14401
|
Xu J, Liu X, Yang S, Shi Y. Apatinib Monotherapy or Combination Therapy for Non-Small Cell Lung Cancer Patients With Brain Metastases. Oncol Res 2019; 28:127-133. [PMID: 31610827 PMCID: PMC7851530 DOI: 10.3727/096504019x15707896762251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Apatinib, an oral small molecular receptor tyrosine kinase inhibitor (TKI) developed first in China, exerts antiangiogenic and antineoplastic function through selectively binding and inhibiting vascular endothelial growth factor receptor 2 (VEGFR-2). In this study, we aimed to explore the efficacy and safety profile of apatinib monotherapy, or combined with chemotherapy or endothelial growth factor receptor (EGFR)-TKI in heavily pretreated non-small cell lung cancer (NSCLC) patients with brain metastases. We performed a retrospective analysis for relapsed NSCLC patients with brain metastases from our institute, who received apatinib (250 mg or 500 mg p.o. qd) monotherapy, or combination with EGFR-TKI or chemotherapy as second or more line systemic therapy until disease progression or unacceptable toxicity occurred. The objective response rate (ORR), disease control rate (DCR), median progression-free survival (mPFS), median overall survival (mOS), and safety were analyzed. A total of 26 eligible patients were included: 24 patients diagnosed with adenocarcinoma, 2 with squamous carcinoma, and 14 patients harboring EGFR sensitizing mutations. The mPFS and mOS were 4.93 (range, 0.27–32.91; 95% CI 3.64–6.22) and 14.70 (range, 0.27–32.91; 95% CI 0.27–43.60) months for the whole group. The ORR and DCR were 7.7% (2/26) and 69.2% (18/26) for the entire lesions, and 7.7% (2/26) and 79.6% (20/26) for brain metastases, respectively. Compared with patients who received apatinib monotherapy, patients who received apatinib combination treatment had more favorable mPFS (11.77 vs. 2.27 months, p < 0.05) and mOS (24.03 vs. 6.07 months, p < 0.05). Treatment-related toxicities were tolerable including grade 1/2 hypertension, hand-and-foot syndrome, fatigue, nausea, liver dysfunction, myelosuppression, skin rash, and palpitation. In conclusion, apatinib exhibited high activity and good tolerance for NSCLC patients with brain metastasis, and it might become a potential choice for metastatic brain tumors in NSCLC patients.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP.R. China
| | - Xiaoyan Liu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP.R. China
| | - Sheng Yang
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP.R. China
| | - Yuankai Shi
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP.R. China
| |
Collapse
|
14402
|
Sanomachi T, Suzuki S, Togashi K, Sugai A, Seino S, Okada M, Yoshioka T, Kitanaka C, Yamamoto M. Spironolactone, a Classic Potassium-Sparing Diuretic, Reduces Survivin Expression and Chemosensitizes Cancer Cells to Non-DNA-Damaging Anticancer Drugs. Cancers (Basel) 2019; 11:cancers11101550. [PMID: 31614999 PMCID: PMC6826935 DOI: 10.3390/cancers11101550] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/22/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Spironolactone, a classical diuretic drug, is used to treat tumor-associated complications in cancer patients. Spironolactone was recently reported to exert anti-cancer effects by suppressing DNA damage repair. However, it currently remains unclear whether spironolactone exerts combinational effects with non-DNA-damaging anti-cancer drugs, such as gemcitabine and epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). Using the cancer cells of lung cancer, pancreatic cancer, and glioblastoma, the combinational effects of spironolactone with gemcitabine and osimertinib, a third-generation EGFR-TKI, were examined in vitro with cell viability assays. To elucidate the underlying mechanisms, we investigated alterations induced in survivin, an anti-apoptotic protein, by spironolactone as well as the chemosensitization effects of the suppression of survivin by YM155, an inhibitor of survivin, and siRNA. We also examined the combinational effects in a mouse xenograft model. The results obtained revealed that spironolactone augmented cell death and the suppression of cell growth by gemcitabine and osimertinib. Spironolactone also reduced the expression of survivin in these cells, and the pharmacological and genetic suppression of survivin sensitized cells to gemcitabine and osimertinib. This combination also significantly suppressed tumor growth without apparent adverse effects in vivo. In conclusion, spironolactone is a safe candidate drug that exerts anti-cancer effects in combination with non-DNA-damaging drugs, such as gemcitabine and osimertinib, most likely through the suppression of survivin.
Collapse
Affiliation(s)
- Tomomi Sanomachi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Keita Togashi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
- Department of Ophthalmology and Visual Sciences, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Asuka Sugai
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Takashi Yoshioka
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
- Research Institute for Promotion of Medical Sciences, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | - Masahiro Yamamoto
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| |
Collapse
|
14403
|
Wang J, Sun P, Wang Q, Zhang P, Wang Y, Zi C, Wang X, Sheng J. (-)-Epigallocatechin-3-gallate derivatives combined with cisplatin exhibit synergistic inhibitory effects on non-small-cell lung cancer cells. Cancer Cell Int 2019; 19:266. [PMID: 31636509 PMCID: PMC6791019 DOI: 10.1186/s12935-019-0981-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/27/2019] [Indexed: 01/04/2023] Open
Abstract
Background Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide. The inhibition of epidermal growth factor receptor (EGFR) signaling by tyrosine kinase inhibitors or monoclonal antibodies plays a key role in NSCLC treatment. Unfortunately, these treatment strategies are limited by eventual resistance and cell lines with differential EGFR status. Therefore, new therapeutic strategies for NSCLC are urgently required. Methods To improve the stability and absorption of (−)-epigallocatechin-3-gallate (EGCG), we synthesized a series of EGCG derivatives. The antitumor activities of EGCG derivatives with or without cisplatin were investigated in vitro and vivo. Cell proliferation, cell cycle distribution and apoptosis were measured in NSCLC cell lines and in vivo in a NCI-H441 xenograft model. Results We found that the EGCG derivatives inhibited cell viability and colony formation, caused cell cycle redistribution, and induced apoptosis. More importantly, the combination of the EGCG derivative and cisplatin led to increased growth inhibition, caused cell cycle redistribution, and enhanced the apoptosis rate compared to either compound alone. Consistent with the experiments in vitro, EGCG derivatives plus cisplatin significantly reduced tumor growth. Conclusions The combination treatment was found to inhibit the EGFR signaling pathway and decrease the expression of p-EGFR, p-AKT, and p-ERK in vitro and vivo. Our results suggest that compound 3 is a novel potential compound for NSCLC patients.
Collapse
Affiliation(s)
- Jing Wang
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,2College of Science, Yunnan Agricultural University, Kunming, 650201 Yunnan China.,3College of Food Science and Technology, Yunnan Agricultural University, Kunming, Yunnan China
| | - Peiyuan Sun
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,3College of Food Science and Technology, Yunnan Agricultural University, Kunming, Yunnan China
| | - Qi Wang
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China
| | - Pan Zhang
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China
| | - Yuna Wang
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,3College of Food Science and Technology, Yunnan Agricultural University, Kunming, Yunnan China
| | - Chengting Zi
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,2College of Science, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Xuanjun Wang
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,2College of Science, Yunnan Agricultural University, Kunming, 650201 Yunnan China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, Yunnan China
| | - Jun Sheng
- 1Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, Yunnan China.,2College of Science, Yunnan Agricultural University, Kunming, 650201 Yunnan China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, Yunnan China
| |
Collapse
|
14404
|
Oliveira Pinho J, Matias M, Gaspar MM. Emergent Nanotechnological Strategies for Systemic Chemotherapy against Melanoma. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1455. [PMID: 31614947 PMCID: PMC6836019 DOI: 10.3390/nano9101455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Abstract
Melanoma is an aggressive form of skin cancer, being one of the deadliest cancers in the world. The current treatment options involve surgery, radiotherapy, targeted therapy, immunotherapy and the use of chemotherapeutic agents. Although the last approach is the most used, the high toxicity and the lack of efficacy in advanced stages of the disease have demanded the search for novel bioactive molecules and/or efficient drug delivery systems. The current review aims to discuss the most recent advances on the elucidation of potential targets for melanoma treatment, such as aquaporin-3 and tyrosinase. In addition, the role of nanotechnology as a valuable strategy to effectively deliver selective drugs is emphasized, either incorporating/encapsulating synthetic molecules or natural-derived compounds in lipid-based nanosystems such as liposomes. Nanoformulated compounds have been explored for their improved anticancer activity against melanoma and promising results have been obtained. Indeed, they displayed improved physicochemical properties and higher accumulation in tumoral tissues, which potentiated the efficacy of the compounds in pre-clinical experiments. Overall, these experiments opened new doors for the discovery and development of more effective drug formulations for melanoma treatment.
Collapse
Affiliation(s)
- Jacinta Oliveira Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
14405
|
Hendrickson PG, Olson M, Luetkens T, Weston S, Han T, Atanackovic D, Fine GC. The promise of adoptive cellular immunotherapies in hepatocellular carcinoma. Oncoimmunology 2019; 9:1673129. [PMID: 32002284 PMCID: PMC6959455 DOI: 10.1080/2162402x.2019.1673129] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/22/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide. Current systemic therapies result only in modest benefits and new therapeutic options are critically needed. Some patients show promising clinical responses to immune checkpoint inhibitors, however, additional immunotherapeutic approaches, such as adoptive cell therapies (ACT), need to be developed. This review summarizes recent ACT studies and discusses the promise and obstacles of this approach. We further discuss ways of improving the efficacy of ACT in HCC including the use of combination therapies and locoregional delivery methods.
Collapse
Affiliation(s)
- Peter G. Hendrickson
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Michael Olson
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Tim Luetkens
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Siani Weston
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Tiffany Han
- Department of Radiology, Norwalk Hospital, Norwalk, CT, USA
| | - Djordje Atanackovic
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
14406
|
Breast Cancer Cells and PD-1/PD-L1 Blockade Upregulate the Expression of PD-1, CTLA-4, TIM-3 and LAG-3 Immune Checkpoints in CD4 + T Cells. Vaccines (Basel) 2019; 7:vaccines7040149. [PMID: 31614877 PMCID: PMC6963740 DOI: 10.3390/vaccines7040149] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype, and it exhibits resistance to common breast cancer therapies. Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1 (PD-1) and its ligand, PD-L1, have been approved to treat various cancers. However, the therapeutic efficacy of targeting PD-1/PD-L1 axis in breast cancer is under clinical investigation. In addition, the mechanisms of action of drugs targeting PD-1 and PD-L1 have not been fully elucidated. In this study, we investigated the effect of human TNBC cell lines, MDA-MB-231 and MDA-MB-468, and the non-TNBC cell line, MCF-7, on the expression of immune checkpoints (ICs) on CD4+ T cell subsets, including regulatory T cells (Tregs), using a co-culture system. We also examined the effect of blocking PD-1 or PD-L1 separately and in combination on IC expression by CD4+ T cell subsets. We found that breast cancer cells upregulate the expression of ICs including PD-1, cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and lymphocyte activation gene-3 (LAG-3) in CD4+ T cell subsets. We also found that the co-blockade of PD-1 and PD-L1 further upregulates the co-expression of TIM-3 and LAG-3 on CD4+CD25+ T cells and CD4+CD25+FoxP3+Helios+ Tregs in the presence of TNBC cells, but not in non-TNBC cells. Our results indicate the emergence of compensatory inhibitory mechanisms, most likely mediated by Tregs and activated non-Tregs, which could lead to the development of TNBC resistance against PD-1/PD-L1 blockade.
Collapse
|
14407
|
Changing trends in liver cancer incidence by race/ethnicity and sex in the US: 1992-2016. Cancer Causes Control 2019; 30:1377-1388. [PMID: 31606852 DOI: 10.1007/s10552-019-01237-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/01/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Liver cancer incidence continues to increase while incidence of most other cancers is decreasing. We analyze recent and long-term trends of US liver cancer incidence by race/ethnicity and sex to best understand where to focus preventive efforts. METHODS Liver cancer incidence rates from 1992 to 2016 were obtained from the Surveillance, Epidemiology, and End Results registry. Delay-adjusted age-standardized incidence trends by race/ethnicity and sex were analyzed using joinpoint regression. Age-specific incidence was analyzed using age-period-cohort models. Hepatitis C seroprevalence by cohort was calculated using National Health and Nutrition Examination Survey data. RESULTS Liver cancer incidence has peaked in males and Asian or Pacific Islanders. Hispanic males, a high-incidence population, are experiencing a decrease in incidence, although not yet statistically significant. In contrast, incidence continues to increase in females, although at lower rates than in the 1990s, and American Indian/Alaska Natives (AI/ANs). Liver cancer incidence continues to be higher in males. Non-Hispanic Whites have the lowest incidence among racial/ethnic groups. Trends largely reflect differences in incidence by birth-cohort, which increased considerably, particularly in males, for those born around the 1950s, and continues to increase in females and AI/ANs. The patterns in males are likely driven by cohort variations in Hepatitis C infection. CONCLUSIONS Liver cancer incidence appears to have peaked among males. However, important differences in liver cancer trends by race/ethnicity and sex remain, highlighting the need for monitoring trends across different groups. Preventive interventions should focus on existing liver cancer disparities, targeting AI/ANs, females, and high-incidence groups.
Collapse
|
14408
|
Modeling drug exposure in rodents using e-cigarettes and other electronic nicotine delivery systems. J Neurosci Methods 2019; 330:108458. [PMID: 31614162 DOI: 10.1016/j.jneumeth.2019.108458] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/05/2019] [Accepted: 10/06/2019] [Indexed: 12/25/2022]
Abstract
Smoking tobacco products is the leading cause of preventable death worldwide. Coordinated efforts have successfully reduced tobacco cigarette smoking in the United States; however, electronic cigarettes (e-cigarette) and other electronic nicotine delivery systems (ENDS) recently have replaced traditional cigarettes for many users. While the clinical risks associated with long-term ENDS use remain unclear, advancements in preclinical rodent models will enhance our understanding of their overall health effects. This review examines the peripheral and central effects of ENDS-mediated exposure to nicotine and other drugs of abuse in rodents and evaluates current techniques for implementing ENDS in preclinical research.
Collapse
|
14409
|
Zhang Z, Tang H, Chen P, Xie H, Tao Y. Demystifying the manipulation of host immunity, metabolism, and extraintestinal tumors by the gut microbiome. Signal Transduct Target Ther 2019; 4:41. [PMID: 31637019 PMCID: PMC6799818 DOI: 10.1038/s41392-019-0074-5] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
The trillions of microorganisms in the gut microbiome have attracted much attention recently owing to their sophisticated and widespread impacts on numerous aspects of host pathophysiology. Remarkable progress in large-scale sequencing and mass spectrometry has increased our understanding of the influence of the microbiome and/or its metabolites on the onset and progression of extraintestinal cancers and the efficacy of cancer immunotherapy. Given the plasticity in microbial composition and function, microbial-based therapeutic interventions, including dietary modulation, prebiotics, and probiotics, as well as fecal microbial transplantation, potentially permit the development of novel strategies for cancer therapy to improve clinical outcomes. Herein, we summarize the latest evidence on the involvement of the gut microbiome in host immunity and metabolism, the effects of the microbiome on extraintestinal cancers and the immune response, and strategies to modulate the gut microbiome, and we discuss ongoing studies and future areas of research that deserve focused research efforts.
Collapse
Affiliation(s)
- Ziying Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
- Department of Oncology, Third Xiangya Hospital, Central South University, 410013 Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Peng Chen
- Department of Urology, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Hui Xie
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| |
Collapse
|
14410
|
Li F, Li X, Li Z, Ji W, Lu S, Xia W. βKlotho is identified as a target for theranostics in non-small cell lung cancer. Theranostics 2019; 9:7474-7489. [PMID: 31695781 PMCID: PMC6831461 DOI: 10.7150/thno.35582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a great challenge, calling for the identification of novel molecular targets with diagnostic/therapeutic value. Here, we sought to characterize the expression of βKlotho and its anti-tumor roles in NSCLC. Methods: The expression of βKlotho was examined in NSCLC cells and tissues by western blot, qRT-PCR and immunohistochemistry staining respectively. Biological roles of βKlotho were revealed by a series of functional in vitro and in vivo studies. Serum βKlotho concentrations of patients were measured using specific ELISA methods. Results: Serum βKlotho concentrations of NSCLC patients were significantly lower than the control group. Moreover, βKlotho expression was negatively associated with lymph node metastasis, overall survival and progression-free survival. Overexpression of βKlotho or exogenous βKlotho administration inhibited the proliferation and migration of NSCLC cells, accompanied by induction of apoptosis, G1 to S phase arrest, and inactivation of ERK1/2, AKT and STAT3 signaling. Furthermore, βKlotho overexpression inhibited NSCLC tumor growth in vivo. Conclusions: βKlotho serves as a novel target for theranostics in NSCLC, which has potential clinical applications in the future.
Collapse
Affiliation(s)
- Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiyao Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14411
|
Slade AN, Dahman B, Chang MG. Racial differences in the PSA bounce in predicting prostate cancer outcomes after brachytherapy: Evidence from the Department of Veterans Affairs. Brachytherapy 2019; 19:6-12. [PMID: 31611160 DOI: 10.1016/j.brachy.2019.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/14/2019] [Accepted: 08/19/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE African American men have historically had poorer prostate cancer biochemical and survival outcomes than Caucasians. However, emerging data suggest nononcologic factors drive much of this disparity. Prior evidence has suggested an association between a transient prostate specific antigen (PSA) bounce and improved biochemical control. However, racial differences in this relationship have remained relatively unexplored. METHODS AND MATERIALS We identified 4477 men treated for low- or intermediate-risk prostate cancer within the U.S. Department of Veterans Affairs (VA) from 2000 to 2010 with brachytherapy alone or in combination with external beam radiotherapy without androgen deprivation. Longitudinal PSA data were used to define to biochemical failure and PSA bounce. Cox proportional hazard models were used explore racial differences in the relationship between the PSA bounce and time to biochemical failure. RESULTS Thirty-one percent of our sample experienced a PSA bounce, with African Americans more likely to experience a bounce (42%) compared with Caucasians (29%); p < 0.001. Despite this, African Americans had a higher likelihood of biochemical failure (hazard ratio [HR] 1.4; p = 0.006). However, African American men experiencing a PSA bounce were less likely to experience a biochemical failure (HR = 0.64; p = 0.046), whereas this relationship was not statistically significant for Caucasians (HR = 0.78; p = 0.092). On multivariate analysis, African Americans receiving brachytherapy alone were most sensitive to the protective benefit of the PSA bounce (HR = 0.64). CONCLUSIONS A PSA bounce was associated with improved biochemical control among patients receiving brachytherapy as part of their treatment for low- or intermediate-risk prostate cancer at the VA. African American men treated with brachytherapy had a particularly pronounced biochemical control benefit of a PSA bounce.
Collapse
Affiliation(s)
- Alexander N Slade
- Department of Radiation Oncology, Virginia Commonwealth University Health System, Massey Cancer Center, Richmond, VA.
| | - Bassam Dahman
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, VA
| | - Michael G Chang
- Department of Radiation Oncology, Virginia Commonwealth University Health System, Massey Cancer Center, Richmond, VA; Department of Radiation Oncology, Hunter Holmes McGuire VA Medical Center, Richmond VA
| |
Collapse
|
14412
|
Swords DS, Mulvihill SJ, Brooke BS, Firpo MA, Scaife CL. Size and Importance of Socioeconomic Status-Based Disparities in Use of Surgery in Nonadvanced Stage Gastrointestinal Cancers. Ann Surg Oncol 2019; 27:333-341. [DOI: 10.1245/s10434-019-07922-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Indexed: 02/06/2023]
|
14413
|
Chen R, Gong Y, Zou D, Wang L, Yuan L, Zhou Q. Correlation between subsets of tumor-infiltrating immune cells and risk stratification in patients with cervical cancer. PeerJ 2019; 7:e7804. [PMID: 31616592 PMCID: PMC6791348 DOI: 10.7717/peerj.7804] [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: 06/14/2019] [Accepted: 08/31/2019] [Indexed: 12/15/2022] Open
Abstract
Aim To investigate the correlation between clinicopathological features and risk stratification in cervical cancer patients, and evaluate the feasibility of tumor-infiltrating immune cells as prognostic biomarkers in clinical practice. Methods CD3+ tumor infiltrating T cells (TILs), CD45RO+ TILs, CD4+ TILs, CD8+ TILs, FOXP3+ TILs (regulatory T cells, Tregs), CD68+ tumor associated macrophages (TAMs), CD163+ TAMs, and PD-L1+ tumor cells were immunostained in formalin-fixed paraffin-embedded (PPFE) tissues from 96 cervical cancer patients. Immunostaining density and other clinicopathological features such as age, FIGO stage, histopathologic type, Ki67 index, HPV status, lymhovasular invasion status (LVI), lymph node metastasis, tumor size, stromal invasion status, surgical margin status, and parametrial invasion, were evaluated for their roles in risk stratification of cervical cancer patients. Results The results showed that significant differences of lymph node metastasis (p = 0.003), surgical margin status (p = 0.020), and stromal invasion status (p = 0.004) existed between lVI(−) and LVI(+) patients. CD3+ TILs in the central tumor area (p = 0.010), CD4+ TILs in the central tumor area (p = 0.045), CD8 + TILs in the central tumor area (p = 0.033), and CD8+ TILs in the invasive margin area (p = 0.004) showed significant differences between lVI(−) and LVI(+) patients. When patients were grouped by status of lymph node metastasis, significant differences of FIGO stage (p = 0.005), LVI status (p = 0.003), CD3+ TILs in the central tumor area (p = 0.045), CD45RO+ TILs in the central tumor area (p = 0.033), and CD45RO+ TILs in the invasive margin area (p = 0.028) were also observed. After the patients were stratified into low-, intermediate-, and high risk groups, significant differences of FIGO stage (p = 0.018), status of lymph node metastasis (p = 0.000), LVI status (p = 0.000), parametrial invasion status (p=0.012), stromal invasion status (p = 0.000), tumor growth pattern (p = 0.015) and tumor size (p = 0.000) were identified among 3 groups of patients, while only CD45RO+ TILs in the invasive margin area (p = 0.018) and FOXP3+ TILs in the central tumor area (p = 0.009) were statistically different among three groups of patients. Spearman’s correlation analysis demonstrated that FIGO stage, LVI status, status of lymph node metastasis, parametrial invasion, stromal invasion status, and tumor size positively correlated with risk stratification (P = 0.005, 0.020, 0.000, 0.022, 0.000, and 0.000 respectively), while CD45RO+ TILs in the invasive margin area and FOXP3+ TILs in the central tumor area showed statistically negative correlation with risk stratification (P = 0.031, 0.009 respectively). Conclusion Our study suggested that CD45RO+ TILs in the invasive margin area and FOXP3+ TILs in the central tumor area might be useful biomarkers for risk stratification in cervical cancer patients. Large cohort studies of cervical cancer patients are required to validate our hypothesis.
Collapse
Affiliation(s)
- Rui Chen
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Yi Gong
- Department of Hematology-Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Dongling Zou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Lifeng Wang
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Li Yuan
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Qi Zhou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
14414
|
Bing Z, Yao Y, Xiong J, Tian J, Guo X, Li X, Zhang J, Shi X, Zhang Y, Yang K. Novel Model for Comprehensive Assessment of Robust Prognostic Gene Signature in Ovarian Cancer Across Different Independent Datasets. Front Genet 2019; 10:931. [PMID: 31681404 PMCID: PMC6798149 DOI: 10.3389/fgene.2019.00931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Different analytical methods or models can often find completely different prognostic biomarkers for the same cancer. In the study of prognostic molecular biomarkers of ovarian cancer (OvCa), different studies have reported a variety of prognostic gene signatures. In the current study, based on geometric concepts, the linearity-clustering phase diagram with integrated P-value (LCP) method was used to comprehensively consider three indicators that are commonly employed to estimate the quality of a prognostic gene signature model. The three indicators, namely, concordance index, area under the curve, and level of the hazard ratio were determined via calculation of the prognostic index of various gene signatures from different datasets. As evaluation objects, we selected 13 gene signature models (Cox regression model) and 16 OvCa genomic datasets (including gene expression information and follow-up data) from published studies. The results of LCP showed that three models were universal and better than other models. In addition, combining the three models into one model showed the best performance in all datasets by LCP calculation. The combination gene signature model provides a more reliable model and could be validated in various datasets of OvCa. Thus, our method and findings can provide more accurate prognostic biomarkers and effective reference for the precise clinical treatment of OvCa.
Collapse
Affiliation(s)
- Zhitong Bing
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Yuxiang Yao
- School of Physical Science and Technology, Lanzhou University, Lanzhou, China
| | - Jie Xiong
- Department of Applied Mathematics, Changsha University, Changsha, China
| | - Jinhui Tian
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Xiangqian Guo
- Medical Bioinformatics Institute, School of Basic Medicine, Henan University, Henan, China
| | - Xiuxia Li
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,School of Public Health, Lanzhou University, Lanzhou, China
| | - Jingyun Zhang
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Xiue Shi
- Institute for Evidence Based Rehabilitation Medicine of Gansu Province, Lanzhou, China
| | - Yanying Zhang
- Department of Pharmacology and Toxicology of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kehu Yang
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,Institute for Evidence Based Rehabilitation Medicine of Gansu Province, Lanzhou, China.,Department of Pharmacology and Toxicology of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
14415
|
Contextual Regulation of TGF-β Signaling in Liver Cancer. Cells 2019; 8:cells8101235. [PMID: 31614569 PMCID: PMC6829617 DOI: 10.3390/cells8101235] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is one of the leading causes for cancer-related death worldwide. Transforming growth factor beta (TGF-β) is a pleiotropic cytokine that signals through membrane receptors and intracellular Smad proteins, which enter the nucleus upon receptor activation and act as transcription factors. TGF-β inhibits liver tumorigenesis in the early stage by inducing cytostasis and apoptosis, but promotes malignant progression in more advanced stages by enhancing cancer cell survival, EMT, migration, invasion and finally metastasis. Understanding the molecular mechanisms underpinning the multi-faceted roles of TGF-β in liver cancer has become a persistent pursuit during the last two decades. Contextual regulation fine-tunes the robustness, duration and plasticity of TGF-β signaling, yielding versatile albeit specific responses. This involves multiple feedback and feed-forward regulatory loops and also the interplay between Smad signaling and non-Smad pathways. This review summarizes the known regulatory mechanisms of TGF-β signaling in liver cancer, and how they channel, skew and even switch the actions of TGF-β during cancer progression.
Collapse
|
14416
|
TMEFF2 is a novel prognosis signature and target for endometrial carcinoma. Life Sci 2019; 243:116910. [PMID: 31610211 DOI: 10.1016/j.lfs.2019.116910] [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] [Received: 06/12/2019] [Revised: 09/04/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022]
Abstract
AIMS Tomoregulin-2 (TMEFF2) is a single-pass transmembrane protein whose specific functions and mechanisms in endometrial carcinoma (EC) remain unclear. The aim of this study was to investigate the expression, prognostic role, and potential regulatory mechanisms of TMEFF2 in EC. MATERIALS AND METHODS The expression and prognosis of TMEFF2 in EC were analyzed via bioinformatics and verified by immunohistochemistry and survival analysis. Proliferation, invasion, and migration of EC cells in vitro were assessed by cell functional assays, while epithelial-mesenchymal transition (EMT) markers and key signaling pathway proteins were evaluated by western blotting. KEY FINDINGS The expression of TMEFF2 in EC was significantly higher than that in atypical hyperplasia and normal endometrium, the high expression of TMEFF2 was correlated with advanced stage, poor differentiation, and lymph node metastasis, and also predicted a poor prognosis of EC. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that TMEFF2 and its related genes were enriched in the central nervous system, cell adhesion, signal transduction, and several critical signaling pathways. We also elucidated TMEFF2 networks of kinase, microRNA, and transcription factor targets. In vitro, the proliferation, invasion, and migration abilities of EC cells decreased after TMEFF2 downregulation. Downregulation of TMEFF2 reduced the activation of MAPK and PI3K signaling pathways, and inhibited EMT. SIGNIFICANCE TMEFF2 plays an important role in the initiation, development, and malignant behavior of EC and can be a potential target for early diagnosis and treatment in EC.
Collapse
|
14417
|
Liu P, Kong L, Jin H, Wu Y, Tan X, Song B. Differential secretome of pancreatic cancer cells in serum-containing conditioned medium reveals CCT8 as a new biomarker of pancreatic cancer invasion and metastasis. Cancer Cell Int 2019; 19:262. [PMID: 31632196 PMCID: PMC6788113 DOI: 10.1186/s12935-019-0980-1] [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: 04/07/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Background Pancreatic cancer is a malignancy with a very poor prognosis. The emergence of liquid biopsy is expected to achieve accurate early diagnosis through detection of tumor-derived secreted proteins in the blood. Early diagnosis and treatment of pancreatic cancer could help to improve prognosis. Methods The pretreatment approach of samples can have a major effect on downstream analysis. In this study, we used a pair of homologous pancreatic cancer cell supernatants with different capacities for invasion and metastasis to examine secreted proteins in the conditioned media without the removal of fetal bovine serum, namely through size exclusion chromatography combined with high-abundance protein affinity chromatography to enrich low-concentration protein, followed by mass spectrometry using triple dimethyl labeling. Identification of proteins was performed using an online public database and western blot. Results Mass spectrometry data revealed 77 proteins with quantitative properties, of which 12 proteins had over a 1.5-fold difference (in the supernatant of the highly invasive pancreatic cancer cell line PC-1.0, the expression of 8 proteins were increased and the expression of 4 proteins were decreased). Bioinformatics analysis results showed that CCT8, CTSL, SAA1, IGF2 are secreted via the exosome pathway. According to the literature, with the exception of CCT8, the other three proteins can be detected in blood samples of pancreatic cancer patients, and they can be used as prognostic markers. Western blot results were used to validate consistency with MS results. Conclusion This study found that CCT8 can be used as a liquid biopsy marker to assess the prognosis of pancreatic cancer patients.
Collapse
Affiliation(s)
- Peng Liu
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Lingming Kong
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Haoyi Jin
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Yunhao Wu
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Xiaodong Tan
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Bing Song
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China.,2Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, CF14 4XY UK
| |
Collapse
|
14418
|
Yu Y, Hann SS. Novel Tumor Suppressor lncRNA Growth Arrest-Specific 5 (GAS5) In Human Cancer. Onco Targets Ther 2019; 12:8421-8436. [PMID: 31632088 PMCID: PMC6794681 DOI: 10.2147/ott.s221305] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play crucial regulatory roles in fundamental biological processes, and deregulations of lncRNAs have been linked to numerous human diseases, especially cancers. Of particular interest in this regard is lncRNA GAS5, which is mainly identified as a tumor suppressor in several cancers. GAS5 was significantly low expressed in multiple cancers and was associated with clinic-pathological characteristics and patient survival, indicating a novel potential diagnostic and prognostic biomarker, and a therapeutic target for cancer. Functionally, GAS5 is involved in cell proliferation, metastasis, invasion, apoptosis, epithelial-mesenchymal transition (EMT), and drug resistance, among others, via multiple molecular mechanisms, such as binding to DNA sequences, forming RNA-DNA triplex complex, triggering or suppressing the expression of genes, binding proteins to form chromatin-modifying complex, which activates or represses gene expression, and acting as miRNA sponge to suppress miRNA expression, leading to regulation of miRNA target genes. This review provides an overview of the current state of knowledge and role of GAS5 in clinical relevance, biological functions and molecular mechanisms underlying the dysregulation of expression and function of GAS5 in cancer. Finally, the potential prospective role as diagnostic and prognostic biomarker and therapeutic target in cancer is discussed.
Collapse
Affiliation(s)
- Yaya Yu
- Laboratory of Tumor Biology, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, People's Republic of China
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, People's Republic of China
| |
Collapse
|
14419
|
Human Colorectal Cancer from the Perspective of Mouse Models. Genes (Basel) 2019; 10:genes10100788. [PMID: 31614493 PMCID: PMC6826908 DOI: 10.3390/genes10100788] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/25/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease that includes both hereditary and sporadic types of tumors. Tumor initiation and growth is driven by mutational or epigenetic changes that alter the function or expression of multiple genes. The genes predominantly encode components of various intracellular signaling cascades. In this review, we present mouse intestinal cancer models that include alterations in the Wnt, Hippo, p53, epidermal growth factor (EGF), and transforming growth factor β (TGFβ) pathways; models of impaired DNA mismatch repair and chemically induced tumorigenesis are included. Based on their molecular biology characteristics and mutational and epigenetic status, human colorectal carcinomas were divided into four so-called consensus molecular subtype (CMS) groups. It was shown subsequently that the CMS classification system could be applied to various cell lines derived from intestinal tumors and tumor-derived organoids. Although the CMS system facilitates characterization of human CRC, individual mouse models were not assigned to some of the CMS groups. Thus, we also indicate the possible assignment of described animal models to the CMS group. This might be helpful for selection of a suitable mouse strain to study a particular type of CRC.
Collapse
|
14420
|
Kanda M, Koike M, Tanaka C, Kobayashi D, Hattori N, Hayashi M, Yamada S, Omae K, Fujiwara M, Kodera Y. Modified Systemic Inflammation Score is Useful for Risk Stratification After Radical Resection of Squamous Cell Carcinoma of the Esophagus. Ann Surg Oncol 2019; 26:4773-4781. [PMID: 31605344 DOI: 10.1245/s10434-019-07914-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammation plays a critical role in the development and progression of cancers. We evaluated the clinical significance of the preoperative modified systemic inflammation score (mSIS) to predict long-term outcomes of patients with esophageal squamous cell carcinoma (ESCC). METHODS We included 443 patients who underwent curative resection of ESCC. The mSIS was formulated according to the serum albumin level (ALB) and lymphocyte-to-monocyte ratio (LMR) as follows: mSIS 0 (ALB ≥ 4.0 g/dL and LMR ≥ 3.4), mSIS 1 (ALB < 4.0 g/dL or LMR < 3.4), and mSIS 2 (ALB < 4.0 g/dL and LMR < 3.4). RESULTS Patients were categorized into preoperative mSIS 0 (n = 165), mSIS 1 (n = 183), and mSIS 2 (n = 95) groups. Preoperative mSIS was significantly associated with age, preoperative body mass index, and pathological disease stage. The disease-specific survival times of patients in preoperative mSIS 0, 1, and 2 sequentially shortened (P = 0.009), and mSIS 2 was identified as an independent prognostic factor (hazard ratio 2.63, 95% confidence interval 1.33-5.27, P = 0.0053). In most patient subgroups, the mSIS was associated with greater risk of disease-specific death. A stepwise increase in the prevalence of hematogenous recurrences was directly proportion to the mSIS. When patients were subdivided by mSIS before neoadjuvant treatment, there were no significant differences in disease-specific survival. CONCLUSIONS Our findings demonstrate that the preoperative mSIS may serve as a powerful prognosticator of ESCC that definitively stratifies clinical outcomes as well as a tool for selecting treatment strategies.
Collapse
Affiliation(s)
- Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Masahiko Koike
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Suguru Yamada
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kenji Omae
- Department of Innovative Research and Education for Clinicians and Trainees (DiRECT), Fukushima Medical University Hospital, Fukushima, Japan
| | - Michitaka Fujiwara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
14421
|
Macchia G, Lazzari R, Colombo N, Laliscia C, Capelli G, D'Agostino GR, Deodato F, Maranzano E, Ippolito E, Ronchi S, Paiar F, Scorsetti M, Cilla S, Ingargiola R, Huscher A, Cerrotta AM, Fodor A, Vicenzi L, Russo D, Borghesi S, Perrucci E, Pignata S, Aristei C, Morganti AG, Scambia G, Valentini V, Jereczek-Fossa BA, Ferrandina G. A Large, Multicenter, Retrospective Study on Efficacy and Safety of Stereotactic Body Radiotherapy (SBRT) in Oligometastatic Ovarian Cancer (MITO RT1 Study): A Collaboration of MITO, AIRO GYN, and MaNGO Groups. Oncologist 2019; 25:e311-e320. [PMID: 32043791 DOI: 10.1634/theoncologist.2019-0309] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/23/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Recent studies have reported improvement of outcomes (progression-free survival, overall survival, and prolongation of androgen deprivation treatment-free survival) with stereotactic body radiotherapy (SBRT) in non-small cell lung cancer and prostate cancer. The aim of this retrospective, multicenter study (MITO RT-01) was to define activity and safety of SBRT in a very large, real-world data set of patients with metastatic, persistent, and recurrent ovarian cancer (MPR-OC). MATERIALS AND METHODS The endpoints of the study were the rate of complete response (CR) to SBRT and the 24-month actuarial local control (LC) rate on "per-lesion" basis. The secondary endpoints were acute and late toxicities and the 24-month actuarial late toxicity-free survival. Objective response rate (ORR) included CR and partial response (PR). Clinical benefit (CB) included ORR and stable disease (SD). Toxicity was evaluated by the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC) and Common Terminology Criteria for Adverse Events (CTCAE) scales, according to center policy. Logistic and Cox regression were used for the uni- and multivariate analysis of factors predicting clinical CR and actuarial outcomes. RESULTS CR, PR, and SD were observed in 291 (65.2%), 106 (23.8%), and 33 (7.4%) lesions, giving a rate of CB of 96.4%. Patient aged ≤60 years, planning target volume (PTV) ≤18 cm3 , lymph node disease, and biologically effective dose α/β10 > 70 Gy were associated with higher chance of CR in the multivariate analysis. With a median follow-up of 22 months (range, 3-120), the 24-month actuarial LC rate was 81.9%. Achievement of CR and total dose >25 Gy were associated with better LC rate in the multivariate analysis. Mild toxicity was experienced in 54 (20.7%) patients; of 63 side effects, 48 were grade 1, and 15 were grade 2. The 24-month late toxicity-free survival rate was 95.1%. CONCLUSIONS This study confirms the activity and safety of SBRT in patients with MPR-OC and identifies clinical and treatment parameters able to predict CR and LC rate. IMPLICATIONS FOR PRACTICE This study aimed to define activity and safety of stereotactic body radiotherapy (SBRT) in a very large, real life data set of patients with metastatic, persistent, recurrent ovarian cancer (MPR-OC). Patient age <60 years, PTV <18 cm3 , lymph node disease, and biologically effective dose α/β10 >70 Gy were associated with higher chance of complete response (CR). Achievement of CR and total dose >25 Gy were associated with better local control (LC) rate. Mild toxicity was experienced in 20.7% of patients. In conclusion, this study confirms the activity and safety of SBRT in MPR-OC patients and identifies clinical and treatment parameters able to predict CR and LC rate.
Collapse
Affiliation(s)
- Gabriella Macchia
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Roberta Lazzari
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Nicoletta Colombo
- Division of Medical Gynecologic Oncology, IEO European Institute of Oncology, IRCCS and University of Milan-Bicocca, Milan, Italy
| | - Concetta Laliscia
- Department of Translational Medicine, Division of Radiation Oncology, University of Pisa, Pisa, Italy
| | - Giovanni Capelli
- Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, Cassino, Italy
| | - Giuseppe Roberto D'Agostino
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Hospital, IRCSS, Rozzano, Milan, Italy
| | - Francesco Deodato
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | | | - Edy Ippolito
- Department of Radiation Oncology, Campus Bio-Medico University, Rome, Italy
| | - Sara Ronchi
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Fabiola Paiar
- Department of Translational Medicine, Division of Radiation Oncology, University of Pisa, Pisa, Italy
| | - Marta Scorsetti
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Hospital, IRCSS, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Rozzano, Milano, Italy
| | - Savino Cilla
- Fondazione "Giovanni Paolo II," Unità Operativa di Fisica Medica, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | - Rossana Ingargiola
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Alessandra Huscher
- U.O. di Radioterapia Oncologica "Guido Berlucchi," Fondazione Poliambulanza, Brescia, Italy
| | - Anna Maria Cerrotta
- Radiotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrei Fodor
- Department of Radiation Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Lisa Vicenzi
- Department of Oncology and Radiotherapy, Azienda Ospedaliera Universitaria Ospedali Riuniti, Ancona, Italy
| | | | - Simona Borghesi
- Radiotherapy Department, Azienda USL Toscana sud est, San Donato Hospital-Arezzo, Italy
| | - Elisabetta Perrucci
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Sandro Pignata
- Istituto Nazionale Tumori di Napoli, Fondazione Pascale IRCCS, Naples, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Alessio Giuseppe Morganti
- Department of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giovanni Scambia
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Ginecologia e Ostetricia, Roma, Italy
| | - Vincenzo Valentini
- Fondazione "Giovanni Paolo II," Unità Operativa di Radioterapia, Università Cattolica del Sacro Cuore, Campobasso, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Radiologia, Roma, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Gabriella Ferrandina
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Unità Operativa Complessa Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Roma, Italy
- Università Cattolica del Sacro Cuore, Istituto di Ginecologia e Ostetricia, Roma, Italy
| |
Collapse
|
14422
|
Li Y, Gan C, Zhang Y, Yu Y, Fan C, Deng Y, Zhang Q, Yu X, Zhang Y, Wang L, He F, Xie Y, Ye T, Yin W. Inhibition of Stat3 Signaling Pathway by Natural Product Pectolinarigenin Attenuates Breast Cancer Metastasis. Front Pharmacol 2019; 10:1195. [PMID: 31649548 PMCID: PMC6796319 DOI: 10.3389/fphar.2019.01195] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/17/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Breast cancer is the most common female cancer with considerable metastatic potential, which urges the need for developing novel potential drug candidate to inhibit tumor metastasis. Signal transducer and activator of transcription 3 (Stat3) have critical roles in cancer growth and metastasis and have been confirmed as a promising anticancer target. Here, we report our finding with pectolinarigenin, a flavonoid compound isolated from the aerial parts of Cirsium chanroenicum. Methods: The role of Pec. in cell proliferation, cell apoptosis, and cell migration and invasion in three breast cancer cells (4T1, MDA-MB-231, MCF-7) was investigated. Cell proliferation was determined by MTT assay, cell apoptosis was determined by flow cytometry, and protein expression was detected by western blotting. Tumor xenograft mice model and breast tumor metastasis model in vivo were built to further assess the effects of Pec. on 4T1 cells. Results: Intraperitoneal administrations of pectolinarigenin significantly inhibited breast cancer metastasis to lungs without affecting the tumor growth of incubated 4T1 breast cancer cells. Pectolinarigenin could also recruit CD8+ T cells to mediate tumor immune response. Furthermore, pectolinarigenin markedly impaired cancer cell migration and invasion by down-regulating phosphorylated-Stat3, and expression of matrix metalloproteinase (MMP)-2, MMP-9, while up-regulating the expression of TIMP2. We also found that pectolinarigenin inhibited breast cancer cell proliferation and induced apoptosis via mitochondrial-related apoptosis pathway, reduced mitochondrial membrane potential and the expression of Bcl-2, increased expression of Bax, and cleaved caspase-3 as well as disturbed the ROS generation. Conclusions: Pectolinarigenin might potentially be a candidate for metastasis of breast cancer by mediating Stat3 pathway.
Collapse
Affiliation(s)
- Yali Li
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China.,Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Cailing Gan
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yange Zhang
- Cosmetic Plastic and Burn Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yan Yu
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chen Fan
- School of Pharmacy, Southwest University for Nationalities, Chengdu, China
| | - Yuanle Deng
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Qianyu Zhang
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Xi Yu
- Carey Business School, Johns Hopkins University, Baltimore, MD, United States
| | - Yiwen Zhang
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Liqun Wang
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Fang He
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Yongmei Xie
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tinghong Ye
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenya Yin
- West China School of Public Health and West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| |
Collapse
|
14423
|
Schwartz GG, Klug MG. Thyroid Cancer Incidence Rates in North Dakota are Associated with Land and Water Use. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16203805. [PMID: 31658605 PMCID: PMC6843260 DOI: 10.3390/ijerph16203805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/29/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
Objective: The increasing rate of thyroid cancer diagnoses in the U.S. reflects the increasing use of ultrasonography and of specialist medical care. North Dakota is a rural state with limited access to specialist care, yet its incidence of thyroid cancer is significantly greater than that of the U.S. overall. We sought to identify factors responsible for the high incidence of thyroid cancer in North Dakota. Methods: We examined county-specific incidence rates for thyroid cancer in North Dakota in relation to demographic and geographic factors, including median household income, percent of land fertilized, cattle density per capita, and source of drinking water (city or well water), using structural equation modeling. We included county level data on residential radon levels and estimates of radioactive iodine in milk following nuclear weapons testing in the 1950s. Results: Thyroid cancer incidence rates were significantly associated with median income (p < 0.05); percent of land fertilized (p < 0.05); the use of city water (p < 0.01), and cattle density per capita (p < 0.001). Conclusions: The risk of thyroid cancer in North Dakota is positively associated with income and with factors related to land and water use. Our finding that thyroid cancer incidence rates are associated with the use of city water was unexpected and merits examination in other locations with a mix of city and well water use.
Collapse
Affiliation(s)
- Gary G Schwartz
- Department of Population Health, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202-9037, USA.
| | - Marilyn G Klug
- Department of Population Health, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202-9037, USA.
| |
Collapse
|
14424
|
Anderson CF, Grimmett ME, Domalewski CJ, Cui H. Inhalable nanotherapeutics to improve treatment efficacy for common lung diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1586. [PMID: 31602823 DOI: 10.1002/wnan.1586] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022]
Abstract
Respiratory illnesses are prevalent around the world, and inhalation-based therapies provide an attractive, noninvasive means of directly delivering therapeutic agents to their site of action to improve treatment efficacy and limit adverse systemic side effects. Recent trends in medicine and nanoscience have prompted the development of inhalable nanomedicines to further enhance effectiveness, patient compliance, and quality of life for people suffering from lung cancer, chronic pulmonary diseases, and tuberculosis. Herein, we discuss recent advancements in the development of inhalable nanomaterial-based drug delivery systems and analyze several representative systems to illustrate their key design principles that can translate to improved therapeutic efficacy for prevalent respiratory diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease.
Collapse
Affiliation(s)
- Caleb F Anderson
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Maria E Grimmett
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Christopher J Domalewski
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14425
|
Zheng ZH, Wu DM, Fan SH, Wen X, Han XR, Wang S, Wang YJ, Zhang ZF, Shan Q, Li MQ, Hu B, Zheng YL, Lu J. LncRNA AB209371 up-regulated Survivin gene by down-regulating miR-203 in ovarian carcinoma. J Ovarian Res 2019; 12:92. [PMID: 31601255 PMCID: PMC6785849 DOI: 10.1186/s13048-019-0559-4] [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: 05/27/2019] [Accepted: 08/27/2019] [Indexed: 11/10/2022] Open
Abstract
AB209371 gene has been characterized as an oncogenic lncRNA in liver cancer. However, its involvement in ovarian carcinoma (OC) is unknown. In the present study, we analyzed the roles of AB209371 in OC. We found that AB209371 gene and Survivin gene were up-regulated in OC and positively correlated with OC development. AB209371 over-expression led to up-regulated Survivin in OC cells, while Survivin over-expression failed to affect AB209371. In addition, AB209371 over-expression led to down-regulated miR-203. However, miR-203 over-expression failed to affect AB209371, but down-regulated the expression of Survivin. In addition, over-expressions of AB209371 and Survivin resulted in the increased proliferation rate of OC cells. Over-expression MiR-203 played the opposite role and attenuated the effects of AB209371 over-expression. Therefore, AB209371 may down-regulate miR-203 to up-regulate Survivin, thereby promoting OC cell proliferation. Our study provided novel insights into the pathogenesis of OC.
Collapse
Affiliation(s)
- Zi-Hui Zheng
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China. .,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China.
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science; College of Health Sciences, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan District, Xuzhou, 221116, Jiangsu Province, People's Republic of China. .,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, People's Republic of China.
| |
Collapse
|
14426
|
Zhang P, Wu X, Tang M, Nie X, Li L. Detection of EGFR gene mutation status from pleural effusions and other body fluid specimens in patients with lung adenocarcinoma. Thorac Cancer 2019; 10:2218-2224. [PMID: 31602787 PMCID: PMC6885423 DOI: 10.1111/1759-7714.13201] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/04/2019] [Accepted: 09/04/2019] [Indexed: 12/25/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) gene mutation status is essential to the optimal management of lung adenocarcinoma. Liquid biopsy has advantages such as noninvasiveness, speediness, and convenience. This study aimed to detect EGFR gene mutations using next‐generation sequencing (NGS) from different types of body fluids from patients with lung adenocarcinoma. Methods This was a prospective study of 20 patients with lung adenocarcinoma recruited between January 2017 and December 2018 at the Beijing Hospital. All patients had adenocarcinoma with confirmed sensitizing EGFR mutations. Body fluid specimens included pleural effusion, ascites, pericardial effusion, and cerebrospinal fluid. NGS was conducted to test for nine lung cancer‐related gene in body fluid supernatant free DNA, sedimentary tumor cells, and plasma free DNA. Results The EGFR gene mutation abundance of body fluid supernatant free DNA was higher than that of body fluid sedimentary tumor cells and plasma free DNA specimens (100% vs. 90% vs. 80%, respectively, all P < 0.05). The results of EGFR mutation from the body fluid supernatants were consistent with the results from the tissue biopsy. Conclusions This study showed that compared with body fluid sediment tumor cells and plasma free DNA samples, body fluid supernatant free DNA has a higher detection rate and sensitivity of tumor‐specific mutations. Free DNA obtained from body fluid supernatants could be used as high‐quality specimens for gene mutation detection in patients with lung cancer. This could be applied in treatment decisions and patient management.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xiaonan Wu
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Min Tang
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xin Nie
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Lin Li
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| |
Collapse
|
14427
|
Rezaei S, Mahjoubin Tehran M, Sahebkar A, Jalili A, Aghaee‐Bakhtiari SH. Androgen receptor‐related micro RNAs in prostate cancer and their role in antiandrogen drug resistance. J Cell Physiol 2019; 235:3222-3234. [DOI: 10.1002/jcp.29275] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/27/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Mahjoubin Tehran
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Amin Jalili
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | | |
Collapse
|
14428
|
Zang X, Qian C, Ruan Y, Xie J, Luo T, Xu B, Jiang J. Higher maternal embryonic leucine zipper kinase mRNA expression level is a poor prognostic factor in non-small-cell lung carcinoma patients. Biomark Med 2019; 13:1349-1361. [PMID: 31599664 DOI: 10.2217/bmm-2019-0052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: To elucidate potential prognostic significance of MELK mRNA expression in non-small-cell lung carcinoma patients. Methods: A loop algorithm based on R software was used to select genes with the best prognostic value. Mantel-Haenszel method and functional enrichment analysis were used to perform this analysis. Results: MELK mRNA expression level in tumor tissue is significantly higher than that in normal/benign tissue (p < 0.001), and gradually increases from stage I to IV (lung adenocarcinoma: p = 0.011; lung squamous cell carcinoma: p = 0.002), and is negatively correlated with prognosis in lung adenocarcinoma patients (HR: 2.025 in univariate analysis; HR: 2.162 in multivariate analysis). However, it does not show a significant correlation in lung squamous cell carcinoma patients. Conclusion: MELK is a poor biomarker for non-small-cell lung carcinoma patients and can potentially be used as a therapeutic target.
Collapse
Affiliation(s)
- Xuefeng Zang
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Chunyan Qian
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Ye Ruan
- Department of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Junwen Xie
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Ting Luo
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Bin Xu
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| | - Jingting Jiang
- Department of Clinical Trials Research Center, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province 213000, PR China
| |
Collapse
|
14429
|
D'Souza MJ, Li RC, Gannon ML, Wentzien DE. 1997-2017 Leading Causes of Death Information Due to Diabetes, Neoplasms, and Diseases of the Circulatory System, Issues Cautionary Weight-Related Lesson to the US Population at Large. IEEE NETWORK 2019; 2019:1-6. [PMID: 31631939 PMCID: PMC6800725 DOI: 10.1109/icesi.2019.8863033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In the US, cardiovascular disease, cancer, and diabetes are in the top ten leading causes of death categories. The diseases compromise US life-expectancy and account for significant US health-care costs. This observational study investigates the US population's 1997-2017 Centers for Disease Control and Prevention (CDC) WONDER ICD-10 mortality records to extract the prevalence rates for leading causes of death by diabetes, neoplasms (cancers), and diseases of the circulatory system. The variables of race and age are examined for each disease in order to evaluate demographic and age-group risks. To document the public health burden from these three chronic conditions, mortality data from CDC WONDER was analyzed using MS-Excel and Statistical Analysis System (SAS) software. The general trend of deaths by diabetes, neoplasms, and diseases of the circulatory system has been progressively decreasing nationally; however, a significantly higher trend in mortality rates is observed for the Black or African American populations. Furthermore, over the 1997-2017 observational period, the crude mortality rates for the 45-54 (middle-age) and lower age-groups are below national mortality rate averages but are troublingly increasing for diabetes and notably, for the diseases of the circulatory system, the (younger) 25-34 age-group had a crude mortality rate increase of 6.78%.
Collapse
Affiliation(s)
- Malcolm J D'Souza
- Professor of Chemistry & Dean of Interdisciplinary/ Collaborative Sponsored Research at Wesley College, Dover, Delaware, 19901, USA. He is the principal lead on the listed grants and the Wesley College STEM Undergraduate Research Center for Analytics, Talent, and Success
| | - Riza C Li
- Wesley College 2016 summa cum laude graduate in mathematics. Currently, she is a PhD candidate in the Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware, 19711, USA
| | - Morgan L Gannon
- 2017 Wesley College biology graduate. Currently, she is a Physician Assistant Student Extern at the Advanced Plastic Surgery Center, Delaware, 19711, USA
| | - Derald E Wentzien
- Professor of Mathematics & Data Science at Wesley College, Dover, Delaware, 19901, USA. He is the faculty lead on the Wesley College Informatics certificate and minor programs sponsored through the STEM UR-CATS
| |
Collapse
|
14430
|
Schenk EL, Mandrekar SJ, Dy GK, Aubry MC, Tan AD, Dakhil SR, Sachs BA, Nieva JJ, Bertino E, Lee Hann C, Schild SE, Wadsworth TW, Adjei AA, Molina JR. A Randomized Double-Blind Phase II Study of the Seneca Valley Virus (NTX-010) versus Placebo for Patients with Extensive-Stage SCLC (ES SCLC) Who Were Stable or Responding after at Least Four Cycles of Platinum-Based Chemotherapy: North Central Cancer Treatment Group (Alliance) N0923 Study. J Thorac Oncol 2019; 15:110-119. [PMID: 31605793 DOI: 10.1016/j.jtho.2019.09.083] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/09/2019] [Accepted: 09/21/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION The Seneca Valley virus (NTX-010) is an oncolytic picornavirus with tropism for SCLC. This phase II double-blind, placebo-controlled trial evaluated NTX-010 in patients with extensive-stage (ES) SCLC after completion of first-line chemotherapy. METHODS Patients with ES SCLC who did not progress after four or more cycles of platinum-based chemotherapy were randomized 1:1 to a single dose of NTX-010 or placebo within 12 weeks of chemotherapy. The primary end point was progression-free survival (PFS). A prespecified interim analysis for futility was performed after 40 events. Viral clearance and the development of neutralizing antibodies were followed. RESULTS From January 15, 2010, to January 10, 2013, a total of 50 patients were randomized and received therapy on study (26 received NTX-010 and 24 received placebo). At the specified interim analysis, the median PFS was 1.7 months (95% confidence interval [CI]: 1.4-3.1 months) for the NTX-010 group versus 1.7 months (95% CI: 1.4-4.3 months) for the placebo group (hazard ratio = 1.03, p = 0.92), and the trial was terminated owing to futility. In the NTX-010 group, PFS was shorter in patients with detectable virus at days 7 and 14 versus in those in whom it was not detected after treatment (1.0 month [95% CI: 0.4-1.5 months] versus 1.8 months [95% CI: 1.3-5.5 months, p = 0.008] and 0.9 months [95% CI: 0.4-2.6 months] versus 1.3 months [95% CI: 1.0-5.3 months], respectively [p = 0.04]). CONCLUSIONS Patients with ES SCLC did not benefit from NTX-010 treatment after chemotherapy with a platinum doublet. Persistence of NTX-010 in the blood 1 or 2 weeks after treatment was associated with a shorter PFS.
Collapse
Affiliation(s)
| | - Sumithra J Mandrekar
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota; Mayo Clinic, Rochester, Minnesota
| | - Grace K Dy
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Angelina D Tan
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota; Mayo Clinic, Rochester, Minnesota
| | | | | | - Jorge J Nieva
- University of Southern California, Los Angeles, California
| | - Erin Bertino
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | - Troy W Wadsworth
- Northwest NCORP, Multicare Regional Cancer Center, Tacoma, Washington
| | | | | |
Collapse
|
14431
|
Gupta PK, Tripathi SK, Pappuru S, Chabattula SC, Govarthanan K, Gupta S, Biswal BK, Chakraborty D, Verma RS. Metal-free semi-aromatic polyester as nanodrug carrier: A novel tumor targeting drug delivery vehicle for potential clinical application. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110285. [PMID: 31761245 DOI: 10.1016/j.msec.2019.110285] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/09/2019] [Accepted: 10/05/2019] [Indexed: 12/19/2022]
Abstract
Polyester nanomaterials have been widely used in drug delivey application from a longer period of time. This study reports the synthesis of metal-free semi-aromatic polyester (SAP) nanomaterial for drug delivery and evaluate its in vivo acute and systemic toxicity for potential clinical application. The ring opening coplymerization of commercially available cyclohexene oxide (CHO) and phthalic anhydride (PA) monomers was carried out to synthesize fully alternating poly(CHO-co-PA) copolymer using metal-free activators. The obtained low Mn SAP was found to be biocompatible, hemocompataible and biodegradable nature. This copolymer was first-time used to fabricate curcumin (CUR) loaded nanoparticles (NPs). These NPs were physicochemically characterized by thermogravimetric analyzer (TGA), X-ray diffraction (XRD), and UV/visible spectrophotometer analysis. Further, these negatively charged core-shell spherical NPs exhibited slow sustained release behavior of CUR with anomalous transport and further displayed its higher intracellular uptake in SiHa cells at different time-periods compared to free CUR. In vitro anti-cancer therapeutic effects of free CUR and poly(CHO-alt-PA)-CUR NPs were evaluated on different cancer cells. We observed the increased cytotoxicity of CUR NPs with low IC50 values compared to free CUR. These results were further substantiated with ex vivo data where, a significant reduction was observed in CUR NPs treated tumor spheroid's size as compared to free CUR. Furthermore, the different doses of metal-free poly(CHO-alt-PA) nanomaterial were tested for its acute and systemic toxicity in BALB/c mice. We did not observe any significant toxicity of tested nanomaterial on vital organs, blood cells and the body weight of mice. Our study suggest that this metal-free SAP nanomaterial can be used for potential clinical application.
Collapse
Affiliation(s)
- Piyush Kumar Gupta
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology-Rourkela, Rourkela, 769008, Odisha, India
| | - Sreenath Pappuru
- Organometallic and Polymer Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Siva Chander Chabattula
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Kavitha Govarthanan
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Santosh Gupta
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology-Rourkela, Rourkela, 769008, Odisha, India
| | - Debashis Chakraborty
- Organometallic and Polymer Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, Tamilnadu, India.
| |
Collapse
|
14432
|
Debnath R, Majumder D, Nath P, Ghosh D, Maiti D. Bromelain plus peroxidase reduces non-Hodgkin lymphoma progression in invivo via up-regulation of antioxidant enzymes and modulating apoptotic protein expression. Nutr Cancer 2019; 72:1200-1210. [PMID: 31591915 DOI: 10.1080/01635581.2019.1670217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Aim: Pineapple (Ananas comosus (L.) Merr.) is a good source of bromelain (B) and also contain peroxidase. The objective of this study is isoaltion of bromelain plus peroxidase (BP) from the pineapple fruit to evaluate the anticancer activity of BP from the pineapple fruit of Tripura, compared to commercial bromelain against ascitic Dalton's lymphoma cells (DLA) in mice. Methods: By acetone precipitation BP was isolated from the pineapple. Animals bearing DLA, receive B and BP orally for 15 alternative days. Apoptotic proteins are assayed using western blot. Results: BP treated mice showed recover of hemoglobin and WBC count compared to control lymphoma animal. The animal showed significant reduction of body weight due to reduced tunor load and elevated reactive oxygen species (ROS) production, elevated levels of vitamin C and vitamin E and other antioxidants in blood after BP treatment. Histology of liver and kidney also shows restored architecture in BP treated animal compared to only B treated group. BP treatment upregulates the cytochrome C, BAD, and BAX protein and downregulates the Bcl-2 and NF-kβ occuring upon BP treatment in the DLA cells collected from lymphoma animal. This induce the apoptosis of DLA cells in lymphoma animal and reduce the tumor load. Conclusion: The present findings suggest that BP from pineapple improves the survival of the induced lymphoma animal compared to only B which may be used as therapeutic target.
Collapse
Affiliation(s)
- Rahul Debnath
- Department of Human Physiology, Tripura University, Immunology Microbiology Lab, Suryamaninagar, Tripura, India
| | - Debabrata Majumder
- Department of Human Physiology, Tripura University, Immunology Microbiology Lab, Suryamaninagar, Tripura, India
| | - Priyatosh Nath
- Department of Human Physiology, Tripura University, Immunology Microbiology Lab, Suryamaninagar, Tripura, India
| | - Durgadas Ghosh
- Department of Zoology, Tripura University, Suryamaninagar, Tripura, India
| | - Debasish Maiti
- Department of Human Physiology, Tripura University, Immunology Microbiology Lab, Suryamaninagar, Tripura, India
| |
Collapse
|
14433
|
Xu CB, Chen ZW, Mei YJ. Association of TMPRSS4 expression with prognosis in gastric cancer based on data from Oncomine and Kaplan-Meier plotter databases. Shijie Huaren Xiaohua Zazhi 2019; 27:1193-1200. [DOI: 10.11569/wcjd.v27.i19.1193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer related death worldwide and the most frequently diagnosed malignancy of the digestive system. However, the factors related to GC prognosis are not clear yet. In this study, we investigated a potential biomarker for GC prognosis through bioinformatics analysis.
AIM To investigate the transmembrane protease serine 4 (TMPRSS4) mRNA expression in GC through data mining of the Oncomine and Kaplan-Meier Plotter databases.
METHODS TMPRSS4 gene expression in GC was investigated and compared between cancer and normal gastric tissues in the Oncomie and Kaplan-Meier Plotter data platforms. The survival curves were drawn to compare the survival time of patients with high and low expression of TMPRSS4, and to explore the feasibility of TMPRSS4 as a prognostic molecular marker in patients with GC. Meanwhile, the clinical data of 50 patients with GC treated by surgery were retrospectively analyzed. The expression of TMPRSS4 protein in these 50 patients with GC was examined by immunohistochemistry. The correlation between TMPRSS4 expression and clinical characteristics was analyzed.
RESULTS The Oncomine database contained 185 datasets on the TMPRSS4 expression in GC, breast cancer, and other common cancer tissues and corresponding normal tissues. There are 10 datasets on differentially expressed TMPRSS4, of which 9 showed highly expressed TMPRSS4 in cancer tissues, and 1 showed lowly expressed TMPRSS4 in cancer tissues. Hierarchical cluster analysis showed that genes co-expressed with TMPRSS4 in GC were LAMB3, LAD1, ANXA4, etc. The co-expression correlation coefficients of TMPRSS4 with LAMB3, LAD1, and ANXA4 were 0.57, 0.51, and 0.43, respectively. TMPRSS4 may have similar functions to LAMB3, LAD1, ANXA4, and other co-expressed genes. In the Oncomine database, three gene expression microarray datasets were used to study the differential expression of TMPRSS4 gene in GC tissues and normal tissues. The results showed that the expression level of TMPRSS4 in GC tissues was higher than that in normal gastric tissues (P < 0.05). Kaplan-Meier Plotter analysis showed that the total median survival time of the high and low TMPRSS4 expression groups was 22.0 mo and 32.6 mo, respectively, and the median survival time of the high expression group was significantly lower than that of the low expression group (HR = 1.31, 95%CI: 1.09-1.57, P < 0.05). The median disease-free survival time of the high expression group was also significantly lower than that of the low expression group (13.87 mo vs 22.40 mo, HR = 1.27, 95%CI: 1.03-1.58, P < 0.05). Immunohistochemistry showed that the positive expression of TMPRSS4 was correlated with vascular invasion. The percentage of TMPRSS4 positive patients with vascular invasion was significantly higher than that of TMPRSS4 negative patients (P < 0.05).
CONCLUSION Compared with normal gastric tissues, the expression level of TMPRSS4 gene in GC tissues is significantly up-regulated. The high expression of TMPRSS4 gene is associated with vascular invasion and a poor prognosis in GC patients.
Collapse
Affiliation(s)
- Chao-Bo Xu
- Department of Gastrointestinal Surgery, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Zheng-Wei Chen
- Department of Gastrointestinal Surgery, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Yi-Jun Mei
- Department of Gastrointestinal Surgery, Lishui People's Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
14434
|
Woodford MR, Hughes M, Sager RA, Backe SJ, Baker-Williams AJ, Bratslavsky MS, Jacob JM, Shapiro O, Wong M, Bratslavsky G, Bourboulia D, Mollapour M. Mutation of the co-chaperone Tsc1 in bladder cancer diminishes Hsp90 acetylation and reduces drug sensitivity and selectivity. Oncotarget 2019; 10:5824-5834. [PMID: 31645902 PMCID: PMC6791385 DOI: 10.18632/oncotarget.27217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023] Open
Abstract
The molecular chaperone Heat shock protein 90 (Hsp90) is essential for the folding, stability, and activity of several drivers of oncogenesis. Hsp90 inhibitors are currently under clinical evaluation for cancer treatment, however their efficacy is limited by lack of biomarkers to optimize patient selection. We have recently identified the tumor suppressor tuberous sclerosis complex 1 (Tsc1) as a new co-chaperone of Hsp90 that affects Hsp90 binding to its inhibitors. Highly variable mutations of TSC1 have been previously identified in bladder cancer and correlate with sensitivity to the Hsp90 inhibitors. Here we showed loss of TSC1 leads to hypoacetylation of Hsp90-K407/K419 and subsequent decreased binding to the Hsp90 inhibitor ganetespib. Pharmacologic inhibition of histone deacetylases (HDACs) restores acetylation of Hsp90 and sensitizes Tsc1-mutant bladder cancer cells to ganetespib, resulting in apoptosis. Our findings suggest that TSC1 status may predict response to Hsp90 inhibitors in patients with bladder cancer, and co-targeting HDACs can sensitize tumors with Tsc1 mutations to Hsp90 inhibitors.
Collapse
Affiliation(s)
- Mark R. Woodford
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Michael Hughes
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Rebecca A. Sager
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
- College of Medicine, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Sarah J. Backe
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Alexander J. Baker-Williams
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Michael S. Bratslavsky
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Joseph M. Jacob
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Oleg Shapiro
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Gennady Bratslavsky
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University Syracuse, NY 13210, USA
| |
Collapse
|
14435
|
Lu C, Jiang W, Hui B, Rong D, Fu K, Dong C, Tang W, Cao H. The circ_0021977/miR-10b-5p/P21 and P53 regulatory axis suppresses proliferation, migration, and invasion in colorectal cancer. J Cell Physiol 2019; 235:2273-2285. [PMID: 31595500 DOI: 10.1002/jcp.29135] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
An in-depth understanding of circular RNAs (circRNAs) indicates that they are abundant in the eukaryotic transcriptome. Many circRNAs act as microRNA sponges; thus, they represent a new type of regulatory factor. However, the role of circRNA in colorectal cancer (CRC) remains largely unknown. Low circ_0021977 expression in patients with CRC is associated with higher tug-lymph node metastasis (TNM) stage and poorer prognosis compared with patients with high circ_0021977 expression. Moreover, miR-10b-5p was shown to be a target of circ_0021977, and p21 and p53 are suggested to be putative target genes of miR-10b-5p. The results showed that the circ_0021977/miR-10b-5p/p21&p53 regulatory axis suppresses proliferation, migration, and invasion by CRC cells. This evidence reveals new relationships and brings new highlights to the treatment of CRC.
Collapse
Affiliation(s)
- Chen Lu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.,The Third Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Jiang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.,The Third Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bingqing Hui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Rong
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kai Fu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chaoxi Dong
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.,The Third Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Tang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
14436
|
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: 77] [Impact Index Per Article: 12.8] [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
|
14437
|
Role of lymphadenectomy in endometrial cancer with nonbulky lymph node metastasis: Comparison of comprehensive surgical staging and sentinel lymph node algorithm. Gynecol Oncol 2019; 155:177-185. [PMID: 31604668 DOI: 10.1016/j.ygyno.2019.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To compare survival and progression outcomes between 2 nodal assessment approaches in patients with nonbulky stage IIIC endometrial cancer (EC). METHODS Patients with stage IIIC EC treated at 2 institutions were retrospectively identified. At 1 institution, a historical series (2004-2008) was treated with systematic pelvic and para-aortic lymphadenectomy (LND cohort). At the other institution, more contemporary patients (2006-2013) were treated using a sentinel lymph node algorithm (SLN cohort). Outcomes (hazard ratios [HRs]) within the first 5 years after surgery were compared between cohorts using Cox models adjusted for type of adjuvant therapy. RESULTS The study included 104 patients (48 LND, 56 SLN). The use of chemoradiotherapy was similar in the 2 cohorts (46% LND vs 50% SLN), but the use of chemotherapy alone (19% vs 36%) or radiotherapy alone (15% vs 2%) differed. Although there was evidence of higher risk of cause-specific death (HR, 2.10; 95% CI, 0.79-5.58; P = 0.14) and lower risk of para-aortic progression (HR, 0.27; 95% CI, 0.05-1.42; P = 0.12) for the LND group, the associations did not meet statistical significance. The risk of progression was not significantly different between the groups (HR, 1.27; 95% CI, 0.60-2.67; P =0 .53). In parsimonious multivariable models, high-risk tumor characteristics and nonendometrioid type were independently associated with lower cause-specific survival and progression-free survival. CONCLUSIONS In EC patients with nonbulky positive lymph nodes, use of the SLN algorithm with limited nodal dissection does not compromise survival compared with LND. Aggressive pathologic features of the primary tumor are the strongest determinants of prognosis.
Collapse
|
14438
|
Xia C, Jiang H, Ye F, Zhuang Z. The Multifunction Of miR-218-5p-Cx43 Axis In Breast Cancer. Onco Targets Ther 2019; 12:8319-8328. [PMID: 31632081 PMCID: PMC6790128 DOI: 10.2147/ott.s218524] [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] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/10/2019] [Indexed: 01/07/2023] Open
Abstract
Background Gemcitabine is proven to be the first-line standard treatment of breast cancers. Yet, little is known involving gemcitabine resistance and remains largely to be elucidated. Materials and methods We evaluated the expression of Cx43 in gemcitabine-resistant cells and parental cells by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analyses. Dual-luciferase reporter assay was applied to examine the epigenetic regulator of Cx43. The role of miR-218-5p-Cx43 axis on cell cytotoxicity, cell proliferation, colony formation, chemoresistance and migration was detected via mammalian expression vector and small short RNA (shRNA) transfection in vitro. Results In this study, we found that Cx43 expression levels were significantly lower in gemcitabine-resistant cells than in the parental cells. On deep investigation of the epigenetic regulation of Cx43, a few miRNA candidates targeting Cx43 were derived. Through dual-luciferase reporter assay, Cx43 was proved to be a direct target of miR-218-5p. Besides, qPCR, Western blot demonstrated an inverse correlation between miR-218-5p and Cx43 expression in breast cancer cells, thus forming the miR-218-5p-Cx43 axis. Notably, miR-218-5p-Cx43 axis was found to be involved in the process of gemcitabine chemoresistance, cell proliferation and migration in breast cancer cells. Conclusion Our findings suggested that miR-218-5p-Cx43 axis was versatile and indicated significant potency in breast cancer cells. More importantly, miR-218-5p-Cx43 axis might be valuable in translational medicine, with therapeutic and prognostic information.
Collapse
Affiliation(s)
- Chen Xia
- Department of Medical Oncology, Hepatobiliary and Pancreatic Unit, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, People's Republic of China
| | - Hong Jiang
- Medical Department, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, People's Republic of China
| | - Fugui Ye
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Zhigang Zhuang
- Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People's Republic of China
| |
Collapse
|
14439
|
Ku SY, Gleave ME, Beltran H. Towards precision oncology in advanced prostate cancer. Nat Rev Urol 2019; 16:645-654. [PMID: 31591549 DOI: 10.1038/s41585-019-0237-8] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2019] [Indexed: 12/19/2022]
Abstract
Metastatic biopsy programmes combined with advances in genomic sequencing have provided new insights into the molecular landscape of castration-resistant prostate cancer (CRPC), identifying actionable targets, and emerging resistance mechanisms. The detection of DNA repair aberrations, such as mutation of BRCA2, could help select patients for poly(ADP-ribose) polymerase (PARP) inhibitor or platinum chemotherapy, and mismatch repair gene defects and microsatellite instability have been associated with responses to checkpoint inhibitor immunotherapy. Poor prognostic features, such as the presence of RB1 deletion, might help guide future therapeutic strategies. Our understanding of the molecular features of CRPC is now being translated into the clinic in the form of increased molecular testing for use of these agents and for clinical trial eligibility. Genomic testing offers opportunities for improving patient selection for systemic therapies and, ultimately, patient outcomes. However, challenges for precision oncology in advanced prostate cancer still remain, including the contribution of tumour heterogeneity, the timing and potential cooperation of multiple driver gene aberrations, and diverse resistant mechanisms. Defining the optimal use of molecular biomarkers in the clinic, including tissue-based and liquid biopsies, is a rapidly evolving field.
Collapse
Affiliation(s)
- Sheng-Yu Ku
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Martin E Gleave
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Himisha Beltran
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
14440
|
Li W, Li L, Wu M, Lang J, Bi Y. The Prognosis of Stage IA Mixed Endometrial Carcinoma. Am J Clin Pathol 2019; 152:616-624. [PMID: 31318970 DOI: 10.1093/ajcp/aqz083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To explore the survival and definition of stage IA mixed endometrial carcinoma. METHODS From June 1, 2010, to June 1, 2017, cases with stage IA endometrial cancer were included in this study. The survival outcomes were compared among patients with endometrioid (group A), nonendometrioid (group B), and mixed subtypes (group C) and among patients with different proportions of nonendometrioid components (<5%, >50%, and others). RESULTS In total, 890 cases were included, comprising 808 (90.8%), 33 (3.7%), and 47 (5.3%) cases in groups A, B, and C, respectively. After a median follow-up of 55.9 months, groups B and C had significantly more inferior disease-free survival, overall survival, and cancer-specific overall survival. Patients with a nonendometrioid proportion of more than 50% and serous subtype also had a significantly more inferior prognosis. Adjuvant therapy could improve the prognosis in mixed endometrial carcinomas. CONCLUSIONS Patients with endometrial cancer of mixed subtypes had inferior survival outcomes.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Ming Wu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yalan Bi
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
14441
|
Yamamoto TN, Kishton RJ, Restifo NP. Developing neoantigen-targeted T cell-based treatments for solid tumors. Nat Med 2019; 25:1488-1499. [PMID: 31591590 DOI: 10.1038/s41591-019-0596-y] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
Stimulating an immune response against cancer through adoptive transfer of tumor-targeting lymphocytes has shown great promise in hematological malignancies, but clinical efficacy against many common solid epithelial cancers remains low. Targeting 'neoantigens'-the somatic mutations expressed only by tumor cells-might enable tumor destruction without causing undue damage to vital healthy tissues. Major challenges to targeting neoantigens with T cells include heterogeneity and variability in antigen processing and presentation of targets by tumors, and an incomplete understanding of which T cell qualities are essential for clinically effective therapies. Finally, the prospect of targeting somatic tumor mutations to promote T cell destruction of cancer must contend with the biology that not all tumor-expressed 'neoepitopes' actually generate neoantigens that can be functionally recognized and provoke an effective immune response. In this Review, we discuss the promise, progress and challenges for improving neoantigen-targeted T cell-based immunotherapies for cancer.
Collapse
Affiliation(s)
- Tori N Yamamoto
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Rigel J Kishton
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA. .,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA. .,Lyell Immunopharma, South San Francisco, CA, USA.
| |
Collapse
|
14442
|
Preoperative metabolic classification of thyroid nodules using mass spectrometry imaging of fine-needle aspiration biopsies. Proc Natl Acad Sci U S A 2019; 116:21401-21408. [PMID: 31591199 DOI: 10.1073/pnas.1911333116] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Thyroid neoplasia is common and requires appropriate clinical workup with imaging and fine-needle aspiration (FNA) biopsy to evaluate for cancer. Yet, up to 20% of thyroid nodule FNA biopsies will be indeterminate in diagnosis based on cytological evaluation. Genomic approaches to characterize the malignant potential of nodules showed initial promise but have provided only modest improvement in diagnosis. Here, we describe a method using metabolic analysis by desorption electrospray ionization mass spectrometry (DESI-MS) imaging for direct analysis and diagnosis of follicular cell-derived neoplasia tissues and FNA biopsies. DESI-MS was used to analyze 178 tissue samples to determine the molecular signatures of normal, benign follicular adenoma (FTA), and malignant follicular carcinoma (FTC) and papillary carcinoma (PTC) thyroid tissues. Statistical classifiers, including benign thyroid versus PTC and benign thyroid versus FTC, were built and validated with 114,125 mass spectra, with accuracy assessed in correlation with clinical pathology. Clinical FNA smears were prospectively collected and analyzed using DESI-MS imaging, and the performance of the statistical classifiers was tested with 69 prospectively collected clinical FNA smears. High performance was achieved for both models when predicting on the FNA test set, which included 24 nodules with indeterminate preoperative cytology, with accuracies of 93% and 89%. Our results strongly suggest that DESI-MS imaging is a valuable technology for identification of malignant potential of thyroid nodules.
Collapse
|
14443
|
Metz EP, Wilder PJ, Dong J, Datta K, Rizzino A. Elevating SOX2 in prostate tumor cells upregulates expression of neuroendocrine genes, but does not reduce the inhibitory effects of enzalutamide. J Cell Physiol 2019; 235:3731-3740. [PMID: 31587305 DOI: 10.1002/jcp.29267] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/18/2019] [Indexed: 01/02/2023]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer deaths in men. In this cancer, the stem cell transcription factor SOX2 increases during tumor progression, especially as the cancer progresses to the highly aggressive neuroendocrine-like phenotype. Other studies have shown that knockdown of RB1 and TP53 increases the expression of neuroendocrine markers, decreases the sensitivity to enzalutamide, and increases the expression of SOX2. Importantly, knockdown of SOX2 in the context of RB1 and TP53 depletion restored sensitivity to enzalutamide and reduced the expression of neuroendocrine markers. In this study, we examined whether elevating SOX2 is not only necessary, but also sufficient on its own to promote the expression of neuroendocrine markers and confer enzalutamide resistance. For this purpose, we engineered LNCaP cells for inducible overexpression of SOX2 (i-SOX2-LNCaP). As shown previously for other tumor cell types, inducible elevation of SOX2 in i-SOX2-LNCaP inhibited cell proliferation. SOX2 elevation also increased the expression of several neuroendocrine markers, including several neuropeptides and synaptophysin. However, SOX2 elevation did not decrease the sensitivity of i-SOX2-LNCaP cells to enzalutamide, which indicates that elevating SOX2 on its own is not sufficient to confer enzalutamide resistance. Furthermore, knocking down SOX2 in C4-2B cells, a derivative of LNCaP cells which is far less sensitive to enzalutamide and which expresses much higher levels of SOX2 than LNCaP cells, did not alter the growth response to this antiandrogen. Thus, our studies indicate that NE marker expression can increase independently of the sensitivity to enzalutamide.
Collapse
Affiliation(s)
- Ethan P Metz
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Phillip J Wilder
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Biochemistry and Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
14444
|
Vicari M, Guidobaldi L, Perrella E, Bianchi A, Ferrari S, Martucci M, Petitti T, Rabitti C, Crescenzi A. Morphometric analysis of atypical glandular cells correctly classifies normal, reactive, and atypical cells in cervical smears. Diagn Cytopathol 2019; 48:10-16. [DOI: 10.1002/dc.24320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/29/2019] [Accepted: 09/06/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Marco Vicari
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| | - Leo Guidobaldi
- Cytodiagnostic Service, Microbiology and Virology Unit“Sandro Pertini” Hospital Rome Italy
| | - Eleonora Perrella
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| | - Antonella Bianchi
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| | - Simona Ferrari
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| | | | - Tommasangelo Petitti
- Hygiene, Public Health and statistics Research UnitCampus Bio‐Medico University Rome Italy
| | - Carla Rabitti
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| | - Anna Crescenzi
- Pathology UnitUniversity Hospital Campus Bio‐Medico of Rome Rome Italy
| |
Collapse
|
14445
|
Mining expression and prognosis of FOLR1 in ovarian cancer by using Oncomine and Kaplan-Meier plotter. Pteridines 2019. [DOI: 10.1515/pteridines-2019-0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abstract
Objective: To further explore folate receptor 1 (FOLR1) gene expression in ovarian cancer and its association with patients’ prognosis by deep mining the Oncomine and Kaplan-Meier plotter databases.
Methods:
FOLR1 mRNA expression data of ovarian cancer were retrieved from the Oncomine database and further analyzed by comparing tumor to healthy tissue. The prognostic value of FOLR1 in ovarian cancer was analyzed by Kaplan-Meier Plotter, an online survival analysis database.
Results A total of 439 studies were included in the Oncomine database in multiple types of cancers. Of the 439 studies, there were 54 with statistical differences for the expression of FOLR1, 19 with increased expression of FOLR1 and 35 with decreased expression comparing ovarian cancer to normal ovary tissue. After searching the Oncomine database, six datasets were discovered comparing the mRNA expression in ovarian tumor to healthy tissue. FOLR1 mRNA expression in ovarian tumor was significantly higher than that of normal ovarian tissue (all p<0.05). The Kaplan-Meier Plotter database analyzed the correlation between FOLR1 expression and ovarian cancer patient’s prognosis. A significant difference of progression-free survival between FOLR1 high and low expressing groups was found in ovarian cancer patients (HR=1.14, 95%CI: 1.00-1.29, p=0.043). However, the overall survival was not statistically different between high and low FOLR1 expressing patients (HR=0.95, 95%CI: 0.84-1.09, p=0.48).
Conclusion
FOLR1 mRNA was found to be highly expressed in ovarian tumor compared to normal ovarian tissue. Elevated FOLR1 mRNA expression was associated with the poor progression-free survival.
Collapse
|
14446
|
Association between Itch and Cancer in 3836 Pediatric Pruritus Patients at a Tertiary Care Center. MEDICINES 2019; 6:medicines6040099. [PMID: 31590346 PMCID: PMC6963483 DOI: 10.3390/medicines6040099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
Background: Pruritus is a well-recognized paraneoplastic phenomenon. Previous studies have examined the association of itch with a variety of malignancies in adults. However, no large study has examined this association in a pediatric population. Methods: A retrospective study was conducted of patients age 18 or less seen at Johns Hopkins Health System between 2012 and 2019. Results: A pediatric hospital population of 1,042,976 patients was reviewed. Pruritus was observed in 3836 pediatric patients of whom 130 also had cancer. Pediatric patients with pruritus were significantly more likely to have concomitant malignancy compared to pediatric patients without pruritus (OR 12.84; 95% CI 10.73–15.35, p < 0.001). Malignancies most strongly associated with pruritus included neoplasms of the blood (OR 14.38; 95% CI 11.30–18.29, p < 0.001), bone (OR 29.02, 95% CI 18.28–46.06, p < 0.001) and skin (OR 22.76, 95% CI 9.14–56.72, p < 0.001. Conclusions: Pruritus is significantly associated with malignancy in the pediatric hospital population. Clinicians should also be aware of the high burden of itch in pediatric malignancies and the variation in pruritus across malignancies.
Collapse
|
14447
|
Rao B, Leng X, Zeng Y, Lin Y, Chen R, Zhou Q, Hagemann AR, Kuroki LM, McCourt CK, Mutch DG, Powell MA, Hagemann IS, Zhu Q. Optical Resolution Photoacoustic Microscopy of Ovary and Fallopian Tube. Sci Rep 2019; 9:14306. [PMID: 31586106 PMCID: PMC6778126 DOI: 10.1038/s41598-019-50743-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer is the leading cause of death among gynecological cancers, but is poorly amenable to preoperative diagnosis. In this study, we investigate the feasibility of "optical biopsy," using high-optical-resolution photoacoustic microscopy (OR-PAM) to quantify the microvasculature of ovarian and fallopian tube tissue. The technique is demonstrated using excised human ovary and fallopian tube specimens imaged immediately after surgery. Quantitative parameters are derived using Amira software. The parameters include three-dimensional vascular segment count, total volume and length, which are associated with tumor angiogenesis. Qualitative results of OR-PAM demonstrate that malignant ovarian tissue has larger and more tortuous blood vessels as well as smaller vessels of different sizes, while benign and normal ovarian tissue has smaller vessels of uniform size. Quantitative analysis shows that malignant ovaries have greater tumor vessel volume, length and number of segments, as compared with benign and normal ovaries. The vascular pattern of benign fallopian tube is different than that of benign ovarian tissue. Our initial results demonstrate the potential of OR-PAM as an imaging tool for fast assessment of ovarian tissue and fallopian tube and could avoid unnecessary surgery if the risk of the examined ovary is extremely low.
Collapse
Affiliation(s)
- Bin Rao
- Biomedical Engineering, Washington University, St Louis, MO, 63130, USA
- Applied Bioptics LLC, St Louis, MO, 63146, USA
| | - Xiandong Leng
- Biomedical Engineering, Washington University, St Louis, MO, 63130, USA
| | - Yifeng Zeng
- Biomedical Engineering, Washington University, St Louis, MO, 63130, USA
| | - Yixiao Lin
- Biomedical Engineering, Washington University, St Louis, MO, 63130, USA
| | - Ruimin Chen
- Department of Biomedical Engineering and Ophthalmology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Qifa Zhou
- Department of Biomedical Engineering and Ophthalmology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Andrea R Hagemann
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lindsay M Kuroki
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Carolyn K McCourt
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David G Mutch
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Matthew A Powell
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ian S Hagemann
- Department of Obstetrics & Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Quing Zhu
- Biomedical Engineering, Washington University, St Louis, MO, 63130, USA.
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
14448
|
He Z, Duan X, Zeng G. Identification of potential biomarkers and pivotal biological pathways for prostate cancer using bioinformatics analysis methods. PeerJ 2019; 7:e7872. [PMID: 31598425 PMCID: PMC6779116 DOI: 10.7717/peerj.7872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background Prostate cancer (PCa) is a common urinary malignancy, whose molecular mechanism has not been fully elucidated. We aimed to screen for key genes and biological pathways related to PCa using bioinformatics method. Methods Differentially expressed genes (DEGs) were filtered out from the GSE103512 dataset and subjected to the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The protein–protein interactions (PPI) network was constructed, following by the identification of hub genes. The results of former studies were compared with ours. The relative expression levels of hub genes were examined in The Cancer Genome Atlas (TCGA) and Oncomine public databases. The University of California Santa Cruz Xena online tools were used to study whether the expression of hub genes was correlated with the survival of PCa patients from TCGA cohorts. Results Totally, 252 (186 upregulated and 66 downregulated) DEGs were identified. GO analysis enriched mainly in “oxidation-reduction process” and “positive regulation of transcription from RNA polymerase II promoter”; KEGG pathway analysis enriched mostly in “metabolic pathways” and “protein digestion and absorption.” Kallikrein-related peptidase 3, cadherin 1 (CDH1), Kallikrein-related peptidase 2 (KLK2), forkhead box A1 (FOXA1), and epithelial cell adhesion molecule (EPCAM) were identified as hub genes from the PPI network. CDH1, FOXA1, and EPCAM were validated by other relevant gene expression omnibus datasets. All hub genes were validated by both TCGA and Oncomine except KLK2. Two additional top DEGs (ABCC4 and SLPI) were found to be associated with the prognosis of PCa patients. Conclusions This study excavated the key genes and pathways in PCa, which might be biomarkers for diagnosis, prognosis, and potential therapeutic targets.
Collapse
Affiliation(s)
- Zihao He
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| |
Collapse
|
14449
|
Senapati D, Kumari S, Heemers HV. Androgen receptor co-regulation in prostate cancer. Asian J Urol 2019; 7:219-232. [PMID: 32742924 PMCID: PMC7385509 DOI: 10.1016/j.ajur.2019.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/30/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) progression relies on androgen receptor (AR) action. Preventing AR's ligand-activation is the frontline treatment for metastatic PCa. Androgen deprivation therapy (ADT) that inhibits AR ligand-binding initially induces remission but eventually fails, mainly because of adaptive PCa responses that restore AR action. The vast majority of castration-resistant PCa (CRPC) continues to rely on AR activity. Novel therapeutic strategies are being explored that involve targeting other critical AR domains such as those that mediate its constitutively active transactivation function, its DNA binding ability, or its interaction with co-operating transcriptional regulators. Considerable molecular and clinical variability has been found in AR's interaction with its ligands, DNA binding motifs, and its associated coregulators and transcription factors. Here, we review evidence that each of these levels of AR regulation can individually and differentially impact transcription by AR. In addition, we examine emerging insights suggesting that each can also impact the other, and that all three may collaborate to induce gene-specific AR target gene expression, likely via AR allosteric effects. For the purpose of this review, we refer to the modulating influence of these differential and/or interdependent contributions of ligands, cognate DNA-binding motifs and critical regulatory protein interactions on AR's transcriptional output, which may influence the efficiency of the novel PCa therapeutic approaches under consideration, as co-regulation of AR activity.
Collapse
Affiliation(s)
| | - Sangeeta Kumari
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Hannelore V Heemers
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA.,Department of Urology, Cleveland Clinic, Cleveland, OH, USA.,Department of Hematology/Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
14450
|
Lin X, Kapoor A, Gu Y, Chow MJ, Xu H, Major P, Tang D. Assessment of biochemical recurrence of prostate cancer (Review). Int J Oncol 2019; 55:1194-1212. [PMID: 31638194 PMCID: PMC6831208 DOI: 10.3892/ijo.2019.4893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The assessment of the risk of biochemical recurrence (BCR) is critical in the management of males with prostate cancer (PC). Over the past decades, a comprehensive effort has been focusing on improving risk stratification; a variety of models have been constructed using PC-associated pathological features and molecular alterations occurring at the genome, protein and RNA level. Alterations in RNA expression (lncRNA, miRNA and mRNA) constitute the largest proportion of the biomarkers of BCR. In this article, we systemically review RNA-based BCR biomarkers reported in PubMed according to the PRISMA guidelines. Individual miRNAs, mRNAs, lncRNAs and multi-gene panels, including the commercially available signatures, Oncotype DX and Prolaris, will be discussed; details related to cohort size, hazard ratio and 95% confidence intervals will be provided. Mechanistically, these individual biomarkers affect multiple pathways critical to tumorigenesis and progression, including epithelial-mesenchymal transition (EMT), phosphatase and tensin homolog (PTEN), Wnt, growth factor receptor, cell proliferation, immune checkpoints and others. This variety in the mechanisms involved not only validates their associations with BCR, but also highlights the need for the coverage of multiple pathways in order to effectively stratify the risk of BCR. Updates of novel biomarkers and their mechanistic insights are considered, which suggests new avenues to pursue in the prediction of BCR. Additionally, the management of patients with BCR and the potential utility of the stratification of the risk of BCR in salvage treatment decision making for these patients are briefly covered. Limitations will also be discussed.
Collapse
Affiliation(s)
- Xiaozeng Lin
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anil Kapoor
- The Research Institute of St. Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yan Gu
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mathilda Jing Chow
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pierre Major
- Division of Medical Oncology, Department of Oncology, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Damu Tang
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|