14851
|
Miller DR, Ingersoll MA, Chatterjee A, Baker B, Shrishrimal S, Kosmacek EA, Zhu Y, Cheng PW, Oberley-Deegan RE, Lin MF. p66Shc protein through a redox mechanism enhances the progression of prostate cancer cells towards castration-resistance. Free Radic Biol Med 2019; 139:24-34. [PMID: 31100478 PMCID: PMC6620027 DOI: 10.1016/j.freeradbiomed.2019.05.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) remains the second leading cause of cancer-related deaths in U.S. men due to the development of the castration-resistant (CR) PCa phenotype. A useful cell model for analysis of the molecular mechanism of PCa progression is required for developing targeted therapies toward CR PCa. In this study, we established a PCa cell progressive model in three separate cell lines, of which androgen-independent (AI) cells were derived from respective androgen-sensitive (AS) cells. Those AI PCa cells obtain the biochemical properties of the clinical CR phenotype, including AR and PSA expression as well as enhanced proliferation and tumorigenicity under androgen-deprived conditions. Thus, those AI cells recapitulate CR PCa and exhibit increased oxidant species levels as well as enhanced signaling of proliferation and survival pathways. H2O2 treatment directly enhanced AS cell growth and migration, which was counteracted by antioxidant N-acetyl cysteine (NAC). We further identified p66Shc protein enhances the production of oxidant species which contributes to phenotypic and cell signaling alterations from AS to AI PCa cells. H2O2-treated LNCaP-AS cells had a similar signaling profile to that of LNCaP-AI or p66Shc subclone cells. Conversely, the oxidant species-driven alterations of LNCaP-AI and p66Shc subclone cell signaling is mitigated by p66Shc knockdown. Moreover, LNCaP-AI cells and p66Shc subclones, but not LNCaP-AS cells, develop xenograft tumors with metastatic nodules, correlating with p66Shc protein levels. Together, the data shows that p66Shc enhances oxidant species production that plays a role in promoting PCa progression to the CR stage.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Disease Progression
- Drug Resistance, Neoplasm/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Heterografts
- Humans
- Hydrogen Peroxide/pharmacology
- Kallikreins/genetics
- Kallikreins/metabolism
- Lymphatic Metastasis
- Male
- Mice
- Mice, Nude
- Prostate/drug effects
- Prostate/metabolism
- Prostate/pathology
- Prostate-Specific Antigen/genetics
- Prostate-Specific Antigen/metabolism
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Reactive Oxygen Species/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
- Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
Collapse
Affiliation(s)
- Dannah R Miller
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew A Ingersoll
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brian Baker
- Department of Biology, Clark Atlanta University, Atlanta, GA, USA
| | - Shashank Shrishrimal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yuxiang Zhu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pi-Wan Cheng
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Section of Urology, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
14852
|
Bilgin B, Sendur MAN, Hizal M, Kandil SU, Yaman S, Akıncı MB, Dede DŞ, Neselioglu S, Erel Ö, Yalçın B. Evaluation of dynamic serum thiol-disulphide homeostasis in colorectal cancer. JOURNAL OF ONCOLOGICAL SCIENCES 2019. [DOI: 10.1016/j.jons.2019.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
14853
|
Ansari D, Althini C, Ohlsson H, Andersson R. Early-onset pancreatic cancer: a population-based study using the SEER registry. Langenbecks Arch Surg 2019; 404:565-571. [PMID: 31377855 PMCID: PMC6713682 DOI: 10.1007/s00423-019-01810-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/24/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Early-onset pancreatic cancer (< 50 years, EOPC) is uncommon and limited data exist on clinical presentation and long-term survival. The aim of this study was to compare outcomes between patients with EOPC and those with later-onset pancreatic cancer (≥ 50 years, LOPC) using a large population-based cohort. METHODS The Surveillance, Epidemiology, and End Results (SEER) database was queried to identify patients with a microscopically confirmed pancreatic ductal adenocarcinoma for the period 2004 to 2016. Propensity score matching was used to compare overall survival (OS) and cancer-specific survival (CSS) between patients with EOPC and LOPC. The EOPC and LOPC patients were paired 1:1 on propensity scores based on gender, tumor location, tumor size, AJCC stage, and treatment details. RESULTS The overall cohort included 72,906 patients with pancreatic ductal adenocarcinoma, including 4523 patients with EOPC (6.2%). EOPC patients were diagnosed at a more advanced AJCC stage (p < 0.001) compared with LOPC patients and received significantly more treatment, including surgery (p < 0.001), radiation (p < 0.001), and chemotherapy (p < 0.001). Following propensity score matching, 3172 EOPC patients were matched to 3172 LOPC patients, alleviating any covariate differences between the groups. The matched analysis showed that EOPC was associated with poorer 5-year OS (6.1% vs 8.6%, p = 0.003) and 5-year CSS (6.7% vs 9.7%, p < 0.001). In multivariable Cox regression analysis, EOPC remained significantly associated with adverse OS and CSS. Subgroup analyses showed that EOPC was associated with adverse 5-year OS (17.7% vs 26.9%, p < 0.001) and 5-year CSS (18.9% vs 29.7%, p < 0.001) in operated patients. After multivariable analysis, EOPC remained significantly associated with OS and CSS. For patients that did not undergo surgery, the OS and CSS remained dismal without any significant differences between the groups. CONCLUSION To our knowledge, this is the largest study to compare the outcome of EOPC vs LOPC, as well as the first to use propensity score matching methodology for this purpose. The findings demonstrate that EOPC is diagnosed at a later stage and the matched survival analysis demonstrated reduced OS and CSS. We suggest that pancreatic cancer in young patients may have a unique tumor biology, which may be of importance for risk stratification and patient counseling.
Collapse
Affiliation(s)
- Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 85, Lund, Sweden.
| | - Carl Althini
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 85, Lund, Sweden
| | - Henrik Ohlsson
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 85, Lund, Sweden
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, SE-221 85, Lund, Sweden
| |
Collapse
|
14854
|
Whole-lesion histogram and texture analyses of breast lesions on inline quantitative DCE mapping with CAIPIRINHA-Dixon-TWIST-VIBE. Eur Radiol 2019; 30:57-65. [PMID: 31372782 DOI: 10.1007/s00330-019-06365-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/17/2019] [Accepted: 07/10/2019] [Indexed: 01/01/2023]
Abstract
PURPOSE To investigate the diagnostic capability of whole-lesion (WL) histogram and texture analysis of dynamic contrast-enhanced (DCE) MRI inline-generated quantitative parametric maps using CAIPIRINHA-Dixon-TWIST-VIBE (CDTV) to differentiate malignant from benign breast lesions and breast cancer subtypes. MATERIALS AND METHODS From February 2018 to November 2018, DCE MRI using CDTV was performed on 211 patients. The inline-generated parametric maps included Ktrans, kep, Ve, and IAUGC60. Histogram and texture features were extracted from the above parametric maps respectively based on a WL analysis. Student's t tests, one-way ANOVAs, Mann-Whitney U tests, Jonckheere-Terpstra tests, and ROC curves were used for statistical analysis. RESULTS Compared with benign breast lesions, malignant breast lesions showed significantly higher Ktrans_median, 5th percentile, entropy, and diff-entropy, IAUGC60_median, 5th percentile, entropy, and diff-entropy, kep_mean, median, 5th percentile, entropy, and diff-entropy, and Ve_95th percentile, diff-variance, and contrast, and significantly lower kep_skewness and Ve_SD, entropy, diff-entropy, and skewness (all p ≤ 0.011). The combination of all the extracted parameters yielded an AUC of 0.85 (sensitivity 76%, specificity 86%). kep_contrast showed a significant difference among different subtypes of breast cancer (p = 0.006). kep_skewness showed a significant difference between lymph node-positive and lymph node-negative breast cancer (p = 0.007). The IAGC60_5th percentile had an AUC of 0.71 (sensitivity 50%, specificity 91%) for differentiating between high- and low-proliferation groups of breast cancer. CONCLUSIONS The WL histogram and texture analyses of CDTV-DCE-derived parameters may give additional information for further evaluation of breast cancer. KEY POINTS • Inline DCE mapping with CDTV is effective and time-saving. • WL histogram and texture-extracted features could distinguish breast cancer from benign lesions accurately. • kep_contrast, kep_skewness, and IAUGC60_5th percentile could predict breast cancer subtypes, lymph node metastasis, and proliferation abilities, respectively.
Collapse
|
14855
|
Leuva H, Sigel K, Zhou M, Wilkerson J, Aggen DH, Park YHA, Anderson CB, Hsu TCM, Langhoff E, McWilliams G, Drake CG, Simon R, Bates SE, Fojo T. A novel approach to assess real-world efficacy of cancer therapy in metastatic prostate cancer. Analysis of national data on Veterans treated with abiraterone and enzalutamide. Semin Oncol 2019; 46:351-361. [DOI: 10.1053/j.seminoncol.2019.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Indexed: 11/11/2022]
|
14856
|
Lee JK, Agrawal D, Thosani N, Al-Haddad M, Buxbaum JL, Calderwood AH, Fishman DS, Fujii-Lau LL, Jamil LH, Jue TL, Khashab MA, Law JK, Naveed M, Qumseya BJ, Sawhney MS, Storm AC, Yang J, Wani SB. ASGE guideline on the role of endoscopy for bleeding from chronic radiation proctopathy. Gastrointest Endosc 2019; 90:171-182.e1. [PMID: 31235260 DOI: 10.1016/j.gie.2019.04.234] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 04/22/2019] [Indexed: 12/11/2022]
Abstract
Chronic radiation proctopathy is a common sequela of radiation therapy for malignancies in the pelvic region. A variety of medical and endoscopic therapies have been used for the management of bleeding from chronic radiation proctopathy. In this guideline, we reviewed the results of a systematic search of the literature from 1946 to 2017 to formulate clinical questions and recommendations on the role of endoscopy for bleeding from chronic radiation proctopathy. The following endoscopic modalities are discussed in our document: argon plasma coagulation, bipolar electrocoagulation, heater probe, radiofrequency ablation, and cryoablation. Most studies were small observational studies, and the evidence for effectiveness of endoscopic therapy for chronic radiation proctopathy was limited because of a lack of controlled trials and comparative studies. Despite this limitation, our systematic review found that argon plasma coagulation, bipolar electrocoagulation, heater probe, and radiofrequency ablation were effective in the treatment of rectal bleeding from chronic radiation proctopathy.
Collapse
Affiliation(s)
- Jeffrey K Lee
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California, USA
| | - Deepak Agrawal
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nirav Thosani
- Division of Gastroenterology, Hepatology and Nutrition, McGovern Medical School, UTHealth, Houston, Texas, USA
| | - Mohammad Al-Haddad
- Division of Gastroenterology/Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - James L Buxbaum
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Audrey H Calderwood
- Department of Gastroenterology and Hepatology, Dartmouth Hitchcock Medical Center, Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Douglas S Fishman
- Department of Gastroenterology, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, USA
| | | | - Laith H Jamil
- Pancreatic and Biliary Diseases Program, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Terry L Jue
- The Permanente Medical Group, Kaiser Permanente San Francisco Medical Center, San Francisco, California, USA
| | - Mouen A Khashab
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joanna K Law
- Digestive Disease Institute, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Mariam Naveed
- Division of Gastroenterology and Hepatology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, USA
| | - Bashar J Qumseya
- Department of Gastroenterology, Archbold Medical Group, Thomasville, Georgia, USA
| | - Mandeep S Sawhney
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew C Storm
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Julie Yang
- Division of Gastroenterology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sachin B Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| |
Collapse
|
14857
|
|
14858
|
Derry HM, Reid MC, Prigerson HG. Advanced cancer patients' understanding of prognostic information: Applying insights from psychological research. Cancer Med 2019; 8:4081-4088. [PMID: 31199597 PMCID: PMC6675734 DOI: 10.1002/cam4.2331] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/06/2019] [Accepted: 05/22/2019] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Informed medical decision-making at the end of life often requires engaging in highly emotional, potentially upsetting discussions about prognosis, while ensuring that patients grasp its personal meaning. Behavioral science offers insights into ways to promote prognostic understanding among patients with advanced cancer. SUMMARY In this literature review, we synthesize complementary findings from basic behavioral science and applied clinical research, which suggest that psychological factors can significantly influence both patients' clinical interactions and their prognostic understanding. For example, stress and emotion can affect cognition, which may shape how patients process complex medical information. Additionally, clinicians may be less likely to share prognostic information with distressed patients who, in turn, may be hesitant to ask about their prognosis for fear of the answer. Although traditional approaches for increasing advanced cancer patients' understanding focus on improving information delivery, these efforts may not be sufficient without corresponding interventions that assist patients in managing distress. CONCLUSIONS Psychological barriers may limit opportunities for patients to fully understand their prognosis and to receive high quality of end-of-life care that is linked with an accurate understanding of their disease and treatment options. Failure to attend to patients' emotional distress may undermine efforts to improve medical communication. This underscores the importance of increased attention to the psychological factors that impede patients' comprehension of material shared in cancer clinic visits, in order to inform interventions that address patient distress both before and after receiving "bad news." Integrating findings from psychological research into prognostic discussions may not only improve advanced cancer patients' mental health, but may also promote their ability to make informed, value-consistent medical decisions.
Collapse
|
14859
|
Arafat M, Fouladian P, Blencowe A, Albrecht H, Song Y, Garg S. Drug-eluting non-vascular stents for localised drug targeting in obstructive gastrointestinal cancers. J Control Release 2019; 308:209-231. [DOI: 10.1016/j.jconrel.2019.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023]
|
14860
|
Zeng X, Peng L, Tan C, Wang Y. Cost-effectiveness analysis of positron-emission tomography-computed tomography in preoperative staging for nonsmall-cell lung cancer with resected monometastatic disease. Medicine (Baltimore) 2019; 98:e16843. [PMID: 31415409 PMCID: PMC6831239 DOI: 10.1097/md.0000000000016843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The aim of this study was, from the Chinese healthcare perspective, to assess the cost-effectiveness of positron-emission tomography-computed tomography (PET-CT) with F-fluorodeoxyglucose (F-FDG) in preoperation staging for nonsmall-cell lung cancer (NSCLC) with resected monometastatic disease based on a retrospective study. This study was conducted from January 2017 to February 2019 at an academic hospital. METHODS A Markov model and 3 decision-tree models were designed to calculate the long-term medical costs, outcomes, and incremental cost-effectiveness ratios (ICERs) of the 2 diagnostic strategies (PET-CT and conventional CT). Model robustness was assessed in sensitivity analyses. RESULTS For the base-case analysis, preoperative PET-CT evaluation for NSCLC with resected monometastatic disease provided an additional 1.475, 2.129, and 2.412 life-years (LYs), in the time horizon of 10-, 20-, and 30-year, respectively, and the ICERs for the PET-CT group compared with the conventional CT group were $1153, $1393, and $1430 per LY, separately. The acceptability curves demonstrated that when the willingness-to-pay (WTP) thresholds ranged from $500 to $3000/LY, the probability of cost-effectiveness changed varied dramatically, and at WTP > $3000, the probability that the PET-CT group achieved cost-effectiveness was 100%. Sensitivity analyses suggested that the models we designed were robust. CONCLUSION Compared with conventional CT scan, preoperative F-FDG PET-CT evaluation for patients with resected monometastatic NSCLC is cost-effective from the Chinese healthcare perspective. Preoperative F-FDG PET-CT evaluation should be popularized for patients with resected monometastatic NSCLC.
Collapse
Affiliation(s)
| | - Liubao Peng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chongqing Tan
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | |
Collapse
|
14861
|
Abou Dalle I, Ravandi F. Moxetumomab pasudotox for the treatment of relapsed and/or refractory hairy cell leukemia. Expert Rev Hematol 2019; 12:707-714. [PMID: 31298972 DOI: 10.1080/17474086.2019.1643231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Introduction: Hairy cell leukemia is a rare indolent B-cell malignancy, characterized by pancytopenia, recurrent infections, and splenomegaly. After initial therapy with purine nucleoside analogs, up to 50% of patients relapse after several years of remission. The number of relapsed patients is increasing and, until recently, there was no approved therapy with durable responses for hairy cell leukemia patients in the relapsed setting, thus the need for new non-chemotherapy approach with significant efficacy and less myelosuppression. Areas covered: Moxetumomab pasudotox is a recombinant immunotoxin containing a Fv fragment of an anti-CD22 monoclonal antibody and truncated Pseudomonas exotoxin (PE38). The authors reviewed pre-clinical and clinical studies that led to the FDA approval of the drug in patients with relapsed and/or refractory hairy cell leukemia, who received at least two prior therapies, including at least one purine nucleoside analog. Expert opinion: Moxetumomab pasudotox demonstrated a durable complete remission rate of 30% in heavily pretreated patients with hairy cell leukemia, and MRD eradication in 85% of responding patients. Moxetumomab pasudotox got a global FDA approval in September 2018. The US prescribing information carries boxed warnings regarding the risk of capillary leak syndrome and hemolytic uremic syndrome. Long-term follow-up of the pivotal study is ongoing (NCT01829711).
Collapse
Affiliation(s)
- Iman Abou Dalle
- Department of leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Farhad Ravandi
- Department of leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
14862
|
Sun C, Tian X, Liu Z, Li W, Li P, Chen J, Zhang W, Fang Z, Du P, Duan H, Liu P, Wang L, Chen C, Tian J. Radiomic analysis for pretreatment prediction of response to neoadjuvant chemotherapy in locally advanced cervical cancer: A multicentre study. EBioMedicine 2019; 46:160-169. [PMID: 31395503 PMCID: PMC6712288 DOI: 10.1016/j.ebiom.2019.07.049] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/05/2019] [Accepted: 07/18/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND We aimed to investigate whether pre-therapeutic radiomic features based on magnetic resonance imaging (MRI) can predict the clinical response to neoadjuvant chemotherapy (NACT) in patients with locally advanced cervical cancer (LACC). METHODS A total of 275 patients with LACC receiving NACT were enrolled in this study from eight hospitals, and allocated to training and testing sets (2:1 ratio). Three radiomic feature sets were extracted from the intratumoural region of T1-weighted images, intratumoural region of T2-weighted images, and peritumoural region of T2-weighted images before NACT for each patient. With a feature selection strategy, three single sequence radiomic models were constructed, and three additional combined models were constructed by combining the features of different regions or sequences. The performance of all models was assessed using receiver operating characteristic curve. FINDINGS The combined model of the intratumoural zone of T1-weighted images, intratumoural zone of T2-weighted images,and peritumoural zone of T2-weighted images achieved an AUC of 0.998 in training set and 0.999 in testing set, which was significantly better (p < .05) than the other radiomic models. Moreover, no significant variation in performance was found if different training sets were used. INTERPRETATION This study demonstrated that MRI-based radiomic features hold potential in the pretreatment prediction of response to NACT in LACC, which could be used to identify rightful patients for receiving NACT avoiding unnecessary treatment.
Collapse
Affiliation(s)
- Caixia Sun
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, School of Computer Science and Technology, Guizhou University, Guiyang, China; CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Xin Tian
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Weili Li
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengfei Li
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaming Chen
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weifeng Zhang
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziyu Fang
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peiyan Du
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Duan
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Liu
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lihui Wang
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, School of Computer Science and Technology, Guizhou University, Guiyang, China.
| | - Chunlin Chen
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China; Engineering Research Center of Molecular and NeSuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China.
| |
Collapse
|
14863
|
Faria S, Devine C, Rao B, Sagebiel T, Bhosale P. Imaging and Staging of Endometrial Cancer. Semin Ultrasound CT MR 2019; 40:287-294. [DOI: 10.1053/j.sult.2019.04.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14864
|
Blundon MA, Dasgupta S. Metabolic Dysregulation Controls Endocrine Therapy-Resistant Cancer Recurrence and Metastasis. Endocrinology 2019; 160:1811-1820. [PMID: 31157867 PMCID: PMC6620757 DOI: 10.1210/en.2019-00097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/24/2019] [Indexed: 01/16/2023]
Abstract
Cancer recurrence and metastasis involves many biological interactions, such as genetic, transcription, environmental, endocrine signaling, and metabolism. These interactions add a complex understanding of cancer recurrence and metastatic progression, delaying the advancement in therapeutic opportunities. We highlight the recent advances on the molecular complexities of endocrine-related cancers, focusing on breast and prostate cancer, and briefly review how endocrine signaling and metabolic programs can influence transcriptional complexes for metastasis competence. Nuclear receptors and transcriptional coregulators function as molecular nodes for the crosstalk between endocrine signaling and metabolism that alter downstream gene expression important for tumor progression and metastasis. This exciting regulatory axis may provide insights to the development of cancer therapeutics important for these desensitized endocrine-dependent cancers.
Collapse
Affiliation(s)
- Malachi A Blundon
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Correspondence: Subhamoy Dasgupta, PhD, Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14263. E-mail:
| |
Collapse
|
14865
|
Mitra AK, Yang-Hartwich Y. Tumor microenvironment and immunology of ovarian cancer: 12th Biennial Rivkin Center Ovarian Cancer Research Symposium. Int J Gynecol Cancer 2019; 29:s12-s15. [PMID: 31462543 DOI: 10.1136/ijgc-2019-000666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 11/03/2022] Open
Abstract
The 12th Biennial Ovarian Cancer Research Symposium organized by the Rivkin Center for Ovarian Cancer and the American Association for Cancer Research held on September 13-15, 2018 covered cutting edge and relevant research topics in ovarian cancer biology and therapy. Sessions included detection and prevention, genomics and molecular mechanisms, tumor microenvironment and immunology, novel therapeutics, and an education session. In this article we provide an overview of the key findings presented in the tumor microenvironment and immunology session.
Collapse
Affiliation(s)
- Anirban K Mitra
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Yang Yang-Hartwich
- Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14866
|
Di Corpo M, Schlottmann F, Strassle PD, Nurczyk K, Patti MG. Treatment Modalities for Esophageal Adenocarcinoma in the United States: Trends and Survival Outcomes. J Laparoendosc Adv Surg Tech A 2019; 29:989-994. [DOI: 10.1089/lap.2019.0350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Marco Di Corpo
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Francisco Schlottmann
- Department of Surgery, Hospital Alemán of Buenos Aires, University of Buenos Aires, Buenos Aires, Argentina
| | - Paula D. Strassle
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kamil Nurczyk
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Marco G. Patti
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
14867
|
Wong C, Chen F, Alirezaie N, Wang Y, Cuggia A, Borgida A, Holter S, Lenko T, Domecq C, Petersen GM, Syngal S, Brand R, Rustgi AK, Cote ML, Stoffel E, Olson SH, Roberts NJ, Akbari MR, Majewski J, Klein AP, Greenwood CMT, Gallinger S, Zogopoulos G. A region-based gene association study combined with a leave-one-out sensitivity analysis identifies SMG1 as a pancreatic cancer susceptibility gene. PLoS Genet 2019; 15:e1008344. [PMID: 31469826 PMCID: PMC6742418 DOI: 10.1371/journal.pgen.1008344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/12/2019] [Accepted: 08/03/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic adenocarcinoma (PC) is a lethal malignancy that is familial or associated with genetic syndromes in 10% of cases. Gene-based surveillance strategies for at-risk individuals may improve clinical outcomes. However, familial PC (FPC) is plagued by genetic heterogeneity and the genetic basis for the majority of FPC remains elusive, hampering the development of gene-based surveillance programs. The study was powered to identify genes with a cumulative pathogenic variant prevalence of at least 3%, which includes the most prevalent PC susceptibility gene, BRCA2. Since the majority of known PC susceptibility genes are involved in DNA repair, we focused on genes implicated in these pathways. We performed a region-based association study using the Mixed-Effects Score Test, followed by leave-one-out characterization of PC-associated gene regions and variants to identify the genes and variants driving risk associations. We evaluated 398 cases from two case series and 987 controls without a personal history of cancer. The first case series consisted of 109 patients with either FPC (n = 101) or PC at ≤50 years of age (n = 8). The second case series was composed of 289 unselected PC cases. We validated this discovery strategy by identifying known pathogenic BRCA2 variants, and also identified SMG1, encoding a serine/threonine protein kinase, to be significantly associated with PC following correction for multiple testing (p = 3.22x10-7). The SMG1 association was validated in a second independent series of 532 FPC cases and 753 controls (p<0.0062, OR = 1.88, 95%CI 1.17-3.03). We showed segregation of the c.4249A>G SMG1 variant in 3 affected relatives in a FPC kindred, and we found c.103G>A to be a recurrent SMG1 variant associating with PC in both the discovery and validation series. These results suggest that SMG1 is a novel PC susceptibility gene, and we identified specific SMG1 gene variants associated with PC risk.
Collapse
Affiliation(s)
- Cavin Wong
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Fei Chen
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Najmeh Alirezaie
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Yifan Wang
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Adeline Cuggia
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Ayelet Borgida
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Spring Holter
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Tatiana Lenko
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | - Celine Domecq
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| | | | - Gloria M. Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sapna Syngal
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Gastroenterology Division, Brigham and Women’s Hospital, Harvard Medical Schozol, Boston, Massachusetts, United States of America
| | - Randall Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anil K. Rustgi
- Division of Gastroenterology, Departments of Medicine and Genetics, Pancreatic Cancer Translation Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michele L. Cote
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Elena Stoffel
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sara H. Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Nicholas J. Roberts
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mohammad R. Akbari
- Women’s College Hospital Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jacek Majewski
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Alison P. Klein
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Celia M. T. Greenwood
- Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, and Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - George Zogopoulos
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- The Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14868
|
Vansteenkiste JF, Van De Kerkhove C, Wauters E, Van Mol P. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther 2019; 19:659-671. [PMID: 31368815 DOI: 10.1080/14737140.2019.1643239] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: Activation of the MET pathway through MET amplifications or mutations is present in 3-4% of stage IV non-squamous non-small cell lung cancers (NSCLC). High MET amplifications and exon 14 skipping mutations are associated with poor prognosis: new treatments are needed for these patients. Capmatinib is a highly selective, potent small-molecule MET inhibitor with antitumor activity in NSCLC in vitro and in vivo. Areas covered: This article provides an overview of the capmatinib clinical development program in NSCLC, both as monotherapy in NSCLC with a dysregulated MET pathway, and in combination with epidermal growth factor receptor (EGFR) inhibitor therapy in EGFR-mutant NSCLC with MET-based acquired resistance to previous EGFR inhibition. Expert opinion: In the GEOMETRY Mono-1 study, treatment with capmatinib resulted in high response rates in stage IV NSCLC with MET exon 14 skipping mutations, particularly in first line, supporting testing for this biomarker at the time of diagnosis. Durable responses have been reported and results in MET-amplified NSCLC are eagerly anticipated. In EGFR-mutant NSCLC, notable responses have been observed in combination with an EGFR-tyrosine kinase inhibitor (TKI) in case of acquired resistance to EGFR-TKIs based on high MET amplification.
Collapse
Affiliation(s)
| | | | - Els Wauters
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| | - Pierre Van Mol
- Respiratory Oncology Unit (Respiratory Diseases), University Hospital KU Leuven , Leuven , Belgium
| |
Collapse
|
14869
|
Alijabbari N, Alshahrani SS, Pattyn A, Mehrmohammadi M. Photoacoustic Tomography with a Ring Ultrasound Transducer: A Comparison of Different Illumination Strategies. APPLIED SCIENCES (BASEL, SWITZERLAND) 2019; 9:10.3390/app9153094. [PMID: 32095283 PMCID: PMC7039403 DOI: 10.3390/app9153094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Photoacoustic (PA) imaging is a methodology that uses the absorption of short laser pulses by endogenous or exogenous chromophores within human tissue, and the subsequent generation of acoustic waves acquired by an ultrasound (US) transducer, to form an image that can provide functional and molecular information. Amongst the various types of PA imaging, PA tomography (PAT) has been proposed for imaging pathologies such as breast cancer. However, the main challenge for PAT imaging is the deliverance of sufficient light energy horizontally through an imaging cross-section as well as vertically. In this study, three different illumination methods are compared for a full-ring ultrasound (US) PAT system. The three distinct illumination setups are full-ring, diffused-beam, and point source illumination. The full-ring system utilizes a cone mirror and parabolic reflector to create the ringed-shaped beam for PAT, while the diffuse scheme uses a light diffuser to expand the beam, which illuminates tissue-mimicking phantoms. The results indicate that the full-ring illumination is capable of providing a more uniform fluence irrespective of the vertical depth of the imaged cross-section, while the point source and diffused illumination methods provide a higher fluence at regions closer to the point of entry, which diminishes with depth. In addition, a set of experiments was conducted to determine the optimum position of ring-illumination with respect to the position of the acoustic detectors to achieve the highest signal-to-noise ratio.
Collapse
Affiliation(s)
- Naser Alijabbari
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Suhail S. Alshahrani
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Alexander Pattyn
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Mohammad Mehrmohammadi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA
- Department of Electrical and Computer Engineering, Wayne State University, Detroit, MI 48201, USA
- Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA
| |
Collapse
|
14870
|
Caspa Gokulan R, Garcia-Buitrago MT, Zaika AI. From genetics to signaling pathways: molecular pathogenesis of esophageal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2019; 1872:37-48. [PMID: 31152823 PMCID: PMC6692203 DOI: 10.1016/j.bbcan.2019.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Esophageal adenocarcinoma (EAC) has one of the fastest rising incidence rates in the U.S. and many other Western countries. One of the unique risk factors for EAC is gastroesophageal reflux disease (GERD), a chronic digestive condition in which acidic contents from the stomach, frequently mixed with duodenal bile, enter the esophagus resulting in esophageal tissue injury. At the cellular level, progression to EAC is underlined by continuous DNA damage caused by reflux and chronic inflammatory factors that increase the mutation rate and promote genomic instability. Despite recent successes in cancer diagnostics and treatment, EAC remains a poorly treatable disease. Recent research has shed new light on molecular alterations underlying progression to EAC and revealed novel treatment options. This review focuses on the genetic and molecular studies of EAC. The molecular changes that occur during the transformation of normal Barrett's esophagus to esophageal adenocarcinoma are also discussed.
Collapse
Affiliation(s)
| | | | - Alexander I Zaika
- Department of Surgery, University of Miami, Miami, FL, United States of America; Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, United States of America.
| |
Collapse
|
14871
|
Wang X, Lyu J, Ji A, Zhang Q, Liao G. Jarid2 enhances the progression of bladder cancer through regulating PTEN/AKT signaling. Life Sci 2019; 230:162-168. [DOI: 10.1016/j.lfs.2019.05.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
|
14872
|
Bertuccio P, Alicandro G, Malvezzi M, Carioli G, Boffetta P, Levi F, La Vecchia C, Negri E. Cancer mortality in Europe in 2015 and an overview of trends since 1990. Ann Oncol 2019; 30:1356-1369. [DOI: 10.1093/annonc/mdz179] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
14873
|
Zheng Y, Lan T, Wei D, Zhang G, Hou G, Yuan J, Yan F, Wang F, Meng P, Yang X, Chen G, Zhu Z, Lu Z, He W, Yuan J. Coupling the near-infrared fluorescent dye IR-780 with cabazitaxel makes renal cell carcinoma chemotherapy possible. Biomed Pharmacother 2019; 116:109001. [DOI: 10.1016/j.biopha.2019.109001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 01/22/2023] Open
|
14874
|
Bhagirath D, Yang TL, Tabatabai ZL, Majid S, Dahiya R, Tanaka Y, Saini S. BRN4 Is a Novel Driver of Neuroendocrine Differentiation in Castration-Resistant Prostate Cancer and Is Selectively Released in Extracellular Vesicles with BRN2. Clin Cancer Res 2019; 25:6532-6545. [PMID: 31371344 DOI: 10.1158/1078-0432.ccr-19-0498] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/21/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Neuroendocrine prostate cancer (NEPC), an aggressive variant of castration-resistant prostate cancer (CRPC), often emerges after androgen receptor-targeted therapies such as enzalutamide or de novo, via trans-differentiation process of neuroendocrine differentiation. The mechanistic basis of neuroendocrine differentiation is poorly understood, contributing to lack of effective predictive biomarkers and late disease recognition. The purpose of this study was to examine the role of novel proneural Pit-Oct-Unc-domain transcription factors (TF) in NEPC and examine their potential as noninvasive predictive biomarkers.Experimental Design: Prostate cancer patient-derived xenograft models, clinical samples, and cellular neuroendocrine differentiation models were employed to determine the expression of TFs BRN1 and BRN4. BRN4 levels were modulated in prostate cancer cell lines followed by functional assays. Furthermore, extracellular vesicles (EV) were isolated from patient samples and cell culture models, characterized by nanoparticle tracking analyses, Western blotting, and real-time PCR. RESULTS We identify for the first time that: (i) BRN4 is amplified and overexpressed in NEPC clinical samples and that BRN4 overexpression drives neuroendocrine differentiation via its interplay with BRN2, a TF that was previously implicated in NEPC; (ii) BRN4 and BRN2 mRNA are actively released in prostate cancer EVs upon neuroendocrine differentiation induction; and (iii) enzalutamide treatment augments release of BRN4 and BRN2 in prostate cancer EVs, promoting neuroendocrine differentiation induction. CONCLUSIONS Our study identifies a novel TF that drives NEPC and suggests that as adaptive mechanism to enzalutamide treatment, prostate cancer cells express and secrete BRN4 and BRN2 in EVs that drive oncogenic reprogramming of prostate cancer cells to NEPC. Importantly, EV-associated BRN4 and BRN2 are potential novel noninvasive biomarkers to predict neuroendocrine differentiation in CRPC.
Collapse
Affiliation(s)
- Divya Bhagirath
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Thao Ly Yang
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Z Laura Tabatabai
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Shahana Majid
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Rajvir Dahiya
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Yuichiro Tanaka
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California
| | - Sharanjot Saini
- Department of Urology, Veterans Affairs Medical Center San Francisco and University of California San Francisco, San Francisco, California.
| |
Collapse
|
14875
|
Shinchi Y, Komohara Y, Yonemitsu K, Sato K, Ohnishi K, Saito Y, Fujiwara Y, Mori T, Shiraishi K, Ikeda K, Suzuki M. Accurate expression of PD-L1/L2 in lung adenocarcinoma cells: A retrospective study by double immunohistochemistry. Cancer Sci 2019; 110:2711-2721. [PMID: 31294893 PMCID: PMC6726681 DOI: 10.1111/cas.14128] [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: 04/05/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
The percentage of programmed death ligand 1 (PD‐L1) positivity in cancer cells, named as the tumor proportion score, is considered to be a predictive biomarker for anti‐PD‐1/PD‐L1 therapy in lung cancer. PD‐L1 is expressed on not only cancer cells but also on immune cells, including macrophages. Although previous studies related to PD‐L1/2 expression in cancer tissues have been generally based on single immunohistochemistry (IHC), in the present study, we attempted to evaluate accurate PD‐L1/2 expression in cancer cells in lung adenocarcinoma cells using double IHC to also evaluate macrophages. Of the 231 patients, PD‐L1 expression was negative in 169 patients (73.2%), 1%‐49% positive in 47 patients (20.3%), and ≥50% positive in 15 patients (6.5%). Interestingly, PD‐L1 positivity was decreased when using double IHC compared with the estimation by single IHC. High PD‐L1 expression was associated with high‐grade cancer cells and in higher stage cancer. PD‐L2 was negative in 109 patients (47.2%), 1%‐49% positive in 50 patients (21.6%), and ≥50% positive in 72 patients (31.2%). The number of PD‐L2‐positive patients was increased in cases that had an epidermal growth factor receptor (EGFR) mutation and in lower stage cancer. Thirty‐five patients (15.2%) were positive for both PD‐L1 and PD‐L2, whereas 81 patients (35.1%) were negative for both PD‐L1 and PD‐L2. Log‐rank analysis showed that progression‐free survival and overall survival were significantly the longest in the PD‐L1‐negative and PD‐L2‐positive groups (P < .0001 and P = .0120). We observed lower PD‐L1 or PD‐L2 expression in lung adenocarcinoma than previously reported. Double IHC for macrophages may help clinicians to evaluate PD‐L1 or PD‐L2 expression specifically in cancer cells.
Collapse
Affiliation(s)
- Yusuke Shinchi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| | - Kimihiro Yonemitsu
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensaku Sato
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoichi Saito
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Mori
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenji Shiraishi
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koei Ikeda
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
14876
|
Liu T, Yao R, Pang Y, Sun W. Review on biofabrication and applications of heterogeneous tumor models. J Tissue Eng Regen Med 2019; 13:2101-2120. [PMID: 31359625 DOI: 10.1002/term.2949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 07/08/2019] [Accepted: 07/19/2019] [Indexed: 11/12/2022]
Abstract
Resolving the origin and development of tumor heterogeneity has proven to be a crucial challenge in cancer research. In vitro tumor models have been widely used for both scientific and clinical research. Currently, tumor models based on 2D cell culture, animal models, and 3D cell-laden constructs are widely used. Heterogeneous tumor models, which consist of more than one cell type and mimic cell-cell as well as cell-matrix interactions, are attracting increasing attention. Heterogeneous tumor models can serve as pathological models to study the microenvironment and tumor development such as tumorigenesis, invasiveness, and malignancy. They also provide disease models for drug screening and personalized therapy. In this review, the current techniques, models, and oncological applications regarding 3D heterogeneous tumor models are summarized and discussed.
Collapse
Affiliation(s)
- Tiankun Liu
- Tsinghua University, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China.,Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China.,Tsinghua University, 111 "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base, Beijing, People's Republic of China.,Key Laboratory of Advanced Forming and Manufacturing, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Rui Yao
- Tsinghua University, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China.,Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China.,Tsinghua University, 111 "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base, Beijing, People's Republic of China.,Key Laboratory of Advanced Forming and Manufacturing, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Yuan Pang
- Tsinghua University, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China.,Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China.,Tsinghua University, 111 "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base, Beijing, People's Republic of China.,Key Laboratory of Advanced Forming and Manufacturing, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Wei Sun
- Tsinghua University, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China.,Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China.,Tsinghua University, 111 "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base, Beijing, People's Republic of China.,Key Laboratory of Advanced Forming and Manufacturing, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China.,Department of Mechanical Engineering, Drexel University, Philadelphia, PA
| |
Collapse
|
14877
|
Severe Vaginal Bleeding in a Case of Renal Cell Carcinoma. Case Rep Obstet Gynecol 2019; 2019:2174051. [PMID: 31360564 PMCID: PMC6644278 DOI: 10.1155/2019/2174051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/08/2019] [Indexed: 11/17/2022] Open
Abstract
Renal Cell Carcinoma (RCC) accounts for approximately 2-3% of all adult cancers and carries the highest mortality of the genitourinary cancers. Metastatic disease is seen in approximately 16% of cases and when present represents an advanced status. Metastasis of RCC to the vagina has rarely been cited in literature and when present can mimic primary vaginal cancer in clinical presentation and symptoms. Biopsy is performed to delineate the etiology and, in the presence of clear cells and certain immunohistochemistry markers, RCC needs to be included in the differential diagnosis. Treatment protocols are limited due to the rarity of the condition, with retrospective and comparative studies alongside cervical cancer treatment protocols serving as the basis. Herein, we describe a unique case of profuse vaginal bleeding secondary to vaginal metastases of RCC and discuss the relevant aspects of diagnosis and treatment.
Collapse
|
14878
|
Khunger A, Buchwald ZS, Lowe M, Khan MK, Delman KA, Tarhini AA. Neoadjuvant therapy of locally/regionally advanced melanoma. Ther Adv Med Oncol 2019; 11:1758835919866959. [PMID: 31391869 PMCID: PMC6669845 DOI: 10.1177/1758835919866959] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/08/2019] [Indexed: 11/18/2022] Open
Abstract
Locally/regionally advanced melanoma confers a major challenge in terms of surgical and medical management. Surgical treatment carries the risks of surgical morbidities and potential complications that could be lasting. In addition, these patients continue to have a high risk of relapse and death despite the use of standard adjuvant therapy. Neoadjuvant therapy has the potential to significantly improve the clinical outcome of these patients, particularly in this era of newer and effective targeted and immunotherapeutic agents. Previous neoadjuvant studies tested chemotherapy with temozolomide where the clinical activity was limited. Biochemotherapy (BCT) was tested in two studies in the neoadjuvant setting and showed high tumor response rates; however, BCT was ultimately abandoned following its failure to demonstrate survival benefits in randomized trials of metastatic disease. Success of immunotherapy and targeted therapy in prolonging the lives of patients with metastatic melanoma generated considerable interest to investigate these novel strategies in the adjuvant and neoadjuvant settings. A number of neoadjuvant targeted and immunotherapy studies have been completed in melanoma to date and have yielded promising clinical activity. Given these encouraging results, a number of studies with other molecularly targeted and immunotherapeutic agents and their combinations are ongoing in the neoadjuvant setting; long-term outcome data are eagerly awaited. Such studies also provide access to biospecimens before and during therapy, allowing for the conduct of biomarker and mechanistic studies that may have a significant impact in guiding adjuvant therapy choices and drug development.
Collapse
Affiliation(s)
- Arjun Khunger
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, USA
| | - Zachary S. Buchwald
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Lowe
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammad K. Khan
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Keith A. Delman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Ahmad A. Tarhini
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Comprehensive Cancer Center, 1365 Clifton Rd Atlanta, GA 30322, USA
| |
Collapse
|
14879
|
Sutaria DS, Jiang J, Azevedo-Pouly AC, Wright L, Bray JA, Fredenburg K, Liu X, Lu J, Torres C, Mancinelli G, Grippo PJ, Coppola V, Schmittgen TD. Knockout of Acinar Enriched microRNAs in Mice Promote Duct Formation But Not Pancreatic Cancer. Sci Rep 2019; 9:11147. [PMID: 31367007 PMCID: PMC6668398 DOI: 10.1038/s41598-019-47566-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
The pancreatic acinar-enriched miR-216a, miR-216b and miR-217 are encoded within the miR217HG. These miRNAs have been purported to play a tumor suppressive role as their expression is reduced in both human and mouse pancreatic ductal adenocarcinoma (PDAC). To examine this possibility, we generated individual, germline knockout (KO) mice of miR-216a, miR-216b or miR-217. Unlike our previous study showing germline deletion of the miR217HG was embryonic lethal, CRISPR-Cas9 deleted portions of the 5' seed region of the miRNAs produced live births. To investigate possible phenotypes during pancreatic acinar ductal metaplasia (ADM), pancreatic acini from wild type and KO mice were plated on collagen and allowed to transdifferentiate over 4 days. Acini from each of the three miRNA KO mice produced greater numbers of ducts compared to controls. Evaluation of the gene expression during in vitro ADM demonstrated an increase in Krt19 and a reduction in acinar genes (Carboxypeptidase A1, Amylase2a) on day 4 of the transdifferentiation. Recovery was delayed for the miR-216a and miR-216b KOs following caerulein-induced acute pancreatitis. Also predominate in the caerulein treated miR-216a and miR-216b KO mice was the presence of pancreatic duct glands (PDGs). To further establish a phenotype, miRNA KO mice were crossed with EL-KRASG12D (EK) mice and followed up to 13 months of age. While all mice developed severe dysplasia and cystic papillary neoplasms, there existed no apparent phenotypic difference in the miRNA KO/EK mice compared to EK mice. Our data does not support a tumor suppressor role for miR-216a, miR-216b or miR-217 in PDAC and emphasizes the need for phenotypic evaluation of miRNAs in complex in vivo models beyond that performed using cell culture.
Collapse
Affiliation(s)
- Dhruvitkumar S Sutaria
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Jinmai Jiang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ana Clara Azevedo-Pouly
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA
| | - Lais Wright
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie A Bray
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | | | - Xiuli Liu
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Jun Lu
- Department of Pathology, Beijing Chaoyang Hospital, Capital University, Beijing, China
| | - Carolina Torres
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | | | - Paul J Grippo
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - Thomas D Schmittgen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14880
|
Deng JL, Zhang R, Zeng Y, Zhu YS, Wang G. Statins induce cell apoptosis through a modulation of AKT/FOXO1 pathway in prostate cancer cells. Cancer Manag Res 2019; 11:7231-7242. [PMID: 31839714 PMCID: PMC6681436 DOI: 10.2147/cmar.s212643] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In recent years, statins have been frequently investigated in neoplasms. However, the potential roles of statins on prostate cancer cells and the underlying mechanisms have not been fully elucidated. In current study, we explored the effect and molecular mechanism of statins on cell proliferation and apoptosis in prostate cancer cells. METHODS Prostate cancer cell were treated with gradient doses of simvastatin and fluvastatin for 24-72 h. Cell proliferation was analyzed by using MTS assay and colony formation. Cell apoptosis was measured by Hoechst staining, flow cytometry and caspase-3 activity. Western blotting was used to evaluate the proteins levels. RESULTS Both simvastatin and fluvastatin produced a dose- and time-dependent inhibition of cell viability and colony formation while a promotion of cell apoptosis as evident with increases in caspase-3 activity, cleaved-caspase-3, cleaved-caspase-8 and cleaved-PARP levels in PC3 cells. Similar statin effects were observed in DU145 prostate cancer cells. Furthermore, statins produced a time- and dose-dependent reduction of phosphorylated-AKT and phosphorylated-FOXO1 levels in PC3 cells, and pretreatment of cells with an AKT phosphorylation inhibitor, MK2206, potentiated statins' effect. CONCLUSION Statins decrease cell proliferation and induce cell apoptosis, probably mediated via a downregulation of AKT/FOXO1 phosphorylation in prostate cancer cells, which may have a potential benefit in prostate cancer prevention and therapy.
Collapse
Affiliation(s)
- Jun-Li Deng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha410078, People’s Republic of China
| | - Rui Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha410078, People’s Republic of China
| | - Ying Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha410078, People’s Republic of China
| | - Yuan-Shan Zhu
- Department of Medicine, Weill Cornell Medicine, NY, NY10065, USA
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha410078, People’s Republic of China
| |
Collapse
|
14881
|
Shen T, Chen Z, Qiao J, Sun X, Xiao Q. Neutralizing monoclonal antibody against Dickkopf2 impairs lung cancer progression via activating NK cells. Cell Death Discov 2019; 5:123. [PMID: 31372243 PMCID: PMC6668384 DOI: 10.1038/s41420-019-0204-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/03/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
Adenomatous polyposis coli (APC) and KRAS proto-oncogene (KRAS) mutations frequently co-occur in non-small cell lung cancer. Inactivating APC mutations in colorectal carcinoma has been well characterized, leading to the approaches targeting on dysregulated APC pathway. However, it remains undetermined whether such approaches are also applicable to non-small cell lung cancer patients harboring similar mutations of APC. Dickkopf-related protein 2 (DKK2) is a Wnt antagonist. Our previous study has proved that anti-DKK2 antibody 5F8 suppressed the growth of colorectal carcinoma with APC mutations, illustrating a new target agent of APC-mutated tumors. This study aimed to investigate the potential of applying anti-DKK2 antibody to non-small cell lung cancer with APC mutations. We found significant upregulation of Dkk2 expression in APC-mutated lung cancers. Administration of DKK2 antibody inhibited cancer growth via modulating tumor immune microenvironment in lung cancer mouse models. Our study provided strong evidence supporting APC mutations-directed applications of anti-DKK2 targeted therapy in a wide range of cancer types, including lung cancer.
Collapse
Affiliation(s)
- Tianli Shen
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
| | - Zhengxi Chen
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
- Department of Orthodontics, Shanghai Ninth People׳s Hospital, School of Stomatology, Shanghai key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Ju Qiao
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA USA
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Qian Xiao
- Department of Pharmacology, School of Medicine, Yale University, 10 Amistad St, New Haven, CT USA
| |
Collapse
|
14882
|
Lee TH, Sung WW, Chan L, Lee HL, Chen SL, Huang YH, Kwan AL. The Association between Mortality-to-Incidence Ratios and Health Expenditures in Brain and Nervous System Cancers. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:2739. [PMID: 31370357 PMCID: PMC6696604 DOI: 10.3390/ijerph16152739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 07/27/2019] [Indexed: 12/24/2022]
Abstract
Mortality-to-incidence ratios (MIRs) are alternative parameters used to evaluate the prognosis of a disease. In addition, MIRs are associated with the ranking of health care systems and expenditures for certain types of cancer. However, a lack of association between MIRs and pancreatic cancer has been noted. Given the poor prognosis of brain and nervous system cancers, similar to pancreatic cancer, the relation of MIRs and health care disparities is worth investigating. We used the Spearman's rank correlation coefficient (CC) to analyze the correlation between the MIRs in brain and nervous system cancers and inter-country disparities, including expenditures on health and human development index. Interestingly, the MIRs in brain and nervous system cancers are associated with the human development index score (N = 157, CC = -0.394, p < 0.001), current health expenditure (CHE) per capita (N = 157, CC = -0.438, p < 0.001), and CHE as percentage of gross domestic product (N = 157, CC = -0.245, p = 0.002). In conclusion, the MIRs in the brain and nervous system cancer are significantly associated with health expenditures and human development index. However, their role as an indicator of health disparity warrants further investigation.
Collapse
Affiliation(s)
- Tsung-Han Lee
- Division of Neurosurgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wen-Wei Sung
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Lung Chan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Hsiang-Lin Lee
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Sung-Lang Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Yu-Hui Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- Department of Physical Medicine & Rehabilitation, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Aij-Lie Kwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.
| |
Collapse
|
14883
|
Nevala-Plagemann C, Moser J, Gilcrease GW, Garrido-Laguna I. Survival of patients with metastatic HER2 positive gastro-oesophageal cancer treated with second-line chemotherapy plus trastuzumab or ramucirumab after progression on front-line chemotherapy plus trastuzumab. ESMO Open 2019; 4:e000539. [PMID: 31423338 PMCID: PMC6677976 DOI: 10.1136/esmoopen-2019-000539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 12/16/2022] Open
Abstract
Background The role of continuing anti-HER2 therapy beyond progression on front-line therapy in patients with metastatic HER2 positive gastro-oesophageal cancer (GEC) is unclear. Continued chemotherapy plus trastuzumab (CT) has never been compared with the current standard second-line treatment, chemotherapy plus ramucirumab (CR). Methods The Flatiron Health electronic health record derived database, a nationwide database comprising patient-level structured and unstructured data, curated via technology-enabled abstraction, was reviewed for patients with metastatic HER2 positive GEC who received first-line CT, followed by second-line CT or CR. Survival from second-line therapy (SST) and time to next therapy or death (TTNTD) were compared using Kaplan-Meier curves and logrank analysis. Results 133 patients with metastatic HER2 positive GEC who received first-line CT were identified. 32 received second-line CR and 101 received CT. Median SST for patients treated with CT versus CR was 10.2 months (IQR 5.1–20.8) and 6.8 months (IQR 2.4–20.2), respectively (p=0.29). Median TTNTD for second-line CT versus CR was 4.9 months (IQR 2.8–9.8) and 5.1 months (IQR 2.3–7.5), respectively (p=0.65). Patients who received second-line CT were more likely to receive a multiagent chemotherapy backbone (76% vs 3%, p≤0.001). Conclusions This analysis showed no significant difference in SST for patients treated with second-line CT versus CR. Further studies are needed to clarify the role of trastuzumab in the second line, especially in patients with confirmed retention of HER2 positivity following progression.
Collapse
Affiliation(s)
| | - Justin Moser
- Oncology, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | | |
Collapse
|
14884
|
Wortman BG, Nout RA, Bosse T, Creutzberg CL. Selecting Adjuvant Treatment for Endometrial Carcinoma Using Molecular Risk Factors. Curr Oncol Rep 2019; 21:83. [PMID: 31367798 PMCID: PMC6669195 DOI: 10.1007/s11912-019-0825-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of common molecular risk factors in endometrial cancer (EC) with the possibility to improve adjuvant treatment selection. RECENT FINDINGS Recent studies have discovered and confirmed four different molecular subclasses in EC, with each having a distinct prognosis; POLE-ultramutated, microsatellite unstable, copy-number low, and copy-number high. Subsequent studies have shown that combining both molecular with clinicopathological risk factors can potentially improve adjuvant treatment selection for women with high-intermediate risk EC. For high risk and advanced stage EC, several molecular alterations are being explored for targeted therapy. Molecular alterations are frequently found in endometrial cancer and have currently not been implemented in the treatment guidelines for EC. Assessment of molecular alterations can distinguish patients that require less or more intensified adjuvant treatment. Trials investigating targeted therapies in EC are ongoing and have shown some promising results, however, more evidence is needed and results of randomized trials have to be awaited.
Collapse
Affiliation(s)
- Bastiaan G Wortman
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Remi A Nout
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
14885
|
Jia Y, Duan Y, Liu T, Wang X, Lv W, Wang M, Wang J, Liu L. LncRNA TTN-AS1 promotes migration, invasion, and epithelial mesenchymal transition of lung adenocarcinoma via sponging miR-142-5p to regulate CDK5. Cell Death Dis 2019; 10:573. [PMID: 31363080 PMCID: PMC6667499 DOI: 10.1038/s41419-019-1811-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/21/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Emerging evidence suggests that long noncoding RNA (lncRNA) plays pivotal roles in regulating various biological process in human cancers. Titin-antisense RNA1 (TTN-AS1) has been regarded as a tumor promoting lncRNA in numerous cancers. However, the clinical significance and biological function of TTN-AS1 in lung adenocarcinoma (LUAD) remain unclear. In the present study, we revealed that the expression of TTN-AS1 was upregulated in LUAD tissues and cell lines. High TTN-AS1 expression was associated with TNM stage and lymph node metastasis of LUAD patients. In addition, high expression of TTN-AS1 was correlated with poor postoperative prognosis of LUAD patients. Knockdown of TTN-AS1 significantly inhibited the growth, proliferation, migration, and invasion ability of LUAD cells in vitro. Then, by using bioinformation analysis and luciferase reporter experiment, we identified that TTN-AS1 could function as a competing endogenous RNA (ceRNA) by sponging miR-142-5p to regulate the expression of cyclin-dependent kinase 5 (CDK5) in LUAD. Since CDK5 is a key regulator in the process of epithelial mesenchymal transition (EMT), we detected the expression of EMT-related proteins, consequently, EMT was suppressed by knockdown of TTN-AS1 while this phenomenon was rescued by miR-142-5p inhibitor. Taken above, our study revealed that TTN-AS1 played an important role in LUAD progression. TTN-AS1/miR-142-5p/CDK5 regulatory axis may serve as a novel therapeutic target in the treatment of LUAD.
Collapse
Affiliation(s)
- Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Tianxu Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Xuexiao Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Wei Lv
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Mengjie Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, 050035, Hebei, China.
| |
Collapse
|
14886
|
Xia H, Chen Y, Meng J, Liang C. Effect of polymorphism on IL1A to cancer susceptibility: Evidence based on 34,016 subjects. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3138-3152. [PMID: 31359795 DOI: 10.1080/21691401.2019.1646750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Haoran Xia
- Department of Urology, the First Affiliated Hospital of Anhui Medical University; Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Yiding Chen
- The First Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Jialin Meng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University; Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University; Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
14887
|
Wang D, Zheng X, Fu B, Nian Z, Qian Y, Sun R, Tian Z, Wei H. Hepatectomy promotes recurrence of liver cancer by enhancing IL-11-STAT3 signaling. EBioMedicine 2019; 46:119-132. [PMID: 31375423 PMCID: PMC6711863 DOI: 10.1016/j.ebiom.2019.07.058] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023] Open
Abstract
Background Patients undergoing surgical resection of hepatocellular carcinoma (HCC) are at risk of recurrence; however, the underlying mechanism remains poorly understood. Methods Through the analysis of gene expression profiles in tumour and matched normal tissues from patients with hepatocellular carcinoma (HCC), we identified differences in interleukin-11 (IL-11) expression. Further, we used genetic mouse, orthotopic tumour, chemically induced, and orthotopic allograft models to study the correlation between IL-11 and postsurgical recurrence. Additionally, we conducted a series of experiments, including histology and immunohistochemistry analysis, three-dimensional culture, immunofluorescence, western blotting, enzyme-linked immunosorbent assay (ELISA) and flow cytometry to investigate the role of IL-11-signal transducer and activator of transcription 3 (STAT3) signaling in HCC recurrence. Findings We demonstrate that IL-11 levels increase after surgery, triggering HCC outgrowth. Accordingly, pharmacological blocking of IL-11-STAT3 signaling in model systems significantly alleviates tumour cell proliferation and suppresses postsurgical recurrence of HCC tumours. Interpretation These data demonstrate that IL-11 has a central role in postsurgical HCC recurrence, and that inhibition of IL-11-STAT3 signaling is a potential therapeutic strategy to prevent recurrence. Fund Natural Science Foundation of China.
Collapse
Affiliation(s)
- Dongyao Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaohu Zheng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Binqing Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Nian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yeben Qian
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230027, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
14888
|
MiR-1261/circ-PTPRZ1/PAK1 pathway regulates glioma cell growth and invasion. Hum Cell 2019; 32:540-547. [DOI: 10.1007/s13577-019-00271-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
|
14889
|
Roberts CM, Cardenas C, Tedja R. The Role of Intra-Tumoral Heterogeneity and Its Clinical Relevance in Epithelial Ovarian Cancer Recurrence and Metastasis. Cancers (Basel) 2019; 11:E1083. [PMID: 31366178 PMCID: PMC6721439 DOI: 10.3390/cancers11081083] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/22/2019] [Accepted: 07/27/2019] [Indexed: 12/14/2022] Open
Abstract
Epithelial ovarian cancer is the deadliest gynecologic cancer, due in large part to recurrent tumors. Recurrences tend to have metastasized, mainly in the peritoneal cavity and developed resistance to the first line chemotherapy. Key to the progression and ultimate lethality of ovarian cancer is the existence of extensive intra-tumoral heterogeneity (ITH). In this review, we describe the genetic and epigenetic changes that have been reported to give rise to different cell populations in ovarian cancer. We also describe at length the contributions made to heterogeneity by both linear and parallel models of clonal evolution and the existence of cancer stem cells. We dissect the key biological signals from the tumor microenvironment, both directly from other cell types in the vicinity and soluble or circulating factors. Finally, we discuss the impact of tumor heterogeneity on the choice of therapeutic approaches in the clinic. Variability in ovarian tumors remains a major barrier to effective therapy, but by leveraging future research into tumor heterogeneity, we may be able to overcome this barrier and provide more effective, personalized therapy to patients.
Collapse
Affiliation(s)
- Cai M Roberts
- Obstetrics, Gynecology and Reproductive Sciences Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Carlos Cardenas
- Obstetrics, Gynecology and Reproductive Sciences Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Roslyn Tedja
- Obstetrics, Gynecology and Reproductive Sciences Department, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
14890
|
Rosas S, Hughes RT, Farris M, Lee H, McTyre ER, Plate JF, Shi L, Emory CL, Blackstock AW, Kerr BA, Willey JS. Cartilage oligomeric matrix protein in patients with osteoarthritis is independently associated with metastatic disease in prostate cancer. Oncotarget 2019; 10:4776-4785. [PMID: 31413818 PMCID: PMC6677668 DOI: 10.18632/oncotarget.27113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/29/2019] [Indexed: 11/25/2022] Open
Abstract
Metastatic prostate cancer has a 5-year survival rate of 30%. Identifying predictors of metastasis outcome could potentially reduce patient mortality. The objective of this study was to determine whether osteoarthritis had an impact on outcomes of prostate cancer including death, local recurrence and/or metastasis and to determine whether cartilage oligomeric matrix protein was involved. We performed a retrospective case-control study of patients with prostate cancer with and without the diagnosis of osteoarthritis and completed immunohistochemistry (IHC) analysis of prostate (n=20) and lymph node (n=7) surgical specimens. We evaluated death, local recurrence and metastatic disease by various IHC biomarkers including prostate specific membrane antigen (PSMA), cartilage oligomeric matrix protein (COMP), CD31, and Ki-67. Our model identified osteoarthritis as an independent risk factor for metastatic disease (OR 5.24, 95% CI 1.49 - 18.41). Most notably, when joint arthroplasty was included in the model, osteoarthritis was no longer an independent risk factor for this outcome (p=0.071). IHC demonstrated that those with osteoarthritis, had greater expression of COMP in the prostate samples (mean 23.9% vs 5.84%, p<0.05) but not of Ki-67, CD31, or PSMA. This study identified and quantified increased metastatic disease in patients with osteoarthritis. Also, patients with osteoarthritis expressed increased COMP levels in the prostate and most likely in distant lymphatic nodes. Moreover, our findings suggest that joint arthroplasty may affect the ability of osteoarthritis to promote metastasis, which could impact treatment protocols and survival outcomes of the most common cause of cancer-related death (metastasis) in the United States.
Collapse
Affiliation(s)
- Samuel Rosas
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ryan T. Hughes
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Michael Farris
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hwajin Lee
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Emory R. McTyre
- Radiation Oncology, Greenville Health System Cancer Institute, Greenville, SC, USA
| | - Johannes F. Plate
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cynthia L. Emory
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - A. William Blackstock
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Bethany A. Kerr
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jeffrey S. Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
14891
|
Judge SJ, Lata‐Arias K, Yanagisawa M, Darrow MA, Monjazeb AM, Kirane AR, Bold RJ, Canter RJ, Canter DJ. Morbidity, mortality and temporal trends in the surgical management of retroperitoneal sarcoma: An ACS‐NSQIP follow up analysis. J Surg Oncol 2019; 120:753-760. [DOI: 10.1002/jso.25649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/18/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Sean J. Judge
- Department of SurgeryUniversity of California Davis Medical CenterSacramento California
| | | | - Mio Yanagisawa
- Department of SurgeryUniversity of California Davis Medical CenterSacramento California
| | - Morgan A. Darrow
- Department of PathologyUniversity of California Davis Medical CenterSacramento California
| | - Arta M. Monjazeb
- Department of Radiation OncologyUniversity of California Davis Medical CenterSacramento California
| | - Amanda R. Kirane
- Department of SurgeryUniversity of California Davis Medical CenterSacramento California
| | - Richard J. Bold
- Department of SurgeryUniversity of California Davis Medical CenterSacramento California
| | - Robert J. Canter
- Department of SurgeryUniversity of California Davis Medical CenterSacramento California
| | | |
Collapse
|
14892
|
Sun W, Yu W, Shen L, Huang T. MLKL is a potential prognostic marker in gastric cancer. Oncol Lett 2019; 18:3830-3836. [PMID: 31516595 DOI: 10.3892/ol.2019.10687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
The mixed lineage kinase domain-like protein (MLKL), which is a major mediator of the necroptosis pathway, is involved in a certain cancers. The present study aimed to explore the expression patterns and exact role of MLKL in gastric cancer (GC) tumorigenesis and progression. In Cancer Cell Line Encyclopedia analysis, the MLKL mRNA expression levels in GC cell lines were not higher compared with that in other cancer cell lines. The results of the present study demonstrated that MLKL expression was decreased in gastric cancer tissues compared with that in normal tissues. In the Kaplan-Meier Plotter database survival analyses, decreased MLKL expression was associated with poor overall survival and first progression in patients with gastric cancer. In Oncomine gene co-expression analysis, MLKL expression was significantly associated with fatty acid 2-hydroxylase (FA2H) expression, which also exhibited similar effects on the prognosis of patients with GC in the survival analysis. This result suggested that FA2H may be a downstream molecule of MLKL. The results of the present study indicated that MLKL may be a novel prognostic biomarker for patients with GC.
Collapse
Affiliation(s)
- Wei Sun
- Department of General Surgery, Xiangcheng People's Hospital, Suzhou, Jiangsu 215131, P.R. China
| | - Wenyan Yu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Lili Shen
- Department of Oncology, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Tieao Huang
- Department of General Surgery, Xiangcheng People's Hospital, Suzhou, Jiangsu 215131, P.R. China
| |
Collapse
|
14893
|
Patel HD, Singla N, Ghandour RA, Freifeld Y, Cheaib JG, Woldu SL, Pierorazio PM, Bagrodia A. Site of extranodal metastasis impacts survival in patients with testicular germ cell tumors. Cancer 2019; 125:3947-3952. [PMID: 31355922 DOI: 10.1002/cncr.32427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/28/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Using a large, nationally representative, population-based cancer registry, this study systematically evaluated the impact of the location and burden of extranodal testicular germ cell tumor (TGCT) metastases on survival. METHODS Men with stage III TGCTs captured by the Surveillance, Epidemiology, and End Results registry from 2010 to 2015 with distant extranodal metastases were identified. Clinicopathologic information was collected, and patients were subdivided according to the specific organ site or sites of metastatic involvement (lung, liver, bone, and/or brain). Kaplan-Meier analysis and multivariable Cox regression were used to evaluate cancer-specific survival (CSS), and model performance was assessed with Harrell's C statistic. RESULTS Nine hundred sixty-nine patients with stage III TGCTs were included with predominantly nonseminomatous histology (84%). Most patients (91%) had pulmonary metastases, whereas 20%, 10%, and 10% had liver, bone, and brain metastases, respectively. Over a median follow-up of 21 months, 19% of these men died of TGCTs. When they were grouped by the primary site of metastasis, patients with more than 1 extrapulmonary metastasis exhibited the worst CSS (hazard ratio [HR] vs isolated pulmonary involvement, 4.27; 95% confidence interval [CI], 2.60-7.00; P < .01). Among patients with isolated extrapulmonary involvement, those with brain metastases had the poorest survival (HR, 3.24; 95% CI, 1.98-5.28; P < .01), and they were followed by patients with liver (HR, 2.29; 95% CI, 1.56-3.35; P < .01) and bone metastases (HR, 1.97; 95% CI, 1.11-3.50; P = .02). Harrell's C statistic (multivariable) was 0.71. CONCLUSIONS The site of metastatic involvement affects survival outcomes for patients with TGCTs, and this may reflect both the aggressive biology and the challenging treatment of these tumors. Further incorporation of organotropism into current prognostic models for metastatic TGCTs warrants attention.
Collapse
Affiliation(s)
- Hiten D Patel
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nirmish Singla
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Rashed A Ghandour
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Yuval Freifeld
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Joseph G Cheaib
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Solomon L Woldu
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Phillip M Pierorazio
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
14894
|
Bercow AS, Eisenhauer EL. Screening and surgical prophylaxis for hereditary cancer syndromes with high risk of endometrial and ovarian cancer. J Surg Oncol 2019; 120:864-872. [PMID: 31355450 DOI: 10.1002/jso.25645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/10/2019] [Indexed: 01/23/2023]
Abstract
In the era of advanced cancer genomics, our recognition of hereditary cancer mutations continues to increase. Two of these conditions, which carry an increased risk of female cancers including endometrial, ovarian, breast, are hereditary breast and ovarian cancer syndrome and Lynch syndrome. Risk-reducing surgery, such as mastectomy, salpingo-oophorectomy, and hysterectomy may decrease cancer risk for mutation carriers. Background, indications, techniques, and consequences of these surgical procedures are reviewed.
Collapse
Affiliation(s)
- Alexandra S Bercow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eric L Eisenhauer
- Division of Gynecologic Oncology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14895
|
Soluble SLAMF7 promotes the growth of myeloma cells via homophilic interaction with surface SLAMF7. Leukemia 2019; 34:180-195. [PMID: 31358854 DOI: 10.1038/s41375-019-0525-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/24/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022]
Abstract
SLAMF7 is expressed mainly on multiple myeloma (MM) cells and considered an ideal target for immunotherapeutic approaches. Indeed, elotuzumab, an anti-SLAMF7 antibody, is used for the treatment of MM in combination with immunomodulatory drugs. SLAMF7 is cleaved via unknown mechanisms and detected as a soluble form (sSLAMF7) exclusively in the serum of MM patients; however, little is known about the role of sSLAMF7 in MM biology. In this study, we found that sSLAMF7 enhanced the growth of MM cells via homophilic interaction with surface SLAMF7 and subsequent activation of the SHP-2 and ERK signaling pathways. Elotuzumab suppressed sSLAMF7-induced MM cell growth both in vitro and in vivo. Promoter analyses identified IKZF1 (Ikaros) as a pivotal transcriptional activator of the SLAMF7 gene. Pharmacological targeting of Ikaros by lenalidomide and its analog pomalidomide downregulated SLAMF7 expression and ameliorated the response of MM cells to sSLAMF7. Elotuzumab blocked the growth-promoting function of sSLAMF7 when combined with lenalidomide in a murine xenograft model. Neutralization of sSLAMF7 is a novel antimyeloma mechanism of elotuzumab, which is enhanced by immunomodulatory drugs via downregulation of surface SLAMF7 expression on MM cells. These findings may provide important information for the optimal use of elotuzumab in MM treatment.
Collapse
|
14896
|
Amorim BJ, Hong TS, Blaszkowsky LS, Ferrone CR, Berger DL, Bordeianou LG, Ricciardi R, Clark JW, Ryan DP, Wo JY, Qadan M, Vangel M, Umutlu L, Groshar D, Cañamaques LG, Gervais DA, Mahmood U, Rosen BR, Catalano OA. Clinical impact of PET/MR in treated colorectal cancer patients. Eur J Nucl Med Mol Imaging 2019; 46:2260-2269. [PMID: 31359108 DOI: 10.1007/s00259-019-04449-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE The primary aim of the present study was to evaluate if PET/MR induced management changes versus standard of care imaging (SCI) in treated colorectal cancer patients. The secondary aim was to assess the staging performance of PET/MR and of SCI versus the final oncologic stage. METHODS Treated CRC patients who underwent PET/MR with 18F-FDG and SCI between January 2016 and October 2018 were enrolled in this retrospective study. Their medical records were evaluated to ascertain if PET/MR had impacted on their clinical management versus SCI. The final oncologic stage, as reported in the electronic medical record, was considered the true stage of disease. RESULTS A total of 39 patients who underwent 42 PET/MR studies were included, mean age 56.7 years (range 39-75 years), 26 males, and 13 females. PET/MR changed clinical management 15/42 times (35.7%, standard error ± 7.4%); these 15 changes in management were due to upstaging in 9/42 (21.5%) and downstaging in 6/42 (14.2%). The differences in management prompted by SCI versus PET/MR were statistically significant, and PET/MR outperformed SCI (P value < 0.001; odds ratio = 2.8). In relation to the secondary outcome, PET/MR outperformed the SCI in accuracy of oncologic staging (P value = 0.016; odds ratio = 4.6). CONCLUSIONS PET/MR is a promising imaging tool in the evaluation of treated CRC and might change the management in these patients. However, multicenter prospective studies with larger patient samples are required in order to confirm these preliminary results.
Collapse
Affiliation(s)
- Barbara J Amorim
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA.,Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Nuclear Medicine, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lawrence S Blaszkowsky
- Department of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David L Berger
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liliana G Bordeianou
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rocco Ricciardi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey W Clark
- Department of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David P Ryan
- Department of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark Vangel
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA.,Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - David Groshar
- Department of Nuclear Medicine, Assuta Medical Centers, Tel Aviv, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Debra A Gervais
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA
| | - Umar Mahmood
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA.,Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce R Rosen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Onofrio A Catalano
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, White 270, Boston, MA, 02114, USA. .,Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Radiology, University of Naples "Parthenope", Naples, Italy.
| |
Collapse
|
14897
|
Xu L, Cheng Z, Cui C, Wu X, Yu H, Guo J, Kong Y. Frequent genetic aberrations in the cell cycle related genes in mucosal melanoma indicate the potential for targeted therapy. J Transl Med 2019; 17:245. [PMID: 31358010 PMCID: PMC6664769 DOI: 10.1186/s12967-019-1987-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022] Open
Abstract
Background Melanoma is one of the most aggressive cancers with extremely poor prognosis, and the median survival time for stage IV patients is approximately 6 to 8 months. Unlike cutaneous melanoma, mucosal melanoma is a rare melanoma subtype among Caucasian patients but its incidence remains as high as 22.6% among Chinese patients. Screening specific genetic variations is the guideline to select targeted drugs for the treatment of advanced melanoma, whereas the genetic variation spectrum and potential therapeutic targets for mucosal melanoma are largely unclear. It is urgent to identify promising genetic variants for mucosal melanoma so as to develop effective targeted therapies for this disease. Methods Tumor samples from 213 Chinese mucosal melanoma patients were involved in this study. P16INK4a/Cyclin D1/CDK4 copy number was examined using the QuantiGene Plex DNA assay and the correlation between abnormal copy number and clinicopathological parameters was analyzed. Patient-derived xenograft models (PDX) were performed to detect the effects of CDK4/6 inhibitors on the proliferation of mucosal melanoma cells with altered copy number of CDK4 pathway (CDK4, Cyclin D1 and P16INK4a). The molecular mechanisms of CDK4/6 inhibitors on the proliferation of mucosal melanoma were analyzed by RNAseq. Results Among the 213 samples, the amplification rate of CDK4 and CCND1 was 47.0% and 27.7%, respectively, and the deletion rate of P16INK4a was 57.7%. Patients with more than one genetic abnormality were up to 81.7%. CDK4 pathway gene copy number variation was not associated with the prognosis of patients with mucosal melanoma (P > 0.05). Drug sensitivity tests showed that AT7519, a broad-spectrum CDK inhibitor, and PD0332991, a specific CDK4/6 inhibitor, exhibited higher inhibitory effect on CDK4 signaling pathway abnormal mucosal melanoma cells-derived PDX tumors growth than CDK4 signaling pathway normal ones. RNA-seq analysis showed that CDK4 inhibitors may affect tumor proliferation through multiple signaling pathways. Conclusions Abnormal copy number of cell cycle related genes is frequently found in mucosal melanoma. CDK4/6 inhibitors significantly suppress the PDX tumor growth with abnormal CDK4 pathway. CDK4 signaling variations predict the effectiveness of CDK4 inhibitors in mucosal melanoma. Electronic supplementary material The online version of this article (10.1186/s12967-019-1987-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Longwen Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China
| | - Zhiyuan Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China
| | - Huan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China.
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
14898
|
Meng J, Liu Y, Guan S, Fan S, Zhou J, Zhang M, Liang C. The establishment of immune infiltration based novel recurrence predicting nomogram in prostate cancer. Cancer Med 2019; 8:5202-5213. [PMID: 31355524 PMCID: PMC6718526 DOI: 10.1002/cam4.2433] [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: 04/18/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 12/29/2022] Open
Abstract
Prostate cancer (PCa), a severe health burden for males, accounts for the second frequent cancer and fifth tumor specific death cancer around the world. Several studies on tumor-infiltrating immune cells (TIICs) have shown inconsistent and controversial results to PCa. We downloaded a gene expression matrix and clinical information from TCGA, and CIBERSORT was used to identify the proportion of TIICs. Wilcoxon's Sign Rank Test evaluated different gene expression levels in PCa and normal tissues. Kaplan-Meier curves were used to evaluate the associations of TIICs and recurrence-free survival (RFS). Finally, based on the preset P-value of .05, the distribution of TIICs in 73 PCa tissues and 11 normal tissues was illustrated. Activated CD4+ T cells and M0 macrophages account for a high proportion in PCa tissues, while neutrophils and monocytes were found at a high density in normal tissues. Further results showed that the density of plasma cells, Treg cells and resting mast cells were associated with advanced PCa. Additionally, M2 macrophages affected the RFS of PCa patients, and AR was also involved. In the current study, we first evaluated the immune infiltration among PCa and revealed that M2 macrophages could predict the prognosis of PCa patients. Meanwhile, based on the LASSO regression analysis, we established a novel nomogram to assess the recurrence risk of PCa based on immune cell proportions and clinical features.
Collapse
Affiliation(s)
- Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Shiyang Guan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute of Urology, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
14899
|
Cleversey C, Robinson M, Willerth SM. 3D Printing Breast Tissue Models: A Review of Past Work and Directions for Future Work. MICROMACHINES 2019; 10:E501. [PMID: 31357657 PMCID: PMC6723606 DOI: 10.3390/mi10080501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer often results in the removal of the breast, creating a need for replacement tissue. Tissue engineering offers the promise of generating such replacements by combining cells with biomaterial scaffolds and serves as an attractive potential alternative to current surgical repair methods. Such engineered tissues can also serve as important tools for drug screening and provide in vitro models for analysis. 3D bioprinting serves as an exciting technology with significant implications and applications in the field of tissue engineering. Here we review the work that has been undertaken in hopes of generating the recognized in-demand replacement breast tissue using different types of bioprinting. We then offer suggestions for future work needed to advance this field for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Chantell Cleversey
- Doctor of Medicine (MD), Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Meghan Robinson
- Department of Urological Sciences, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Mechanical Engineering and Division of Medical Science, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Stephanie M Willerth
- Department of Urological Sciences, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
- Department of Mechanical Engineering and Division of Medical Science, University of Victoria, Victoria, BC V8W 2Y2, Canada.
| |
Collapse
|
14900
|
Sun D, Lu J, Zhang L, Chen Z. Aptamer-based electrochemical cytosensors for tumor cell detection in cancer diagnosis: A review. Anal Chim Acta 2019; 1082:1-17. [PMID: 31472698 DOI: 10.1016/j.aca.2019.07.054] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells, a type of viable cancer cell circulating from primary or metastatic tumors in the blood stream, can lead to the parallel development of primary tumors and metastatic lesions. Highly selective and sensitive detection of tumor cells has become a hot research topic and can provide a basis for early diagnosis of cancers and anticancer drug evaluation to develop the best treatment plan. Aptamers are single-stranded oligonucleotides that can bind to target tumor cells in unique three-dimensional structures with high specificity and affinity. Aptamer-based methods or signal amplification methods using aptamers show great potential in improving the selectivity and sensitivity of electrochemical (EC) cytosensors for tumor cell detection. This review covers the remarkable developments in aptamer-based EC cytosensors for the identification of cell type, cell counting and detection of crucial proteins on the cell surface. Various EC techniques have been developed for cancer cell detection, including common voltammetry or impedance, electrochemiluminescence and photoelectrochemistry in a direct approach (aptamer-target cell), sandwich approach (capture probe-target cell-signaling probe) or other approach. The current challenges and promising opportunities in the establishment of EC aptamer cytosensors for tumor cell detection are also discussed.
Collapse
Affiliation(s)
- Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Jing Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zuanguang Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|