14451
|
Li Q, Chen Y, Zhang D, Grossman J, Li L, Khurana N, Jiang H, Grierson PM, Herndon J, DeNardo DG, Challen GA, Liu J, Ruzinova MB, Fields RC, Lim KH. IRAK4 mediates colitis-induced tumorigenesis and chemoresistance in colorectal cancer. JCI Insight 2019; 4:130867. [PMID: 31527315 DOI: 10.1172/jci.insight.130867] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/04/2019] [Indexed: 01/05/2023] Open
Abstract
Aberrant activation of the NF-κB transcription factors underlies chemoresistance in various cancer types, including colorectal cancer (CRC). Targeting the activating mechanisms, particularly with inhibitors to the upstream IκB kinase (IKK) complex, is a promising strategy to augment the effect of chemotherapy. However, clinical success has been limited, largely because of low specificity and toxicities of tested compounds. In solid cancers, the IKKs are driven predominantly by the Toll-like receptor (TLR)/IL-1 receptor family members, which signal through the IL-1 receptor-associated kinases (IRAKs), with isoform 4 (IRAK4) being the most critical. The pathogenic role and therapeutic value of IRAK4 in CRC have not been investigated. We found that IRAK4 inhibition significantly abrogates colitis-induced neoplasm in APCMin/+ mice, and bone marrow transplant experiments showed an essential role of IRAK4 in immune cells during neoplastic progression. Chemotherapy significantly enhances IRAK4 and NF-κB activity in CRC cells through upregulating TLR9 expression, which can in turn be suppressed by IRAK4 and IKK inhibitors, suggesting a feed-forward pathway that protects CRC cells from chemotherapy. Lastly, increased tumor phospho-IRAK4 staining or IRAK4 mRNA expression is associated with significantly worse survival in CRC patients. Our results support targeting IRAK4 to improve the effects of chemotherapy and outcomes in CRC.
Collapse
Affiliation(s)
- Qiong Li
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yali Chen
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daoxiang Zhang
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Lin Li
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Namrata Khurana
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hongmei Jiang
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Herndon
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David G DeNardo
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Grant A Challen
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingxia Liu
- Division of Public Health Sciences, Department of Surgery, and
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
14452
|
Lu H, Lu Y, Xie Y, Qiu S, Li X, Fan Z. Rational combination with PDK1 inhibition overcomes cetuximab resistance in head and neck squamous cell carcinoma. JCI Insight 2019; 4:131106. [PMID: 31578313 DOI: 10.1172/jci.insight.131106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/31/2019] [Indexed: 12/28/2022] Open
Abstract
Cetuximab, an EGFR-blocking antibody, is currently approved for treatment of metastatic head and neck squamous cell carcinoma (HNSCC), but its response rate is limited. In addition to blocking EGFR-stimulated cell signaling, cetuximab can induce endocytosis of ASCT2, a glutamine transporter associated with EGFR in a complex, leading to glutathione biosynthesis inhibition and cellular sensitization to ROS. Pyruvate dehydrogenase kinase-1 (PDK1), a key mitochondrial enzyme overexpressed in cancer cells, redirects glucose metabolism from oxidative phosphorylation toward aerobic glycolysis. In this study, we tested the hypothesis that targeting PDK1 is a rational approach to synergize with cetuximab through ROS overproduction. We found that combination of PDK1 knockdown or inhibition by dichloroacetic acid (DCA) with ASCT2 knockdown or with cetuximab treatment induced ROS overproduction and apoptosis in HNSCC cells, and this effect was independent of effective inhibition of EGFR downstream pathways but could be lessened by N-acetyl cysteine, an anti-oxidative agent. In several cetuximab-resistant HNSCC xenograft models, DCA plus cetuximab induced marked tumor regression, whereas either agent alone failed to induce tumor regression. Our findings call for potentially novel clinical trials of combining cetuximab and DCA in patients with cetuximab-sensitive EGFR-overexpressing tumors and patients with cetuximab-resistant EGFR-overexpressing tumors.
Collapse
Affiliation(s)
- Haiquan Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Yang Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yangyiran Xie
- Program in Neuroscience, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Songbo Qiu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xinqun Li
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Fan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
14453
|
Zhang L, Huang P, Li Q, Wang D, Xu CX. miR-134-5p Promotes Stage I Lung Adenocarcinoma Metastasis and Chemoresistance by Targeting DAB2. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:627-637. [PMID: 31689617 PMCID: PMC6838973 DOI: 10.1016/j.omtn.2019.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/15/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
Despite surgery and adjuvant therapy, early-stage lung adenocarcinoma (LUAD) treatment often fails due to local or metastatic recurrence. However, the mechanism is largely unknown. Here, we report that increased expression levels of miR-134-5p and decreased levels of disabled-2 (DAB2) were significantly correlated with recurrence in stage I LUAD patients. Our data show that miR-134-5p overexpression or DAB2 silencing strongly stimulated LUAD cell metastasis and chemoresistance. In contrast, inhibition of miR-134-5p or overexpression of DAB2 strongly suppressed LUAD cell metastasis and overcame the insensitivity of chemoresistant LUAD cells to chemotherapy. In addition, we demonstrated that DAB2 is a target of miR-134-5p and that miR-134-5p stimulates chemoresistance and metastasis through DAB2 in LUAD. Taken together, these findings suggest that miR-134-5p and its target gene DAB2 have potential as a biomarker for predicting recurrence in stage I LUAD patients. Additionally, miR-134-5p inhibition or DAB2 restoration may be a novel strategy for inhibiting LUAD metastasis and overcoming LUAD cell resistance to chemotherapy.
Collapse
Affiliation(s)
- Liang Zhang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Ping Huang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Qing Li
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| | - Cheng-Xiong Xu
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
14454
|
Guo F, Long L, Wang J, Wang Y, Liu Y, Wang L, Luo F. Insights on CXC chemokine receptor 2 in breast cancer: An emerging target for oncotherapy. Oncol Lett 2019; 18:5699-5708. [PMID: 31788042 PMCID: PMC6865047 DOI: 10.3892/ol.2019.10957] [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: 04/23/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common malignant neoplasm in women worldwide, and the treatment regimens currently available are far from optimal. Targeted therapy, based on molecular typing of breast cancer, is the most precise form of treatment, and CXC chemokine receptor 2 (CXCR2) is one of the molecular markers used in targeted therapies. As a member of the seven transmembrane G-protein-coupled receptor family, CXCR2 and its associated ligands have been increasingly implicated in tumor-associated processes. These processes include proliferation, angiogenesis, invasion, metastasis, chemoresistance, and stemness and phenotypic maintenance of cancer stem cells. Thus, the inhibition of CXCR2 or its downstream signaling pathways could significantly attenuate tumor progression. Therefore, studies on the biological functions of CXCR2 and its association with neoplasia may help improve the prognosis of breast cancer. Furthermore, the targeting of CXCR2 could supplement the present clinical approaches of breast cancer treatment strategies. The present review discusses the structures and mechanisms of CXCR2 and its ligands. Additionally, the contribution of CXCR2 to the development of breast cancer and its potential therapeutic benefits are also discussed.
Collapse
Affiliation(s)
- Fengzhu Guo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lang Long
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiantao Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuyi Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanyang Liu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Wang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Luo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
14455
|
Association of rs8444 polymorphism in the LASS2 3'-UTR and bladder cancer risk in Chinese population. Eur J Cancer Prev 2019; 29:329-337. [PMID: 31577563 DOI: 10.1097/cej.0000000000000551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of the present study was to explore the correlations between single nucleotide polymorphisms in LASS2 gene 3'-untranslated regions and bladder cancer risk in Chinese population. We first performed PCR and sequence for LASS2-3'-UTR in 105 bladder cancer patients and 100 control subjects. Next, multivariate logistic regression analysis was used to determine the relationship between single nucleotide polymorphisms frequency and susceptibility of bladder cancer, and clinical features in 105 cases. In addition, survival curves and Cox Regression analysis were used to investigate the effect of single nucleotide polymorphisms on clinical outcome in 58 cases. Finally, quantitative reverse-transcription PCR and immunohistochemical were performed to explore the influence of single nucleotide polymorphisms on LASS2 expression. We found that a single nucleotide polymorphism (rs8444 C>T) located in the 3'-UTR of LASS2 was significantly associated with the risk of bladder cancer. We also showed the frequency of rs8444 T genotype was higher in bladder cancer group and correlated with the risk of clinical prognosis. Yet, there were no significant correlations between T/C allele frequencies and the distributions of rs8444 genotype and tumor-node-metastasis stage, histological grade and distant metastasis in bladder cancer. Furthermore, we demonstrated that rs8444 C>T could affect LASS2 expression by single nucleotide polymorphism-related mRNA stability. Our results showed that LASS2-3'-UTR rs8444 C>T polymorphism was significantly associated with the individual risk and the poor overall survival of bladder cancer, suggesting that rs8444 TT genotype maybe act as an independent risk factor of susceptibility and clinical prognosis for bladder cancer in Chinese population.
Collapse
|
14456
|
Nocon CC, Ajmani GS, Bhayani MK. A contemporary analysis of racial disparities in recommended and received treatment for head and neck cancer. Cancer 2019; 126:381-389. [PMID: 31580491 DOI: 10.1002/cncr.32342] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/21/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Racial disparities in squamous cell carcinoma of the head and neck (HNSCC) negatively affect non-Hispanic black (NHB) patients. This study was aimed at understanding how treatment is prescribed and received across all HNSCC subsites. METHODS With the National Cancer Database, patients from 2004 to 2014 with surgically resectable HNSCCs, including tumors of the oral cavity (OC), oropharynx (OP), hypopharynx (HP), and larynx (LX), were studied. The treatment received was either upfront surgery or nonsurgical treatment. Treatment patterns were compared according to race and subsite, and how these differences changed over time was evaluated. RESULTS NHB patients were less likely than non-Hispanic white (NHW) patients to receive surgery across all subsites (relative risk [RR] for OC, 0.87; RR for OP, 0.75; RR for HP, 0.73; RR for LX, 0.87; all P values <.05). They were also more likely to refuse a recommended surgery (RR for OC, 1.50; RR for OP, 1.23; RR for HP, 1.23; RR for LX, 1.34), and this difference was significant except for HP. NHB patients were more likely to not be offered surgery across all subsites (RR for OC, 1.38; RR for OP, 1.07; RR for HP, 1.05; RR for LX, 1.03; all P values <.05). Rates of surgery increased and rates of not being offered surgery declined for both NHB and NHW patients from 2004 to 2014, but the absolute disparities persisted in 2014. CONCLUSIONS Across all HNSCC subsites, NHB patients were less likely than NHW patients to be recommended for and receive surgery and were more likely to refuse surgery. These differences have closed over time but persist. Enhanced shared decision making may improve these disparities.
Collapse
Affiliation(s)
- Cheryl C Nocon
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois.,Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, Illinois
| | - Gaurav S Ajmani
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mihir K Bhayani
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois.,Department of Surgery, Pritzker School of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
14457
|
Xu L, Shi L, Qiu S, Chen S, Lin M, Xiang Y, Zhao C, Zhu J, Shen L, Zuo Z. Design, Synthesis, And Evaluation Of Cyanopyridines As Anti-Colorectal Cancer Agents Via Inhibiting STAT3 Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3369-3381. [PMID: 31576111 PMCID: PMC6768128 DOI: 10.2147/dddt.s217800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022]
Abstract
Background Colorectal cancer is one of the common malignant tumors. Cyanopyridine and aminocyanopyridine having a carbon-nitrogen bond have been shown to have significant anticancer effects. STAT3 is a promising therapeutic target in multiple cancers. However, there are currently no effective STAT3 inhibitors in clinical practice for the treatment of colorectal cancer. Materials and methods We screened 27 cyanopyridines for their anticancer activity by cell viability. The HCT-116, RKO, and DLD-1 cell lines were used to evaluate the anti-colorectal cancer effect of 3n. Scratch experiments and colony formation assays were used for the assessment of cell migration and proliferation capacity. Phosphorylated STAT3, STAT3, MCL-1, and Survivin levels were assessed by Western blot analysis. Results In this study, we synthesized 27 cyanopyridines and screened their anticancer activities in three human tumor cells, HCT-116, Hela229, and A375. We found that 2-amino-3-cyanopyridine 3n has better anticancer activity with IC50 values in the low micromolar range. Furthermore, 3n significantly inhibited the migration and colony formation of colorectal cancer cells. Mechanistically, 3n inhibited the expression of STAT3 phosphorylation in a dose- and time-dependent manner. Conclusion 3n is worth of further investigations toward the discovery of STAT3 inhibitor as a drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Lingyuan Xu
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China.,Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China.,Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Lingxi Shi
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China.,Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China.,Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Sensen Qiu
- Department of Pharmaceutical Sciences, College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning 530006, People's Republic of China
| | - Siyu Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China.,Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China.,Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Mengsha Lin
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China.,Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China.,Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Youqun Xiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Chengguang Zhao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jiandong Zhu
- Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China
| | - Liqun Shen
- Department of Pharmaceutical Sciences, College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning 530006, People's Republic of China
| | - Zhigui Zuo
- Department of Colorectal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China.,Department of Surgical Oncology, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China
| |
Collapse
|
14458
|
Nguyen D, vanSonnenberg E, Kang P, Mueller PR. Urologic and interventional radiology treatment of renal cell carcinomas-similarities and differences. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S113. [PMID: 31576320 DOI: 10.21037/atm.2019.05.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Diep Nguyen
- Department of Student Affairs, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - Eric vanSonnenberg
- Department of Student Affairs, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA.,Departments of Radiology & Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Paul Kang
- Department of Student Affairs, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - Peter R Mueller
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14459
|
Wang J, Shen T, Wang Q, Zhang T, Li L, Wang Y, Fang Y. The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma: a meta-analysis. Immunotherapy 2019; 11:1325-1335. [PMID: 31578914 DOI: 10.2217/imt-2019-0079] [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: 01/30/2023] Open
Abstract
Aim: The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma patients after curative treatments remains controversial. Methods: A meta-analysis was conducted, and the outcomes were the recurrence rate and overall survival. Results: Eight randomized clinical trials with 1038 participants were included. Compared with the control group, cytokine-induced killer cellular therapy group could reduce 1-year, 3-year recurrence rates, as well as improve 1-5 years overall survival for hepatocellular carcinoma patients (p < 0.05). However, it failed to affect the 5-year recurrence rate and 6-year overall survival (p > 0.05). Conclusion: Cytokine-induced killer cellular adjuvant therapy exerted a favorable role in improving early and long-term efficacy for hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Jiaxue Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tiantian Shen
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qi Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tan Zhang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Lujin Li
- The Center for Drug Clinical Research of Shanghai University of TCM, Shanghai, China
| | - Yu Wang
- Immunotech Applied Science Limited, Beijing, China
| | - Yi Fang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
14460
|
Sindeeva OA, Verkhovskii RA, Sarimollaoglu M, Afanaseva GA, Fedonnikov AS, Osintsev EY, Kurochkina EN, Gorin DA, Deyev SM, Zharov VP, Galanzha EI. New Frontiers in Diagnosis and Therapy of Circulating Tumor Markers in Cerebrospinal Fluid In Vitro and In Vivo. Cells 2019; 8:E1195. [PMID: 31581745 PMCID: PMC6830088 DOI: 10.3390/cells8101195] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/21/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
One of the greatest challenges in neuro-oncology is diagnosis and therapy (theranostics) of leptomeningeal metastasis (LM), brain metastasis (BM) and brain tumors (BT), which are associated with poor prognosis in patients. Retrospective analyses suggest that cerebrospinal fluid (CSF) is one of the promising diagnostic targets because CSF passes through central nervous system, harvests tumor-related markers from brain tissue and, then, delivers them into peripheral parts of the human body where CSF can be sampled using minimally invasive and routine clinical procedure. However, limited sensitivity of the established clinical diagnostic cytology in vitro and MRI in vivo together with minimal therapeutic options do not provide patient care at early, potentially treatable, stages of LM, BM and BT. Novel technologies are in demand. This review outlines the advantages, limitations and clinical utility of emerging liquid biopsy in vitro and photoacoustic flow cytometry (PAFC) in vivo for assessment of CSF markers including circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNA (miRNA), proteins, exosomes and emboli. The integration of in vitro and in vivo methods, PAFC-guided theranostics of single CTCs and targeted drug delivery are discussed as future perspectives.
Collapse
Affiliation(s)
- Olga A. Sindeeva
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
| | - Roman A. Verkhovskii
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
| | - Mustafa Sarimollaoglu
- Arkansas Nanomedicine Center & Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Galina A. Afanaseva
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Saratov State Medical University, 112 Bolshaya Kazachia St., 410012 Saratov, Russia
| | - Alexander S. Fedonnikov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Saratov State Medical University, 112 Bolshaya Kazachia St., 410012 Saratov, Russia
| | - Evgeny Yu. Osintsev
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Saratov State Medical University, 112 Bolshaya Kazachia St., 410012 Saratov, Russia
| | - Elena N. Kurochkina
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Saratov State Medical University, 112 Bolshaya Kazachia St., 410012 Saratov, Russia
| | - Dmitry A. Gorin
- Laboratory of Biophotonics, Skolkovo Institute of Science and Technology, 3 Nobelya Str., 121205 Moscow, Russia;
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia;
| | - Vladimir P. Zharov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Arkansas Nanomedicine Center & Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Ekaterina I. Galanzha
- Laboratory of Biomedical Photoacoustics, Saratov State University, 83 Astrakhanskaya St, 410012 Saratov, Russia; (O.A.S.); (R.A.V.); (G.A.A.); (A.S.F.); (E.Y.O.); (E.N.K.); (V.P.Z.)
- Laboratory of Lymphatic Research, Diagnosis and Therapy (LDT), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
14461
|
Petrioli R, Marrelli D, Roviello F, D'Ignazio A, Torre P, Chirra M, Savelli V, Ambrosio MR, Francini G, Calomino N, Farsi M, Vernillo R, Francini E. Pathological response and outcome after neoadjuvant chemotherapy with DOC (docetaxel, oxaliplatin, capecitabine) or EOF (epirubicin, oxaliplatin, 5-fluorouracil) for clinical T3-T4 non-metastatic gastric cancer. Surg Oncol 2019; 32:2-7. [PMID: 31670056 DOI: 10.1016/j.suronc.2019.10.002] [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] [Received: 07/10/2019] [Revised: 08/29/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE In this prospective observational study, we sought to compare the efficacy and safety of docetaxel + oxaliplatin + capecitabine (DOC) with epirubicin + oxaliplatin + 5-fluouracil (EOF) as neoadjuvant chemotherapy (NAC) for clinical T3 or T4 non-metastatic gastric cancer (GC) patients. METHODS The DOC NAC consisted of docetaxel 35 mg/m2 (days 1-8), oxaliplatin 85 mg/m2 (day 1), and capecitabine 750 mg/m2 twice daily (days 1-14), every 3 weeks. The EOF NAC consisted of intravenous (IV) epirubicin 50 mg/m2 combined with IV oxaliplatin 130 mg/m2 on day 1 and continuous infusion 5-fluouracil 750 mg/m2 on days 1-5, every 3 weeks. After 4 cycles of NAC or upon progression during chemotherapy, patients underwent gastrectomy with standard D2 or D3 lymphadenectomy. Pathological complete response rate per Becker tumor regression grading system was the primary endpoint and the secondary endpoints included progression-free survival (2-yr PFS) and 2-year overall survival (2-yr OS) and tolerability. RESULTS Overall, we identified 63 patients with T3-4 non-metastatic GC starting either NAC regimen between January 2010 and December 2017 at our Institution: 34 in the DOC group and 29 in EOF group. Thirty patients (88%) in the DOC group and 22 (76%) in the EOF group completed the 4 planned cycles of NAC. Fifty-seven patients received surgery. Results indicated no statistical significant differences between the two groups, and only a trend for some better data in favour of the DOC group. The R0 resection rate was 90.6% and 88.0% for the DOC and EOF cohorts, respectively. The pathological complete response rate was 6.2% in the DOC group and 4.0% in the EOF group. Becker 1-2 pathological response was found in 46.8% of the DOC cohort and 28.0% of the EOF cohort (p = .14). The 2-yr PFS rate was 54.1% for DOC vs. 41.4% for EOF (p = .14) and the 2-yr OS rate was 80.8% for DOC vs. 58.6% for EOF (p = .05). Neutropenia was the most common grade ≥3 toxicity and occurred in 8 (23.5%) patients of the DOC group and 10 (34.4%) patients of the EOF group (p = .33). CONCLUSIONS These findings seem to confirm the feasibility of NAC for clinically T3 and T4 non-metastatic GC and, despite no statistical significant difference was documented, suggest a trend for better activity and tolerability for the docetaxel-based regimen (DOC) compared to the epirubicin-based combination (EOF).
Collapse
Affiliation(s)
- Roberto Petrioli
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy.
| | - Daniele Marrelli
- General Surgery and Surgical Oncology Department, University of Siena, Italy
| | - Franco Roviello
- General Surgery and Surgical Oncology Department, University of Siena, Italy
| | - Alessia D'Ignazio
- General Surgery and Surgical Oncology Department, University of Siena, Italy
| | - Pamela Torre
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Martina Chirra
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Vinno Savelli
- General Surgery and Surgical Oncology Department, University of Siena, Italy
| | | | - Guido Francini
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Natale Calomino
- Clinical Surgery, Department of Surgery and Bioengineering, University of Siena, Siena, Italy
| | - Marco Farsi
- Dept Medical Biotechnology, Section of Pathology, University of Siena, Italy
| | - Remo Vernillo
- Clinical Surgery and Surgical Endoscopy, University of Siena, Siena, Italy
| | - Edoardo Francini
- La Sapienza University, Rome, Italy; Oncology Unit, Misericordia Hospital, Grosseto, Italy
| |
Collapse
|
14462
|
Liu W, Peng JF, Tang MJ. Individualized Treatment Analysis Of Breast Cancer With Chronic Renal Failure. Onco Targets Ther 2019; 12:7767-7772. [PMID: 31571926 PMCID: PMC6759282 DOI: 10.2147/ott.s223729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/10/2019] [Indexed: 12/20/2022] Open
Abstract
We report the case of a breast cancer patient with chronic renal failure (CRF). The clinical pharmacist adjusted the chemotherapy regimen and dosage according to the patient’s renal function after reviewing the literature and analyzing the pharmacological and pharmacokinetic characteristics of the patient’s antineoplastic drugs. To the best of our knowledge, this is the first report of successful multimodal treatment of breast cancer in a patient with CRF in China. The purpose of this case report is to optimize breast cancer therapy in patients with CRF and provide a reference for clinicians and clinical pharmacists to use antineoplastic drugs rationally.
Collapse
Affiliation(s)
- Wei Liu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, People's Republic of China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Jin-Fu Peng
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, People's Republic of China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Meng-Jie Tang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, People's Republic of China
| |
Collapse
|
14463
|
Katta S, Srivastava A, Thangapazham RL, Rosner IL, Cullen J, Li H, Sharad S. Curcumin-Gene Expression Response in Hormone Dependent and Independent Metastatic Prostate Cancer Cells. Int J Mol Sci 2019; 20:ijms20194891. [PMID: 31581661 PMCID: PMC6801832 DOI: 10.3390/ijms20194891] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/25/2019] [Accepted: 09/29/2019] [Indexed: 12/25/2022] Open
Abstract
The androgen receptor is one of the key targets for prostate cancer treatment. Despite its less satisfactory effects, chemotherapy is the most common treatment option for metastatic and/or castration-resistant patients. There are constant needs for novel anti-prostate cancer therapeutic/prevention agents. Curcumin, a known chemo-preventive agent, was shown to inhibit prostate cancer cell growth. This study aimed to unravel the inhibitory effect of curcumin in prostate cancer through analyzing the alterations of expressions of curcumin targeting genes clusters in androgen-dependent LNCaP cells and androgen-independent metastatic C4-2B cells. Hierarchical clustering showed the highest number of differentially expressed genes at 12 h post treatment in both cells, suggesting that the androgen-dependent/independent manner of curcumin impacts on prostate cancer cells. Evaluation of significantly regulated top canonical pathways highlighted that Transforming growth factor beta (TGF-β), Wingless-related integration site (Wnt), Phosphoinositide 3-kinase/Protein Kinase B/ mammalian target of rapamycin (PIK3/AKT(PKB)/mTOR), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling were primarily inhibited, and Phosphatase and tensin homolog (PTEN) dependent cell cycle arrest and apoptosis pathways were elevated with curcumin treatment. The short term (3–24 h) and long term (48 h) effect of curcumin treatment revealed 31 and four genes modulated in both cell lines. TGF-β signaling, including the androgen/TGF-β inhibitor Prostate transmembrane protein androgen-induced 1 (PMEPA1), was the only pathway impacted by curcumin treatment after 48 h. Our findings also established that MYC Proto-Oncogene, basic helix-loop-helix (bHLH) Transcription Factor (MYC) signaling was down-regulated in curcumin-treated cell lines. This study established, for the first time, novel gene-networks and signaling pathways confirming the chemo-preventive and cancer-growth inhibitory nature of curcumin as a natural anti-prostate cancer compound.
Collapse
Affiliation(s)
- Shilpa Katta
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| | - Arun Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| | - Rajesh L Thangapazham
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| | - Inger L Rosner
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Department of Urology, Walter Reed National Military Medical Center, 8901 Rockville Pike, Bethesda, MD 20889, USA.
| | - Jennifer Cullen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| | - Hua Li
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| | - Shashwat Sharad
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Suite 300, Bethesda, MD 20817, USA.
| |
Collapse
|
14464
|
Synchronous and metachronous bilateral breast cancer: clinicopathologic characteristics and prognostic outcomes. Hum Pathol 2019; 92:1-9. [DOI: 10.1016/j.humpath.2019.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 11/18/2022]
|
14465
|
Eich ML, Rodriguez Pena MDC, Springer S, Taheri D, Tregnago AC, Salles DC, Bezerra SM, Cunha IW, Fujita K, Ertoy D, Bivalacqua TJ, Tomasetti C, Papadopoulos N, Kinzler KW, Vogelstein B, Netto GJ. Incidence and distribution of UroSEEK gene panel in a multi-institutional cohort of bladder urothelial carcinoma. Mod Pathol 2019; 32:1544-1550. [PMID: 31028363 PMCID: PMC6872189 DOI: 10.1038/s41379-019-0276-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 11/22/2022]
Abstract
Noninvasive approaches for early detection of bladder cancer are actively being investigated. We recently developed a urine- based molecular assay for the detection and surveillance of bladder neoplasms (UroSEEK). UroSEEK is designed to detect alterations in 11 genes that include most common genetic alterations in bladder cancer. In this study, we analyzed 527 cases, including 373 noninvasive and 154 invasive urothelial carcinomas of bladder from transurethral resections or cystectomies performed at four institutions (1991-2016). Two different mutational analysis assays of a representative tumor area were performed: first, a singleplex PCR assay for evaluation of the TERT promoter region (TERTSeqS) and second, a multiplex PCR assay using primers designed to amplify regions of interest of 10 (FGFR3, PIK3CA, TP53, HRAS, KRAS, ERBB2, CDKN2A, MET, MLL, and VHL) genes (UroSeqS). Overall, 92% of all bladder tumors were positive for at least one genetic alteration in the UroSEEK panel. We found TERT promoter mutations in 77% of low-grade noninvasive papillary carcinomas, with a relatively lower incidence of 65% in high-grade noninvasive papillary carcinomas and carcinomas in situ; p = 0.017. Seventy-two percent of pT1 and 63% of muscle-invasive bladder tumors harbored TERT promoter mutations with g.1295228C>T alteration being the most common in all groups. FGFR3 and PIK3CA mutations were more frequent in low-grade noninvasive papillary carcinomas compared with high-grade noninvasive papillary carcinomas and carcinomas in situ (p < 0.0001), while the opposite was true for TP53 (p < 0.0001). Significantly higher rates of TP53 and CDKN2A mutation rates (p = 0.005 and 0.035, respectively) were encountered in muscle-invasive bladder tumors compared with those of pT1 stage. The overwhelming majority of all investigated tumors showed at least one mutation among UroSEEK assay genes, confirming the comprehensive coverage of the panel and supporting its potential utility as a noninvasive urine-based assay.
Collapse
Affiliation(s)
- Marie-Lisa Eich
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Simeon Springer
- Howard Hughes Medical Institute, Ludwig Cancer for Cancer Genetics and Therapeutics, Baltimore, United States,Sidney Kimmel Comprehensive Cancer Center, Baltimore, United States
| | - Diana Taheri
- Department of Pathology, Johns Hopkins University, United States,Department of Pathology, Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | - Stephania Martins Bezerra
- Department of Pathology, AC Camargo Cancer Center, Sao Paulo, Brazil,Department of Pathology, Rede D’OR-São Luiz, Sao Paulo, Brazil
| | - Isabela W. Cunha
- Department of Pathology, AC Camargo Cancer Center, Sao Paulo, Brazil
| | | | - Dilek Ertoy
- Department of Pathology, Hacettepe University, Ankara, Turkey
| | | | - Cristian Tomasetti
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States,Division of Biostatistics and Bioinformatics, Department of Oncology, Sidney Kimmel Cancer Center
| | - Nickolas Papadopoulos
- Howard Hughes Medical Institute, Ludwig Cancer for Cancer Genetics and Therapeutics, Baltimore, United States,Sidney Kimmel Comprehensive Cancer Center, Baltimore, United States
| | - Ken W. Kinzler
- Howard Hughes Medical Institute, Ludwig Cancer for Cancer Genetics and Therapeutics, Baltimore, United States,Sidney Kimmel Comprehensive Cancer Center, Baltimore, United States
| | - Bert Vogelstein
- Howard Hughes Medical Institute, Ludwig Cancer for Cancer Genetics and Therapeutics, Baltimore, United States,Sidney Kimmel Comprehensive Cancer Center, Baltimore, United States
| | - George J. Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
14466
|
Unsworth SP, Heisel CJ, Kahana A. A New Paradigm in the Treatment of Advanced Periocular Basal Cell Carcinoma? Am J Ophthalmol 2019; 206:215-216. [PMID: 31957698 DOI: 10.1016/j.ajo.2019.06.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/18/2019] [Accepted: 06/23/2019] [Indexed: 01/17/2023]
|
14467
|
Pu N, Yin L, Habib JR, Gao S, Hu H, Zhu Y, Wu Y, Yu J, Lou W. Optimized modification of the eighth edition of AJCC TNM staging system for resected pancreatic ductal adenocarcinoma. Future Oncol 2019; 15:3457-3465. [PMID: 31460788 DOI: 10.2217/fon-2019-0264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To reassess the prognostic performance of the American Joint Committee on Cancer (AJCC) 8th edition for pancreatic ductal adenocarcinoma (PDAC) and optimize the categorization of PDAC staging. Patients & methods: A total of 11,858 patients with resected PDAC from the Surveillance, Epidemiology and End Results database were retrospectively enrolled by sequential analyses. Results: There was no statistical significance between stage IIA and IIB tumors with hazard ratios of 2.065 and 2.184 (p = 0.620) for stages IIA and IIB, respectively. With the proposed modification, there was a significant difference between the hazard ratios of stages IIIA and IIIB which were 2.481 and 2.715, respectively (p = 0.009). The C-index of modified system was 0.609, slightly higher than AJCC 8th staging system 0.604. Conclusion: We proposed a modified eighth edition of the AJCC staging system by combining stage IIA with IIB and further subclassifying stage III patients in order to lead to better discriminative power.
Collapse
Affiliation(s)
- Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lingdi Yin
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, & Pancreas Institute of Nanjing Medical University, Nanjing, PR China
| | - Joseph R Habib
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shanshan Gao
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Haijie Hu
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biliary Surgery, West China Hospital of Sichuan University, Sichuan, PR China
| | - Yayun Zhu
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Liver Cancer Institute, Zhongshan Hospital, & Key Laboratory of Carcinogenesis & Cancer Invasion (Ministry of Education), Fudan University, Shanghai, PR China
| | - Yong Wu
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Jiangsu, PR China
| | - Jun Yu
- Department of Surgery & The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, PR China
| |
Collapse
|
14468
|
Nikitski AV, Rominski SL, Condello V, Kaya C, Wankhede M, Panebianco F, Yang H, Altschuler DL, Nikiforov YE. Mouse Model of Thyroid Cancer Progression and Dedifferentiation Driven by STRN-ALK Expression and Loss of p53: Evidence for the Existence of Two Types of Poorly Differentiated Carcinoma. Thyroid 2019; 29:1425-1437. [PMID: 31298630 PMCID: PMC6797076 DOI: 10.1089/thy.2019.0284] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Thyroid tumor progression from well-differentiated cancer to poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) involves step-wise dedifferentiation associated with loss of iodine avidity and poor outcomes. ALK fusions, typically STRN-ALK, are found with higher incidence in human PDTC compared with well-differentiated cancer and, as previously shown, can drive the development of murine PDTC. The aim of this study was to evaluate thyroid cancer initiation and progression in mice with concomitant expression of STRN-ALK and inactivation of the tumor suppressor p53 (Trp53) in thyroid follicular cells. Methods: Transgenic mice with thyroid-specific expression of STRN-ALK and biallelic p53 loss were generated and aged on a regular diet or with methimazole and sodium perchlorate goitrogen treatment. Development and progression of thyroid tumors were monitored by using ultrasound imaging, followed by detailed histological and immunohistochemical evaluation. Gene expression analysis was performed on selected tumor samples by using RNA-Seq and quantitative RT-PCR. Results: In mice treated with goitrogen, the first thyroid cancers appeared at 6 months of age, reaching 86% penetrance by the age of 12 months, while a similar rate (71%) of tumor occurrence in mice on regular diet was observed by 18 months of age. Histological examination revealed well-differentiated papillary thyroid carcinomas (PTC) (n = 26), PDTC (n = 21), and ATC (n = 8) that frequently coexisted in the same thyroid gland. The tumors were frequently lethal and associated with the development of lung metastasis in 24% of cases. Histological and immunohistochemical characteristics of these cancers recapitulated tumors seen in humans. Detailed analysis of PDTC revealed two tumor types with distinct cell morphology and immunohistochemical characteristics, designated as PDTC type 1 (PDTC1) and type 2 (PDTC2). Gene expression analysis showed that PDTC1 tumors retained higher expression of thyroid differentiation genes including Tg and Slc5a5 (Nis) as compared with PDTC2 tumors. Conclusions: In this study, we generated a new mouse model of multistep thyroid cancer dedifferentiation with evidence of progression from PTC to PDTC and ATC. Further, PDTC in these mice showed two distinct histologic appearances correlated with levels of expression of thyroid differentiation and iodine metabolism genes, suggesting a possibility of existence of two PDTC types with different functional characteristics and potential implication for therapeutic approaches to these tumors.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase/genetics
- Animals
- Antithyroid Agents/toxicity
- Calmodulin-Binding Proteins/genetics
- Cell Dedifferentiation/genetics
- Cell Differentiation/genetics
- Disease Models, Animal
- Disease Progression
- Membrane Proteins/genetics
- Methimazole/toxicity
- Mice
- Mice, Knockout
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Perchlorates/toxicity
- RNA-Seq
- Sodium Compounds/toxicity
- Symporters/genetics
- Thyroglobulin/genetics
- Thyroid Cancer, Papillary/chemically induced
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/pathology
- Thyroid Carcinoma, Anaplastic/chemically induced
- Thyroid Carcinoma, Anaplastic/genetics
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Neoplasms/chemically induced
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/pathology
- Transcriptome
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
| | - Susan L. Rominski
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vincenzo Condello
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cihan Kaya
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mamta Wankhede
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Hong Yang
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel L. Altschuler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yuri E. Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Address correspondence to: Yuri E. Nikiforov, MD, PhD, Department of Pathology, University of Pittsburgh, CLB Room 8031, 3477 Euler Way, Pittsburgh, PA 15213
| |
Collapse
|
14469
|
Han J, Lv W, Sheng H, Wang Y, Cao L, Huang S, Zhu L, Hu J. Ecliptasaponin A induces apoptosis through the activation of ASK1/JNK pathway and autophagy in human lung cancer cells. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:539. [PMID: 31807521 DOI: 10.21037/atm.2019.10.07] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Non-small cell lung cancer (NSCLC) is one of the causes of carcinomas mortality worldwide. Ecliptasaponin A (ES), a natural product extracted from the plant known as Eclipta prostrata, has been reported as an anti-cancer drug against various cancer cell lines. However, the exact mechanisms of ES have not yet been fully characterized. Methods Numerous studies have been done to support that ES has a powerful inhibiting effect on the growth of cancers via the activation of apoptosis and autophagy. To explore the underlying mechanisms of anti-cancer and investigate the relationships of the apoptosis and autophagy, we used apoptosis signal-regulating kinase 1 (ASK1) inhibitor (GS-4997), c-Jun N-terminal kinase (JNK) inhibitor (SP600125), and autophagy inhibitor [chloroquine (CQ) and 3-methyladenine (3-MA)]. Results ES could potently suppress cell viability and induces apoptotic cell death of human lung cancer cells H460 and H1975. ES activated apoptosis via ASK1/JNK pathway, GS-4997 and SP600125 can attenuated these effects. Furthermore, ES could triggered autophagy in lung cancer cell lines, and the autophagy inhibitor 3-MA and CQ reversed ES-induced apoptosis in H460 and H1975 cells. Furthermore, SP600125 can inhibit autophagy. Conclusions This study showed that ES induces apoptosis in human lung cancer cells by triggering enhanced autophagy and ASK1/JNK pathway, which may thus be a promising agent against lung cancer.
Collapse
Affiliation(s)
- Jia Han
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hongxu Sheng
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yiqing Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Longxiang Cao
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Sha Huang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Linhai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
14470
|
Paller CJ, Piana D, Eshleman JR, Riel S, Denmeade SR, Velho PI, Rowe SP, Pomper MG, Antonarakis ES, Luo J, Eisenberger MA. A pilot study of prostate-specific membrane antigen (PSMA) dynamics in men undergoing treatment for advanced prostate cancer. Prostate 2019; 79:1597-1603. [PMID: 31361358 PMCID: PMC6818502 DOI: 10.1002/pros.23883] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/24/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) is a rational target for noninvasive detection of recurrent prostate cancer (PCa) and for therapy of metastatic castration-resistant prostate cancer (mCRPC) with PSMA-targeted agents. Here we conducted serial measurements of PSMA expression on circulating tumor cells (CTCs) to evaluate patterns of longitudinal PSMA dynamics over the course of multiple sequential therapies. METHODS A retrospective investigation of men with mCRPC undergoing evaluation at medical oncology clinics at our institution assessed the dynamics of PSMA expression in the context of different systemic treatments administered sequentially. Eligibility included patients who began systemic therapies with androgen receptor (AR)-directed agents or taxane agents for whom peripheral blood samples were tested for CTC mRNA of AR splice variant-7 (AR-V7), prostate-specific antigen (PSA), and PSMA (with >2 CTC + results) in a CLIA-accredited laboratory. RESULTS From August 2015 to November 2017, we identified 96 eligible men. Fifteen had greater than or equal to 2 sequential therapies and evaluable CTC samples, greater than or equal to 1 expressing PSMA (PSMA+). Among the 15 patients included in this analysis, a total of 54 PSMA status evaluations were performed in the context of 48 therapies during a median follow-up of 18 months. At baseline, PSMA signal was detected ("positive") in 11 of 15 (73.3%) patients, while for 4 of 15 (26.7%) patients PSMA signal was undetectable ("negative"). In all but two patients, the baseline collection corresponded with a change in treatment. On the second assessment, PSMA increases were detected in all 4/4 (100%) PSMA-negative patients and 8 of 11 (72.7%) PSMA-positive patients. PSMA significantly decreased in a patient treated with 177 Lu-PSMA-617. Serum PSA declines were seen in 7 of 8 (88%) of the treatment periods where PSMA decreased. CONCLUSIONS PSMA expression in CTCs is a dynamic marker. PSMA transcript declines appear to be associated with concurrent decreases in serum PSA. Sequential CTC sampling could provide a noninvasive response assessment to systemic treatment for mCRPC.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, Surface/blood
- Antigens, Surface/genetics
- Bridged-Ring Compounds/therapeutic use
- Dipeptides/therapeutic use
- Glutamate Carboxypeptidase II/blood
- Glutamate Carboxypeptidase II/genetics
- Heterocyclic Compounds, 1-Ring/therapeutic use
- Humans
- Lutetium
- Male
- Middle Aged
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/therapy
- Neoplastic Cells, Circulating/chemistry
- Pilot Projects
- Prostate-Specific Antigen/blood
- Prostate-Specific Antigen/genetics
- Prostatic Neoplasms/blood
- Prostatic Neoplasms/therapy
- Prostatic Neoplasms, Castration-Resistant/blood
- Prostatic Neoplasms, Castration-Resistant/therapy
- RNA, Messenger/blood
- Receptors, Androgen/drug effects
- Retrospective Studies
- Taxoids/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Channing J. Paller
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Danilo Piana
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - James R. Eshleman
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutes, Baltimore, Maryland
| | - Stacy Riel
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutes, Baltimore, Maryland
| | - Samuel R. Denmeade
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Pedro Isaacsson Velho
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Steven P. Rowe
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G. Pomper
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emmanuel S. Antonarakis
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mario A. Eisenberger
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
14471
|
Wang G, Liu J, Zhu L, Ma X, Wang X, Yang X, Guo Y, Yang L, Lu J. Self-Destruction of Cancer Induced by Ag 2 S Amorphous Nanodots. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902945. [PMID: 31531961 DOI: 10.1002/smll.201902945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/07/2019] [Indexed: 06/10/2023]
Abstract
Studies on distinctive performances and novel applications of amorphous inorganic nanomaterials are becoming attractive. Herein, Ag2 S amorphous and crystalline nanodots (ANDs and CNDs) are prepared via facile methods. In vitro and in vivo studies indicate that Ag2 S ANDs, rather than CNDs, can induce the self-destruction of tumors, which can be attributed to their distinctive chemical properties, e.g., the higher electrochemical active surface area and lower redox potential well matching with the redox reaction requirement in the tumor microenvironment. Ag2 S ANDs can be oxidized by intracellular reactive oxygen species (ROS) to release Ag+ , which further stimulates high generation of intracellular ROS. This mutual stimulation damages the mitochondria, induces apoptosis, and leads to the self-destruction of the tumor. Moreover, Ag2 S ANDs do not show observable in vitro and in vivo side effects. These findings provide a promising self-destructive strategy for cancer therapy by utilizing distinctive chemical properties of inorganic nanomaterials, while avoiding complicated external assistance.
Collapse
Affiliation(s)
- Ge Wang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Jing Liu
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Lin Zhu
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaoming Ma
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaobing Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaoli Yang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yuming Guo
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Lin Yang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Jun Lu
- Chemical Sciences and Engineering Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| |
Collapse
|
14472
|
Kerdsirichairat T, Narang AK, Thompson E, Kim SH, Rao A, Ding K, Shin EJ. Feasibility of Using Hydrogel Spacers for Borderline-Resectable and Locally Advanced Pancreatic Tumors. Gastroenterology 2019; 157:933-935. [PMID: 31306631 PMCID: PMC7263852 DOI: 10.1053/j.gastro.2019.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 01/11/2023]
Abstract
This article has an accompanying continuing medical education activity, also eligible for MOC credit, on page e14 (https://www.gastrojournal.org/cme/home). Learning Objective: Upon completion of this CME activity, successful learners will be able to describe the pharmacokinetics of hydrogel, identify appropriate candidates for hydrogel injection among patients with pancreatic cancer, and describe key techniques to successfully inject hydrogel as well as the histopathologic findings associated with hydrogel.
Collapse
Affiliation(s)
- Tossapol Kerdsirichairat
- Division of Gastroenterology and Hepatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Amol K. Narang
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Elizabeth Thompson
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Seong-Hun Kim
- Department of Internal Medicine, Chonbuk National University Medical School & Hospital, Jeonju, South Korea
| | - Avani Rao
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Kai Ding
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Eun Ji Shin
- Division of Gastroenterology and Hepatology, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
14473
|
Taylor O, Nijhawan RI. Cells to Surgery Quiz: October 2019. J Invest Dermatol 2019. [DOI: 10.1016/j.jid.2019.08.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14474
|
Quantitative next-generation sequencing-based analysis indicates progressive accumulation of microsatellite instability between atypical hyperplasia/endometrial intraepithelial neoplasia and paired endometrioid endometrial carcinoma. Mod Pathol 2019; 32:1508-1520. [PMID: 31186530 DOI: 10.1038/s41379-019-0298-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 12/30/2022]
Abstract
Atypical hyperplasia/endometrial intraepithelial neoplasia is an accepted precursor to endometrioid-type endometrial carcinoma. Mismatch repair-deficient endometrial carcinomas are also known to be a biologically and clinically distinct subset of tumors. However, the development of microsatellite instability in endometrial carcinogenesis has not yet been evaluated by novel next-generation sequencing-based methods. We examined 17 mismatch repair-deficient endometrioid endometrial carcinomas and their paired atypical hyperplasia/endometrial intraepithelial neoplasia precursors using a next-generation sequencing panel with quantitative microsatellite instability detection at 336 loci. Findings were compared to histological features, polymerase chain reaction-based microsatellite instability testing, immunohistochemical expression of mismatch repair proteins, and tumor mutational burden calculations. All 17 endometrial carcinomas and 8/17 atypical hyperplasia/endometrial intraepithelial neoplasia showed microsatellite instability by next-generation sequencing-based testing. Endometrial carcinoma specimens showed significantly more unstable microsatellite loci than paired atypical hyperplasia/endometrial intraepithelial neoplasia (mean: 40.0% vs 19.9 unstable loci, respectively). Out of nine microsatellite-stable atypical hyperplasia/endometrial intraepithelial neoplasia specimens, four showed mismatch repair loss by immunohistochemistry. All atypical hyperplasia/endometrial intraepithelial neoplasia and endometrial carcinoma specimens with microsatellite instability were also mismatch repair-deficient by immunohistochemistry. Tumor mutational burden was significantly greater in endometrial carcinoma than in paired atypical hyperplasia/endometrial intraepithelial neoplasia specimens, and tumor mutational burden was significantly correlated with percent unstable microsatellite loci. Paired atypical hyperplasia/endometrial intraepithelial neoplasia and endometrial carcinoma specimens show progressive accumulation of unstable microsatellite loci following loss of mismatch repair protein expression. Comprehensive next-generation sequencing-based testing of endometrial carcinomas offers new insights into endometrial carcinogenesis and opportunities for improved tumor surveillance, diagnosis, and management.
Collapse
|
14475
|
han Y, Hu H, zhou J. Knockdown of LncRNA SNHG7 inhibited epithelial-mesenchymal transition in prostate cancer though miR-324-3p/WNT2B axis in vitro. Pathol Res Pract 2019; 215:152537. [DOI: 10.1016/j.prp.2019.152537] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 07/12/2019] [Indexed: 12/30/2022]
|
14476
|
Bijou I, Wang J. Evolving trends in pancreatic cancer therapeutic development. ANNALS OF PANCREATIC CANCER 2019; 2:17. [PMID: 33089149 PMCID: PMC7575122 DOI: 10.21037/apc.2019.09.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite advances in translational research, the overall 5-year survival for pancreatic cancer remains dismal and with rising incidence pancreatic cancer is predicted to be the second leading cause of cancer death for many developed countries. Surgical intervention followed by cytotoxic chemotherapy are currently the best options for treatment, but disease recurrence is very common. Efforts to develop new therapeutic agents and delivery systems are necessary to achieve better clinical efficacy with less toxicity. Promising prospects are arising with new preclinical and clinical therapeutic strategies using small molecule targeted therapies, RNAi, stromal therapies, and immunotherapies. With a better understanding of the biology to aid target selection and discovery of biomarkers to aid precision medicine, better opportunities will evolve to shape the therapeutic landscape, enhance patient quality of life and increase overall survival.
Collapse
Affiliation(s)
- Imani Bijou
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| |
Collapse
|
14477
|
Lieu CH, Golemis EA, Serebriiskii IG, Newberg J, Hemmerich A, Connelly C, Messersmith WA, Eng C, Eckhardt SG, Frampton G, Cooke M, Meyer JE. Comprehensive Genomic Landscapes in Early and Later Onset Colorectal Cancer. Clin Cancer Res 2019; 25:5852-5858. [PMID: 31243121 PMCID: PMC6774873 DOI: 10.1158/1078-0432.ccr-19-0899] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 06/21/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE The incidence rates of colorectal cancers are increasing in young adults. The objective of this study was to investigate genomic differences between tumor samples collected from younger and older patients with colorectal cancer. EXPERIMENTAL DESIGN DNA was extracted from 18,218 clinical specimens, followed by hybridization capture of 3,769 exons from 403 cancer-related genes and 47 introns of 19 genes commonly rearranged in cancer. Genomic alterations (GA) were determined, and association with patient age and microsatellite stable/microsatellite instability high (MSS/MSI-H) status established. RESULTS Overall genomic alteration rates in the younger (<40) and older (≥50) cohorts were similar in the majority of the genes analyzed. Gene alteration rates in the microsatellite stable (MSS) younger and older cohorts were largely similar, with several notable differences. In particular, TP53 (FDR < 0.01) and CTNNB1 (FDR = 0.01) alterations were more common in younger patients with colorectal cancer, and APC (FDR < 0.01), KRAS (FDR < 0.01), BRAF (FDR < 0.01), and FAM123B (FDR < 0.01) were more commonly altered in older patients with colorectal cancer. In the MSI-H cohort, the majority of genes showed similar rate of alterations in all age groups, but with significant differences seen in APC (FDR < 0.01), BRAF (FDR < 0.01), and KRAS (FDR < 0.01). CONCLUSIONS Tumors from younger and older patients with colorectal cancer demonstrated similar overall rates of genomic alteration. However, differences were noted in several genes relevant to biology and response to therapy. Further study will need to be conducted to determine whether the differences in gene alteration rates can be leveraged to provide personalized therapies for young patients with early-onset sporadic colorectal cancer.
Collapse
Affiliation(s)
- Christopher H Lieu
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado.
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ilya G Serebriiskii
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Kazan Federal University, Kazan, Russian Federation
| | | | | | | | - Wells A Messersmith
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - S Gail Eckhardt
- Department of Medical Oncology, University of Texas at Austin Dell Medical School and LIVESTRONG Cancer Institutes, Austin, Texas
| | | | | | - Joshua E Meyer
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
14478
|
Cheng A, Zhao S, FitzGerald LM, Wright JL, Kolb S, Karnes RJ, Jenkins RB, Davicioni E, Ostrander EA, Feng Z, Fan JB, Dai JY, Stanford JL. A four-gene transcript score to predict metastatic-lethal progression in men treated for localized prostate cancer: Development and validation studies. Prostate 2019; 79:1589-1596. [PMID: 31376183 PMCID: PMC6715522 DOI: 10.1002/pros.23882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/24/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND Molecular studies have tried to address the unmet need for prognostic biomarkers in prostate cancer (PCa). Some gene expression tests improve upon clinical factors for prediction of outcomes, but additional tools for accurate prediction of tumor aggressiveness are needed. METHODS Based on a previously published panel of 23 gene transcripts that distinguished patients with metastatic progression, we constructed a prediction model using independent training and testing datasets. Using the validated messenger RNAs and Gleason score (GS), we performed model selection in the training set to define a final locked model to classify patients who developed metastatic-lethal events from those who remained recurrence-free. In an independent testing dataset, we compared our locked model to established clinical prognostic factors and utilized Kaplan-Meier curves and receiver operating characteristic analyses to evaluate the model's performance. RESULTS Thirteen of 23 previously identified gene transcripts that stratified patients with aggressive PCa were validated in the training dataset. These biomarkers plus GS were used to develop a four-gene (CST2, FBLN1, TNFRSF19, and ZNF704) transcript (4GT) score that was significantly higher in patients who progressed to metastatic-lethal events compared to those without recurrence in the testing dataset (P = 5.7 × 10-11 ). The 4GT score provided higher prediction accuracy (area under the ROC curve [AUC] = 0.76; 95% confidence interval [CI] = 0.69-0.83; partial area under the ROC curve [pAUC] = 0.008) than GS alone (AUC = 0.63; 95% CI = 0.56-0.70; pAUC = 0.002), and it improved risk stratification in subgroups defined by a combination of clinicopathological features (ie, Cancer of the Prostate Risk Assessment-Surgery). CONCLUSION Our validated 4GT score has prognostic value for metastatic-lethal progression in men treated for localized PCa and warrants further evaluation for its clinical utility.
Collapse
Affiliation(s)
- Anqi Cheng
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Shanshan Zhao
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Liesel M. FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAZ, Australia
| | - Jonathan L. Wright
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Robert B. Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Elaine A. Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ziding Feng
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jian-Bing Fan
- AnchorDx Corporation, Guangzhou, 510300, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - James Y. Dai
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
14479
|
Grassberger C, Huber K, Jacob NK, Green MD, Mahler P, Prisciandaro J, Dominello M, Joiner MC, Burmeister J. Three discipline collaborative radiation therapy (3DCRT) special debate: The single most important factor in determining the future of SBRT is immune response. J Appl Clin Med Phys 2019; 20:6-12. [PMID: 31573143 PMCID: PMC6807212 DOI: 10.1002/acm2.12728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
| | - Kathryn Huber
- Department of Radiation OncologyTufts Medical CenterBostonMAUSA
| | | | - Michael D. Green
- Department of Radiation OncologyUniversity of MichiganAnn ArborMIUSA
| | - Peter Mahler
- Department of Human OncologyUniversity of WisconsinMadisonWIUSA
| | | | - Michael Dominello
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
| | - Michael C. Joiner
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
| | - Jay Burmeister
- Department of OncologyWayne State University School of MedicineDetroitMIUSA
- Gershenson Radiation Oncology CenterBarbara Ann Karmanos Cancer InstituteDetroitMIUSA
| |
Collapse
|
14480
|
Czogalla B, Kuhn C, Heublein S, Schmöckel E, Mayr D, Kolben T, Trillsch F, Burges A, Mahner S, Jeschke U, Hester A. EP3 receptor is a prognostic factor in TA-MUC1-negative ovarian cancer. J Cancer Res Clin Oncol 2019; 145:2519-2527. [PMID: 31485769 DOI: 10.1007/s00432-019-03017-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Prostaglandin-mediated inflammatory reactions play a major role in different cancers. Recently, it has been observed that prostaglandin E2-receptor 3 (EP3) might be an independent prognostic factor for overall survival in cervical and endometrial cancer. The role of EP3 expression in ovarian cancer is currently unknown. METHODS EP3 expression was analyzed by immunohistochemistry in 156 patient samples using the IR-scoring system. Expression levels were correlated with clinical and pathological parameters and with overall survival (OS) to assess for prognostic relevance. Data analysis was performed using Spearman's correlations, Kruskal-Wallis test and Kaplan-Meier estimates. RESULTS EP3 expression was significantly higher in clear-cell carcinoma (p < 0.001) compared to the other histological subtypes. No further correlations with clinical parameters could be found. EP3 expression correlated significantly with FSH-receptor expression (p < 0.001), galectin-1 expression in the tumor (p = 0.012) and with cytoplasmatic TA-MUC1 expression (p = 0.001). None of these parameters showed significant correlation with OS. In the TA-MUC1 negative subgroup, EP3 negative patients showed significantly longer OS (median OS: 102 months vs. 34 months in EP3 positive patients, p = 0.035), while EP3 did not appear to have prognostic relevance in the TA-MUC1-positive subgroup. CONCLUSION The potential prognostic relevance of EP3 expression for OS in TA-MUC1 negative patients might reflect an interplay between the COX and the MUC1 pathway, as it has been shown that MUC1 could induce COX2 expression. Our findings support the importance of the prostanoid signaling in TA-MUC1 negative ovarian cancer; however, future studies are necessary to characterize specific pathways and possible interactions.
Collapse
Affiliation(s)
- Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Sabine Heublein
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Department of Gynecology and Obstetrics, University of Heidelberg, 69120, Heidelberg, Germany
| | - Elisa Schmöckel
- Department of Pathology, Ludwig-Maximilians University of Munich, 81337, Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians University of Munich, 81337, Munich, Germany
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Anna Hester
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
14481
|
White AJ, Keller JP, Zhao S, Carroll R, Kaufman JD, Sandler DP. Air Pollution, Clustering of Particulate Matter Components, and Breast Cancer in the Sister Study: A U.S.-Wide Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:107002. [PMID: 31596602 PMCID: PMC6867190 DOI: 10.1289/ehp5131] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 05/24/2023]
Abstract
BACKGROUND Particulate matter (PM) is a complex mixture. Geographic variations in PM may explain the lack of consistent associations with breast cancer. OBJECTIVE We aimed to evaluate the relationship between air pollution, PM components, and breast cancer risk in a United States-wide prospective cohort. METHODS We estimated annual average ambient residential levels of particulate matter <2.5 μm and <10 μm in aerodynamic diameter (PM2.5 and PM10, respectively) and nitrogen dioxide (NO2) using land-use regression for 47,433 Sister Study participants (breast cancer-free women with a sister with breast cancer) living in the contiguous United States. Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for risk associated with an interquartile range (IQR) increase in pollutants. Predictive k-means were used to assign participants to clusters derived from PM2.5 component profiles to evaluate the impact of heterogeneity in the PM2.5 mixture. For PM2.5, we investigated effect measure modification by component cluster membership and by geographic region without regard to air pollution mixture. RESULTS During follow-up (mean=8.4 y), 2,225 invasive and 623 ductal carcinoma in situ (DCIS) cases were identified. PM2.5 and NO2 were associated with breast cancer overall [HR=1.05 (95% CI:0.99, 1.11) and 1.06 (95% CI:1.02, 1.11), respectively] and with DCIS but not with invasive cancer. Invasive breast cancer was associated with PM2.5 only in the Western United States [HR=1.14 (95% CI:1.02, 1.27)] and NO2 only in the Southern United States [HR=1.16 (95% CI:1.01, 1.33)]. PM2.5 was associated with a higher risk of invasive breast cancer among two of seven identified composition-based clusters. A higher risk was observed [HR=1.25 (95% CI: 0.97, 1.60)] in a California-based cluster characterized by low S and high Na and nitrate (NO3-) fractions and for another Western United States cluster [HR=1.60 (95% CI: 0.90, 2.85)], characterized by high fractions of Si, Ca, K, and Al. CONCLUSION Air pollution measures were related to both invasive breast cancer and DCIS within certain geographic regions and PM component clusters. https://doi.org/10.1289/EHP5131.
Collapse
Affiliation(s)
- Alexandra J. White
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Joshua P. Keller
- Department of Statistics, Colorado State University, Fort Collins, Colorado, USA
| | - Shanshan Zhao
- Biostatistics Branch, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Rachel Carroll
- Department of Mathematics and Statistics, University of North Carolina at Wilmington, North Carolina, USA
| | - Joel D. Kaufman
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| |
Collapse
|
14482
|
Fatehi Hassanabad A. Current perspectives on statins as potential anti-cancer therapeutics: clinical outcomes and underlying molecular mechanisms. Transl Lung Cancer Res 2019; 8:692-699. [PMID: 31737505 DOI: 10.21037/tlcr.2019.09.08] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Statins have been shown to inhibit cell proliferation in vitro and tumor growth in animal models. Various studies have also shown a decreased cancer-specific mortality rate in patients who were prescribed these medications. Statins inhibit 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), the rate-limiting enzyme of the mevalonate pathway. Statins induce tumour-specific apoptosis through mitochondrial apoptotic signaling pathways, which are activated by the suppression of mevalonate or geranylgeranyl pyrophosphate (GGPP) biosynthesis. However, there is no consensus on the molecular targets of statins for their anti-cancer effects. Several studies have been conducted to further assess the association between statin use and mortality in different types of cancer. In this review, current perspectives on clinical significance of statins in prevention and treatment of various types of cancers and proposed mechanisms are discussed.
Collapse
|
14483
|
Hou A, Zhang Y, Zheng Y, Fan Y, Liu H, Zhou X. LncRNA terminal differentiation-induced ncRNA (TINCR) sponges miR-302 to upregulate cyclin D1 in cervical squamous cell carcinoma (CSCC). Hum Cell 2019; 32:515-521. [PMID: 31388923 DOI: 10.1007/s13577-019-00268-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
This study aimed to investigate the role of lncRNA terminal differentiation-induced ncRNA (TINCR) in cervical squamous cell carcinoma (CSCC). By informatics analysis, we found that miR-302 may bind TINCR. Expression analysis showed that miR-302 was downregulated, while TINCR was upregulated in CSCC. Correlation analysis showed that they were not significantly correlated. In CSCC cells, miR-302 and TINCR failed to affect the expression of each other. However, miR-302 overexpression led to downregulated and TINCR overexpression led to upregulated cyclin D1 expression in CSCC cells. Interestingly, overexpression of cyclin D1 led to upregulated miR-302 and TINCR. Cell proliferation analysis showed that TINCR and cyclin D1 overexpression led to increased, while miR-302 overexpression led to decreased rate of cell proliferation. Moreover, miR-302 overexpression reduced the effects of TINCR overexpression. Therefore, TINCR sponges miR-302 to upregulate cyclin D1 in CSCC, thereby promoting cell proliferation.
Collapse
Affiliation(s)
- Anli Hou
- Department of Gynaecology, University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253 Songbai Road, Matian Street, Guangming District, Shenzhen City, 518106, Guangdong Province, China.
| | - Yali Zhang
- Department of Gynaecology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Yi Zheng
- Department of Central Lab, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen City, 518106, Guangdong Province, China
| | - Yujuan Fan
- Department of Gynaecology, University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253 Songbai Road, Matian Street, Guangming District, Shenzhen City, 518106, Guangdong Province, China
| | - Huilan Liu
- Department of Gynaecology, University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253 Songbai Road, Matian Street, Guangming District, Shenzhen City, 518106, Guangdong Province, China
| | - Xiuying Zhou
- Department of Gynaecology, University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253 Songbai Road, Matian Street, Guangming District, Shenzhen City, 518106, Guangdong Province, China
| |
Collapse
|
14484
|
Oezdemir I, Javed K, Rijal G, Hoyt K. Contrast-enhanced ultrasound imaging of acute changes in pancreatic cancer following targeted hyaluronan treatment. IEEE INTERNATIONAL ULTRASONICS SYMPOSIUM : [PROCEEDINGS]. IEEE INTERNATIONAL ULTRASONICS SYMPOSIUM 2019; 2019:2303-2306. [PMID: 36514673 PMCID: PMC9743975 DOI: 10.1109/ultsym.2019.8925558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to monitor acute changes in pancreatic tumor perfusion with contrast-enhanced ultrasound (CEUS) imaging following targeted hyaluronan (HA) treatment. Intratumoral accumulation of HA is one of contributing factors that can lead to an increased tumor interstitial pressure (TIP). These elevated TIP levels can hinder delivery of chemotherapeutic drugs and cause treatment failure. For this study, pancreatic cancer-bearing mice were imaged at baseline and again at 2 h after intravenous administration of physiological saline (control group) or PEGPH20, which targets HA (therapy group). CEUS data were collected for 5 min and the temporal sequence was first analyzed using a singular value filter (SVF) to remove any background clutter signal. Given the time history of contrast agent flow, a tumor perfusion parametric analysis was performed. A series of morphological image operations was applied to quantify structural features of the tumor angiogenic network including vessel count, density, length, diameter, tortuosity, and branching points. After imaging, animals were euthanized, and tumors excised for histological processing. Acute microvascular changes were found at 2 h after drug administration as confirmed by CEUS imaging. Further, histologic analysis of tumor sections revealed lower HA accumulation in the therapy group animals. Overall, these findings suggest that CEUS imaging of acute changes in tumor perfusion may help identify an optimal window whereby follow-up chemotherapeutic drug dosing would be more effective.
Collapse
Affiliation(s)
- Ipek Oezdemir
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Kulsoom Javed
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Girdhari Rijal
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
14485
|
Kamyabi N, Bernard V, Maitra A. Liquid biopsies in pancreatic cancer. Expert Rev Anticancer Ther 2019; 19:869-878. [PMID: 31533487 PMCID: PMC6824837 DOI: 10.1080/14737140.2019.1670063] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) is a disease of high lethality. Invasive tissue biopsies of primary or metastatic lesions remain the gold standard for diagnosis, but repeated sampling is infeasible. Noninvasive liquid biopsies offer new opportunities for early diagnosis for high-risk cohorts, and for the longitudinal analysis of tumor evolution and progression in patients on therapy. Liquid biopsies can capture tumor-associated components, such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs), each of which provides genomic and molecular information about the underlying PDAC that can potentially inform clinical decisions. Areas covered: Here, we reviewed current knowledge and recent technological advances regarding liquid biopsy in PDAC and mention the pitfalls and benefits in each methodology. We also discuss clinical correlative studies for diagnosis and prognosis in PDAC. Expert opinion:In pancreatic cancer where tissue samples are limited and repeated tissue biopsies are mostly invasive and infeasible, liquid biopsies opened a new window for tumor diagnosis, molecular stratification, and treatment monitoring. While none of the isolation and analysis methods have gained widespread clinical acceptance, it is imperative that the advantages and limitations of each platform for isolation and analysis of tumor associated components are taken into consideration.
Collapse
Affiliation(s)
- Nabiollah Kamyabi
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, UT MD Anderson Cancer Center, Houston, Texas, 77030
| | - Vincent Bernard
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, UT MD Anderson Cancer Center, Houston, Texas, 77030
| | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, UT MD Anderson Cancer Center, Houston, Texas, 77030
| |
Collapse
|
14486
|
Cai H, Yang X, Gao Y, Xu Z, Yu B, Xu T, Li X, Xu W, Wang X, Hua L. Exosomal MicroRNA-9-3p Secreted from BMSCs Downregulates ESM1 to Suppress the Development of Bladder Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:787-800. [PMID: 31734559 PMCID: PMC6861677 DOI: 10.1016/j.omtn.2019.09.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/25/2022]
Abstract
Exosomes, carriers to transfer endogenous molecules, derived from bone marrow-derived mesenchymal stem cells (BMSCs) have been reported to play a role in the progression of bladder cancer. Here we aimed to test the functional mechanism of microRNA-9-3p (miR-9-3p)-containing exosomes derived from BMSCs in bladder cancer. BMSCs were cocultured with bladder cancer cells, and exosomes secreted from BMSCs were identified. Next, the expression of miR-9-3p and endothelial cell-specific molecule 1 (ESM1) in bladder cancer tissues and cells was determined. Then effects of miR-9-3p and ESM1 via BMSC-derived exosomes on bladder cancer cell viability, migration, invasion, and apoptosis were determined by loss- and gain-of-function experiments and on in vivo tumor growth, and metastasis was assessed in nude mice. miR-9-3p expression was decreased and ESM1 was increased in bladder cancer. BMSCs inhibited bladder cancer cell viability, migration, and invasion, and induced apoptosis, whereas the addition of exosome secretion inhibitor GW4869 achieved the opposite effects. Moreover, exosomal miR-9-3p upregulation or ESM1 silencing suppressed bladder cancer cell viability, migration, and invasion; induced cell apoptosis; and inhibited in vivo tumor growth and metastasis. Taken together, BMSC-derived exosomal miR-9-3p suppressed the progression of bladder cancer through ESM1 downregulation, offering a potential novel therapeutic target for bladder cancer therapy.
Collapse
Affiliation(s)
- Hongzhou Cai
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Xuejian Yang
- Department of Urology, Suqian First Hospital, Suqian 223800, P.R. China
| | - Yang Gao
- Department of Radiology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Zicheng Xu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Bin Yu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Ting Xu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Xiao Li
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Weizhang Xu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China
| | - Xinwei Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210009, P.R. China.
| | - Lixin Hua
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P.R. China.
| |
Collapse
|
14487
|
Moreira AF, Rodrigues CF, Jacinto TA, Miguel SP, Costa EC, Correia IJ. Microneedle-based delivery devices for cancer therapy: A review. Pharmacol Res 2019; 148:104438. [DOI: 10.1016/j.phrs.2019.104438] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/28/2022]
|
14488
|
Pontious C, Kaul S, Hong M, Hart PA, Krishna SG, Lara LF, Conwell DL, Cruz-Monserrate Z. Cathepsin E expression and activity: Role in the detection and treatment of pancreatic cancer. Pancreatology 2019; 19:951-956. [PMID: 31582345 PMCID: PMC6829043 DOI: 10.1016/j.pan.2019.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
Cathepsin E (CTSE) is an intracellular, hydrolytic aspartic protease found to be expressed in cells of the immune and gastrointestinal systems, lymphoid tissues, erythrocytes, and cancer cells. The precise functions are not fully understood; however, various studies have investigated its numerous cell-type specific roles. CTSE expression has been shown to be a potential early biomarker for pancreatic ductal adenocarcinoma (PDAC). PDAC patients have low survival rates mostly due to the lack of early detection methods. CTSE-specific activity probes have been developed and tested to assist in tumor imaging and functional studies investigating the role of CTSE expression in PDAC tumors. Furthermore, a CTSE protease-specific, photodynamic therapy pro-drug was developed to explore its potential use to treat tumors that express CTSE. Since CTSE is expressed in pancreatic diseases that are risk factors for PDAC, such as pancreatic cysts and chronic pancreatitis, learning about its function in these disease types could assist in early PDAC detection and in understanding the biology of PDAC progression. Overall, CTSE expression and activity shows potential to detect PDAC and other pancreatic diseases. Further research is needed to fully understand its functions and potential translational applicability.
Collapse
Affiliation(s)
- Corbin Pontious
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sabrina Kaul
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Marcus Hong
- The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Kenyon College, Gambier, OH, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Luis F Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
14489
|
Wang J, Li X, Xiao Z, Wang Y, Han Y, Li J, Zhu W, Leng Q, Wen Y, Wen X. MicroRNA-488 inhibits proliferation and glycolysis in human prostate cancer cells by regulating PFKFB3. FEBS Open Bio 2019; 9:1798-1807. [PMID: 31410981 PMCID: PMC6768114 DOI: 10.1002/2211-5463.12718] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/12/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) remains the second leading cause of cancer-related death among men in the United States, and its molecular mechanism remains to be elucidated. Recent studies have suggested that microRNAs may play an important role in cancer development and progression. By analyzing the Gene Expression Omnibus dataset, we found lower expression for miR-488 in PCa than in normal tissues. Moreover, CCK-8, EdU, glucose uptake, and lactate secrete assays revealed that overexpression of miR-488 in PCa cell lines PC3 and DU145 resulted in inhibition of proliferation and glycolysis. In contrast, downregulation of miR-488 expression promoted proliferation and glycolysis in PCa cells. Using a bioinformatic approach and dual-luciferase reporter assays, we identified 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, isoform3 (PFKFB3), as a direct target of miR-488. Inhibition of PFKFB3 also suppressed PCa cell glycolysis and proliferation. Our study suggests that miR-488 inhibits PCa cell proliferation and glycolysis by targeting PFKFB3, and thus, miR-488 may be a novel therapeutic candidate for PCa.
Collapse
Affiliation(s)
- Jun Wang
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of UrologyFudan University Shanghai Cancer CenterChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiaojuan Li
- Center of Health ManagementShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Zhaoming Xiao
- Department of UrologyNanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Yu Wang
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuefu Han
- Department of UrologyYue Bei People's HospitalShaoguanChina
| | - Jun Li
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Weian Zhu
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Qu Leng
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Yuehui Wen
- Department of UrologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Xinqiao Wen
- Department of UrologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
14490
|
Herbst B, Zheng L. Precision medicine in pancreatic cancer: treating every patient as an exception. Lancet Gastroenterol Hepatol 2019; 4:805-810. [PMID: 31511204 PMCID: PMC9516437 DOI: 10.1016/s2468-1253(19)30175-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/09/2019] [Accepted: 04/24/2019] [Indexed: 01/16/2023]
Abstract
Patients with pancreatic cancer have not benefited from recent improvements in overall survival brought about by precision medicine in other malignancies. This failure is not due to a dearth of precision-medicine research in pancreatic ductal adenocarcinoma (PDAC), the main type of pancreatic cancer. In fact, the stalled progress in precision therapies for this type of cancer is due to the absence of agents that are able to target the common genetic alterations in PDAC. Several studies have attempted to phenotypically stratify PDAC at the transcriptional level. However, the value of such classifications will only be revealed through prospective studies and, crucially, only after development of new treatment options for this disease. Therefore, it is essential to learn from breakthrough discoveries in other cancer types that could benefit subpopulations of patients with PDAC and convert them from ordinary to exceptional responders. Identifying these exceptional patients will help to bring PDAC in line with other cancer types in terms of availability of precision therapies. Thus, the true challenge to precision medicine for PDAC might be the poor consensus on which genetic and phenotypic alterations across the spectrum of this disease are actionable; not the absence of actionable variables themselves. To reach consensus, knowledge and tools must be developed and disseminated for individuals who provide pancreatic cancer care, to enable the real-time identification of exceptional patients, more precise subgroup classifications, and effective disease management strategies; all informed by immediate feedback from clinical outcome data.
Collapse
Affiliation(s)
- Brian Herbst
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Graduate Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Pancreatic Cancer Precision Medicine Center of Excellence (PMCoE) Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Graduate Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14491
|
Rong X, Gao W, Yang X, Guo J. Downregulation of hsa_circ_0007534 restricts the proliferation and invasion of cervical cancer through regulating miR-498/BMI-1 signaling. Life Sci 2019; 235:116785. [DOI: 10.1016/j.lfs.2019.116785] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/27/2022]
|
14492
|
Gupta A, Sonis S, Uppaluri R, Bergmark RW, Villa A. Disparities in Oral Cancer Screening Among Dental Professionals: NHANES 2011-2016. Am J Prev Med 2019; 57:447-457. [PMID: 31443957 DOI: 10.1016/j.amepre.2019.04.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION As early detection of oral cancers is associated with better survival, oral cancer screening should be included in dental visits for adults. This study examines the rate and predictors of oral cancer screening exams among U.S. adults with a recent dental visit. METHODS Individuals aged ≥30 years who received a dental visit in the last 2 years, in the 2011-2016 National Health and Nutrition Examination Survey were analyzed in December 2018. Weighted multivariable logistic regression models examined the likelihood of intraoral and extraoral oral cancer screening exams, adjusting for age, sex, race/ethnicity, education, marital status, poverty income ratio, health insurance, tobacco smoking, and alcohol consumption. Subgroup analyses were conducted among races/ethnicities, smokers, and alcohol consumers. Statistical significance was set at p<0.01. RESULTS A total of 37.6% and 31.3% reported receiving an intraoral and extraoral oral cancer screening exam, respectively. Minority racial/ethnic groups versus white, non-Hispanics, less-educated versus more-educated, uninsured and Medicaid-insured versus privately insured, and low-income versus high-income participants were less likely to have received intraoral or extraoral oral cancer screening exams. There was no difference in the likelihood of being screened based on smoking status. Alcohol consumers were more likely to be screened. Among subgroups, less-educated and low-income individuals were less likely to be screened. CONCLUSIONS A significantly higher proportion of minority race/ethnicity and low SES individuals report not receiving an oral cancer screening exam, despite a recent dental visit. This selective screening by dental professionals is incompliant with guidelines and concerning because these groups are more likely to present with an advanced stage of oral cancer at diagnosis. An understanding of the reasons for discriminatory oral cancer screening practices could help develop effective interventions.
Collapse
Affiliation(s)
- Avni Gupta
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Stephen Sonis
- Division of Oral Medicine, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Ravindra Uppaluri
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
| | - Regan W Bergmark
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, Massachusetts; Division of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts; Patient Reported Outcomes, Value, and Experience Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alessandro Villa
- Division of Oral Medicine, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
14493
|
An Executive Summary of Reports From an International Multidisciplinary Roundtable on Exercise and Cancer: Evidence, Guidelines, and Implementation. REHABILITATION ONCOLOGY 2019. [DOI: 10.1097/01.reo.0000000000000186] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14494
|
Diversity and inclusion: the role of unconscious bias on patient care, health outcomes and the workforce in obstetrics and gynaecology. Curr Opin Obstet Gynecol 2019; 31:356-362. [DOI: 10.1097/gco.0000000000000566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14495
|
Bardia A, Aftimos P, Bihani T, Anderson-Villaluz AT, Jung J, Conlan MG, Kaklamani VG. EMERALD: Phase III trial of elacestrant (RAD1901) vs endocrine therapy for previously treated ER+ advanced breast cancer. Future Oncol 2019; 15:3209-3218. [PMID: 31426673 DOI: 10.2217/fon-2019-0370] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Elacestrant is a novel, nonsteroidal, orally bioavailable selective estrogen receptor degrader (SERD) that has demonstrated activity in patients with estrogen receptor (ER)-positive/HER2-negative breast cancer previously treated with endocrine therapies including fulvestrant and/or CDK 4/6 inhibitor therapy, and in those with ESR1 mutations (ESR1-mut) known to confer endocrine resistance. Herein, we describe the design and methodology of EMERALD, an international, multicenter, randomized, open-label, active-controlled, Phase III clinical study comparing the efficacy and safety of elacestrant to standard-of-care endocrine monotherapy treatment (fulvestrant or an aromatase inhibitor, per investigator's choice) in patients with ER-positive/HER2-negative advanced breast cancer. Primary end points are progression-free survival in ESR1-mut patients and in all patients (NCT03778931; EudraCT 2018-002990-24).
Collapse
Affiliation(s)
- Aditya Bardia
- Massachusetts General Hospital Cancer Center, Departments of Medicine and Hematology/Oncology, 55 Fruit Street Boston, MA 02114-3411, USA
| | - Philippe Aftimos
- Clinical Trials Conduct Unit, Institut Jules Bordet, Université Libre de Bruxelles, Rue Heger-Bordet1, 1000 Brussels, Belgium
| | - Teeru Bihani
- Radius Health, Inc., 950 Winter Street Waltham, MA 02451, USA
| | | | - JungAh Jung
- Radius Health, Inc., 950 Winter Street Waltham, MA 02451, USA
| | | | - Virginia G Kaklamani
- UT Health San Antonio MD Anderson Cancer Center, Division of Hematology/Oncology 7979 Wurzbach Road, San Antonio, TX 78229, USA
| |
Collapse
|
14496
|
Deziel NC, Alfonso-Garrido J, Warren JL, Huang H, Sjodin A, Zhang Y. Exposure to Polybrominated Diphenyl Ethers and a Polybrominated Biphenyl and Risk of Thyroid Cancer in Women: Single and Multi-Pollutant Approaches. Cancer Epidemiol Biomarkers Prev 2019; 28:1755-1764. [PMID: 31387967 PMCID: PMC6774868 DOI: 10.1158/1055-9965.epi-19-0526] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/30/2019] [Accepted: 07/30/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Thyroid cancer incidence is the most rapidly increasing malignancy; rates are three times higher in women than men. Thyroid hormone-disrupting flame-retardant chemicals, including polybrominated diphenyl ethers (PBDE) and polybrominated biphenyls (PBB), may contribute to this trend. METHODS We investigated the relationship between PBDE/PBB exposure and papillary thyroid cancer (PTC) in 250 incident female papillary thyroid cancer cases and 250 female controls frequency-matched on age. Interviews and postdiagnostic serum samples were collected from 2010 to 2013. Serum samples were analyzed for 11 congeners. We calculated ORs and 95% confidence intervals (95% CI) using single-pollutant logistic regression models for continuous and categorical lipid-adjusted serum concentrations of PBDE/PBB, adjusted for age, alcohol consumption, and education. We applied three multi-pollutant approaches [standard multipollutant regression models, hierarchical Bayesian logistic regression modeling (HBLR), principal components analysis (PCA)] to investigate associations with PBDE/PBB mixtures. RESULTS In single-pollutant models, a decreased risk was observed at the highest (>90th percentile) versus lowest ( CONCLUSIONS Our results using single- and multi-pollutant modeling do not generally support a positive association with PBDE/PBB and PTC risk. IMPACT Prospective studies with more advanced statistical approaches to analyze mixtures and populations with higher exposures could reveal new insights.
Collapse
Affiliation(s)
- Nicole C Deziel
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut.
| | - Javier Alfonso-Garrido
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut
| | - Joshua L Warren
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Huang Huang
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Andreas Sjodin
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yawei Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
14497
|
Eich ML, Chandrashekar DS, Rodriguez Pen A MDC, Robinson AD, Siddiqui J, Daignault-Newton S, Chakravarthi BVSK, Kunju LP, Netto GJ, Varambally S. Characterization of glycine-N-acyltransferase like 1 (GLYATL1) in prostate cancer. Prostate 2019; 79:1629-1639. [PMID: 31376196 DOI: 10.1002/pros.23887] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/09/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent microarray and sequencing studies of prostate cancer showed multiple molecular alterations during cancer progression. It is critical to evaluate these molecular changes to identify new biomarkers and targets. We performed analysis of glycine-N-acyltransferase like 1 (GLYATL1) expression in various stages of prostate cancer in this study and evaluated the regulation of GLYATL1 by androgen. METHOD We performed in silico analysis of cancer gene expression profiling and transcriptome sequencing to evaluate GLYATL1 expression in prostate cancer. Furthermore, we performed immunohistochemistry using specific GLYATL1 antibody using high-density prostate cancer tissue microarray containing primary and metastatic prostate cancer. We also tested the regulation of GLYATL1 expression by androgen and ETS transcription factor ETV1. In addition, we performed RNA-sequencing of GLYATL1 modulated prostate cancer cells to evaluate the gene expression and changes in molecular pathways. RESULTS Our in silico analysis of cancer gene expression profiling and transcriptome sequencing we revealed an overexpression of GLYATL1 in primary prostate cancer. Confirming these findings by immunohistochemistry, we show that GLYATL1 is overexpressed in primary prostate cancer compared with metastatic prostate cancer and benign prostatic tissue. Low-grade cancers had higher GLYATL1 expression compared to high-grade prostate tumors. Our studies showed that GLYATL1 is upregulated upon androgen treatment in LNCaP prostate cancer cells which harbors ETV1 gene rearrangement. Furthermore, ETV1 knockdown in LNCaP cells showed downregulation of GLYATL1 suggesting potential regulation of GLYATL1 by ETS transcription factor ETV1. Transcriptome sequencing using the GLYATL1 knockdown prostate cancer cell lines LNCaP showed regulation of multiple metabolic pathways. CONCLUSIONS In summary, our study characterizes the expression of GLYATL1 in prostate cancer and explores the regulation of its regulation in prostate cancer showing role for androgen and ETS transcription factor ETV1. Future studies are needed to decipher the biological significance of these findings.
Collapse
Affiliation(s)
- Marie-Lisa Eich
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | - Alyncia D Robinson
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Javed Siddiqui
- Department of Pathology, The University of Michigan, Ann Arbor, Michigan
| | | | | | | | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Sooryanarayana Varambally
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
14498
|
Franzoni MS, Brandi A, de Oliveira Matos Prado JK, Elias F, Dalmolin F, de Faria Lainetti P, Prado MCM, Leis-Filho AF, Fonseca-Alves CE. Tumor-infiltrating CD4 + and CD8 + lymphocytes and macrophages are associated with prognostic factors in triple-negative canine mammary complex type carcinoma. Res Vet Sci 2019; 126:29-36. [PMID: 31425936 DOI: 10.1016/j.rvsc.2019.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
This study aimed to evaluate the association of CD3+, CD4+, and CD8+ T cells and tumor-infiltrating macrophages (TIMs) with the clinical parameters of female dogs harboring mammary gland tumors. Thirty female dogs affected with mammary carcinomas were used, and all tumors were histologically classified as complex carcinoma and were triple-negative phenotype determined by immunohistochemistry. Freshly frozen sections were used to determine CD3+, CD4+ and CD8+ T cells by immunohistochemistry, and TIMs were determined by immunofluorescence assays. Ten out of the 30 dogs showed lymph node metastasis at diagnosis. Fifteen dogs had a tumor of grade I (15/30), nine (9/30) had a tumor of grade II and six (6/30) had a tumor of grade III. The mean overall survival was 680.5 days (± 200.4). Dogs with sentinel lymph node positivity (10/30) (P = .0035) and dogs that developed metastasis (P = .0001) showed a shorter survival time. In addition, dogs with a high level of inflammatory infiltrate in tumor tissues presented a shorter survival time (P = .0001) than that of other dogs. Dogs with tumors containing higher numbers of CD3+ T cells (P = .001), CD4+ T cells (P = .001), or TIM cells (P < .0001) showed a shorter survival time than that of other dogs. Our results suggested that characteristics of immune cell infiltrates, including CD3+ T cells, CD4+ T cells, and TIMs, can be used as potential prognostic indicators for predicting clinical outcomes in dogs with mammary gland tumors, particularly tumors with a complex histological subtype and triple-negative phenotype.
Collapse
Affiliation(s)
- Mayara Simão Franzoni
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Andressa Brandi
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil
| | | | - Fabiana Elias
- Superintendencia Unidade Hospitalar Veterinaria Universitaria, Federal University of the Fronteira Sul, Realeza, Brazil
| | - Fabíola Dalmolin
- Superintendencia Unidade Hospitalar Veterinaria Universitaria, Federal University of the Fronteira Sul, Realeza, Brazil
| | - Patricia de Faria Lainetti
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Maria Carolina Mangini Prado
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Antonio Fernando Leis-Filho
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science São Paulo State University - UNESP, Botucatu, SP, Brazil; Universidade Paulista - UNIP, Bauru, SP, Brazil.
| |
Collapse
|
14499
|
HER2 somatic mutation analysis in breast cancer: correlation with clinicopathological features. Hum Pathol 2019; 92:32-38. [DOI: 10.1016/j.humpath.2019.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/13/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
|
14500
|
He Y, Jia K, Dziadziuszko R, Zhao S, Zhang X, Deng J, Wang H, Hirsch FR, Zhou C. Galectin-9 in non-small cell lung cancer. Lung Cancer 2019; 136:80-85. [DOI: 10.1016/j.lungcan.2019.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/11/2022]
|