14751
|
|
14752
|
Petrillo A, Pappalardo A, Pompella L, Tirino G, Calabrese F, Laterza MM, Caterino M, Ventriglia A, Orditura M, Conzo G, Molino C, Ciardiello F, Biglietto M, De Vita F. Nab-paclitaxel plus gemcitabine as first line therapy in metastatic pancreatic cancer patients relapsed after gemcitabine adjuvant treatment. Med Oncol 2019; 36:83. [PMID: 31444639 DOI: 10.1007/s12032-019-1306-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/15/2019] [Indexed: 12/27/2022]
Abstract
Nab-paclitaxel plus gemcitabine (Nab-Gem) represents one of the standard regimen for first-line treatment of metastatic pancreatic adenocarcinoma (mPDAC). However, few data are available in mPDAC relapsed after gemcitabine as adjuvant treatment. Our study aims to evaluate the efficacy and feasibility of Nab-Gem as first-line treatment for mPDAC patients previously treated with adjuvant treatment. We retrospectively analyzed the safety and efficacy data of 36 patients, who received first-line Nab-Gem after gemcitabine as adjuvant treatment. All patients received gemcitabine after radical surgery. Median disease-free survival was 12 months (95% CI 9.7-14.3); at relapse, all patients received Nab-Gem. We observed an objective response rate and disease control rate of 11.1% and 63.9%, respectively. With a median follow-up of 47 months, median progression-free survival was 5 months (95% CI 1.0-9.0), whereas median overall survival (OS) was 13 months (95% CI 5.5-20.5). Median OS was higher in patients with a relapse ≥ 7 months after the end of adjuvant treatment than in patients relapsed < 7 months (14 vs. 8 months, respectively, p: 0.52). Our results show that first-line Nab-Gem is feasible and effective in patients previously treated with gemcitabine as adjuvant treatment.
Collapse
Affiliation(s)
- Angelica Petrillo
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy.
| | - Annalisa Pappalardo
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Luca Pompella
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Giuseppe Tirino
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | | | - Maria Maddalena Laterza
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Marianna Caterino
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Anna Ventriglia
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Giovanni Conzo
- Department of Cardiothoracic Sciences, University of Study of Campania "L. Vanvitelli", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Carlo Molino
- Department of Oncological Surgery, A.O.R.N. "A. Cardarelli", Naples, Italy
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy
| | - Maria Biglietto
- Division of Medical Oncology, AORN, "A Cardarelli", Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Study of Campania "L. Vanvitelli", via Pansini n.5, 80131, Naples, Italy.
| |
Collapse
|
14753
|
Wang Z, Zhang M, Shan R, Wang YJ, Chen J, Huang J, Sun LQ, Zhou WB. MTMR3 is upregulated in patients with breast cancer and regulates proliferation, cell cycle progression and autophagy in breast cancer cells. Oncol Rep 2019; 42:1915-1923. [PMID: 31485632 PMCID: PMC6775797 DOI: 10.3892/or.2019.7292] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022] Open
Abstract
As a member of the myotubularin family, myotubularin related protein 3 (MTMR3) has been demonstrated to participate in tumor development, including oral and colon cancer. However, little is known about its functional roles in breast cancer. In the present study, the expression of MTMR3 in breast cancer was evaluated by immunohistochemical staining of tumor tissues from 172 patients. Online data was then used for survival analysis from the PROGgeneV2 database. In vitro, MTMR3 expression was silenced in MDA-MB-231 cells via lentiviral shRNA transduction. MTT, colony formation and flow cytometry assays were performed in the control and MTMR3-silenced cells to evaluate the cell growth, proliferation and cell cycle phase distribution, respectively. Western blotting was used to evaluate the protein expression levels of autophagy-related markers. The results demonstrated that the expression of MTMR3 in breast cancer tissues was significantly increased compared with adjacent normal tissues. MTMR3 was highly expressed in triple-negative breast cancer and was associated with disease recurrence. MTMR3 knockdown in MDA-MB-231 cells inhibited cell proliferation and induced cell cycle arrest and autophagy. The present results indicated that MTMR3 may have an important role in promoting the progression of breast cancer, and its inhibition may serve as a promising therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Min Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Rong Shan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu-Jie Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Juan Huang
- Hunan Province Clinic Meditech Research Center for Breast Cancer, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei-Bing Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
14754
|
Ferriss JS, Williams-Brown MY. Immunotherapy: Checkpoint Inhibitors in Lynch-Associated Gynecologic Cancers. Curr Treat Options Oncol 2019; 20:75. [PMID: 31444655 DOI: 10.1007/s11864-019-0676-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Research into novel therapies for gynecologic cancers is underfunded, and as a result, we are still playing catchup with other solid tumors in the realm of immune checkpoint inhibition. This is despite the fact that two of the most common gynecologic cancers in the USA have strong biologic rationales for response to these agents. Work is now underway to demonstrate safe and effective therapies for our patients. As we better understand the immune system, and more specifically the tumor microenvironment, we will be able to achieve complete responses. The immune system can learn, adapt, and provide ongoing surveillance; if only we could mimic its abilities.
Collapse
Affiliation(s)
- J Stuart Ferriss
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Phipps 281, Baltimore, MD, 21287, USA.
| | - M Yvette Williams-Brown
- Departments of Women's Health and Oncology, The University of Texas at Austin Dell Medical School, 1301 W 38th St, Suite 705, Austin, TX, 78705, USA
| |
Collapse
|
14755
|
Treatment of locally advanced cervical cancer in a patient with a bicornuate uterus with MRI-guided intracavitary/interstitial brachytherapy. J Contemp Brachytherapy 2019; 11:285-291. [PMID: 31435436 PMCID: PMC6701377 DOI: 10.5114/jcb.2019.85738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/14/2019] [Indexed: 12/01/2022] Open
Abstract
Purpose Definitive treatment for locally advanced cervical cancer (LACC) includes external beam radiation therapy (EBRT) with concurrent cisplatin and brachytherapy. Image-guided intracavitary brachytherapy utilizes CT and/or MRI for target and organ at risk delineation and has been shown to improve local control rates and decrease toxicity. Hybrid intracavitary/interstitial applicators can be used to improve tumor coverage in certain cases. We describe the use of combined intracavitary/interstitial brachytherapy to treat a cervical cancer patient with a bicornuate uterus. Case presentation A 30-year-old female with bicornuate uterus and FIGO IB2 adenocarcinoma of the cervix was treated with 45 Gy of EBRT to the pelvis with concurrent weekly cisplatin. An examination after EBRT demonstrated a persistent bulky tumor covering most of the cervix. Brachytherapy was delivered via a combined intracavitary/interstitial approach, using a tandem and ring applicator and an interstitial needle. The placement of the tandem was alternated between uterine cornua with each fraction, and the interstitial needle was placed on the side opposite the tandem. CT and MRI were obtained with each fraction. The patient completed the treatment without complications. Three-month restaging PET-CT showed significant interval improvement in the cervix, with a complete clinical response on physical examination. The patient is without evidence of disease 18 months after the treatment. Conclusions We found that the use of the hybrid applicator allowed for adequate target coverage in a patient with unusual anatomy. The patient tolerated treatment well and demonstrated favorable response on follow-up exam and imaging. The long-term curative result needs to be further evaluated.
Collapse
|
14756
|
Santoni M, Cimadamore A, Massari F, Piva F, Aurilio G, Martignetti A, Scarpelli M, Di Nunno V, Gatto L, Battelli N, Cheng L, Lopez-Beltran A, Montironi R. Key Role of Obesity in Genitourinary Tumors with Emphasis on Urothelial and Prostate Cancers. Cancers (Basel) 2019; 11:E1225. [PMID: 31443386 PMCID: PMC6770077 DOI: 10.3390/cancers11091225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Background: In human populations, a certain amount of data correlate obesity/body mass index (BMI) with urothelial cancer (UC) and prostate cancer (PCa) occurrence, however this is not fully elucidated at all stages of disease. In an attempt to shed light on uncertain areas in such field, in the present review we illustrate the main molecular mechanisms linking obesity and cancer, focusing on the correlation between obesity and tumor risk, disease progression and response to chemo- and immunotherapy in patients with UC and the predictive/prognostic role of obesity in PCa patients treated with the currently available therapeutic approaches. Methods: We did a large-scale literature search on existing scientific websites focusing on keywords "obesity", "body mass index (BMI)", "urothelial cancer", "prostate cancer", "docetaxel", "cabazitaxel", "abiraterone acetate", "enzalutamide", and "radium223". Results: Many adipocytes-induced molecules support tumor proliferation through activation of various cellular pathways. The available evidence in the postoperative setting do the role of BMI in oncological outcomes prediction still not completely clear. Likewise, in metastatic UC patients controversial results link the role of obesity/BMI with clinical outcomes of tumor response to chemotherapy. Adipose stromal cells recruitment, induced by PCa cells, from white adipose tissue to the tumor sites inducing cell invasiveness was associated with poor survival. Conflicting data, although more oriented towards a better survival outcome, resulted in obese patients treated with docetaxel. In PCa cell-lines a certain cabazitaxel chemo resistance adipose stromal cells (ASC)-mediated was demonstrated. In metastatic castration-resistant PCa patients with high BMI (>25 kg/m2) receiving abiraterone acetate there were significant worse survival outcomes, while in enzalutamide patients BMI did not affect survival outcome. In radium 223 patients higher BMI significantly correlated with favorable overall survival. Conclusions: The main focus of this review was to understand the interplay between obesity/BMI and UC/PCa. Several pathogenic cellular pathways exploring the issue are discussed, opening the way to challenging tailored treatments on the basis of BMI. Improving the knowledge of molecular connections between obesity and UC and PCa could favor the development of new therapies likely reducing chemo- and immunotherapy drug resistance.
Collapse
Affiliation(s)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy
| | - Francesco Massari
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Gaetano Aurilio
- Medical Division of Urogenital and Head & Neck Cancer, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Angelo Martignetti
- Dipartimento Oncologico USL Sud-Est Toscana-Area Senese, 53036 Poggibonsi, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy
| | - Vincenzo Di Nunno
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Lidia Gatto
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, Cordoba University Medical School, 14004 Cordoba, Spain
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy.
| |
Collapse
|
14757
|
Yang J, Zheng W, Xu Z, Chen J. MAP3K1 rs889312 genotypes influence survival outcomes of Chinese gastric cancer patients who received adjuvant chemotherapy based on platinum and fluorouracil regimes. Onco Targets Ther 2019; 12:6843-6855. [PMID: 31686841 PMCID: PMC6709816 DOI: 10.2147/ott.s205438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/30/2019] [Indexed: 12/23/2022] Open
Abstract
Background For patients with gastric cancer (GC), adjuvant chemotherapy is a standard therapy. However, the responses to the treatment are quite different. Mitogen-activated protein kinase (MAPK) pathway is a core pathway that modulates the efficacy of anticancer drugs. The purpose of our study was to investigate the clinical significance of one pivotal functional gene polymorphism in the MAPK pathway – MAP3K1 rs889312 – in patients with stage II GC to stage III GC. Methods The genotypes of MAP3K1 rs889312 were analyzed in 591 GC patients enrolled in this study who had received radical gastrectomy. Among them, 204 patients accepted adjuvant chemotherapy based on platinum and fluorouracil (PF) regimens after an operation. Cox regression analysis, log-rank test and Kaplan–Meier method were used to explore the link between MAP3K1 rs889312 variant and overall survival (OS) of GC. Results Compared with the AA genotype (mean OS of 68.12 months), MAP3K1 rs889312 AC/CC significantly reduced the mean OS of 56.83 months in patients who received adjuvant chemotherapy only. In addition, AC/CC genotype had a negative impact on OS of patients who received oxaliplatin-based therapy (HR, 8.253; 95% CI: 1.119–60.853, log-rank p=0.013). Stratification analysis showed that MAP3K1 rs889312 AC/CC significantly reduced OS of patients with tumors smaller than or equal to 5 cm in size (HR, 3.706; 95% CI: 1.329–10.335, p=0.012), poorly differentiated tumors (HR, 3.002; 95% CI: 1.076–8.377, p=0.036) and intestinal tumors (HR, 4.780; 95% CI: 1.138–20.073, p=0.033). Conclusion Our findings suggested that MAP3K1 rs889312 single-nucleotide polymorphism may be considered as a biomarker for adjuvant chemotherapy reaction and can predict prognosis of GC patients who received PF-based therapy.
Collapse
Affiliation(s)
- Jian Yang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,Department of Oncology, The Affiliated Yixing Hospital of Jiangsu University, Yixing 214200, People's Republic of China
| | - Wei Zheng
- Department of General Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing 214200, People's Republic of China
| | - Zhi Xu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,ICR Medical Affairs, ICON Plc, Shanghai 200003, People's Republic of China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,Cancer Center, TaiKang Xianlin Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210046, People's Republic of China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210006, People's Republic of China
| |
Collapse
|
14758
|
Kim SI, Kim SJ, Suh DH, Kim K, No JH, Kim YB. Pathologic discrepancies between colposcopy-directed biopsy and loop electrosurgical excision procedure of the uterine cervix in women with cytologic high-grade squamous intraepithelial lesions. J Gynecol Oncol 2019; 31:e13. [PMID: 31912671 PMCID: PMC7044015 DOI: 10.3802/jgo.2020.31.e13] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/05/2022] Open
Abstract
Objective To investigate pathologic discrepancies between colposcopy-directed biopsy (CDB) of the cervix and loop electrosurgical excision procedure (LEEP) in women with cytologic high-grade squamous intraepithelial lesions (HSILs). Methods We retrospectively identified 297 patients who underwent both CDB and LEEP for HSILs in cervical cytology between 2015 and 2018, and compared their pathologic results. Considering the LEEP to be the gold standard, we evaluated the diagnostic performance of CDB for identifying cervical intraepithelial neoplasia (CIN) grades 2 and 3, adenocarcinoma in situ, and cancer (HSIL+). We also performed age subgroup analyses. Results Among the study population, 90.9% (270/297) had pathologic HSIL+ using the LEEP. The diagnostic performance of CDB for identifying HSIL+ was as follows: sensitivity, 87.8%; specificity, 59.3%; balanced accuracy, 73.6%; positive predictive value, 95.6%; and negative predictive value, 32.7%. Thirty-three false negative cases of CDB included CIN2,3 (n=29) and cervical cancer (n=4). The pathologic HSIL+ rate in patients with HSIL− by CDB was 67.3% (33/49). CDB exhibited a significant difference in the diagnosis of HSIL+ compared to LEEP in all patients (p<0.001). In age subgroup analyses, age groups <35 years and 35–50 years showed good agreement with the entire data set (p=0.496 and p=0.406, respectively), while age group ≥50 years did not (p=0.036). Conclusion A significant pathologic discrepancy was observed between CDB and LEEP results in women with cytologic HSILs. The diagnostic inaccuracy of CDB increased in those ≥50 years of age.
Collapse
Affiliation(s)
- Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Se Jeong Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University School of Medicine, Seoul, Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea.
| | - Yong Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
14759
|
Li W, Martinez-Useros J, Garcia-Carbonero N, Fernandez-Aceñero MJ, Ortega-Medina L, Garcia-Botella S, Perez-Aguirre E, Diez-Valladares L, Garcia-Foncillas J. The Prognosis Value of PIWIL1 and PIWIL2 Expression in Pancreatic Cancer. J Clin Med 2019; 8:1275. [PMID: 31443431 PMCID: PMC6780139 DOI: 10.3390/jcm8091275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 08/12/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is a highly aggressive manifestation of cancer, and currently presents poor clinical outcome due to its late diagnosis with metastasic disease. Surgery is the only approach with a curative intend; however, the survival rates seen in this type of patient are still low. After surgery, there is a lack of predictive prognosis biomarkers to predict treatment response and survival to establish a personalized medicine. Human P-element-induced wimpy testis 1 (PIWIL1) and P-element-induced wimpy testis 2 (PIWIL2) proteins act as protectors of germline, and their aberrant expression has been described in several types of tumors. In this study, we aimed to assess an association between PIWIL1 and PIWIL2 expression and the prognosis of biliopancreatic cancer patients. For this, we analyzed protein expression in complete resected tumor samples, and found a significant association between PIWIL2 expression and both progression-free and overall survival (p = 0.036 and p = 0.012, respectively). However, PIWIL2 expression was significantly associated with progression-free survival (p = 0.029), and overall survival (p = 0.025) of such tumors originated in the pancreas, but not in the bile duct or ampulla of Vater. Further analysis revealed that PIWIL1 and PIWIL2, at both mRNA and protein expression levels, correlated positively with factors associated to the progenitor molecular subtype of pancreatic cancer. Based on these findings, PIWIL1 and PIWIL2 expression may be considered a potential prognostic biomarker for resectable pancreatic cancer and may serve to guide subsequent adjuvant treatment decisions.
Collapse
Affiliation(s)
- Weiyao Li
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Av. Reyes Católicos 2, 28040 Madrid, Spain.
| | - Nuria Garcia-Carbonero
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Maria J Fernandez-Aceñero
- Pathology Department, University Hospital Gregorio Marañon, C/del Dr. Esquerdo 46, 28007 Madrid, Spain
| | - Luis Ortega-Medina
- Pathology Department, Clinico San Carlos University Hospital, C/Profesor Martin Lagos, 28040 Madrid, Spain
| | - Sandra Garcia-Botella
- Surgery Department (Pancreatobiliary Unit), Hospital Clínico San Carlos, C/Profesor Martin Lagos, 28040 Madrid, Spain
| | - Elia Perez-Aguirre
- Surgery Department (Pancreatobiliary Unit), Hospital Clínico San Carlos, C/Profesor Martin Lagos, 28040 Madrid, Spain
| | - Luis Diez-Valladares
- Surgery Department (Pancreatobiliary Unit), Hospital Clínico San Carlos, C/Profesor Martin Lagos, 28040 Madrid, Spain
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Av. Reyes Católicos 2, 28040 Madrid, Spain.
| |
Collapse
|
14760
|
Lan CY, Chen SY, Kuo CW, Lu CC, Yen GC. Quercetin facilitates cell death and chemosensitivity through RAGE/PI3K/AKT/mTOR axis in human pancreatic cancer cells. J Food Drug Anal 2019; 27:887-896. [PMID: 31590760 PMCID: PMC9306979 DOI: 10.1016/j.jfda.2019.07.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/05/2023] Open
Abstract
The triggering of gemcitabine (GEM) drug resistance in pancreatic cancer by the receptor for advanced glycation end products (RAGE) has been demonstrated. Hence, finding a safe and effective adjuvant for preventing pancreatic cancer progression is imperative. Quercetin is a flavonoid that is abundant in apples, grapes, red raspberry, and onions and has been reported to inhibit RAGE. This research aimed to investigate the mechanisms of quercetin in regulating cell death and enhancing drug effects through RAGE reduction, especially in GEM-resistant pancreatic cancer cells. Our results showed that silencing RAGE expression by RAGE-specific siRNA transfection significantly increased cell death by apoptosis, autophagy and GEM-induced cytotoxicity by suppressing the PI3K/AKT/mTOR axis in MIA Paca-2 and MIA Paca-2 GEMR cells (GEM-resistant cells). Notably, quercetin showed a dramatic effect similar to RAGE silencing that effectively attenuated RAGE expression to facilitate cell cycle arrest, autophagy, apoptosis, and GEM chemosensitivity in MIA Paca-2 GEMR cells, suggesting that an additional reaction occurred under combined quercetin and GEM treatment. In conclusion, the results demonstrated that the molecular mechanisms of quercetin in regulating apoptosis and autophagy-related pathways and increasing GEM chemosensitivity in pancreatic cancer cells involved inhibition of RAGE expression.
Collapse
Affiliation(s)
- Chieh-Yu Lan
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Sheng-Yi Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Chia-Wen Kuo
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan
| | - Chi-Cheng Lu
- Department of Sport Performance, National Taiwan University of Sport, 16, Sec. 1, Shuang-Shih Road, Taichung 40404, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung 40227, Taiwan.
| |
Collapse
|
14761
|
Witt RG, Baldini EH, Raut CP. Screening populations at high risk for soft tissue sarcoma and surveillance following soft tissue sarcoma resection. J Surg Oncol 2019; 120:882-890. [PMID: 31432526 DOI: 10.1002/jso.25676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 08/07/2019] [Indexed: 11/12/2022]
Abstract
Soft tissue sarcomas (STS) are a rare and diverse group of tumors that affect both adult and pediatric populations. This review discusses current screening recommendations for populations at increased risk for STS, including those with genetic predispositions. We also review surveillance guidelines for those at risk for recurrence following curative-intent surgery.
Collapse
Affiliation(s)
- Russell G Witt
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth H Baldini
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14762
|
Ding H, Luo Y, Hu K, Liu P, Xiong M. Linc00467 promotes lung adenocarcinoma proliferation via sponging miR-20b-5p to activate CCND1 expression. Onco Targets Ther 2019; 12:6733-6743. [PMID: 31686834 PMCID: PMC6709798 DOI: 10.2147/ott.s207748] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/16/2019] [Indexed: 01/15/2023] Open
Abstract
Background Recently, numerous studies have demonstrated the emerging role of long non-coding RNAs (lncRNAs) in human cancers. Linc00467 is a newly defined lncRNA and was reported to promote cell survival in neuroblastoma. However, the function of linc00467 in lung cancer is still unclear. Material and methods We analyzed linc00467 expression and survival data derived from The Cancer Genome Altas lung adenocarcinoma (LUAD) dataset as well as in collected LUAD tissues. Then, we silenced linc00467 expression in two lung cancer cell lines using small interfering RNAs and explored the effect of linc00467 knockdown on cell growth in vitro and in vivo. Moreover, we revealed a novel target gene of linc00467 and elucidated the underlying competitive endogenous RNA regulatory mechanism in lung cancer cells. Results Our data suggested that linc00467 expression was elevated in LUAD tissues and correlated with overall survival of LUAD patients. Linc00467 knockdown resulted in reduced proliferation rate in lung cancer cells. Furthermore, we elucidated that linc00467 promoted CCND1 expression in lung cancer cells via functioning as a molecular sponge for miR-20b-5p. Conclusion Linc00467/miR-20b-5p/CCND1 signaling pathway may provide new insights into lung cancer treatment.
Collapse
Affiliation(s)
- Hao Ding
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yuchuan Luo
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Ke Hu
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Pei Liu
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Mengqing Xiong
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
14763
|
Shao C, Yu B, Liu Y. Androgen receptor splicing variant 7: Beyond being a constitutively active variant. Life Sci 2019; 234:116768. [PMID: 31445027 DOI: 10.1016/j.lfs.2019.116768] [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: 07/17/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 01/01/2023]
Abstract
In prostate cancer development, the androgen receptor (AR) signaling plays a crucial role during both formation of early prostate lesions and progression to the lethal, incurable castration resistant stage. Accordingly, numerous approaches have been developed to inhibit AR activity including androgen deprivation therapy, application of the AR antagonists as well as the use of taxanes. However, these treatments, although effective initially, resistance inevitably occur for most of the patients within several years and limiting the therapeutic efficacy. Of note, alterations and reactivation of the AR signaling pathway have been demonstrated as the major reasons for the observed resistance. Accumulating evidences have suggested that synthesis of AR splicing variants, in particular, the constitutively active AR-V7, is one of the most important mechanisms that contribute to the abnormal AR signaling. In addition, clinical data also highlight the potential of using AR-V7 as a predictive biomarker and a therapeutic target in metastatic castration resistant prostate cancer (mCRPC). In this review, we summarize the recent findings concerning the specific role of AR-V7 in CRPC progression, drug resistance and its potential value in clinical assessment.
Collapse
Affiliation(s)
- Chen Shao
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Bingbing Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yanan Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
14764
|
Yang DY, Frank I, Avant RA, Miller AR, Thapa P, Boorjian SA, Tollefson MK. Incidence and risk factors for peritoneal carcinomatosis following open radical cystectomy. Urol Oncol 2019; 37:886-892. [PMID: 31427158 DOI: 10.1016/j.urolonc.2019.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/30/2019] [Accepted: 06/21/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To characterize the frequency and risk factors of peritoneal carcinomatosis (PC) in patients undergoing open radical cystectomy (RC). METHODS We identified 3,285 patients with urothelial carcinoma treated with RC for curative intent between 1980 and 2016. At last follow-up, 72.1% (2,370/3,285) of patients had died, with a median follow-up of 8.6 years (Interquartile Range, (IQR) 3.7, 14.1). PC was defined as any recurrence involving the omentum, small bowel, and mesentery. Overall-specific survival (OSS) and cancer-specific survival (CSS) was evaluated using Kaplan-Meier methodology and log-rank test. Risk factors for mortality and recurrence were performed using Cox proportional hazards regression models. RESULTS One hundred and twenty nine (3.9%) patients were diagnosed with PC, while a total of 1,148 (34.9%) patients experienced recurrence at other sites. Median time to PC vs. other-site recurrence was 1.3 (IQR 1.3, 2.3) and 0.9 (IQR 0.5, 2.1) years, respectively (P= 0.04). Only increasing pathologic stage on multivariable analysis was associated with developing PC (pT1 HR 2.51, 95CI 1.14-5.55, P= 0.02; pT2 OR 2.82, 95CI 1.47-5.43, P= 0.002; pT3+ 2.40, 95CI 1.31-4.42, P= 0.005) over other recurrence patterns. Nodal status and tumor margin status were not associated. Patients with PC experienced worse OSS and CSS than other types of recurrence (P< 0.001). CONCLUSION PC was identified in almost 4% of patients undergoing open RC. PC is a rare occurrence after RC and primarily impacts patients with locally advanced disease.
Collapse
Affiliation(s)
- David Y Yang
- Mayo Clinic, Department of Urology, Rochester, MN
| | - Igor Frank
- Mayo Clinic, Department of Urology, Rochester, MN
| | - Ross A Avant
- Mayo Clinic, Department of Urology, Rochester, MN
| | | | - Prabin Thapa
- Mayo Clinic, Division of Biomedical Statistics and Informatics, Rochester, MN
| | | | | |
Collapse
|
14765
|
Pagano E, Izzo AA. Potential of phytochemicals in breast cancer prevention and therapy. Phytother Res 2019; 33:2795-2797. [DOI: 10.1002/ptr.6480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Ester Pagano
- Department of PharmacyUniversity of Naples Federico II Naples Italy
| | - Angelo A. Izzo
- Department of PharmacyUniversity of Naples Federico II Naples Italy
| |
Collapse
|
14766
|
Zhu X, Li H, Li S, Zhou M. Isolated Rare Urethral Metastasis From Primary Lung Adenocarcinoma: Case Report and Literature Review. Front Oncol 2019; 9:784. [PMID: 31482069 PMCID: PMC6710319 DOI: 10.3389/fonc.2019.00784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/02/2019] [Indexed: 01/27/2023] Open
Abstract
Background: Lung adenocarcinoma is a common respiratory malignancy, however urethral metastasis of lung adenocarcinoma has not yet been reported. This study aims to present a rare case of isolated urethral metastasis in a male patient with a history of primary lung adenocarcinoma. Case Presentation: A 69 year-old male patient was admitted complaining of dysuria and nocturia for 3 months, with a history of lung adenocarcinoma after surgery. The patient was diagnosed as benign prostatic hyperplasia (BPH) and received holmium laser enucleation of the prostate, an effective transurethral procedure to treat bladder outflow obstruction due to BPH. Four months after surgery for BPH, the patient had no improvement in symptoms and continued to complain of dysuria and perineum pain. An MRI of the pelvis indicated posterior urethral mass without any regional lymphadenopathy or other sites of lesion. Urethrocystoscopy found the mass in the membranous urethra near the verumontanum, and pathology combined with immunohistochemical staining confirmed the isolated urethral metastasis of lung adenocarcinoma. The further therapeutic regimen consisting of chemotherapy (pemetrexed combined with nedaplatin) and bevacizumab was well-tolerated, and obviously relieved the patient from dysuria and perineum pain. Conclusion: This study reported the first case of isolated rare urethral metastasis from primary lung adenocarcinoma and underlined the necessity for clinicians to remain vigilant to metastasis during follow-up of primary cancer.
Collapse
Affiliation(s)
- Xin Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Heqiu Li
- Department of Pathology, Molecular Medicine Testing Center, Chongqing Medical University, Chongqing, China
| | - Shuang Li
- Department of General Surgery, Jinshan Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mi Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14767
|
Di Nunno V, Mollica V, Schiavina R, Nobili E, Fiorentino M, Brunocilla E, Ardizzoni A, Massari F. Improving IMDC Prognostic Prediction Through Evaluation of Initial Site of Metastasis in Patients With Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2019; 18:e83-e90. [PMID: 31753738 DOI: 10.1016/j.clgc.2019.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/29/2019] [Accepted: 08/10/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Several models are adopted in clinical practice to estimate prognosis of patients with metastatic renal cell carcinoma (mRCC); however, none of these models have evaluated patients treated by immune-checkpoint inhibitors. The aim of this study was to investigate if the site of initial metastasis could be a parameter able to stratified prognosis among patients with mRCC among different risk groups defined by the International Metastatic Renal Cell Database Consortium (IMDC) model. The site of initial metastasis was defined as the primary tissue or organ in which metastasis was diagnosed in the course of the medical history of the disease. PATIENTS AND METHODS A total of 134 patients treated between January 2010 and December 2018 in our institution were retrospectively evaluated. The primary outcome was overall survival (OS) defined as the time from initiation of first-line therapy to death from any cause. Of note, 26 (19.4%) patients received immune-checkpoint inhibitors. Univariable analysis was performed through the log-rank test to estimate the effect of number of metastatic sites and site of initial metastasis on OS. Subsequently, a Cox regression proportional hazards model was employed in multivariable analysis. RESULTS Of the 12 variables analyzed, 4 were statistically associated to worse OS in univariable analysis (number of metastases and liver, bone, or central nervous system metastases). Multivariate analysis confirmed that bone (hazard ratio [HR], 1.92; 95% confidence interval [CI], 1.17-3.13), liver (HR, 2.65; 95% CI, 1.59-4.42), and central nervous system (HR, 3.3; 95% CI, 1.62-6.74) initial metastases were independent parameters related to worse OS. The presence of 1 or more of the selected sites recognized specific populations of patients associated to worse prognosis in both good (P = .003) and intermediate (P = .047) risk groups. CONCLUSION The site of initial metastasis defines specific populations of patients associated with worse prognosis in the good and intermediate IMDC groups.
Collapse
Affiliation(s)
| | - Veronica Mollica
- Division of Oncology, S-Orsola-Malpighi Hospital, Bologna, Italy
| | - Riccardo Schiavina
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, Italy
| | - Eugenio Brunocilla
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, Bologna, Italy
| | - Andrea Ardizzoni
- Division of Oncology, S-Orsola-Malpighi Hospital, Bologna, Italy
| | | |
Collapse
|
14768
|
Tarver WL, Robb BW, Haggstrom DA. Usefulness and Usability of a Personal Health Record and Survivorship Care Plan for Colorectal Cancer Survivors: Survey Study. JMIR Cancer 2019; 5:e10692. [PMID: 31432780 PMCID: PMC6719487 DOI: 10.2196/10692] [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: 06/11/2018] [Revised: 01/30/2019] [Accepted: 02/18/2019] [Indexed: 12/29/2022] Open
Abstract
Background As a result of improvements in cancer screening, treatment, and supportive care, nearly two-thirds of individuals diagnosed with colorectal cancer (CRC) live for 5 years after diagnosis. An ever-increasing population of CRC survivors creates a need for effective survivorship care to help manage and mitigate the impact of CRC and its treatment. Personal health records (PHRs) and survivorship care plans provide a means of supporting the long-term care of cancer survivors. Objective The purpose of this study is to characterize the usefulness of a CRC PHR and survivorship care plan and to describe the usability of these technologies in a population of CRC survivors. To our knowledge, this is the first study to assess a PHR and survivorship care plan specifically targeting CRC survivors. Methods Twenty-two patients with CRC were recruited from surgery clinics of an academic medical center and Veterans Affairs hospital in Indianapolis and provided access to an online Colorectal Cancer Survivor’s Personal Health Record (CRCS-PHR). Survey data were collected to characterize the usefulness of the CRCS-PHR and describe its usability in a population of CRC survivors. CRC survivors were surveyed 6 months after being provided online access. Means and proportions were used to describe the usefulness and ease of using the CRC website. Open-ended questions were qualitatively coded using the constant comparative method. Results CRC survivors perceived features related to their health care (ie, summary of cancer treatment history, follow-up care schedule, description of side effects, and list of community resources) to be more useful than communication features (ie, creating online relationships with family members or caregivers, communicating with doctor, and secure messages). CRC survivors typically described utilizing traditional channels (eg, via telephone or in person) to communicate with their health care provider. Participants had overall positive perceptions with respect to ease of use and overall satisfaction. Major challenges experienced by participants included barriers to system log-in, lack of computer literacy or experience, and difficulty entering their patient information. Conclusions For CRC, survivors may find the greater value in a PHR’s medical content than the communication functions, which they have available elsewhere. These findings regarding the usefulness and usability of a PHR for the management of CRC survivorship provide valuable insights into how best to tailor these technologies to patients’ needs. These findings can inform future design and development of PHRs for purposes of both cancer and chronic disease management.
Collapse
Affiliation(s)
- Will L Tarver
- VA Health Services Research and Development, Center for Health Information & Communication, Richard L Roudebush VA Medical Center, Indianapolis, IN, United States.,Department of Health Policy & Management, Fairbanks School of Public Health, Indiana University, Indianapolis, IN, United States
| | - Bruce W Robb
- Department of Surgery, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - David A Haggstrom
- VA Health Services Research and Development, Center for Health Information & Communication, Richard L Roudebush VA Medical Center, Indianapolis, IN, United States.,Division of General Internal Medicine and Geriatrics, School of Medicine, Indiana University, Indianapolis, IN, United States.,Center for Health Services Research, Regenstrief Institute, Indianapolis, IN, United States
| |
Collapse
|
14769
|
Abstract
PURPOSE OF REVIEW This review presents an overview of the diagnostic approach to esophageal dysphagia and summarizes recent epidemiological trends and technical advancements. RECENT FINDINGS The evaluation of dysphagia begins with a detailed history followed by endoscopy to evaluate for any structural abnormalities including malignancy. This is especially true given the emergence of eosinophilic esophagitis (EoE) as a dominant cause of esophageal dysphagia. In fact, it is now standard practice to obtain esophageal biopsies during endoscopy performed to evaluate dysphagia, since EoE can present without the characteristic mucosal features of rings, furrows, and exudate. Achalasia is also more frequently encountered since the introduction of high-resolution manometry (HRM) and the Chicago Classification into clinical practice. The Chicago Classification provides a stepwise diagnostic algorithm for evaluating HRM studies and systematically diagnosing esophageal motility disorders. Lastly, the functional lumen imaging probe (FLIP) is a novel technology that has added insight into both achalasia and EoE. Measuring esophageal distensibility with FLIP has useful prognostic implications for both diseases, and FLIP can identify motility abnormalities in achalasics not detected with HRM. A careful history is key to the efficient evaluation of dysphagia, and endoscopy is usually the first diagnostic study to obtain. For patients with prominent reflux symptoms, an empiric trial with proton pump inhibitors is reasonable then because reflux disease is such a common cause of dysphagia. Thereafter, patients should undergo HRM to evaluate for a motility disorder, and FLIP can provide complementary data to guide management.
Collapse
Affiliation(s)
- Jooho P Kim
- Feinberg School of Medicine, Department of Medicine, Northwestern University, 676 St Clair St, 14th floor, Chicago, IL, 60611-2951, USA
| | - Peter J Kahrilas
- Feinberg School of Medicine, Department of Medicine, Northwestern University, 676 St Clair St, 14th floor, Chicago, IL, 60611-2951, USA.
| |
Collapse
|
14770
|
Santoro A, Angelico G, Piermattei A, Inzani F, Valente M, Arciuolo D, Spadola S, Mulè A, Zorzato P, Fagotti A, Scambia G, Zannoni GF. Pathological Chemotherapy Response Score in Patients Affected by High Grade Serous Ovarian Carcinoma: The Prognostic Role of Omental and Ovarian Residual Disease. Front Oncol 2019; 9:778. [PMID: 31482065 PMCID: PMC6709655 DOI: 10.3389/fonc.2019.00778] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/31/2019] [Indexed: 02/05/2023] Open
Abstract
Background: The chemotherapy response score (CRS) has emerged as a simple and reproducible histopathological grading system for assessing chemotherapy response in patients affected by ovarian high-grade serous carcinoma. Objective: To evaluate the prognostic impact of histological tumor response in ovarian and omental surgical specimens from patients with advanced stage ovarian high-grade serous carcinoma. Study Design: A cohort of 161 women were identified from the database of Department of Gynecology, “Fondazione Policlinico Universitario Agostino Gemelli IRCCS” of Rome, Italy between January 2014 and December 2017 with a follow-up of 65 months. All the omentum, the ovarian tissue and peritoneal samples, defined as “other sites,” were reviewed by gynecological pathologists to assign a CRS of 1–3 to the omentum and ovarian sites and a score of 0–1 to the peritoneal tissue. The Cox proportional hazards regression and the log-rank test were used to assess the survival pattern and the prognostic value of the CRS adjusting for age and stage. The Kaplan-Meier method was applied to estimate the progression free and overall survival. Results: The evaluation of adnexal disease showed significant differences in PFS, both in univariate and in multivariate analyses. On PFS univariate analysis, ovCRS1 vs. ovCRS3: HR, 2.27; 95% CI, 1.37–3.77; p = 0.001; ovCRS2 vs. ovCRS3: HR, 1.83; 95% CI, 1.03–3.23; p = 0.04, and on PFS multivariate model ovCRS1 vs. ovCRS3; HR, 2.53; 95% CI, 1.5–4.24; p = 0.001 and ovCRS2 vs. ovCRS3; HR, 1.90; 95% CI, 1.08–3.37; p = 0.03. Regarding the omental residual disease, as expected, CRS showed a significant prognostic value for OS and PFS; in detail the median PFS of patients with CRS1, 2 and 3 was 15, 15, and 22 months, respectively, the median OS was 41 and >50 months, respectively. Moreover, the univariate analysis for OS suggested that in our cohort the “other sites” score of 0 was significantly associated with an improvement in overall survival compared to score 1. Conclusions: We demonstrated for the first time the prognostic significance of adnexal CRS confirming also the prognostic role of omental CRS.
Collapse
Affiliation(s)
- Angela Santoro
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Angelico
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessia Piermattei
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Frediano Inzani
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michele Valente
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Damiano Arciuolo
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Saveria Spadola
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonino Mulè
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Piercarlo Zorzato
- Unità di Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Fagotti
- Unità di Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Scambia
- Unità di Ginecologia Oncologica, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Franco Zannoni
- Unità di Gineco-patologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
14771
|
Low colorectal cancer survival in the Mountain West state of Nevada: A population-based analysis. PLoS One 2019; 14:e0221337. [PMID: 31425558 PMCID: PMC6699684 DOI: 10.1371/journal.pone.0221337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third greatest cancer burden in the United States. The remarkably diverse Mountain West state of Nevada has uncharacteristically high CRC mortality compared to other Western states. We aimed to study the determinants of the CRC excess burden by using data from the Nevada Central Cancer Registry from 2003-2013. Five-year cause-specific age-adjusted survival from colorectal cancer was calculated and stratified by sex, race/ethnicity and region of Nevada. Cox Proportional Hazards regression modelling was used to study the impact of demographic, social, and clinical factors on CRC survival in Nevada, assessing follow-up as accurately as possible. The extent to which differences in survival can be explained by receipt of stage-appropriate treatment was also assessed. 12,413 CRC cases from 2003-2013 in Nevada were analyzed. Five-year CRC survival was low: 56.0% (95% CI: 54.6-57.5) among males and 59.5% (95% CI: 58.0-61.1) among females; significantly lower than national 5-year survival of 65.1% and 66.5%, respectively. Low survival was driven by populous Southern Nevada; after adjustment for all covariates, Southern Nevadans were at 17% higher risk of death than their counterparts in Northwestern Nevada (HR:1.17; 95% CI:1.08-1.27). Many patients did not receive stage-appropriate treatment, although this only partly explained the poor survival, uniformly low for every race/ethnicity in Nevada. The observed disparity for this one state within a single nation merits public health attention; regardless of the state or region of residence, all Americans deserve equal opportunity for optimum health outcomes in the face of a cancer diagnosis. The current study provides baseline information critical to clinicians, public health professionals, and all relevant stakeholders as they attempt to discern why Nevada's outcomes are vastly divergent from its neighboring Western states and make plans for remediation.
Collapse
|
14772
|
Wang D, Guo D, Shi F, Zhu Y, Li A, Kong L, Teng F, Yu J. The predictive effect of the systemic immune-inflammation index for patients with small-cell lung cancer. Future Oncol 2019; 15:3367-3379. [PMID: 31424272 DOI: 10.2217/fon-2019-0288] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: The purpose of this study was to investigate the predictive power of the systemic immune inflammation index (SII) based on neutrophil (N), platelet (P) and lymphocyte (L) on the clinical outcomes of patients with SCLC. Patients & methods: Blood samples of 228 patients were obtained 1 week before treatment to measure the SII (SII = P × N/L). Overall survival (OS) and progression-free survival (PFS) were estimated using the Kaplan-Meier curves and Cox regression models. Results: Higher SII was associated with poorer OS (p < 0.001) and poorer PFS (p < 0.001). Multivariable analyses further revealed SII as an independent prognostic factor for OS (p < 0.001) and PFS (p < 0.001). Conclusion: Pretreatment SII was a valuable prognostic factor for PFS and OS in SCLC patients.
Collapse
Affiliation(s)
- Duoying Wang
- Department of Clinical Medicine, Weifang Medical University, Weifang, PR China.,Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Dong Guo
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Fang Shi
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Ying Zhu
- Department of Clinical Medicine, Weifang Medical University, Weifang, PR China
| | - Aijie Li
- Department of Clinical Medicine, Weifang Medical University, Weifang, PR China.,Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Li Kong
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Feifei Teng
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Jinming Yu
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| |
Collapse
|
14773
|
Xing D, Liu Y, Park HJ, Baek I, Tran H, Cheang G, Novo J, Dillon J, Matoso A, Farmer E, Cheng MA, Tsai YC, Lombardo K, Conner MG, Vang R, Hung CF, Wu TC, Song W. Recurrent genetic alterations and biomarker expression in primary and metastatic squamous cell carcinomas of the vulva. Hum Pathol 2019; 92:67-80. [PMID: 31437519 DOI: 10.1016/j.humpath.2019.08.003] [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: 06/18/2019] [Revised: 07/30/2019] [Accepted: 08/07/2019] [Indexed: 12/26/2022]
Abstract
Using a comprehensive next-generation sequencing pipeline (143 genes), Oncomine Comprehensive v.2, we analyzed genetic alterations on a set of vulvar squamous cell carcinomas (SCCs) with emphasis on the primary and metastatic samples from the same patient, to identify amenable therapeutic targets. Clinicopathologic features were reported and genomic DNA was extracted from 42 paraffin-embedded tumor tissues of 32 cases. PD-L1 expression was evaluated in 20 tumor tissues (10 cases with paired primary and metastatic tumors). Fifteen (88%) of 17 successfully analyzed HPV-unrelated SCCs harbored TP53 mutations. 2 different TP53 mutations had been detected in the same tumor in 4 of 15 cases. Other recurrent genetic alterations in this group of tumors included CDKN2a mutations (41%), HRAS mutations (12%), NOTCH1 mutations (12%) and BIRC3 (11q22.1-22.2) amplification (12%). Six HPV-related tumors harbored PIK3CA, BAP1, PTEN, KDR, CTNNB1, and BRCA2 mutations, of which, one case also contained TP53 mutation. Six cases showed identical mutations in paired primary site and distant metastatic location and four cases displayed different mutational profiles. PD-L1 expression was seen in 6 of 10 primary tumors and all 6 paired cases showed discordant PD-L1 expression in the primary and metastatic sites. Our results further confirmed the genetic alterations that are amenable to targeted therapy, offering the potential for individualized management strategies for the treatment of these aggressive tumors with different etiology. Discordant PD-L1 expression in the primary and metastatic vulvar SCCs highlights the importance of evaluation of PD-L1 expression in different locations to avoid false negative information provided for immunotherapy.
Collapse
Affiliation(s)
- Deyin Xing
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD.
| | - Yuehua Liu
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Hyeon Jin Park
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, NY
| | - Inji Baek
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, NY
| | - Hung Tran
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, NY
| | - Gloria Cheang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, NY
| | - Jorge Novo
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Jessica Dillon
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Andres Matoso
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Emily Farmer
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Max A Cheng
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Ya-Chea Tsai
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Kara Lombardo
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Michael G Conner
- Department of Pathology, The University of Alabama at Birmingham, Birmingham 35233, AL
| | - Russell Vang
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD; Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Tzyy-Choou Wu
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore 21231, MD; Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore 21231, MD
| | - Wei Song
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York 10065, NY.
| |
Collapse
|
14774
|
The Role of the HOXA Gene Family in Acute Myeloid Leukemia. Genes (Basel) 2019; 10:genes10080621. [PMID: 31426381 PMCID: PMC6723066 DOI: 10.3390/genes10080621] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/06/2019] [Accepted: 08/14/2019] [Indexed: 01/12/2023] Open
Abstract
The HOXA gene family is associated with various cancer types. However, the role of HOXA genes in acute myeloid leukemia (AML) have not been comprehensively studied. We compared the transcriptional expression, survival data, and network analysis of HOXA-associated signaling pathways in patients with AML using the ONCOMINE, GEPIA, LinkedOmics, cBioPortal, and Metascape databases. We observed that HOXA2-10 mRNA expression levels were significantly upregulated in AML and that high HOXA1-10 expression was associated with poor AML patient prognosis. The HOXA genes were altered in ~18% of the AML samples, either in terms of amplification, deep deletion, or elevated mRNA expression. The following pathways were modulated by HOXA gene upregulation: GO:0048706: embryonic skeletal system development; R-HSA-5617472: activation of HOX genes in anterior hindbrain development during early embryogenesis; GO:0060216: definitive hemopoiesis; hsa05202: transcriptional mis-regulation in cancer; and GO:0045638: negative regulation of myeloid cell differentiation, and they were significantly regulated due to alterations affecting the HOXA genes. This study identified HOXA3-10 genes as potential AML therapeutic targets and prognostic markers.
Collapse
|
14775
|
Zhang L, Wang S, Zhou Y, Wang C, Zhang X, Deng H. Covalent Organic Frameworks as Favorable Constructs for Photodynamic Therapy. Angew Chem Int Ed Engl 2019; 58:14213-14218. [DOI: 10.1002/anie.201909020] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Liang Zhang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Shibo Wang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Yi Zhou
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| | - Chao Wang
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| | - Xian‐Zheng Zhang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Hexiang Deng
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| |
Collapse
|
14776
|
Zhang L, Wang S, Zhou Y, Wang C, Zhang X, Deng H. Covalent Organic Frameworks as Favorable Constructs for Photodynamic Therapy. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201909020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Liang Zhang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Shibo Wang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Yi Zhou
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| | - Chao Wang
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| | - Xian‐Zheng Zhang
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
| | - Hexiang Deng
- The Institute For Advanced Studies Wuhan University Luojiashan Wuhan 430072 China
- Key Laboratory of Biomedical Polymers-Ministry of Education College of Chemistry and Molecular Sciences Wuhan University Luojiashan Wuhan 430072 China
| |
Collapse
|
14777
|
Lokman NA, Price ZK, Hawkins EK, Macpherson AM, Oehler MK, Ricciardelli C. 4-Methylumbelliferone Inhibits Cancer Stem Cell Activation and Overcomes Chemoresistance in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081187. [PMID: 31443261 PMCID: PMC6721459 DOI: 10.3390/cancers11081187] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/07/2019] [Accepted: 08/11/2019] [Indexed: 01/01/2023] Open
Abstract
We have recently shown that the extracellular matrix molecule hyaluronan (HA) plays a role in the development of ovarian cancer chemoresistance. This present study determined if HA production is increased in chemotherapy-resistant ovarian cancers and if the HA inhibitor 4-methylubelliferone (4-MU) can overcome chemoresistance to the chemotherapeutic drug carboplatin (CBP) and inhibit spheroid formation and the expression of cancer stem cell (CSC) markers. We additionally assessed whether 4-MU could inhibit in vivo invasion of chemoresistant primary ovarian cancer cells in the chicken embryo chorioallantoic membrane (CAM) assay. The expression of the HA synthases HAS2 and HAS3 was significantly increased in chemoresistant compared to chemosensitive primary ovarian cancer cells isolated from patient ascites. 4-MU significantly inhibited HA production, cell survival, and spheroid formation of chemoresistant serous ovarian cancer cells. In combination with CBP, 4-MU treatment significantly decreased ovarian cancer cell survival and increased apoptosis of chemoresistant primary cells compared to CBP alone. 4-MU significantly reduced spheroid formation, expression of CSC markers ALDH1A1 and ABCG2 in primary cell spheroid cultures, and ALDH1 immunostaining in patient-derived tissue explant assays following treatment with CBP. Furthermore, 4-MU was very effective at inhibiting in vivo invasion of chemoresistant primary cells in CAM assays. Inhibition of HA is therefore a promising new strategy to overcome chemoresistance and to improve ovarian cancer survival.
Collapse
Affiliation(s)
- Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Zoe K Price
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Emily K Hawkins
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Anne M Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5005, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
14778
|
Wang WJ, Guo CA, Li R, Xu ZP, Yu JP, Ye Y, Zhao J, Wang J, Wang WA, Zhang A, Li HT, Wang C, Liu HB. Long non-coding RNA CASC19 is associated with the progression and prognosis of advanced gastric cancer. Aging (Albany NY) 2019; 11:5829-5847. [PMID: 31422382 PMCID: PMC6710062 DOI: 10.18632/aging.102190] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/10/2019] [Indexed: 12/24/2022]
Abstract
Evidence indicates that aberrantly expressed long non-coding RNAs (lncRNAs) are involved in the development and progression of advanced gastric cancer (AGC). Using RNA sequencing data and clinical information obtained from The Cancer Gene Atlas, we combined differential lncRNA expression profiling and weighted gene co-expression network analysis to identify key lncRNAs associated with AGC progression and prognosis. Cancer susceptibility 19 (CASC19) was the top hub lncRNA among the lncRNAs included in the gene module most significantly correlated with AGC’s pathological variables. CASC19 was upregulated in AGC clinical samples and was significantly associated with higher pathologic TNM stage, pathologic T stage, lymph node metastasis, and poor overall survival. Multivariable Cox analysis confirmed that CASC19 overexpression is an independent prognostic factor for overall survival. Furthermore, quantitative real-time PCR assay confirmed that CASC19 expression in four human gastric cancer cells (AGS, BGC-823, MGC-803, and HGC-27) was significantly upregulated compared with human normal gastric mucosal epithelial cell line (GES-1). Functionally, CASC19 knockdown inhibited GC cell proliferation and migration in vitro. These findings suggest that CASC19 may be a novel prognostic biomarker and a potential therapeutic target for AGC.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China
| | - Chang-An Guo
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China.,Department of Emergency, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, P.R. China
| | - Rui Li
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, P.R. China
| | - Zi-Peng Xu
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China
| | - Jian-Ping Yu
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China
| | - Yan Ye
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China
| | - Jun Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, P.R. China
| | - Jing Wang
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China.,Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730030, Gansu, P.R. China
| | - Wen-An Wang
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China.,Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730030, Gansu, P.R. China
| | - An Zhang
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou 730050, Gansu, China.,Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730030, Gansu, P.R. China
| | - Hong-Tao Li
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China
| | - Chen Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, P.R. China
| | - Hong-Bin Liu
- Second Clinical Medical College, Lanzhou University, Lanzhou 730030, Gansu, P.R. China.,Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou 730050, Gansu, P.R. China
| |
Collapse
|
14779
|
Shao L, Wang W, Song Z, Zhang Y. The efficacy and safety of anlotinib treatment for advanced lung cancer. Onco Targets Ther 2019; 12:6549-6554. [PMID: 31616163 PMCID: PMC6699585 DOI: 10.2147/ott.s205674] [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: 02/16/2019] [Accepted: 07/21/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Anlotinib is an oral novel multi-target tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor, fibroblast growth factor receptor, platelet-derived growth factor receptor, and stem cell factor receptor (c-Kit). The aim of this study was to evaluate the efficacy and safety of anlotinib treatment in advanced lung cancer in the real world. Methods We evaluated the efficacy and toxicity of apatinib in patients with previously treated advanced lung cancer from 2018 to 2019 in Zhejiang Cancer Hospital. Survival analysis was performed by the Kaplan-Meier method. Results Fifty-eight patients were included in the present study. Thirty-one of these patients received anlotinib treatment as a third line and 27 patients received further therapy. All 58 patients had therapeutic evaluation and 46 patients acquired progression-free survival evaluation. Ten patients achieved partial response (PR), and 36 achieved stable disease (SD), representing a response rate of 17.2% and a disease control rate of 77.6%. Median progression-free survival was 3.3 months (95% CI 1.595-5.071). The toxicities associated with anlotinib were generally acceptable with a total grade 3/4 toxicity of 5.2%. The toxicities of anlotinib were generally tolerated and the common toxicities were hand-foot syndrome and hypertension. Conclusion In the third-line or more-line treatment of advanced lung cancer, anlotinib appears to have some activity when utilized as a salvage treatment. Adverse reactions are controllable.
Collapse
Affiliation(s)
- Lan Shao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Wenxian Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Yiping Zhang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
14780
|
TIMP-1-Mediated Chemoresistance via Induction of IL-6 in NSCLC. Cancers (Basel) 2019; 11:cancers11081184. [PMID: 31443242 PMCID: PMC6721590 DOI: 10.3390/cancers11081184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/05/2019] [Accepted: 08/11/2019] [Indexed: 01/28/2023] Open
Abstract
Elevated tissue inhibitor of metalloproteinase-1 (TIMP-1) is a negative prognosticator in non-small cell lung carcinoma NSCLC patients. This study sought to identify mechanisms whereby TIMP-1 impacts anticancer therapy. Using NSCLC cells and their TIMP-1 knockdown clones, we examined the chemoresistance against two chemotherapeutic agents, Gemcitabine and Cisplatin, as identified by increased apoptosis in the knockdown clones. A bead-based cytokine screening assay identified interleukin-6 (IL-6) as a key factor in chemoresistance. Exogenous human recombinant rhTIMP-1 or rhIL-6 resulted in reduced apoptosis. IL-6 expression was closely correlated with TIMP-1 kinetics and was upregulated by the addition of exogenous TIMP-1 while TIMP-1 neutralizing antibodies delayed IL-6 elevation. IL-6 production was regulated by TIMP-1, exerting its effect via activation of downstream signal transducer and activator of transcription 3 (STAT3) signaling. Both molecules and their documented transcription factors were upregulated and activated in chemoresistant NSCLC cells, confirming the roles of TIMP-1 and IL-6 in chemoresistance. To examine the role of these genes in patients, survival data from lung adenocarcinoma (LUAD) patients was curated from the cancer genome atlas (TCGA) database. Kaplan-Meier analysis found that individuals expressing low TIMP-1 and IL-6 have a higher survival rate and that the two-gene signature was more significant than the single-gene status. We define for the first time, a regulatory relationship between TIMP-1 and IL-6 in NSCLCs, suggesting that the TIMP-1/IL6 axis may be a valuable prognostic biomarker. Therapeutic interventions directed at this dual target may improve overall prognosis while negatively affecting the development of chemoresistance in NSCLC.
Collapse
|
14781
|
Harrington BS, Annunziata CM. NF-κB Signaling in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081182. [PMID: 31443240 PMCID: PMC6721592 DOI: 10.3390/cancers11081182] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
The NF-κB signaling pathway is a master and commander in ovarian cancer (OC) that promotes chemoresistance, cancer stem cell maintenance, metastasis and immune evasion. Many signaling pathways are dysregulated in OC and can activate NF-κB signaling through canonical or non-canonical pathways which have both overlapping and distinct roles in tumor progression. The activation of canonical NF-κB signaling has been well established for anti-apoptotic and immunomodulatory functions in response to the tumor microenvironment and the non-canonical pathway in cancer stem cell maintenance and tumor re-initiation. NF-κB activity in OC cells helps to create an immune-evasive environment and to attract infiltrating immune cells with tumor-promoting phenotypes, which in turn, drive constitutive NF-κB activation in OC cells to promote cell survival and metastasis. For these reasons, NF-κB is an attractive target in OC, but current strategies are limited and broad inhibition of this major signaling pathway in normal physiological and immunological functions may produce unwanted side effects. There are some promising pre-clinical outcomes from developing research to target and inhibit NF-κB only in the tumor-reinitiating cancer cell population of OC and concurrently activate canonical NF-κB signaling in immune cells to promote anti-tumor immunity.
Collapse
|
14782
|
Akil H, Rouanet J, Viallard C, Besse S, Auzeloux P, Chezal JM, Miot-Noirault E, Quintana M, Degoul F. Targeted Radionuclide Therapy Decreases Melanoma Lung Invasion by Modifying Epithelial-Mesenchymal Transition-Like Mechanisms. Transl Oncol 2019; 12:1442-1452. [PMID: 31421458 PMCID: PMC6704444 DOI: 10.1016/j.tranon.2019.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Melanin-radiolabeled molecules for targeted radionuclide therapy (TRT) provide a promising approach for the treatment of pigmented melanoma. Among these radiolabeled molecules, the iodinated melanin-specific binding molecule ([131I]ICF01012) has shown a significant antitumor effect on metastatic melanoma preclinical models. We report herein that [131I]ICF01012 decreases the epithelial-mesenshymal transition-like (EMT-like) markers in both in vivo and in vitro three-dimensional (3D) melanoma spheroid models. [131I]ICF01012 spheroids irradiation resulted in reduced clonogenic capacity of all pigmented spheroids accompanied by increased protein expression levels of phosphorylated H2A.X, p53 and its downstream target p21. In addition, [131I]ICF01012 treatment leads to a significant increase of cell pigmentation as demonstrated in SK-MEL3 human xenograft model. We also showed that [131I]ICF01012 decreases the size and the number of melanoma lung colonies in the syngeneic murine B16BL6 in vivo model assessing its potentiality to kill circulating tumor cells. Taken together, these results indicate that [131I]ICF01012 reduces metastatic capacity of melanoma cells presumably through EMT-like reduction and cell differentiation induction.
Collapse
Affiliation(s)
- Hussein Akil
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Jacques Rouanet
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France; Department of Dermatology and Oncodermatology, CHU Estaing, Clermont-Ferrand, France; Centre Jean Perrin, Clermont-Ferrand, France.
| | - Claire Viallard
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Sophie Besse
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Philippe Auzeloux
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Jean-Michel Chezal
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | | | - Mercedes Quintana
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Françoise Degoul
- UMR 1240 INSERM, University of Clermont Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
14783
|
Wang S, Shao J, Li Z, Ren Q, Yu XF, Liu S. Black Phosphorus-Based Multimodal Nanoagent: Showing Targeted Combinatory Therapeutics against Cancer Metastasis. NANO LETTERS 2019; 19:5587-5594. [PMID: 31260628 DOI: 10.1021/acs.nanolett.9b02127] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In breast cancer chemophotothermal therapy, it is a great challenge for the development of multifunctional nanoagents for precision targeting and the effective treatment of tumors, especially for metastasis. Herein, we successfully design and synthesize a multifunctional black phosphorus (BP)-based nanoagent, BP/DTX@PLGA, to address this challenge. In this composite nanoagent, BP quantum dots (BPQDs) are loaded into poly(lactic-co-glycolic acid) (PLGA) with additional conjugation of a chemotherapeutic agent, docetaxel (DTX). The in vivo distribution results demonstrate that BP/DTX@PLGA shows striking tropism for targeting both primary tumors and lung metastatic tumors. Moreover, BP/DTX@PLGA exhibits outstanding controllable chemophotothermal combinatory therapeutics, which dramatically improves the efficacy of photothermal tumor ablation when combined with near-light irradiation. Mechanistically, accelerated DTX release from the nanocomplex upon heating and thermal treatment per se synergistically incurs apoptosis-dependent cell death, resulting in the elimination of lung metastasis. Meanwhile, in vitro and in vivo results further confirm that BP/DTX@PLGA possesses good biocompatibility. This study provides a promising BP-based multimodal nanoagent to constrain cancer metastasis.
Collapse
Affiliation(s)
- Shunhao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Jundong Shao
- Materials and Interfaces Center , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Zhibin Li
- Materials and Interfaces Center , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Xue-Feng Yu
- Materials and Interfaces Center , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| |
Collapse
|
14784
|
Assessment of Food Safety Knowledge and Behaviors of Cancer Patients Receiving Treatment. Nutrients 2019; 11:nu11081897. [PMID: 31416193 PMCID: PMC6722877 DOI: 10.3390/nu11081897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 11/17/2022] Open
Abstract
Cancer patients receiving treatment are at a higher risk for the acquisition of foodborne illness than the general population. Despite this, few studies have assessed the food safety behaviors, attitudes, risk perceptions, and food acquisition behaviors of this population. Further, no studies have, yet, quantified the food safety knowledge of these patients. This study aims to fill these gaps in the literature by administering a thorough questionnaire to cancer patients seeking treatment in three hospitals in a Midwest, metropolitan area. Demographic, treatment, food security, and food safety knowledge, behaviors, attitudes, risk perceptions, and acquisition information was assessed for 288 patients. Specific unsafe attitudes, behaviors, and acquisition practices were identified. Most notable is that 49.4% (n = 139) of participants were not aware that they were at increased risk of foodborne infection, due to their disease and treatment. Additionally, though patients exhibited a general understanding of food safety, the participant average for correctly answering the food safety questions was 74.77% ± 12.24%. The section concerning food storage showed lowest participant knowledge, with an average score of 69.53% ± 17.47%. Finally, patients reporting low food security also reported a higher incidence of unsafe food acquisition practices (P < 0.05). These findings will help healthcare providers to better educate patients in the food safety practices necessary to decrease risk of foodborne infection, and to provide targeted food safety education to low-food-security patients.
Collapse
|
14785
|
Pretreatment prognostic nutritional index as a novel biomarker in non-small cell lung cancer patients treated with immune checkpoint inhibitors. Lung Cancer 2019; 136:45-51. [PMID: 31437663 DOI: 10.1016/j.lungcan.2019.08.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/26/2019] [Accepted: 08/05/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICIs) have been established as a novel strategy for non-small cell lung cancer (NSCLC) therapy. However, a definitive biomarker that can predict response to ICI therapy remains unestablished. The prognostic nutritional index (PNI) is used to assess immune-nutritional conditions and is a prognostic factor in patients with various malignancies; however, its usefulness as a biomarker of response to ICI therapy and survival outcomes in NSCLC patients is unknown. Thus, we retrospectively analyzed the clinicopathological features of advanced-stage or recurrent NSCLC patients treated with ICI therapy to identify predictors of response to ICI therapy and investigate the effects of pretreatment PNI levels on survival after ICI therapy. MATERIALS AND METHODS We selected 102 consecutive NSCLC patients who were treated with ICI therapy from November 2015 to February 2019. We measured their pretreatment PNI levels and performed univariate and multivariate Cox regression analyses of progression-free survival (PFS) or overall survival (OS) after ICI therapy. RESULTS Pretreatment PNI levels were significantly associated with response to ICI therapy (objective response rate:P = 0.0131; disease control rate: P = 0.0002), PFS (P = 0.0013), and OS (P = 0.0053). In univariate and multivariate analyses of the associations between PNI, C-reactive protein (CRP) or neutrophil-lymphocyte ratio (NLR) and PFS or OS, NLR and PNI, but not CRP, are independent prognostic factors for PFS (NLR: relative risk [RR]=1.655, 95% confidence interval [CI]: 1.012-2.743, P = 0.0449, PNI: RR=1.704, 95% CI: 1.039-2.828, P = 0.0346). Only PNI showed a trend towards being an independent prognostic factor for OS (RR=1.606, 95% CI: 0.952-2.745, P = 0.0761). CONCLUSION The pretreatment PNI has the potential to be a simple and novel predictive biomarker of ICI response in NSCLC patients and might help to identify patients who will obtain a survival benefit from ICI therapy.
Collapse
|
14786
|
Targeting Cellular Metabolism Modulates Head and Neck Oncogenesis. Int J Mol Sci 2019; 20:ijms20163960. [PMID: 31416244 PMCID: PMC6721038 DOI: 10.3390/ijms20163960] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/24/2022] Open
Abstract
Considering the great energy and biomass demand for cell survival, cancer cells exhibit unique metabolic signatures compared to normal cells. Head and neck squamous cell carcinoma (HNSCC) is one of the most prevalent neoplasms worldwide. Recent findings have shown that environmental challenges, as well as intrinsic metabolic manipulations, could modulate HNSCC experimentally and serve as clinic prognostic indicators, suggesting that a better understanding of dynamic metabolic changes during HNSCC development could be of great benefit for developing adjuvant anti-cancer schemes other than conventional therapies. However, the following questions are still poorly understood: (i) how does metabolic reprogramming occur during HNSCC development? (ii) how does the tumorous milieu contribute to HNSCC tumourigenesis? and (iii) at the molecular level, how do various metabolic cues interact with each other to control the oncogenicity and therapeutic sensitivity of HNSCC? In this review article, the regulatory roles of different metabolic pathways in HNSCC and its microenvironment in controlling the malignancy are therefore discussed in the hope of providing a systemic overview regarding what we knew and how cancer metabolism could be translated for the development of anti-cancer therapeutic reagents.
Collapse
|
14787
|
Song M, Giovannucci E. Preventable incidence of carcinoma associated with adiposity, alcohol and physical inactivity according to smoking status in the United States. Int J Cancer 2019; 146:2960-2967. [PMID: 31369145 DOI: 10.1002/ijc.32602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 01/11/2023]
Abstract
The changing profile of lifestyles and their intricate relationships with smoking indicate the importance of accounting for smoking status when assessing cancer preventability. We assessed the association of body mass index, weight change, alcohol intake and physical activity with risk of total carcinoma among 53,195 smokers and 62,842 nonsmokers in two prospective cohorts. Then, leveraging the national prevalence estimates, we calculated the population attributable risk (PAR) for healthy lifestyle defined as body mass index ≥18.5 and <27.5 kg/m2 , mid-life weight change of ≤20 pounds, no or moderate alcohol drinking (≤1 and 2 drinks/day for women and men, respectively) and weekly moderate or vigorous physical activity of at least 150 min. The PAR (95% CI) for healthy lifestyle was 18% (14-22%) in nonsmokers and 14% (10-19%) in smokers among women, and 20% (12-27%) in nonsmokers and 11% (5-17%) in smokers among men. While adiposity accounted for a substantially higher proportion of carcinoma cases in nonsmokers than smokers (16% vs. 2% in women, 15% vs. 2% in men), alcohol contributed more in smokers than nonsmokers (7% vs. 3% in women, 8% vs. 1% in men). When more strict criteria were used to define healthy lifestyle, the PAR estimates further increased (for women: 37% in smokers and 32% in nonsmokers; for men: 15% and 24%, respectively). In conclusion, lifestyle modification has great potential to reduce cancer risk in both smokers and nonsmokers. Weight control and reducing alcohol consumption should be prioritized for cancer prevention in nonsmokers and smokers, respectively.
Collapse
Affiliation(s)
- Mingyang Song
- Department of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Edward Giovannucci
- Department of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
14788
|
Yang J, Zhang Z, Zhao Y, Cheng J, Zhao C, Wang Z. CCT α is a novel biomarker for diagnosis of laryngeal squamous cell cancer. Sci Rep 2019; 9:11823. [PMID: 31413263 PMCID: PMC6694151 DOI: 10.1038/s41598-019-47895-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
Choline phosphate-based delivery systems can target the acidic tumor microenvironment. In this study, we set out to evaluate the diagnostic value of Choline phosphate cytidylyltransferase-α (CCTα) in laryngeal squamous cell cancer (LSCC). The expression of CCTα was detected using immunohistochemistry in 50 LSCC patients’ tissues and 16 vocal polyps as control group. Then, clinical data was collected and we used receiver operating characteristic curve (ROC) to estimate the potential of CCTα as diagnostic biomarker. We found CCTα levels to be significantly high in the tissues derived from LSCC patients, (p < 0.001). Further, we observed a positive correlation of CCTα with tumor size (p < 0.001), TNM stage (p < 0.001), lymph node metastasis (p < 0.001) as well as the grade of LSCC malignancy (p < 0.001). Furthermore, AUC was determined to be 0.939 by ROC, and the optimal cutoff value 3.100, with 76.0% sensitivity and 100% specificity. We also found an epigenetic basis of CCTα over-expression in LSCC tissues with significantly reduced methylation of CCTα in LSCC tissues, compared to vocal polyps (p < 0.001). These results support epigenetically-induced over-expression of CCTα as a potential diagnostic marker for LSCC.
Collapse
Affiliation(s)
- Jingpu Yang
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Zhuping Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Central Hospital of Wuhan, 26 Shengli Street, Wuhan, 430014, China
| | - Yin Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Jinzhang Cheng
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Chang Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Zonggui Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
14789
|
Desai PB, Bukatko AR, Simpson MC, Adjei Boakye E, Greenberg JW, Ward GM, Walker RJ, Antisdel JL, Osazuwa Peters N. Comorbidity burden and nonclinical factors associated with sinonasal cancer all‐cause mortality. Laryngoscope 2019; 130:1443-1449. [DOI: 10.1002/lary.28223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/24/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Premal B. Desai
- Saint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Aleksandr R. Bukatko
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Matthew C. Simpson
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Eric Adjei Boakye
- the Department of Population Science and PolicySouthern Illinois University School of Medicine Springfield Illinois U.S.A
| | | | - Greg M. Ward
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Ronald J. Walker
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Jastin L. Antisdel
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
| | - Nosayaba Osazuwa Peters
- Department of Otolaryngology–Head and Neck SurgerySaint Louis University School of Medicine St. Louis Missouri U.S.A
- Saint Louis University Cancer Center St. Louis Missouri U.S.A
| |
Collapse
|
14790
|
Liang Y, Zhang H, Song X, Yang Q. Metastatic heterogeneity of breast cancer: Molecular mechanism and potential therapeutic targets. Semin Cancer Biol 2019; 60:14-27. [PMID: 31421262 DOI: 10.1016/j.semcancer.2019.08.012] [Citation(s) in RCA: 526] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most common malignancies among women throughout the world and is the major cause of most cancer-related deaths. Several explanations account for the high rate of mortality of breast cancer, and metastasis to vital organs is identified as the principal cause. Over the past few years, intensive efforts have demonstrated that breast cancer exhibits metastatic heterogeneity with distinct metastatic precedence to various organs, giving rise to differences in prognoses and responses to therapy in breast cancer patients. Bone, lung, liver, and brain are generally accepted as the primary target sites of breast cancer metastasis. However, the underlying molecular mechanism of metastatic heterogeneity of breast cancer remains to be further elucidated. Recently, the advent of novel genomic and pathologic approaches as well as technological breakthroughs in imaging analysis and animal modelling have yielded an unprecedented change in our understanding of the heterogeneity of breast cancer metastasis and provided novel insight for establishing more effective therapeutics. This review summarizes recent molecular mechanisms and emerging concepts on the metastatic heterogeneity of breast cancer and discusses the potential of identifying specific molecules against tumor cells or tumor microenvironments to thwart the development of metastatic disease and improve the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Hanwen Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaojin Song
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China; Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
14791
|
Dudzinski SO, Cameron BD, Wang J, Rathmell JC, Giorgio TD, Kirschner AN. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J Immunother Cancer 2019; 7:218. [PMID: 31412954 PMCID: PMC6694548 DOI: 10.1186/s40425-019-0704-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/07/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Prostate cancer is poorly responsive to immune checkpoint inhibition, yet a combination with radiotherapy may enhance the immune response. In this study, we combined radiotherapy with immune checkpoint inhibition (iRT) in a castration-resistant prostate cancer (CRPC) preclinical model. METHODS Two Myc-CaP tumor grafts were established in each castrated FVB mouse. Anti-PD-1 or anti-PD-L1 antibodies were given and one graft was irradiated 20 Gy in 2 fractions. RESULTS In CRPC, a significant increase in survival was found for radiation treatment combined with either anti-PD-1 or anti-PD-L1 compared to monotherapy. The median survival for anti-PD-L1 alone was 13 days compared to 30 days for iRT (p = 0.0003), and for anti-PD-1 alone was 21 days compared to 36 days for iRT (p = 0.0009). Additional treatment with anti-CD8 antibody blocked the survival effect. An abscopal treatment effect was observed for iRT in which the unirradiated graft responded similarly to the irradiated graft in the same mouse. At 21 days, the mean graft volume for anti-PD-1 alone was 2094 mm3 compared to iRT irradiated grafts 726 mm3 (p = 0.04) and unirradiated grafts 343 mm3 (p = 0.0066). At 17 days, the mean graft volume for anti-PD-L1 alone was 1754 mm3 compared to iRT irradiated grafts 284 mm3 (p = 0.04) and unirradiated grafts 556 mm3 (p = 0.21). Flow cytometry and immunohistochemistry identified CD8+ immune cell populations altered by combination treatment in grafts harvested at the peak effect of immunotherapy, 2-3 weeks after starting treatment. CONCLUSIONS These data provide preclinical evidence for the use of iRT targeting PD-1 and PD-L1 in the treatment of CRPC. Immune checkpoint inhibition combined with radiotherapy treats CPRC with significant increases in median survival compared to drug alone: 70% longer for anti-PD-1 and 130% for anti-PD-L1, and with an abscopal treatment effect. PRECIS Castration-resistant prostate cancer in a wild-type mouse model is successfully treated by X-ray radiotherapy combined with PD-1 or PD-L1 immune checkpoint inhibition, demonstrating significantly increased median overall survival and robust local and abscopal treatment responses, in part mediated by CD8 T-cells.
Collapse
Affiliation(s)
- Stephanie O. Dudzinski
- 0000 0001 2264 7217grid.152326.1Vanderbilt University School of Medicine, Nashville, TN 37232 USA ,0000 0001 2264 7217grid.152326.1Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232 USA
| | - Brent D. Cameron
- 0000 0004 1936 9916grid.412807.8Department of Radiation Oncology, Vanderbilt University Medical Center, B1003 PRB, 2220 Pierce Avenue, Nashville, TN 37232 USA
| | - Jian Wang
- 0000 0004 1936 9916grid.412807.8Department of Radiation Oncology, Vanderbilt University Medical Center, B1003 PRB, 2220 Pierce Avenue, Nashville, TN 37232 USA
| | - Jeffrey C. Rathmell
- 0000 0004 1936 9916grid.412807.8Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232 USA ,0000 0001 2264 7217grid.152326.1Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Todd D. Giorgio
- 0000 0001 2264 7217grid.152326.1Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232 USA
| | - Austin N. Kirschner
- 0000 0004 1936 9916grid.412807.8Department of Radiation Oncology, Vanderbilt University Medical Center, B1003 PRB, 2220 Pierce Avenue, Nashville, TN 37232 USA
| |
Collapse
|
14792
|
Mina LA, Arun B. Polygenic Risk Scores in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00320-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14793
|
Eskra JN, Dodge A, Schlicht MJ, Bosland MC. Effects of Black Raspberries and Their Constituents on Rat Prostate Carcinogenesis and Human Prostate Cancer Cell Growth In Vitro. Nutr Cancer 2019; 72:672-685. [PMID: 31402717 DOI: 10.1080/01635581.2019.1650943] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prostate cancer patients often use dietary supplements, such as black raspberries, which are a rich source of compounds with antioxidant and anticancer activity, particularly on gastrointestinal cancers. Feeding black raspberries inhibited mammary cancer induction in rats and growth of cancer cells in nude mice, indicating systemic bioavailability of bioactive compounds. We tested whether feeding black raspberries and its constituents would inhibit prostate cancer development. However, we did not find preventive effects in two rat prostate carcinogenesis models, even though the berry anthocyanin metabolite protocatechuic acid was detectable in their prostates. Black raspberry extract, the anthocyanin cyanidin-3-rutinoside and protocatechuic acid did not inhibit prostate cancer cell growth in vitro, but ellagic acid and its urolithin A metabolite did at high concentrations. Prostate cancer cell migration was not affected by these agents nor was growth in soft agar, except that ellagic acid reduced colony formation at physiological concentrations and protocatechuic acid at high concentrations. Low bioavailability of bioactive berry compounds and metabolites may limit exposure of tissues such as the prostate, since we found that cyanidin-3-rutinoside was not bioavailable to prostate cancer cells, but its aglycone cyanidin was and inhibited their growth. Thus, black raspberries are unlikely to prevent prostate cancer.
Collapse
Affiliation(s)
- Jillian N Eskra
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Alaina Dodge
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael J Schlicht
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Maarten C Bosland
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
14794
|
Patton MC, Zubair H, Khan MA, Singh S, Singh AP. Hypoxia alters the release and size distribution of extracellular vesicles in pancreatic cancer cells to support their adaptive survival. J Cell Biochem 2019; 121:828-839. [PMID: 31407387 DOI: 10.1002/jcb.29328] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
Abstract
Pancreatic tumors are highly desmoplastic and poorly-vascularized, and therefore must develop adaptive mechanisms to sustain their survival under hypoxic condition. Extracellular vesicles (EV) play vital roles in pancreatic tumor pathobiology by facilitating intercellular communication. Here we studied the effect of hypoxia on the release of EVs and examined their role in adaptive survival of pancreatic cancer (PC) cells. Hypoxia promoted the release of EV in PC cell lines, MiaPaCa and AsPC1, wherein former exhibited a far greater induction. Moreover, a time-dependent, measurable and significant increase was recorded for small EV (SEV) in both the cell lines with only minimal induction observed for medium (MEV) and large EVs (LEV). Similarly, noticeable changes in size distribution of SEV were also recorded with a shift toward smaller average size under extreme hypoxia. Thrombospondin (apoptotic bodies marker) was exclusively detected on LEVs, while Arf6 (microvesicles marker) was mostly present on MEV with some expression in LEV as well. However, CD9 and CD63 (exosome markers) were expressed in both SEV and MEVs with a decreased expression recorded under hypoxia. Among all subfractions, SEV was the most bioactive in promoting the survival of hypoxic PC cells and hypoxia-inducible factor-1α stabilization was involved in heightened EV release under hypoxia and for their potency to promote hypoxic cell survival. Altogether, our findings provide a novel mechanism for the adaptive hypoxic survival of PC cells and should serve as the basis for future investigations on broader functional implications of EV.
Collapse
Affiliation(s)
- Mary C Patton
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Haseeb Zubair
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Mohammad Aslam Khan
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Seema Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Ajay P Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| |
Collapse
|
14795
|
Manucha V, Henegan J. Clinicopathologic Diagnostic Approach to Aggressive Variant Prostate Cancer. Arch Pathol Lab Med 2019; 144:18-23. [PMID: 31403335 DOI: 10.5858/arpa.2019-0124-ra] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Aggressive variant prostate cancer (AVPCa) develops in a subset of patients with metastatic castration-resistant prostate cancer. The clinical and histologic overlap of AVPCa with other neuroendocrine carcinomas of the prostate has resulted in a lack of consensus on its terminology and treatment. OBJECTIVE.— To review AVPCa to familiarize pathologists with this entity so they can actively participate in the detection, ongoing research, and evolving management of AVPCa. DATA SOURCES.— The English language literature was reviewed. CONCLUSIONS.— The current review summarizes the pathologic features of AVPCa, describes how it has been defined clinically, and discusses how biomarkers may inform treatment strategies in the future.
Collapse
Affiliation(s)
- Varsha Manucha
- From the Department of Pathology (Dr Manucha) and the Division of Hematology/Oncology, Department of Medicine (Dr Henegan), University of Mississippi Medical Center, Jackson
| | - John Henegan
- From the Department of Pathology (Dr Manucha) and the Division of Hematology/Oncology, Department of Medicine (Dr Henegan), University of Mississippi Medical Center, Jackson
| |
Collapse
|
14796
|
Barabadi H, Vahidi H, Damavandi Kamali K, Hosseini O, Mahjoub MA, Rashedi M, Jazayeri Shoushtari F, Saravanan M. Emerging Theranostic Gold Nanomaterials to Combat Lung Cancer: A Systematic Review. J CLUST SCI 2019. [DOI: 10.1007/s10876-019-01650-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
14797
|
Benitez MLR, Bender CB, Oliveira TL, Schachtschneider KM, Collares T, Seixas FK. Mycobacterium bovis BCG in metastatic melanoma therapy. Appl Microbiol Biotechnol 2019; 103:7903-7916. [PMID: 31402426 DOI: 10.1007/s00253-019-10057-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Melanoma is the most aggressive form of skin cancer, with a high mortality rate and with 96,480 new cases expected in 2019 in the USS. BRAFV600E, the most common driver mutation, is found in around 50% of melanomas, contributing to tumor growth, angiogenesis, and metastatic progression. Dacarbazine (DTIC), an alkylate agent, was the first chemotherapeutic agent approved by the US Food and Drug Administration (FDA) used as a standard treatment. Since then, immunotherapies have been approved for metastatic melanoma (MM) including ipilimumab and pembrolizumab checkpoint inhibitors that help decrease the risk of progression. Moreover, Mycobacterium bovis Bacillus Calmette-Guerin (BCG) serves as an adjuvant therapy that induces the recruitment of natural killer NK, CD4+, and CD8+ T cells and contributes to antitumor immunity. BCG can be administered in combination with chemotherapeutic and immunotherapeutic agents and can be genetically manipulated to produce recombinant BCG (rBCG) strains that express heterologous proteins or overexpress immunogenic proteins, increasing the immune response and improving patient survival. In this review, we highlight several studies utilizing rBCG immunotherapy for MM in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Martha Lucia Ruiz Benitez
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Bonnemann Bender
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thaís Larré Oliveira
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
14798
|
Liu F, Lin S, Zhang C, Ma J, Han Z, Jia F, Xie W, Li X. The Novel Nature Microtubule Inhibitor Ivalin Induces G2/M Arrest and Apoptosis in Human Hepatocellular Carcinoma SMMC-7721 Cells In Vitro. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E470. [PMID: 31409007 PMCID: PMC6723664 DOI: 10.3390/medicina55080470] [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] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/02/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022]
Abstract
Background and Objectives: Microtubules are an attractive target for cancer chemotherapy. Previously, we reported that Ivalin exhibited excellent anti-migration and anti-invasion activities in human breast cancer cells. Here, we examined the microtubule inhibition effect of Ivalin in human hepatocellular carcinoma SMMC-7721 cells. Materials and Methods: We used the 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide (MTT) assay to evaluate the cell proliferation effect of Ivalin and flow cytometry analysis to detect the apoptotic and cell cycle arrest effects of Ivalin. Immunofluorescence staining was used to measure the effect of Ivalin on the cytoskeleton network, and Western blotting was used to detect the expression levels of Bax, Bcl-2, Cdc2, phosphor-Cdc2, Cdc25A, Cyclin B1, and tubulin. Results: Ivalin induced cell cycle G2/M arrest and subsequent triggered apoptosis in human hepatocellular carcinoma SMMC-7721 cells. Furthermore, microtubules were shown to be involved in Ivalin-meditated apoptosis. In this connection, Ivalin treatment suppressed cellular microtubule network formation by regulating microtubule depolymerization. Moreover, Western blotting revealed Cdc25A and Cyclin B1 were upregulated in Ivalin-meditated cell cycle arrest. Subsequently, the induction of Bax (a proapoptotic protein) and reduction of Bcl-2 (an anti-apoptotic protein) expression were observed in Ivalin-treated SMMC-7721 cells. Conclusion: Ivalin induced microtubule depolymerization, then blocked cells in mitotic phase, and eventually resulted in apoptosis in SMMC-7721 cells. Collectively, these data indicate that Ivalin, acting as a novel inhibitor of microtubules, could be considered as a promising lead in anticancer drug development.
Collapse
Affiliation(s)
- Fangyuan Liu
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Shiqi Lin
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Caiyun Zhang
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Jiahui Ma
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Zhuo Han
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Fujuan Jia
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Weidong Xie
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Xia Li
- College of Marine Science, Shandong University, Weihai 264209, China.
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China.
| |
Collapse
|
14799
|
Wang T, McCullough LE, White AJ, Bradshaw PT, Xu X, Cho YH, Terry MB, Teitelbaum SL, Neugut AI, Santella RM, Chen J, Gammon MD. Prediagnosis aspirin use, DNA methylation, and mortality after breast cancer: A population-based study. Cancer 2019; 125:3836-3844. [PMID: 31402456 DOI: 10.1002/cncr.32364] [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: 07/13/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The authors hypothesized that epigenetic changes may help to clarify the underlying biologic mechanism linking aspirin use to breast cancer prognosis. To the authors' knowledge, this is the first epidemiologic study to examine whether global methylation and/or tumor promoter methylation of breast cancer-related genes interact with aspirin use to impact mortality after breast cancer. METHODS Prediagnosis aspirin use was assessed through in-person interviews within a population-based cohort of 1508 women diagnosed with a first primary breast cancer in 1996 and 1997. Global methylation in peripheral blood was assessed by long interspersed elements-1 (LINE-1) and the luminometric methylation assay. Promoter methylation of 13 breast cancer-related genes was measured in tumor by methylation-specific polymerase chain reaction and the MethyLight assay. Vital status was determined by the National Death Index through December 31, 2014 (N = 202/476 breast cancer-specific/all-cause deaths identified among 1266 women with any methylation assessment and complete aspirin data). Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% CIs, and the likelihood ratio test was used to evaluate multiplicative interactions. RESULTS All-cause mortality was elevated among aspirin users who had methylated promotor of BRCA1 (HR, 1.67; 95% CI, 1.26-2.22), but not among those with unmethylated promoter of BRCA1 (HR, 0.99; 95% CI, 0.67-1.45; P for interaction ≤.05). Decreased breast cancer-specific mortality was observed among aspirin users who had unmethylated promotor of BRCA1 and PR and global hypermethylation of LINE-1 (HR, 0.60, 0.78, and 0.63, respectively; P for interaction ≤.05), although the 95% CIs included the null. CONCLUSIONS The current study suggests that the LINE-1 global methylation and promoter methylation of BRCA1 and PR in tumor may interact with aspirin use to influence mortality after breast cancer.
Collapse
Affiliation(s)
- Tengteng Wang
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | | | - Alexandra J White
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Patrick T Bradshaw
- Division of Epidemiology, University of California, Berkeley, California
| | - Xinran Xu
- Department of Biometrics, Roche Product Development in Asia-Pacific, Shanghai, China
| | - Yoon Hee Cho
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, New York
| | - Susan L Teitelbaum
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alfred I Neugut
- Department of Epidemiology, Columbia University, New York, New York.,Department of Medicine, Columbia University, New York, New York
| | | | - Jia Chen
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
14800
|
Whole Transcriptome Analysis Identifies TNS4 as a Key Effector of Cetuximab and a Regulator of the Oncogenic Activity of KRAS Mutant Colorectal Cancer Cell Lines. Cells 2019; 8:cells8080878. [PMID: 31409052 PMCID: PMC6721647 DOI: 10.3390/cells8080878] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/31/2019] [Accepted: 08/10/2019] [Indexed: 12/21/2022] Open
Abstract
The targeting of activated epidermal growth factor receptor (EGFR) with therapeutic anti-EGFR monoclonal antibodies (mAbs) such as cetuximab and panitumumab has been used as an effective strategy in the treatment of colorectal cancer (CRC). However, its clinical efficacy occurs only in a limited number of patients. Here, we performed whole-transcriptome analysis in xenograft mouse tumors induced by KRASG12D mutation-bearing LS174T CRC cells following treatment with either cetuximab or PBS. Through integrated analyses of differential gene expression with TCGA and CCLE public database, we identified TNS4, overexpressed in CRC patients and KRAS mutation-harboring CRC cell lines, significantly downregulated by cetuximab. While ablation of TNS4 expression via shRNA results in significant growth inhibition of LS174T, DLD1, WiDr, and DiFi CRC cell lines, conversely, its ectopic expression increases the oncogenic growth of these cells. Furthermore, TNS4 expression is transcriptionally regulated by MAP kinase signaling pathway. Consistent with this finding, selumetinib, a MEK1/2 inhibitor, suppressed oncogenic activity of CRC cells, and this effect is more profound in combination with cetuximab. Altogether, we propose that TNS4 plays a crucial role in CRC tumorigenesis, and that suppression of TNS4 would be an effective therapeutic strategy in treating a subset of cetuximab-refractory CRC patients including KRAS activating mutations.
Collapse
|