits proliferation and metastasis of triple-negative breast cancer by modulating miR-421 and RASA1. Aging (Albany NY) 2019; 11:12043-12056. [PMID: 31857500 PMCID: PMC6949091 DOI: 10.18632/aging.102539] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/19/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND There is increasing evidence that circular RNAs (circRNAs) participate in regulating cancer progression. However, the function and potential molecular mechanisms of circRNA in triple negative breast cancer (TNBC) are currently largely unclear. RESULTS We found that circAHNAK1 was significantly down-regulated in TNBC, and its expression was negatively associated with RFS and OS. Overexpression of circAHNAK1 can inhibit TNBC proliferation, migration and invasion in vitro. In vivo studies confirmed that circAHNAK1 inhibited TNBC tumor growth and metastasis. Mechanistic analysis indicated that circAHNAK1 acted as a miR-421 ceRNA (competitive endogenous RNA) to attenuate the inhibitory effect of miR-421 on its target gene RASA1. CONCLUSIONS In conclusion, CircAHNAK1 inhibits proliferation and metastasis of TNBC by modulating miR-421 and RASA1. METHODS CircRNA microarrays were used to screen for differential circRNA expression profiles. qRT-PCR was used to detect the expression levels of circRNAs. The effect of circAHNAK1 on recurrence -free survival (RFS) and overall survival (OS) in patients with TNBC was subsequently analyzed. The role of circAHNKA1 in the progression of TNBC was further evaluated by multiple in vivo and in vitro assays. Finally, we focused on the regulation of circAHNAK1 on miR-421 and its targeted gene RASA1 in TNBC.
Collapse
Affiliation(s)
- Weikai Xiao
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People’s Republic of China
| | - Shaoquan Zheng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People’s Republic of China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People’s Republic of China
| | - Anli Yang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People’s Republic of China
| | - Xinhua 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, Guangzhou 510060, People’s Republic of China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People’s Republic of China
| | - 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, Guangzhou 510060, People’s Republic of China
| |
Collapse
|
2655
|
Fan Y, Yu D, Li D, Wang X. Prevention of Local Tumor Recurrence After Surgery by Thermosensitive Gel-Based Chemophotothermal Therapy in Mice. Lasers Surg Med 2019; 52:682-691. [PMID: 31854013 DOI: 10.1002/lsm.23206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Local recurrence of cancer after surgery has long been a tough problem. In the present study, thermosensitive gel-based chemophotothermal therapy was applied to prevent the recurrence of liver cancer after surgery. STUDY DESIGN/MATERIALS AND METHODS Mesoporous silica nanoparticles (MSNs) were used as first-level carrier to co-load doxorubicin (DOX) and ICG. Then, the drug-loaded MSNs (D-I@MSN) were incorporated into poloxamer gel. A mimic model of liver cancer recurrence after surgery was prepared by subcutaneously injecting H22 cells into the armpit of mice. Then the two-level composite gel (D-I@MSN/gel) was also subcutaneously injected at the same site before the formation of tumor, followed by 808 nm laser irradiation. RESULTS The loading efficiency and entrapment efficiency of DOX were as high as 8.85% and 96.9%, and that of ICG were 9.24% and 99.3%, respectively. The results of in vitro cytotoxicity showed that cell viability in D-I@MSN+Laser group was only 5.8% after being irradiated by 808 nm laser for 5 minutes (0.5 W/cm2 ). In animal studies, tumor formation (tumor recurrence) was greatly inhibited in D-I@MSN+Laser group. CONCLUSIONS The thermosensitive gel-based chemophotothermal therapy showed excellent safety and efficacy when applied in the prevention of mimic local tumor replase after surgery in mice, presenting its great potential clinically. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yanyan Fan
- Department of Gynecology, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Dujuan Yu
- Department of Respiratory, China-Japan Union Hospital, Jilin University, Changchun, 130021, P. R. China
| | - Duan Li
- The First People's Hospital of Tianmen, Tianmen, Hubei, 431700, P. R. China
| | - Xue Wang
- Physical Examination Center, China-Japan Union Hospital, Jilin University, Changchun, 130031, P. R. China
| |
Collapse
|
2656
|
First-line fulvestrant plus anastrozole for hormone-receptor-positive metastatic breast cancer in postmenopausal women: a cost-effectiveness analysis. Breast Cancer 2019; 27:399-404. [PMID: 31853795 DOI: 10.1007/s12282-019-01034-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/04/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE In a recent randomized, open-label trial (S0226), the addition of fulvestrant to anastrozole therapy decreased the risk of progression and death in patients with hormone-receptor-positive metastatic breast cancer. However, the cost-effectiveness of incorporating fulvestrant into the first-line setting is unknown. METHODS We developed a Markov model to assess the costs and clinical outcomes of fulvestrant plus anastrozole compared with anastrozole as a first-line therapy in a cohort of patients with advanced hormone-receptor-positive breast cancer. The transition probabilities were estimated from the fitted survival curves in the S0226 trial. Health care costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios (ICERs) were calculated for fulvestrant plus anastrozole compared with anastrozole from US payer's perspective. RESULTS Fulvestrant plus anastrozole led to an improvement of 0.11 QALYs compared with treatment with anastrozole alone. However, incorporating fulvestrant into the first-line therapy produced significantly higher health care costs ($72,496 vs. $38,959 for all eligible patients, and $73,728 vs. $37,239 for patients with no previous hormonal adjuvant therapy), resulting in ICERs of $300,564 and $194,450/QALY, respectively. Two-way sensitivity analysis showed that when the cost of fulvestrant decreased to $1.5/mg for all eligible patients or $3.5/mg for patients with no previous hormonal adjuvant therapy, at the perfect health in progression-free status, the ICER became $141,320 and $145,543 per QALY. CONCLUSION Substituting fulvestrant as a first-line therapy for hormone-receptor-positive metastatic breast cancer is not cost-effective compared with anastrozole based on the willing-to-pay threshold of $150,000 per QALY.
Collapse
|
2657
|
Zhang J, Li G, Feng L, Lu H, Wang X. Krüppel-like factors in breast cancer: Function, regulation and clinical relevance. Biomed Pharmacother 2019; 123:109778. [PMID: 31855735 DOI: 10.1016/j.biopha.2019.109778] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Breast cancer has accounted for the leading cause of cancer-related mortality among women worldwide. Although the progress in its diagnosis and treatment has come at a remarkable pace during the past several decades, there are still a wide array of problems regarding its progression, metastasis and treatment resistance that have not yet been fully clarified. Recently, an increasing number of studies have revealed that some members of Krüppel-like factors(KLFs) are significantly associated with cell proliferation, apoptosis, metastasis, cancer stem cell regulation and prognostic and predictive value for patients in breast cancer, indicating their promising prognostic and predictive potential for breast cancer survival and outcome. In this review, we will summarize our current knowledge of the functions, regulations and clinical relevance of KLFs in breast cancer.
Collapse
Affiliation(s)
- Jianping Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Guangliang Li
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| |
Collapse
|
2658
|
Duan X, Guo G, Pei X, Wang X, Li L, Xiong Y, Qiu X. Baicalin Inhibits Cell Viability, Migration and Invasion in Breast Cancer by Regulating miR-338-3p and MORC4. Onco Targets Ther 2019; 12:11183-11193. [PMID: 31908485 PMCID: PMC6930519 DOI: 10.2147/ott.s217101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
Background Baicalin is a natural compound from the roots of Scutellaria lateriflora Georgi, which plays anti-cancer role in multiple cancers. However, the exact role and potential underlying mechanism of baicalin in breast cancer (BC) remain poorly understood. Methods Thirty BC patients were recruited in this study. MCF-10A, MCF-7 and MDA-MB-231 cells were used to investigate the anti-cancer role of baicalin in vitro. Cell viability, migration, invasion and apoptosis were measured by MTT, trans-well and flow cytometry, respectively. The expression levels of microRNA-338-3p (miR-338-3p) and microrchidia family CW-type zinc-finger 4 (MORC4) were measured by quantitative real-time polymerase chain reaction or Western blot. The interaction between miR-338-3p and MORC4 was explored by luciferase reporter assay and RNA immunoprecipitation. Results We found that Baicalin treatment inhibited cell viability, migration and invasion but promoted apoptosis of BC cells. The expression of miR-338-3p was decreased in BC tissues and cells and miR-338-3p overexpression suppressed cell viability, migration and invasion but induced apoptosis. MiR-338-3p expression was reversed by baicalin exposure and inhibition of miR-338-3p attenuated the role of baicalin in viability, apoptosis, migration and invasion. MORC4 mRNA level was increased in BC tissues and cells, which was decreased by baicalin exposure. MORC4 was a target of miR-338-3p and its overexpression alleviated the effect of miR-338-3p on cell viability, apoptosis, migration and invasion. Conclusion In conclusion, baicalin suppressed cell viability, migration and invasion but promoted apoptosis in BC cells by regulating miR-338-3p and MORC4, indicating the promising pharmacological value of baicalin in BC treatment.
Collapse
Affiliation(s)
- Xin Duan
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Guangcheng Guo
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xinhong Pei
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xinxing Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Lin Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Youyi Xiong
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xinguang Qiu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
2659
|
Lambertini M, Poggio F, Bruzzone M, Conte B, Bighin C, de Azambuja E, Giuliano M, De Laurentiis M, Cognetti F, Fabi A, Bisagni G, Durando A, Turletti A, Urracci Y, Garrone O, Puglisi F, Montemurro F, Ceppi M, Del Mastro L. Dose-dense adjuvant chemotherapy in HER2-positive early breast cancer patients before and after the introduction of trastuzumab: Exploratory analysis of the GIM2 trial. Int J Cancer 2019; 147:160-169. [PMID: 31724170 DOI: 10.1002/ijc.32789] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022]
Abstract
Dose-dense adjuvant chemotherapy is standard of care in high-risk early breast cancer patients. However, its role in HER2-positive patients is still uncertain. In this exploratory analysis of the GIM2 trial, we investigated the efficacy of dose-dense chemotherapy in HER2-positive breast cancer patients with or without exposure to trastuzumab. In the GIM2 trial, node-positive early breast cancer patients were randomized to receive four cycles of (fluorouracil)epirubicin/cyclophosphamide followed by four cycles of paclitaxel administered every 2 (dose-dense) or 3 (standard-interval) weeks. After approval of adjuvant trastuzumab, protocol was amended in April 2006 to allow use of trastuzumab for 1 year after chemotherapy completion in HER2-positive patients. The efficacy of dose-dense chemotherapy in terms of disease-free survival (DFS) and overall survival (OS) was assessed according to HER2 status and trastuzumab use. Out of 2,003 breast cancer patients, HER2 status was negative/unknown in 1,551 patients; among the 452 patients with HER2-positive breast cancer, chemotherapy alone or followed by trastuzumab was given to 320 and 132 patients, respectively. Median follow-up was 8.1 years. No significant interaction between HER2 status, trastuzumab use and chemotherapy treatment was observed for both DFS (p = 0.698) and OS (p = 0.708). Nevertheless, there was no apparent benefit in the HER2-positive group treated with trastuzumab (DFS: HR, 0.99; 95% CI 0.52-1.89; OS: HR, 0.95; 95% CI 0.37-2.41). Although dose-dense chemotherapy was associated with a significant survival improvement in high-risk breast cancer patients, its benefit appeared to be smaller (if any) in patients with HER2-positive disease who received adjuvant trastuzumab.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
| | - Francesca Poggio
- Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marco Bruzzone
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Benedetta Conte
- Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Bighin
- Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Evandro de Azambuja
- Department of Medical Oncology, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Francesco Cognetti
- La Sapienza University, Rome, Italy.,Department of Medical Oncology 1, Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Alessandra Fabi
- La Sapienza University, Rome, Italy.,Department of Medical Oncology 1, Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Giancarlo Bisagni
- Department of Medical Oncology, Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonio Durando
- Breast Unit, Azienda Ospedaliera Universitaria Città della Salute e delle Scienze, Torino, Italy
| | - Anna Turletti
- Medical Oncology, Martini Hospital, ASL Città' di Torino, Italy
| | - Ylenia Urracci
- Department of Medical Oncology, Hospital Businco, Cagliari, Italy
| | - Ornella Garrone
- Breast Unit, Medical Oncology, Azienda Ospedaliera S. Croce e Carle, Ospedale di Insegnamento, Cuneo, Italy
| | - Fabio Puglisi
- Unit of Medical Oncology and Cancer Prevention, IRCCS Centro di Riferimento Oncologico Aviano-National Cancer Institute, Aviano (Udine), Italy.,Medical Oncology, Department of Medicine, University of Udine, Udine, Italy
| | - Filippo Montemurro
- Day Hospital Oncologico Multidisciplinare, Istituto di Candiolo, FPO-IRCCS, Candiolo, Torino, Italy
| | - Marcello Ceppi
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy.,Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
2660
|
Lam CSP, Arnott C, Beale AL, Chandramouli C, Hilfiker-Kleiner D, Kaye DM, Ky B, Santema BT, Sliwa K, Voors AA. Sex differences in heart failure. Eur Heart J 2019; 40:3859-3868c. [DOI: 10.1093/eurheartj/ehz835] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/08/2019] [Accepted: 11/28/2019] [Indexed: 12/28/2022] Open
Abstract
Abstract
The overall lifetime risk of heart failure (HF) is similar between men and women, however, there are marked sex differences in the landscape of this condition that are both important and under-recognized. Men are predisposed to HF with reduced ejection fraction (HFrEF), whereas women predominate in HF with preserved ejection fraction (HFpEF). Sex differences are also notable in the penetrance of genetic cardiomyopathies, risk factors, e.g. breast cancer which may be associated with cancer treatment-induced cardiomyopathy, as well as sex-specific conditions such as peripartum cardiomyopathy (PPCM). This review outlines the key sex differences with respect to clinical characteristics, pathophysiology, and therapeutic responses to HF treatments. Finally, we address important differences in the prognosis of HF. A central hypothesis is that the higher risk of HFrEF in men compared to women may be attributable to their predisposition to macrovascular coronary artery disease and myocardial infarction, whereas coronary microvascular dysfunction/endothelial inflammation has been postulated to play a key role in HFpEF and maybe the common link among HF syndromes that women are predisposed to Takotsubo cardiomyopathy, PPCM, and breast cancer radiotherapy-induced cardiomyopathy. Under-pinning current sex disparities in HF, there is a paucity of women recruited to HF clinical trials (20–25% of cohorts) and thus treatment guidelines are predominantly based on male-derived data. Large gaps in knowledge exist in sex-specific mechanisms, optimal drug doses for women and sex-specific criteria for device therapy.
Collapse
Affiliation(s)
- Carolyn S P Lam
- National Heart Centre Singapore, 5 Hospital Drive, Singapore, Singapore
- Duke-National University of Singapore, 8 College Rd, Singapore, Singapore
- University Medical Centre Groningen, Hanzeplein 1, GZ Groningen, The Netherlands
- The George Institute, Level 5/1 King St, Newtown NSW, Sydney, Australia
| | - Clare Arnott
- The George Institute, Level 5/1 King St, Newtown NSW, Sydney, Australia
| | - Anna L Beale
- Baker Heart & Diabetes Institute, 75 Commercial Rd, Melbourne VIC, Australia
| | | | | | - David M Kaye
- Baker Heart & Diabetes Institute, 75 Commercial Rd, Melbourne VIC, Australia
| | - Bonnie Ky
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, USA
| | - Bernadet T Santema
- University Medical Centre Groningen, Hanzeplein 1, GZ Groningen, The Netherlands
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Adriaan A Voors
- University Medical Centre Groningen, Hanzeplein 1, GZ Groningen, The Netherlands
| |
Collapse
|
2661
|
Yao QY, Li J, Chen R, Yao Y, Xue JS, Chen WJ, Lu W, Zhou TY. Preclinical PK/PD model for the combinatorial use of dexamethasone and sulpiride in the treatment of breast cancer. Acta Pharmacol Sin 2019; 40:1596-1602. [PMID: 31165782 PMCID: PMC7470835 DOI: 10.1038/s41401-019-0251-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/12/2019] [Indexed: 01/09/2023]
Abstract
Previous studies show that dopamine D2-like receptor (D2DR) antagonist sulpiride (SUL) enhances the antitumor efficacy of dexamethasone (DEX) in drug-resistant breast cancer involving cancer stem-like cells (CSCs). In this study, we investigated the pharmacokinetic (PK) properties of SUL in nude mice and developed a semi-mechanism PK/PD model to quantitatively characterize the synergistic effect of DEX and SUL in preclinical breast cancer xenografts. After nude mice received oral administration of a single dose of SUL (50 mg/kg, ig), plasma concentrations were assessed using LC-MS/MS. A two-compartment model with double first-order absorption rate was developed to describe the PK profiles of SUL. The pharmacodynamic (PD) study was conducted in nude mice bearing human breast cancer MCF-7/Adr xenografts, which received oral administration of DEX (1, 8 mg·kg−1·d−1) or SUL (25, 50 mg·kg−1·d−1) alone or in various combination. Tumor volumes were measured every other day. The PK model of SUL as well as that of DEX with a time-dependent clearance were integrated into the final PK/PD model both using Hill’s function, where DEX exerted its antitumor efficacy by inhibiting the proliferation of tumor cells, and SUL enhanced DEX responses by decreasing the sensitivity parameter EC50. The PK/PD model was evaluated and subjected external validation. Finally, simulations were performed to predict the antitumor efficacy of DEX combined with SUL under various dose regimens, where changing dosing frequency of SUL had little effect, while the antitumor efficacy was predicted to be improved when DEX was given more frequently. The established PK/PD model in this study quantitatively characterizes the antitumor efficacy of the DEX combined with SUL as well as their synergism, and the simulations could provide reference for dose optimization of the combination in future studies.
Collapse
|
2662
|
Wang Z, Chi D, Wu X, Wang Y, Lin X, Xu Z, Liu H, Sun J, He Z, Wang Y. Tyrosine modified irinotecan-loaded liposomes capable of simultaneously targeting LAT1 and ATB0,+ for efficient tumor therapy. J Control Release 2019; 316:22-33. [DOI: 10.1016/j.jconrel.2019.10.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022]
|
2663
|
Guan Q, Zhou LL, Li YA, Li WY, Wang S, Song C, Dong YB. Nanoscale Covalent Organic Framework for Combinatorial Antitumor Photodynamic and Photothermal Therapy. ACS NANO 2019; 13:13304-13316. [PMID: 31689082 DOI: 10.1021/acsnano.9b06467] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite the excellent photodynamic and photothermal properties of organic molecular photosensitizers (PSs) and photothermal agents (PTAs), such as porphyrin and naphthalocyanine, their poor water solubility severely impedes their biological applications. Covalent organic frameworks (COFs), as an emerging class of organic crystalline porous materials, possess free active end groups (bonding defects) and large inner pores, which make them an ideal type of nanocarriers for loading hydrophobic organic molecular PSs and PTAs by both bonding defect functionalization (BDF) and guest encapsulation approaches to obtain multifunctional nanomedicines for PDT/PTT combination therapy. In this work, we report a nanoscale COF (NCOF) prepared via a facile synthetic approach under ambient conditions. Furthermore, a dual-modal PDT/PTT therapeutic nanoagent, VONc@COF-Por (3), is successfully fabricated by stepwise BDF and guest encapsulation processes. The covalently grafted porphyrinic PS (Por) and the noncovalently loaded naphthalocyanine PTA (VONc) are independently responsible for the PDT and PTT functionalities of the nanoagent. Upon visible (red LED) and NIR (808 nm laser) irradiation, VONc@COF-Por (3) displayed high 1O2 generation and photothermal conversion ability (55.9%), consequently providing an excellent combined PDT/PTT therapeutic effect on inhibiting MCF-7 tumor cell proliferation and metastasis, which was well evidenced by in vitro and in vivo experiments. We believe that the results obtained herein can significantly promote the development of NCOF-based multifunctional nanomedicines for biomedical applications.
Collapse
Affiliation(s)
- Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education , Shandong Normal University , Jinan 250014 , P. R. China
| | - Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education , Shandong Normal University , Jinan 250014 , P. R. China
- School of Chemical and Biological Engineering , Qilu Institute of Technology , Jinan 250200 , P. R. China
| | - Yan-An Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education , Shandong Normal University , Jinan 250014 , P. R. China
| | - Wen-Yan Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education , Shandong Normal University , Jinan 250014 , P. R. China
| | - Shumei Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Shandong University , Jinan 250012 , P. R. China
| | - Chun Song
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Shandong University , Jinan 250012 , P. R. China
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education , Shandong Normal University , Jinan 250014 , P. R. China
| |
Collapse
|
2664
|
Punzi S, Balestrieri C, D'Alesio C, Bossi D, Dellino GI, Gatti E, Pruneri G, Criscitiello C, Lovati G, Meliksetyan M, Carugo A, Curigliano G, Natoli G, Pelicci PG, Lanfrancone L. WDR5 inhibition halts metastasis dissemination by repressing the mesenchymal phenotype of breast cancer cells. Breast Cancer Res 2019; 21:123. [PMID: 31752957 PMCID: PMC6873410 DOI: 10.1186/s13058-019-1216-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Development of metastases and drug resistance are still a challenge for a successful systemic treatment in breast cancer (BC) patients. One of the mechanisms that confer metastatic properties to the cell relies in the epithelial-to-mesenchymal transition (EMT). Moreover, both EMT and metastasis are partly modulated through epigenetic mechanisms, by repression or induction of specific related genes. Methods We applied shRNAs and drug targeting approaches in BC cell lines and metastatic patient-derived xenograft (PDX) models to inhibit WDR5, the core subunit of histone H3 K4 methyltransferase complexes, and evaluate its role in metastasis regulation. Result We report that WDR5 is crucial in regulating tumorigenesis and metastasis spreading during BC progression. In particular, WDR5 loss reduces the metastatic properties of the cells by reverting the mesenchymal phenotype of triple negative- and luminal B-derived cells, thus inducing an epithelial trait. We also suggest that this regulation is mediated by TGFβ1, implying a prominent role of WDR5 in driving EMT through TGFβ1 activation. Moreover, such EMT reversion can be induced by drug targeting of WDR5 as well, leading to BC cell sensitization to chemotherapy and enhancement of paclitaxel-dependent effects. Conclusions We suggest that WDR5 inhibition could be a promising pharmacologic approach to reduce cell migration, revert EMT, and block metastasis formation in BC, thus overcoming resistance to standard treatments.
Collapse
Affiliation(s)
- Simona Punzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Balestrieri
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Humanitas University, Pieve Emanuele (MI), 20090, Italy.,Humanitas Clinical and Research Institute, Rozzano (MI), 20089, Italy
| | - Carolina D'Alesio
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Present address: Department of Internal Medicine and Medical Specialties (Di.M.I), University of Genova, Genoa, Italy
| | - Daniela Bossi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Present address: Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Gaetano Ivan Dellino
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Gatti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giancarlo Pruneri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Department of Pathology, Biobank for Translational Medicine Unit, European Institute of Oncology, IRCCS, Milan, Italy.,Present address: Istituto Nazionale dei Tumori - Fondazione IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Lovati
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Marine Meliksetyan
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandro Carugo
- Institute for Applied Cancer Science, UT MD Anderson Cancer Cente, Houston, TX, 77030, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Gioacchino Natoli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Humanitas University, Pieve Emanuele (MI), 20090, Italy.,Humanitas Clinical and Research Institute, Rozzano (MI), 20089, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
2665
|
Güth R, Adamian Y, Geller C, Molnar J, Maddela J, Kutscher L, Bhakta K, Meade K, Kim SL, Agajanian M, Kelber JA. DHPS-dependent hypusination of eIF5A1/2 is necessary for TGFβ/fibronectin-induced breast cancer metastasis and associates with prognostically unfavorable genomic alterations in TP53. Biochem Biophys Res Commun 2019; 519:838-845. [PMID: 31558321 PMCID: PMC6801012 DOI: 10.1016/j.bbrc.2019.09.075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 12/21/2022]
Abstract
Metastasis is the leading cause of mortality in patients with solid tumors. In this regard, we previously reported that Pseudopodium-Enriched Atypical Kinase One (PEAK1) is necessary for non-canonical Transforming Growth Factor β (TGFβ) signaling and TGFβ/fibronectin-induced metastasis. Here, we demonstrate that inhibition of DHPS-dependent eIF5A1/2 hypusination blocks PEAK1 and E-Cadherin expression, breast cancer cell viability and TGFβ/fibronectin-induced PEAK1-dependent breast cancer metastasis. Interestingly, TGFβ stimulation of high-grade metastatic breast cancer cells increases and sustains eIF5A1/2 hypusination. We used a suite of bioinformatics platforms to search biochemical/functional interactions and clinical databases for additional control points in eIF5A1/2 and PEAK1-Epithelial to Mesenchymal Transition (EPE) pathways. This effort revealed that interacting EPE genes were enriched for TP53 transcriptional targets and were commonly co-amplified in breast cancer patients harboring inactivating TP53 mutations. Taken together, these results suggest that combinatorial therapies targeting DHPS and protein activities elevated in TP53-mutant breast cancers may reduce systemic tumor burden and improve patient outcomes.
Collapse
Affiliation(s)
- R Güth
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - Y Adamian
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - C Geller
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - J Molnar
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - J Maddela
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - L Kutscher
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - K Bhakta
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - K Meade
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - S L Kim
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - M Agajanian
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - J A Kelber
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA.
| |
Collapse
|
2666
|
Huang X. Rethinking the combination treatment of fulvestrant and anastrozole for metastatic breast cancer: an integrated reanalysis of aromatase-estrogen receptor axis. Clin Transl Med 2019; 8:29. [PMID: 31741086 PMCID: PMC6861394 DOI: 10.1186/s40169-019-0246-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 10/22/2019] [Indexed: 01/07/2023] Open
Abstract
Aberrant expression or hyperactivation of aromatase (CYP19A1)-estrogen receptor (ESR) axis is well identified as one of the major causes of breast cancer. Lots of drugs have been developed for targeting CYP19A1 or ESR respectively, such as anastrozole and fulvestrant. Recently, Mehta et al. reported in NEJM that the combined treatment of anastrozole and fulvestrant increased long-term survival of patients with metastatic breast cancer, especially for those without receiving endocrine therapy. However, the integrated prognostic analyses of CYP19A1 and ESR1/ESR2 indicated some contradictory outcomes to the recent clinical trial. Moreover, immunological investigation further revealed that targeting the whole CYP19A1-ESR axis might cause the inactivation of anti-tumor immune response, which largely attenuated its application prospects in breast cancer. Considered the pathophysiologic functions of CYP19A1 and ESR1/ESR2-mediated signaling pathway in breast cancer seem as more complicated than what we have already known, more precise evaluation will be needed in urgent.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
2667
|
Wang S, Liu Y, Feng Y, Zhang J, Swinnen J, Li Y, Ni Y. A Review on Curability of Cancers: More Efforts for Novel Therapeutic Options Are Needed. Cancers (Basel) 2019; 11:E1782. [PMID: 31766180 PMCID: PMC6896199 DOI: 10.3390/cancers11111782] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer remains a major cause of death globally. Given its relapsing and fatal features, curing cancer seems to be something hardly possible for the majority of patients. In view of the development in cancer therapies, this article summarizes currently available cancer therapeutics and cure potential by cancer type and stage at diagnosis, based on literature and database reviews. Currently common cancer therapeutics include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, treatment with curative intent by these methods are mainly eligible for patients with localized disease or treatment-sensitive cancers and therefore their contributions to cancer curability are relatively limited. The prognosis for cancer patients varies among different cancer types with a five-year relative survival rate (RSR) of more than 80% in thyroid cancer, melanoma, breast cancer, and Hodgkin's lymphoma. The most dismal prognosis is observed in patients with small-cell lung cancer, pancreatic cancer, hepatocellular carcinoma, oesophagal cancer, acute myeloid leukemia, non-small cell lung cancer, and gastric cancer with a five-year RSR ranging between 7% and 28%. The current review is intended to provide a general view about how much we have achieved in curing cancer as regards to different therapies and cancer types. Finally, we propose a small molecule dual-targeting broad-spectrum anticancer strategy called OncoCiDia, in combination with emerging highly sensitive liquid biopsy, with theoretical curative potential for the management of solid malignancies, especially at the micro-cancer stage.
Collapse
Affiliation(s)
- Shuncong Wang
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yewei Liu
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yuanbo Feng
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Jian Zhang
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China;
| | - Johan Swinnen
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yicheng Ni
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| |
Collapse
|
2668
|
Makena MR, Rao R. Subtype specific targeting of calcium signaling in breast cancer. Cell Calcium 2019; 85:102109. [PMID: 31783287 DOI: 10.1016/j.ceca.2019.102109] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/09/2019] [Accepted: 11/10/2019] [Indexed: 01/16/2023]
Abstract
An important component of breast milk, calcium also appears as radiographically prominent microcalcifications in breast tissue that are often the earliest sign of malignancy. Ionic Ca2+ is a universal second messenger that controls a wide swathe of effector pathways integral to gene transcription, cell cycle control, differentiation, proliferation, cell migration, and apoptosis. Whereas prolonged elevation in resting Ca2+ levels drives proliferation to initiate and sustain tumor growth, depletion of calcium stores and attenuation of calcium influx pathways underlies tumor chemoresistance and evasion of apoptosis. This paradox of Ca2+ homeostasis highlights the challenge of targeting Ca2+ signaling pathways for breast cancer therapy. Furthermore, breast cancer is a heterogeneous disease classified into distinct subtypes based on tumor origin, stage of invasiveness and hormone receptor status. Classification is important for tailoring treatment, and in predicting clinical outcome or response to chemotherapy. There have been numerous reports of dysregulated expression, localization or activity of Ca2+ channels, regulators and pumps in breast cancer. An important aspect of these alterations is that they are specific to breast cancer subtype, as exemplified by a reciprocal switch in secretory pathway Ca2+-ATPase isoforms SPCA1 and SPCA2 depending on receptor status. In this review, we discuss the current knowledge of subtype specific changes in calcium channels and pumps, with a focus on functional insights that may inform new opportunities for breast cancer therapy.
Collapse
Affiliation(s)
- Monish Ram Makena
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
2669
|
Bitencourt AGV, Goldberg J, Pinker K, Thakur SB. Clinical applications of breast cancer metabolomics using high-resolution magic angle spinning proton magnetic resonance spectroscopy (HRMAS 1H MRS): systematic scoping review. Metabolomics 2019; 15:148. [PMID: 31696341 DOI: 10.1007/s11306-019-1611-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Breast cancer is a heterogeneous disease with different prognoses and responses to systemic treatment depending on its molecular characteristics, which makes it imperative to develop new biomarkers for an individualized diagnosis and personalized oncological treatment. Ex vivo high-resolution magic angle spinning proton magnetic resonance spectroscopy (HRMAS 1H MRS) is the most common technique for metabolic quantification in human surgical and biopsy tissue specimens. OBJECTIVE To perform a review of the current available literature on the clinical applications of HRMAS 1H MRS metabolic analysis in tissue samples of breast cancer patients. METHODS This systematic scoping review included original research papers published in the English language in peer-reviewed journals. Study selection was performed independently by two reviewers and preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines were followed. RESULTS The literature search returned 159 studies and 26 papers were included as part of this systematic review. There was considerable variation regarding tissue type, aims, and statistical analysis methods across the different studies. To facilitate the interpretation of the results, the included studies were grouped according to their aims or main outcomes into: feasibility and tumor diagnosis (n = 6); tumor heterogeneity (n = 2); correlation with proteomics/transcriptomics (n = 3); correlation with prognostic factors (n = 11); and response evaluation to NAC (n = 4). CONCLUSION There is a lot of potential in including metabolic information of breast cancer tissue obtained with HRMAS 1H MRS. To date, studies show that metabolic concentrations quantified by this technique can be related to the diagnosis, prognosis, and treatment response in breast cancer patients.
Collapse
Affiliation(s)
- Almir G V Bitencourt
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Imaging, A.C.Camargo Cancer Center, São Paulo, SP, Brazil
| | - Johanna Goldberg
- MSK Library, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katja Pinker
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sunitha B Thakur
- Breast Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.
| |
Collapse
|
2670
|
Benvenuto M, Focaccetti C, Izzi V, Masuelli L, Modesti A, Bei R. Tumor antigens heterogeneity and immune response-targeting neoantigens in breast cancer. Semin Cancer Biol 2019; 72:65-75. [PMID: 31698088 DOI: 10.1016/j.semcancer.2019.10.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/30/2019] [Indexed: 12/25/2022]
Abstract
Breast cancer is both the most common type of cancer and the most frequent cause of cancer mortality in women, mainly because of its heterogeneity and limited immunogenicity. The aim of specific active cancer immunotherapy is to stimulate the host's immune response against cancer cells directly using a vaccine platform carrying one or more tumor antigens. In particular, the ideal tumor antigen should be able to elicit T cell and B cell responses, be specific for the tumor and be expressed at high levels on cancer cells. Neoantigens are ideal targets for immunotherapy because they are exclusive to individual patient's tumors, are absent in healthy tissues and are not subject to immune tolerance mechanisms. Thus, neoantigens should generate a specific reaction towards tumors since they constitute the largest fraction of targets of tumor-infiltrating T cells. In this review, we describe the technologies used for neoantigen discovery, the heterogeneity of neoantigens in breast cancer and recent studies of breast cancer immunotherapy targeting neoantigens.
Collapse
Affiliation(s)
- Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy; Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131, Rome, Italy.
| | - Chiara Focaccetti
- Department of Human Science and Promotion of the Quality of Life, University San Raffaele Rome, Via di Val Cannuta 247, 00166, Rome, Italy.
| | - Valerio Izzi
- Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7C, FI-90230, Oulu, Finland.
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
2671
|
DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, Jemal A, Siegel RL. Breast cancer statistics, 2019. CA Cancer J Clin 2019; 69:438-451. [PMID: 31577379 DOI: 10.3322/caac.21583] [Citation(s) in RCA: 1971] [Impact Index Per Article: 328.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022] Open
Abstract
This article is the American Cancer Society's biennial update on female breast cancer statistics in the United States, including data on incidence, mortality, survival, and screening. Over the most recent 5-year period (2012-2016), the breast cancer incidence rate increased slightly by 0.3% per year, largely because of rising rates of local stage and hormone receptor-positive disease. In contrast, the breast cancer death rate continues to decline, dropping 40% from 1989 to 2017 and translating to 375,900 breast cancer deaths averted. Notably, the pace of the decline has slowed from an annual decrease of 1.9% during 1998 through 2011 to 1.3% during 2011 through 2017, largely driven by the trend in white women. Consequently, the black-white disparity in breast cancer mortality has remained stable since 2011 after widening over the past 3 decades. Nevertheless, the death rate remains 40% higher in blacks (28.4 vs 20.3 deaths per 100,000) despite a lower incidence rate (126.7 vs 130.8); this disparity is magnified among black women aged <50 years, who have a death rate double that of whites. In the most recent 5-year period (2013-2017), the death rate declined in Hispanics (2.1% per year), blacks (1.5%), whites (1.0%), and Asians/Pacific Islanders (0.8%) but was stable in American Indians/Alaska Natives. However, by state, breast cancer mortality rates are no longer declining in Nebraska overall; in Colorado and Wisconsin in black women; and in Nebraska, Texas, and Virginia in white women. Breast cancer was the leading cause of cancer death in women (surpassing lung cancer) in four Southern and two Midwestern states among blacks and in Utah among whites during 2016-2017. Declines in breast cancer mortality could be accelerated by expanding access to high-quality prevention, early detection, and treatment services to all women.
Collapse
Affiliation(s)
- Carol E DeSantis
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| | - Jiemin Ma
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| | - Mia M Gaudet
- Behavioral and Epidemiology Research, American Cancer Society, Atlanta, Georgia
| | - Lisa A Newman
- Department of Surgery, Weill Cornell Medical Center, New York, New York
| | - Kimberly D Miller
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| | - Ann Goding Sauer
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| | - Ahmedin Jemal
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| | - Rebecca L Siegel
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
2672
|
Ding Y, Ding K, Yu K, Zou D, Yang H, He X, Mo W, Yu X, Ding X. Prognosis and endocrine therapy selection for patients with low hormone receptor-positive breast cancer following neoadjuvant chemotherapy: A retrospective study of 570 patients in China. Oncol Lett 2019; 18:6690-6696. [PMID: 31807179 PMCID: PMC6876291 DOI: 10.3892/ol.2019.11033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
The 2010 American Society of Clinical Oncology guidelines have reduced the immunohistochemistry cut-off value for determining estrogen receptor b positivity from 10 to 1% of stained cells in breast cancer. In clinical practice, low-hormone receptor positive (low HR+) tumors are classified in the luminal subtype, although they exhibit aggressive features and poor prognosis. Information regarding the prognosis of patients with breast cancer following treatment with optimal endocrine therapy and neoadjuvant chemotherapy (NAC) is currently lacking. In the present study, the differences in clinical characteristics and survival of patients with breast cancer were compared among those with low and high HR+ breast cancer who received NAC. Furthermore, the effects of different types of endocrine therapies on the prognosis of patients with breast cancer were compared. The study population comprised patients with primary breast cancer who were treated at the Zhejiang Cancer Hospital between January, 2007 and December, 2017. Patients were divided into three groups based on the results of immunohistochemistry: HR+ (positive staining >10%), HR- (positive staining <1%) and low HR+ (positive staining 1–10%). The low HR+ group was further divided into three subgroups according to the different endocrine therapies administered: Tamoxifen, aromatase inhibitor or no treatment. Among the 570 patients included in the present study, 60 (10.53%) patients had low HR+ tumors. With a median follow-up of 48.98 months, patients with low HR+ tumors had reduced survival rates compared with those with HR+ tumors. Furthermore, the pathologic complete response rate (pCR) of patients with low HR+ was comprised between pCR from patients with HR+ and pCR from patients with HR- following NAC treatment. In addition, no significant difference in the overall prognosis was observed among patients with low HR+ following treatment with different endocrine therapies. Subsequently, patients in the low HR+ group were more likely to benefit from NAC compared with patients in the HR+ group. Intensive endocrine therapy may therefore improve the prognosis of patients with breast cancer and low HR+; however, further investigation is required.
Collapse
Affiliation(s)
- Yuqin Ding
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Kaijing Ding
- Department of Child Psychology, Zhejiang University Affiliated Mental Health Center, Hangzhou, Zhejiang 310013, P.R. China
| | - Kun Yu
- Department of Thyroid Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Dehong Zou
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Hongjian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiangming He
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Wenju Mo
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xingfei Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiaowen Ding
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
2673
|
Dou WT, Liu LF, Gao J, Zang Y, Chen GR, Field RA, James TD, Li J, He XP. Fluorescence imaging of a potential diagnostic biomarker for breast cancer cells using a peptide-functionalized fluorogenic 2D material. Chem Commun (Camb) 2019; 55:13235-13238. [PMID: 31621698 DOI: 10.1039/c9cc06399d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein C receptor (PROCR) is a recently discovered transmembrane biomarker for several tissue stem cells and is highly expressed in triple-negative breast cancer (TNBC) patient-derived xenografts. Herein, to enrich the toolbox for the biochemical evaluation of PROCR, we have developed a peptide-functionalized fluorogenic 2D material based on the self-assembly between a fluorescent peptide probe and thin-layer molybdenum disulfide. The material developed was suitable for the sensitive detection of PROCR recombinant protein in buffer solution and the fluorescence imaging of TNBC cells that express high levels of PROCR.
Collapse
Affiliation(s)
- Wei-Tao Dou
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Li-Fang Liu
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China. and National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China.
| | - Jie Gao
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China. and National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China.
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China.
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Robert A Field
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Rd, Shanghai 201203, P. R. China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| |
Collapse
|
2674
|
Papadakos KS, Darlix A, Jacot W, Blom AM. High Levels of Cartilage Oligomeric Matrix Protein in the Serum of Breast Cancer Patients Can Serve as an Independent Prognostic Marker. Front Oncol 2019; 9:1141. [PMID: 31737569 PMCID: PMC6831625 DOI: 10.3389/fonc.2019.01141] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Cartilage oligomeric matrix protein (COMP) is a pentameric cartilage protein also expressed in breast cancer tumors. A high expression of COMP evaluated by immunohistochemical staining is as an independent prognostic marker associated with poor patients' prognosis. Methods: Herein, levels of COMP were analyzed using an IVD approved ELISA in serum samples from 233 well-characterized breast cancer patients; 176 with metastatic breast cancer; and 57 in an early stage of the disease. Results: The metastatic patients had double the concentration of serum COMP compared with those with early breast cancer. High levels of COMP in sera of metastatic patients were associated with the histological subtype (p = 0.025) and estrogen receptor positivity (p = 0.019) at the time of breast cancer diagnosis. Further, correlation was observed between the serum levels of COMP and the presence of liver (p = 0.010) or bone (p = 0.010) metastases in this population. Most importantly, elevated serum levels of COMP appear to serve as an independent prognostic marker of survival as assessed by Cox proportional hazard regression analysis (p = 0.001) for the metastatic patients. Among metastatic patients treated with taxanes (Docetaxel-Paclitaxel) as part of their first metastatic line (n = 25), those with high levels of serum COMP detected in the metastatic stage of the disease had a shorter median survival (0.2 years) compared with those with low levels of serum COMP (1.1 years) (p = 0.001). Conclusions: Taken together, the serum levels of COMP are elevated in the metastatic patients and may be a potential novel biomarker for the evaluation of the prognosis in this population.
Collapse
Affiliation(s)
- Konstantinos S Papadakos
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Amélie Darlix
- Department of Medical Oncology, Institut Régional du Cancer Montpellier ICM, University of Montpellier, Montpellier, France
| | - William Jacot
- Department of Medical Oncology, Institut Régional du Cancer Montpellier ICM, University of Montpellier, Montpellier, France
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
2675
|
Abstract
Critical illness syndromes, including sepsis and the acute respiratory distress syndrome (ARDS), are identified using consensus definitions that are based on broad, clinically available criteria and include patients with heterogeneous biology. This heterogeneity is a barrier to developing and testing effective therapies for these syndromes. Biomarkers identify clinically distinct molecular phenotypes of ARDS and sepsis. These molecular phenotypes are associated with differences in mortality and predict response to several treatments in retrospective analyses of clinical trials. Biomarkers can be used for prognostic and predictive enrichment of clinical trials in critical illness to incorporate precision medicine in critical care.
Collapse
Affiliation(s)
- Aartik Sarma
- Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, Box 0111, San Francisco, CA 94143-0111, USA
| | - Carolyn S Calfee
- Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, Box 0111, San Francisco, CA 94143-0111, USA; Department of Anesthesia, University of California, San Francisco, 505 Parnassus Avenue, Box 0111, San Francisco, CA 94143-0111, USA; Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, Box 0111, San Francisco, CA 94143-0111, USA
| | - Lorraine B Ware
- Department of Medicine, Vanderbilt University School of Medicine, T1218 MCN, 1161 21st Avenue South, Nashville, TN 37232-2650, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA; Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st Avenue South, Nashville, TN 37232-2650, USA.
| |
Collapse
|
2676
|
Cosentino G, Plantamura I, Cataldo A, Iorio MV. MicroRNA and Oxidative Stress Interplay in the Context of Breast Cancer Pathogenesis. Int J Mol Sci 2019; 20:ijms20205143. [PMID: 31627322 PMCID: PMC6829356 DOI: 10.3390/ijms20205143] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is a pathological condition determined by a disturbance in reactive oxygen species (ROS) homeostasis. Depending on the entity of the perturbation, normal cells can either restore equilibrium or activate pathways of cell death. On the contrary, cancer cells exploit this phenomenon to sustain a proliferative and aggressive phenotype. In fact, ROS overproduction or their reduced disposal influence all hallmarks of cancer, from genome instability to cell metabolism, angiogenesis, invasion and metastasis. A persistent state of oxidative stress can even initiate tumorigenesis. MicroRNAs (miRNAs) are small non coding RNAs with regulatory functions, which expression has been extensively proven to be dysregulated in cancer. Intuitively, miRNA transcription and biogenesis are affected by the oxidative status of the cell and, in some instances, they participate in defining it. Indeed, it is widely reported the role of miRNAs in regulating numerous factors involved in the ROS signaling pathways. Given that miRNA function and modulation relies on cell type or tumor, in order to delineate a clearer and more exhaustive picture, in this review we present a comprehensive overview of the literature concerning how miRNAs and ROS signaling interplay affects breast cancer progression.
Collapse
Affiliation(s)
- Giulia Cosentino
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Ilaria Plantamura
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Alessandra Cataldo
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
- IFOM Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy.
| | - Marilena V Iorio
- Molecular Targeting Unit, Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
- IFOM Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy.
| |
Collapse
|
2677
|
Qiao K, Ning S, Wan L, Wu H, Wang Q, Zhang X, Xu S, Pang D. LINC00673 is activated by YY1 and promotes the proliferation of breast cancer cells via the miR-515-5p/MARK4/Hippo signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:418. [PMID: 31623640 PMCID: PMC6796384 DOI: 10.1186/s13046-019-1421-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023]
Abstract
Background An increasing number of studies have shown that long noncoding RNAs (lncRNAs) play essential roles in tumor initiation and progression. LncRNAs act as tumor promoters or suppressors by targeting specific genes via epigenetic modifications and competing endogenous RNA (ceRNA) mechanisms. In this study, we explored the function and detailed mechanisms of long intergenic nonprotein coding RNA 673 (LINC00673) in breast cancer progression. Methods Quantitative real-time PCR (qRT-PCR) was used to examine the expression of LINC00673 in breast cancer tissues and in adjacent normal tissues. Gain-of-function and loss-of function experiments were conducted to investigate the biological functions of LINC00673 in vitro and in vivo. We also explored the potential role of LINC00673 as a therapeutic target using antisense oligonucleotide (ASO) in vivo. RNA sequencing (RNA-seq), dual-luciferase reporter assays, chromatin immunoprecipitation (ChIP) assay, and rescue experiments were performed to uncover the detailed mechanism of LINC00673 in promoting breast cancer progression. Results In the present study, LINC00673 displayed a trend of remarkably increased expression in breast cancer tissues and was associated with poor prognosis in breast cancer patients. Importantly, LINC00673 depletion inhibited breast cancer cell proliferation by inhibiting the cell cycle and increasing apoptosis. Furthermore, ASO therapy targeting LINC00673 substantially suppressed breast cancer cell proliferation in vivo. Mechanistically, LINC00673 was found to act as a ceRNA by sponging miR-515-5p to regulate MARK4 expression, thus inhibiting the Hippo signaling pathway. Finally, ChIP assay showed that the transcription factor Yin Yang 1 (YY1) could bind to the LINC00673 promoter and increase its transcription in cis. Conclusions YY1-activated LINC00673 may exert an oncogenic function by acting as a sponge for miR-515-5p to upregulate the MARK4 and then inhibit Hippo signaling pathway, and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Shipeng Ning
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Lin Wan
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Hao Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Qin Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Xingda Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China.
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150086, China. .,Heilongjiang Academy of Medical Sciences, 157 Baojian Road, Harbin, 150086, China.
| |
Collapse
|
2678
|
Kargbo RB. PROTAC-Mediated Degradation of Estrogen Receptor in the Treatment of Cancer. ACS Med Chem Lett 2019; 10:1367-1369. [PMID: 31620216 DOI: 10.1021/acsmedchemlett.9b00397] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Indexed: 11/30/2022] Open
Affiliation(s)
- Robert B. Kargbo
- Usona Institute, 277 Granada Drive, San Luis Obispo, California 93401-7337, United States
| |
Collapse
|
2679
|
Rahman MM, Brane AC, Tollefsbol TO. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer. Cells 2019; 8:cells8101214. [PMID: 31597272 PMCID: PMC6829616 DOI: 10.3390/cells8101214] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/04/2019] [Accepted: 10/06/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is a sporadic disease with genetic and epigenetic components. Genomic instability in breast cancer leads to mutations, copy number variations, and genetic rearrangements, while epigenetic remodeling involves alteration by DNA methylation, histone modification and microRNAs (miRNAs) of gene expression profiles. The accrued scientific findings strongly suggest epigenetic dysregulation in breast cancer pathogenesis though genomic instability is central to breast cancer hallmarks. Being reversible and plastic, epigenetic processes appear more amenable toward therapeutic intervention than the more unidirectional genetic alterations. In this review, we discuss the epigenetic reprogramming associated with breast cancer such as shuffling of DNA methylation, histone acetylation, histone methylation, and miRNAs expression profiles. As part of this, we illustrate how epigenetic instability orchestrates the attainment of cancer hallmarks which stimulate the neoplastic transformation-tumorigenesis-malignancy cascades. As reversibility of epigenetic controls is a promising feature to optimize for devising novel therapeutic approaches, we also focus on the strategies for restoring the epistate that favor improved disease outcome and therapeutic intervention.
Collapse
Affiliation(s)
- Mohammad Mijanur Rahman
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Andrew C Brane
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Center for Healthy Aging, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA.
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA.
| |
Collapse
|
2680
|
Del Prete S, Caraglia M, Luce A, Montella L, Galizia G, Sperlongano P, Cennamo G, Lieto E, Capasso E, Fiorentino O, Aliberti M, Auricchio A, Iodice P, Addeo R. Clinical and pathological factors predictive of response to neoadjuvant chemotherapy in breast cancer: A single center experience. Oncol Lett 2019; 18:3873-3879. [PMID: 31516598 PMCID: PMC6732960 DOI: 10.3892/ol.2019.10729] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) of breast cancer (BC) improves outcomes, especially in patients with locally advanced and inflammatory cancer. Further insight into clinic-pathological factors influencing outcomes is essential to define the optimal therapeutic strategy for each category of patients and to predict the response to the treatment. In total, 117 patients with BC were treated with NAC with or without trastuzumab between 2010 and 2015. The histologic response to NAC was defined as a pathological complete response (pCR) when there was no evidence of residual invasive tumor in the breast or axillary lymph nodes. Relapse-free survival (RFS) was estimated using the Kaplan-Meier method and compared using log rank analysis. P-value <0.05 was considered statistically significant. The median age of the 117 patients enrolled in the present study was 52 years (age range, 35-85 years). The overall response rate (complete and partial responses) assessed by radiological and pathological evaluation were 76 and 72%, respectively. pCR was achieved in 35 out of 117 patients (~30%). In total, 6 patients (5%) developed progressive disease during chemotherapy. The RFS was 85 months (SE=3; 95% CI 79-91). The median was not reached and the mean follow-up time was 55 months (median 52 months; range 11-100 months). In this time, 20 patients (17%) experienced tumor recurrence. From the univariate analysis, the pathological response was significantly associated with receptor-based subtype, menopausal status and T-stage. From the multivariate analysis by using linear multiple regression and including receptor- menopausal status and T-stage, the model was not significant (P=0.062). However, by using the multiple logistic regression, and including age, pCR was significantly associated with ER+ HER2neg (P=0.006), T2 (P=0.043) and T3 (P=0.018). T-stage, menopausal status and receptor status are significantly associated with the pathological response in patients with inoperable BC treated with NAC.
Collapse
Affiliation(s)
- Salvatore Del Prete
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Amalia Luce
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Liliana Montella
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Gennaro Galizia
- Division of Gastrointestinal Tract Surgical Oncology, Department of Translational Medical Sciences, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Pasquale Sperlongano
- Division of Gastrointestinal Tract Surgical Oncology, Department of Translational Medical Sciences, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Gregorio Cennamo
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Eva Lieto
- Division of Gastrointestinal Tract Surgical Oncology, Department of Translational Medical Sciences, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Elena Capasso
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Olga Fiorentino
- Pathologist Department, Medicina Futura Group, I-80011 Naples, Italy
| | - Maria Aliberti
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Annamaria Auricchio
- Division of Gastrointestinal Tract Surgical Oncology, Department of Translational Medical Sciences, University of Campania ‘L. Vanvitelli’, I-80138 Naples, Italy
| | - Patrizia Iodice
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| | - Raffaele Addeo
- Division of Medical Oncology, ‘San Giovanni Di Dio Hospital’, ASL NA2NORD, I-80027 Naples, Italy
| |
Collapse
|
2681
|
Irradiation partielle et accélérée du sein par une technique de curiethérapie de haut débit de dose chez les patientes âgées : résultats oncologiques et facteurs pronostiques. Cancer Radiother 2019. [DOI: 10.1016/j.canrad.2019.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
2682
|
Mallick A, Taylor S. Therapeutic potential of estradiol in treating breast cancer. BREAST CANCER MANAGEMENT 2019. [DOI: 10.2217/bmt-2019-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Avijit Mallick
- Department of Biology, McMaster University, 1280 Main St West, Hamilton, Ontario L8S4K1, Canada
| | - Shane Taylor
- Department of Biology, McMaster University, 1280 Main St West, Hamilton, Ontario L8S4K1, Canada
| |
Collapse
|
2683
|
Kitagawa Y, Ito T, Mizuno Y, Sudo Y, Kim Y, Tsunoda R, Takai S. Effect of Orthopedics Promotional Activities on Multidisciplinary Management of Patients with Bone Metastases. J NIPPON MED SCH 2019; 86:327-335. [PMID: 31564687 DOI: 10.1272/jnms.jnms.2019_86-609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The rapid aging of the Japanese population is leading to an increase in the number of patients with bone metastases. Since 2014, our orthopedics department has promoted multidisciplinary hospital activities, including offering lectures to hospital staff on multidisciplinary approaches for bone metastases and holding regular cancer board meetings on bone metastases. This study investigated whether these activities were effective in promoting multidisciplinary approaches and improving outcomes of patients with bone metastasis. METHODS To investigate the effects of changes in medical practice on patients with bone metastases, we compared patient clinical characteristics after (January 2014 through December 2017) and before the start of the activities (January 2011 through December 2013). RESULTS The semiannual numbers of first-visit, referral, and orthopedic surgical patients, the number of patients with slower growing primary cancers, and the number of patients with milder pain were significantly higher post-activity than pre-activity. The number of patients without paralysis was higher after the start of the activity than before the activity, but the difference was not significant. Survival after the first visit to the orthopedics department was significantly longer after the start of the activity than before the activity. CONCLUSIONS The potential demand for a multidisciplinary approach to bone metastases is high, and orthopedic specialists should actively participate in this approach.
Collapse
Affiliation(s)
| | - Toshihiko Ito
- Department of Orthopaedic Surgery, Nippon Medical School
| | | | - Yoshihiro Sudo
- Department of Orthopaedic Surgery, Nippon Medical School
| | - Yong Kim
- Department of Orthopaedic Surgery, Nippon Medical School
| | - Ryu Tsunoda
- Department of Orthopaedic Surgery, Nippon Medical School
| | - Shinro Takai
- Department of Orthopaedic Surgery, Nippon Medical School
| |
Collapse
|
2684
|
Risk of Hematologic Malignant Neoplasms after Postoperative Treatment of Breast Cancer. Cancers (Basel) 2019; 11:cancers11101463. [PMID: 31569513 PMCID: PMC6827362 DOI: 10.3390/cancers11101463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 12/31/2022] Open
Abstract
An indirect consequence of the improved long-term survival seen in patients with breast cancer (BC) is the increased risk of hematologic malignant neoplasms (HM). This study aimed to analyze the role of postoperative treatment for BC in the development of subsequent HM. Using the French National Health Data System, we examined the HM risks in patients diagnosed with an incident primary breast cancer between 2007 and 2015, who underwent surgery as first-line treatment for BC. Main outcomes were acute myeloid leukemia (AML), Myelodysplastic syndrome (MDS), myeloproliferative neoplasms (MPNs), multiple myeloma (MM), Hodgkin's lymphoma or non-Hodgkin's lymphoma (HL/NHL), and acute lymphoblastic leukemia or lymphocytic lymphoma (ALL/LL). Analyses were censored at HM occurrence, death, loss to follow up, or December 2017. The risk of each type of HM was compared according to the initial postoperative treatment of breast cancer. Of a total of 324,056 BC survivors, 15.5% underwent surgery only, 46.7% received radiotherapy after surgery, 4.3% received chemotherapy after surgery, and 33.5% received all three modalities. Overall, 2236 cases of hematologic malignancies occurred. Compared to the surgery alone group, AML was significantly increased after surgery plus radiation (aHR, 1.5; 95% CI, 1.0-2.1), surgery plus chemotherapy (aHR, 2.1; 95% CI, 1.2-3.6) and all modalities (aHR, 3.3; 95% CI, 2.3-4.7). MDS was significantly increased after surgery plus chemotherapy (aHR, 1.7; 95% CI, 1.1-2.5) or after all modalities (aHR, 1.4; 95% CI, 1.1-1.8). HL/NHL were significantly increased only in the radiotherapy and surgery group (aHR, 1.3; 95% CI, 1.0-1.6). A nonsignificant increase of ALL/LL (aHR, 1.8; 95% CI, 0.6-3.5) was noted after chemotherapy and with all three modalities (aHR, 1.4; 95% CI, 0.7-2.8). Our population based study revealed increased risks of various HM associated with postoperative BC treatment. The added benefit of chemotherapy and radiation therapy should take into consideration these long-term complications.
Collapse
|
2685
|
Drug repurposing for breast cancer therapy: Old weapon for new battle. Semin Cancer Biol 2019; 68:8-20. [PMID: 31550502 PMCID: PMC7128772 DOI: 10.1016/j.semcancer.2019.09.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
Despite tremendous resources being invested in prevention and treatment, breast cancer remains a leading cause of cancer deaths in women globally. The available treatment modalities are very costly and produces severe side effects. Drug repurposing that relate to new uses for old drugs has emerged as a novel approach for drug development. Repositioning of old, clinically approved, off patent non-cancer drugs with known targets, into newer indication is like using old weapons for new battle. The advances in genomics, proteomics and information computational biology has facilitated the process of drug repurposing. Repositioning approach not only fastens the process of drug development but also offers more effective, cheaper, safer drugs with lesser/known side effects. During the last decade, drugs such as alkylating agents, anthracyclins, antimetabolite, CDK4/6 inhibitor, aromatase inhibitor, mTOR inhibitor and mitotic inhibitors has been repositioned for breast cancer treatment. The repositioned drugs have been successfully used for the treatment of most aggressive triple negative breast cancer. The literature review suggest that serendipity plays a major role in the drug development. This article describes the comprehensive overview of the current scenario of drug repurposing for the breast cancer treatment. The strategies as well as several examples of repurposed drugs are provided. The challenges associated with drug repurposing are discussed.
Collapse
|
2686
|
Synnott NC, O’Connell D, Crown J, Duffy MJ. COTI-2 reactivates mutant p53 and inhibits growth of triple-negative breast cancer cells. Breast Cancer Res Treat 2019; 179:47-56. [DOI: 10.1007/s10549-019-05435-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/05/2019] [Indexed: 10/26/2022]
|
2687
|
Cüce Aydoğmuş EM, İnhan Garip GA. Synergistic Effects of 4-Aminopyridine and Paclitaxel on MCF 7 cell line. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2019. [DOI: 10.33808/clinexphealthsci.557797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
2688
|
Liu Z, Mi M, Li X, Zheng X, Wu G, Zhang L. lncRNA OSTN-AS1 May Represent a Novel Immune-Related Prognostic Marker for Triple-Negative Breast Cancer Based on Integrated Analysis of a ceRNA Network. Front Genet 2019; 10:850. [PMID: 31572452 PMCID: PMC6753250 DOI: 10.3389/fgene.2019.00850] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
The competing endogenous RNA (ceRNA) networks are an effective method for investigating cancer; however, construction of ceRNA networks among different subtypes of breast cancer has not been previously performed. Based on analysis of differentially expressed RNAs between 150 triple-negative breast cancer (TNBC) tissues and 823 non–triple-negative breast cancer (nTNBC) tissues downloaded from TCGA database, a ceRNA network was constructed based on database comparisons using Cytoscape. Survival analysis and receiver operating characteristic curve data were combined to screen out prognostic candidate genes, which were subsequently analyzed using co-expressed functionally related analysis, Gene Set Variation Analysis (GSVA) pathway-related analysis, and immune infiltration and tumor mutational burden immune-related analysis. A total of 190 differentially expressed lncRNAs (DElncRNAs), 48 differentially expressed mRNAs (DEmRNAs), and 13 differentially expressed miRNAs (DEmiRNAs) were included in the ceRNA network between TNBC and nTNBC subtypes. Gene ontology analysis of mRNAs coexpressed with prognostic candidate lncRNAs (AC104472.1, PSORS1C3, DSCR9, OSTN-AS1, AC012074.1, AC005035.1, SIAH2-AS1, and ERVMER61-1) were utilized for functional prediction. Consequently, OSTN-AS1 was primarily related to immunologic function, for instance, immune cell infiltration and immune-related markers coexpression. The GSVA deviation degree was increased with OSTN increased expression. In addition, many important immune molecules, such as PDCD1 and CTLA-4, were strongly correlated in terms of their quantitative expression. Competing endogenous RNA networks may identify candidate therapeutic targets and potential prognostic biomarkers in breast cancer. In particular, OSTN-AS1 serves as a novel immune-related molecule and could be involved in immunotherapy efforts in the future.
Collapse
Affiliation(s)
- Zijian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mi Mi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2689
|
Estrogens Counteract Platinum-Chemosensitivity by Modifying the Subcellular Localization of MDM4. Cancers (Basel) 2019; 11:cancers11091349. [PMID: 31547268 PMCID: PMC6770881 DOI: 10.3390/cancers11091349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/28/2022] Open
Abstract
Estrogen activity towards cancer-related pathways can impact therapeutic intervention. Recent omics data suggest possible crosstalk between estrogens/gender and MDM4, a key regulator of p53. Since MDM4 can either promote cell transformation or enhance DNA damage-sensitivity, we analysed in vivo impact of estrogens on both MDM4 activities. In Mdm4 transgenic mouse, Mdm4 accelerates the formation of fibrosarcoma and increases tumor sensitivity to cisplatin as well, thus confirming in vivo Mdm4 dual mode of action. Noteworthy, Mdm4 enhances chemo- and radio-sensitivity in male but not in female animals, whereas its tumor-promoting activity is not affected by mouse gender. Combination therapy of transgenic females with cisplatin and fulvestrant, a selective estrogen receptor degrader, was able to recover tumor cisplatin-sensitivity, demonstrating the relevance of estrogens in the observed sexual dimorphism. Molecularly, estrogen receptor-α alters intracellular localization of MDM4 by increasing its nuclear fraction correlated to decreased cell death, in a p53-independent manner. Importantly, MDM4 nuclear localization and intra-tumor estrogen availability correlate with decreased platinum-sensitivity and apoptosis and predicts poor disease-free survival in high-grade serous ovarian carcinoma. These data demonstrate estrogen ability to modulate chemo-sensitivity of MDM4-expressing tumors and to impinge on intracellular trafficking. They support potential usefulness of combination therapy involving anti-estrogenic drugs.
Collapse
|
2690
|
LncRNA HEIH regulates cell proliferation and apoptosis through miR-4458/SOCS1 axis in triple-negative breast cancer. Hum Cell 2019; 32:522-528. [DOI: 10.1007/s13577-019-00273-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022]
|
2691
|
Lee D, Lee WY, Jung K, Kwon YS, Kim D, Hwang GS, Kim CE, Lee S, Kang KS. The Inhibitory Effect of Cordycepin on the Proliferation of MCF-7 Breast Cancer Cells, and its Mechanism: An Investigation Using Network Pharmacology-Based Analysis. Biomolecules 2019; 9:E414. [PMID: 31454995 PMCID: PMC6770402 DOI: 10.3390/biom9090414] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022] Open
Abstract
Cordyceps militaris is a well-known medicinal mushroom. It is non-toxic and has clinical health benefits including cancer inhibition. However, the anticancer effects of C. militaris cultured in brown rice on breast cancer have not yet been reported. In this study, we simultaneously investigated the anticancer effects of cordycepin and an extract of C. militaris cultured in brown rice on MCF-7 human breast cancer cells using a cell viability assay, cell staining with Hoechst 33342, and an image-based cytometric assay. The C. militaris concentrate exhibited significant MCF-7 cell inhibitory effects, and its IC50 value was 73.48 µg/mL. Cordycepin also exhibited significant MCF-7 cell inhibitory effects, and its IC50 value was 9.58 µM. We applied network pharmacological analysis to predict potential targets and pathways of cordycepin. The gene set enrichment analysis showed that the targets of cordycepin are mainly associated with the hedgehog signaling, apoptosis, p53 signaling, and estrogen signaling pathways. We further verified the predicted targets related to the apoptosis pathway using western blot analysis. The C. militaris concentrate and cordycepin exhibited the ability to induce apoptotic cell death by increasing the cleavage of caspase-7 -8, and -9, increasing the Bcl-2-associated X protein/ B-cell lymphoma 2 (Bax/Bcl-2) protein expression ratio, and decreasing the protein expression of X-linked inhibitor of apoptosis protein (XIAP) in MCF-7 cells. Consequently, the C. militaris concentrate and cordycepin exhibited significant anticancer effects through their ability to induce apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Dahae Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Won-Yung Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844, Korea
| | - Yong Sam Kwon
- Dong-A Pharmaceutical Co., LTD., Yongin 17073, Korea
| | - Daeyoung Kim
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam, 13120, Korea
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea
| | - Chang-Eop Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam, 13120, Korea.
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea.
| |
Collapse
|
2692
|
Tu Z, Schmöllerl J, Cuiffo BG, Karnoub AE. Microenvironmental Regulation of Long Noncoding RNA LINC01133 Promotes Cancer Stem Cell-Like Phenotypic Traits in Triple-Negative Breast Cancers. Stem Cells 2019; 37:1281-1292. [PMID: 31283068 DOI: 10.1002/stem.3055] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 01/08/2023]
Abstract
The fibrotic tumor microenvironment is a critical player in the pathogenesis of triple-negative breast cancers (TNBCs), with the presence of fibroblastic infiltrates particularly correlating with tumors that are clinically advanced. On this front, we previously demonstrated that TNBCs are highly enriched in fibroblastic stromal progenitor cells called mesenchymal stem/stromal cells (MSCs) and that such cells play critical roles in promoting TNBC initiation and progression. How TNBC cells respond to MSC stimulation, however, is not fully understood, and stands to reveal contextual signals used by TNBC cells during tumor development and provide biomarkers and therapeutic targets of pertinence to TNBC management. Here, we report that MSCs strongly induced the long noncoding RNA (lncRNA) LINC01133 in neighboring TNBC cells. Indeed, although lncRNAs have been tightly associated with cancer development, their contributions to breast cancer in general, and to TNBC pathogenesis in particular, have not been fully elucidated, and we set out to determine if LINC01133 regulated malignant traits in TNBC cells. We establish that LINC01133 is sufficient, on its own, in promoting phenotypic and growth characteristics of cancer stem cell-like cells, and that it is a direct mediator of the MSC-triggered miR-199a-FOXP2 pathway in TNBC models. Furthermore, we show that LINC01133 is a critical regulator of the pluripotency-determining gene Kruppel-Like Factor 4 (KLF4), and that it represents a biomarker and prognosticator of disease outcome in the clinic. Collectively, our findings introduce LINC01133 as a novel functional driver of malignancy and a potential theranostic in TNBC. Stem Cells 2019;37:1281-1292.
Collapse
Affiliation(s)
- Zhenbo Tu
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Johannes Schmöllerl
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin G Cuiffo
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
2693
|
Wu D, Daly HC, Grossi M, Conroy E, Li B, Gallagher WM, Elmes R, O'Shea DF. RGD conjugated cell uptake off to on responsive NIR-AZA fluorophores: applications toward intraoperative fluorescence guided surgery. Chem Sci 2019; 10:6944-6956. [PMID: 31588261 PMCID: PMC6686729 DOI: 10.1039/c9sc02197c] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
The use of NIR-fluorescence imaging to demarcate tumour boundaries for real-time guidance of their surgical resection has a huge untapped potential. However, fluorescence imaging using molecular fluorophores, even with a targeting biomolecule attached, has a major shortcoming of signal interference from non-specific background fluorescence outside the region of interest. This poor selectivity necessitates prolonged time delays to allow clearance of background fluorophore and retention within the tumour prior to image acquisition. In this report, an innovative approach to overcome this issue is described in which cancer targeted off to on bio-responsive NIR-fluorophores are utilised to switch-on first within the tumour. Bio-responsive cRGD, iRGD and PEG conjugates have been synthesised using activated ester/amine or maleimide/thiol couplings to link targeting and fluorophore components. Their off to on emission responses were measured and compared with an always-on non-responsive control with each bio-responsive derivative showing large fluorescence enhancement values. Live cell imaging experiments using metastatic breast cancer cells confirmed in vitro bio-responsive capabilities. An in vivo assessment of MDA-MB 231 tumour imaging performance for bio-responsive and always-on fluorophores was conducted with monitoring of fluorescence distributions over 96 h. As anticipated, the always-on fluorophore gave an immediate, non-specific and very strong emission throughout whereas the bio-responsive derivatives initially displayed very low fluorescence. All three bio-responsive derivatives switched on within tumours at time points consistent with their conjugated targeting groups. cRGD and iRGD conjugates both had effective tumour turn-on in the first hour, though the cRGD derivative had superior specificity for tumour over the iRGD conjugate. The pegylated derivative had similar switch-on characteristics but over a much longer period, taking 9 h before a significant emission was observable from the tumour. Evidence for in vivo active tumour targeting was obtained for the best performing cRGD bio-responsive NIR-AZA derivative from competitive binding studies. Overall, this cRGD-conjugate has the potential to overcome the inherent drawback of targeted always-on fluorophores requiring prolonged clearance times and shows excellent potential for clinical translation for intraoperative use in fluorescence guided tumour resections.
Collapse
Affiliation(s)
- Dan Wu
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Harrison C Daly
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Marco Grossi
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| | - Emer Conroy
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - Bo Li
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - William M Gallagher
- School of Biomolecular and Biomedical Science , Conway Institute, University College Dublin , Belfield , Dublin 4 , Ireland
| | - Robert Elmes
- Department of Chemistry , Maynooth University Human Health Institute , Maynooth University , Maynooth , Ireland
| | - Donal F O'Shea
- Department of Chemistry , RCSI , 123 St. Stephen's Green , Dublin 2 , Ireland .
| |
Collapse
|
2694
|
Pristimerin induces apoptosis and autophagy via activation of ROS/ASK1/JNK pathway in human breast cancer in vitro and in vivo. Cell Death Discov 2019; 5:125. [PMID: 31396402 PMCID: PMC6680048 DOI: 10.1038/s41420-019-0208-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women, and progress toward long-term survival has stagnated. Pristimerin, a natural quinonemethide triterpenoid, exhibits potential anti-tumor effects on various cancers. However, the underlying mechanism remains poorly understood. In this study, we found that pristimerin reduced the viability of breast cancer cells in vitro and the growth of xenografts in vivo, and these reductions were accompanied by thioredoxin-1 (Trx-1) inhibition and ASK1 and JNK activation. The results showed that pristimerin inhibited cell cycle progression and triggered cell apoptosis and autophagy. Furthermore, we found that the generation of reactive oxygen species (ROS) was a critical mediator in pristimerin-induced cell death. Enhanced ROS generation by pristimerin activated the ASK1/JNK signaling pathway. Inhibition of ROS with N-acetyl cysteine (NAC) significantly decreased pristimerin-induced cell death by inhibiting the phosphorylation of ASK1 and JNK. Taken together, these results suggest a critical role for the ROS/ASK1/JNK pathway in the anticancer activity of pristimerin.
Collapse
|
2695
|
Wang CCN, Li CY, Cai JH, Sheu PCY, Tsai JJP, Wu MY, Li CJ, Hou MF. Identification of Prognostic Candidate Genes in Breast Cancer by Integrated Bioinformatic Analysis. J Clin Med 2019; 8:1160. [PMID: 31382519 PMCID: PMC6723760 DOI: 10.3390/jcm8081160] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the most common malignancies. However, the molecular mechanisms underlying its pathogenesis remain to be elucidated. The present study aimed to identify the potential prognostic marker genes associated with the progression of breast cancer. Weighted gene coexpression network analysis was used to construct free-scale gene coexpression networks, evaluate the associations between the gene sets and clinical features, and identify candidate biomarkers. The gene expression profiles of GSE48213 were selected from the Gene Expression Omnibus database. RNA-seq data and clinical information on breast cancer from The Cancer Genome Atlas were used for validation. Four modules were identified from the gene coexpression network, one of which was found to be significantly associated with patient survival time. The expression status of 28 genes formed the black module (basal); 18 genes, dark red module (claudin-low); nine genes, brown module (luminal), and seven genes, midnight blue module (nonmalignant). These modules were clustered into two groups according to significant difference in survival time between the groups. Therefore, based on betweenness centrality, we identified TXN and ANXA2 in the nonmalignant module, TPM4 and LOXL2 in the luminal module, TPRN and ADCY6 in the claudin-low module, and TUBA1C and CMIP in the basal module as the genes with the highest betweenness, suggesting that they play a central role in information transfer in the network. In the present study, eight candidate biomarkers were identified for further basic and advanced understanding of the molecular pathogenesis of breast cancer by using co-expression network analysis.
Collapse
Affiliation(s)
- Charles C N Wang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Chia Ying Li
- Department of Surgery, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Jia-Hua Cai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Phillip C-Y Sheu
- Department of EECS and BME, University of California, Irvine, CA 92697, USA
| | - Jeffrey J P Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research,Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung 807, Taiwan.
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- National Sun Yat-Sen University-Kaohsiung Medical University Joint Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- National Chiao Tung University-Kaohsiung Medical University Joint Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
2696
|
Allocca G, Hughes R, Wang N, Brown HK, Ottewell PD, Brown NJ, Holen I. The bone metastasis niche in breast cancer-potential overlap with the haematopoietic stem cell niche in vivo. J Bone Oncol 2019; 17:100244. [PMID: 31236323 PMCID: PMC6582079 DOI: 10.1016/j.jbo.2019.100244] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Bone metastasis is one of the most common complications of advanced breast cancer. During dissemination to bone, breast cancer cells locate in a putative 'metastatic niche', a microenvironment that regulates the colonisation, maintenance of tumour cell dormancy and subsequent tumour growth. The precise location and composition of the bone metastatic niche is not clearly defined. We have used in vivo models of early breast cancer dissemination to provide novel evidence that demonstrates overlap between endosteal, perivascular, HSC and the metastatic niche in bone. METHODS Estrogen Receptor (ER) +ve and -ve breast cancer cells were labelled with membrane dyes Vybrant-DiD and Vybrant-CM-DiI and injected via different routes in BALBc/nude mice of different ages. Two-photon microscopy was used to detect and quantitate tumour cells and map their location within the bone microenvironment as well as their distance to the nearest bone surface compared to the nearest other tumour cell. To investigate whether the metastatic niche overlapped with the HSC niche, animals were pre-treated with the CXCR4 antagonist AMD3100 to mobilise hematopoietic (HSCs) prior to injection of breast cancer cells. RESULTS Breast cancer cells displayed a characteristic pattern of homing in the long bones, with the majority of tumour cells seeded in the trabecular regions, regardless of the route of injection, cell-line characteristics (ER status) or animal age. Breast cancer cells located in close proximity to the nearest bone surface and the average distance between individual tumour cells was higher than their distance to bone. Mobilisation of HSCs from the niche to the circulation prior to injection of cell lines resulted in increased numbers of tumour cells disseminated in trabecular regions. CONCLUSION Our data provide evidence that homing of breast cancer cells is independent of their ER status and that the breast cancer bone metastasis niche is located within the trabecular region of bone, an area rich in osteoblasts and microvessels. The increased number of breast cancer cells homing to bone after mobilisation of HSCs suggests that the HSC and the bone metastasis niche overlap.
Collapse
Key Words
- ANOVA, Analysis of variance
- Animal models
- Bone metastasis
- Breast cancer
- CTC, Circulating tumour cell
- DAPI, 4′,6-diamidino-2-phenylindole
- DTC, Disseminated tumour cell
- EDTA, Ethylenediaminetetraacetic acid
- ER, Estrogen Receptor
- FBS, Foetal bovine serum
- GFP, Green fluorescent protein
- HSC, Hematopoietic stem cell
- Hematopoietic stem cell
- IC, Intra cardiac
- IV, Intra venous
- Luc2, Luciferase2
- OVX, Ovariectomy
- ROI, Region of interest
- TSP-1, thrombospondin-1
- µCT, Microcomputed tomography
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ingunn Holen
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK
| |
Collapse
|
2697
|
Blundon MA, Dasgupta S. Metabolic Dysregulation Controls Endocrine Therapy-Resistant Cancer Recurrence and Metastasis. Endocrinology 2019; 160:1811-1820. [PMID: 31157867 PMCID: PMC6620757 DOI: 10.1210/en.2019-00097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/24/2019] [Indexed: 01/16/2023]
Abstract
Cancer recurrence and metastasis involves many biological interactions, such as genetic, transcription, environmental, endocrine signaling, and metabolism. These interactions add a complex understanding of cancer recurrence and metastatic progression, delaying the advancement in therapeutic opportunities. We highlight the recent advances on the molecular complexities of endocrine-related cancers, focusing on breast and prostate cancer, and briefly review how endocrine signaling and metabolic programs can influence transcriptional complexes for metastasis competence. Nuclear receptors and transcriptional coregulators function as molecular nodes for the crosstalk between endocrine signaling and metabolism that alter downstream gene expression important for tumor progression and metastasis. This exciting regulatory axis may provide insights to the development of cancer therapeutics important for these desensitized endocrine-dependent cancers.
Collapse
Affiliation(s)
- Malachi A Blundon
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Correspondence: Subhamoy Dasgupta, PhD, Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14263. E-mail:
| |
Collapse
|
2698
|
Yang N, Wang C, Wang J, Wang Z, Huang D, Yan M, Kamran M, Liu Q, Xu B. Aurora kinase A stabilizes FOXM1 to enhance paclitaxel resistance in triple-negative breast cancer. J Cell Mol Med 2019; 23:6442-6453. [PMID: 31359594 PMCID: PMC6714217 DOI: 10.1111/jcmm.14538] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has a relatively poor outcome. Acquired chemoresistance is a major clinical challenge for TNBC patients. Previously, we reported that kinase-dead Aurora kinase A (Aurora-A) could effectively transactivate the FOXM1 promoter. Here, we demonstrate an additional pathway through which Aurora-A stabilizes FOXM1 by attenuating its ubiquitin in TNBC. Specifically, Aurora-A stabilizes FOXM1 in late M phase and early G1 phase of the cell cycle, which promotes proliferation of TNBC cells. Knock-down of Aurora-A significantly suppresses cell proliferation in TNBC cell lines and can be rescued by FOXM1 overexpression. We observe that paclitaxel-resistant TNBC cells exhibit high expression of Aurora-A and FOXM1. Overexpression of Aurora-A offers TNBC cells an additional growth advantage and protection against paclitaxel. Moreover, Aurora-A and FOXM1 could be simultaneously targeted by thiostrepton. Combination of thiostrepton and paclitaxel treatment reverses paclitaxel resistance and significantly inhibits cell proliferation. In conclusion, our study reveals additional mechanism through which Aurora-A regulates FOXM1 and provides a new therapeutic strategy to treat paclitaxel-resistant triple-negative breast cancer.
Collapse
Affiliation(s)
- Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chang Wang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jian Wang
- Department of Pathology, GanZhou Municipal People's Hospital, NanChang University, GanZhou, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Di Huang
- Department of Breast Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Muhammad Kamran
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - BangLao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
2699
|
Stompor M, Świtalska M, Bajek A, Wietrzyk J. Influence of amide versus ester linkages on the anticancer properties of the new flavone-biotin conjugates. ACTA ACUST UNITED AC 2019; 74:193-200. [PMID: 31100057 DOI: 10.1515/znc-2018-0195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/16/2019] [Indexed: 11/15/2022]
Abstract
Novel biotinylated C-6 substituted flavones were synthesised by a one-step method that connects biotin to 6-hydroxyflavone and 6-aminoflavone by esterification and amidation of hydroxyl and amino groups, respectively. The obtained compounds, 6-O-biotinylflavone and 6-biotinylamidoflavone, are the bifunctional molecules composed of a flavone moiety as a fluorescent reporter and biotin as a cancer-targeting unit. Antiproliferative activity was evaluated using SRB assays in MCF-7, MCF-10A, HepG2, MDA-MB-231, 4T1, and Balb/3T3 cell lines. In vitro evaluation revealed that compounds with biotin moiety displayed better cell selectivity between the cancer and normal cells than the parental substrates. These results indicate that anticancer effect is not related to the position of biotin moiety, but it is related to the presence of ester or amide bond. 6-O-Biotinylflavone was more active than 6-hydroxyflavone against human breast (MDA-MB-231) and liver (HepG2) cancer cells with IC50 (concentration of tested agent that inhibits proliferation of the cell population by 50%) values equal to 78.5 ± 18.8 μM and 133.2 ± 14.2 μM, respectively. Non biotinylated 6-aminoflavone was more active than 6-biotinylamidoflavone against all tested cell lines, with IC50 values between 34.3 ± 9.1 μM (4T1) and 173.86 ± 24.3 μM (MCF-7).
Collapse
Affiliation(s)
- Monika Stompor
- Faculty of Medicine, University of Rzeszów, Warzywna 1a, 35-310 Rzeszów, Poland, Phone: +48 17 8516880
| | - Marta Świtalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Oncology, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland
| | - Agata Bajek
- Department of Industrial and Materials Chemistry, Faculty of Chemistry, Rzeszow University of Technology, Al. Powstańców Warszawy 6, 35-959 Rzeszow, Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Oncology, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland
| |
Collapse
|
2700
|
Wang J, Li Y, Fu W, Zhang Y, Jiang J, Zhang Y, Qi X. Prognostic nomogram based on immune scores for breast cancer patients. Cancer Med 2019; 8:5214-5222. [PMID: 31342666 PMCID: PMC6718583 DOI: 10.1002/cam4.2428] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/29/2019] [Accepted: 07/05/2019] [Indexed: 12/29/2022] Open
Abstract
Background Increased attention has been focused on cancer immunity gene signature. However, the threshold of immune scores to predict disease‐free survival (DFS) and overall survival (OS) in breast cancer has not yet been defined. This study aimed to explore the association of immune scores with prognosis and build a clinical nomogram to predict the survival of patients with breast cancer. Methods A total of 986 subjects were analyzed, and clinicopathological characteristics and immune scores were obtained from the TCGA database. Cox proportional hazards regression model was used to estimate the adjusted hazard ratios (HRs). Based on results of multivariate analysis, nomograms were built. The models were subjected to bootstrap internal validation. The predictive accuracy and discriminative ability were measured by concordance index (C‐index) and the calibration curve. Results The patients were divided into three subgroups according to their immune scores. We found that compared with patients with low immune scores, those with intermediate and high immune scores had significantly improved DFS (HR and 95% confidence interval [CI]: 0.439 [0.242‐0.799], 0.541 [0.343‐0.855], respectively), whereas only intermediate immune scores significantly indicated better OS (HR and 95% CI: 0.385 [0.163‐0.910]). The C‐index for DFS and OS prediction was 0.723 (95% CI, 0.661‐0.785) and 0.800 (95% CI, 0.724‐0.877), respectively. The calibration curves for probability of 3‐ and 5‐year DFS showed significant agreement between nomogram predictions and the actual observations. Conclusions High and/or intermediate immune scores are significantly correlated with better DFS and OS in patients with breast cancer. Moreover, the nomograms for predicting prognosis may help to estimate the survival of patients.
Collapse
Affiliation(s)
- Ju Wang
- Department of Epidemiology and Biostatistics, West China School of Public Health, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yanling Li
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Wenying Fu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Ye Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Jun Jiang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P.R. China
| |
Collapse
|