1
|
Palizkaran Yazdi M, Barjasteh A, Moghbeli M. MicroRNAs as the pivotal regulators of Temozolomide resistance in glioblastoma. Mol Brain 2024; 17:42. [PMID: 38956588 PMCID: PMC11218189 DOI: 10.1186/s13041-024-01113-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive nervous system tumor with a poor prognosis. Although, surgery, radiation therapy, and chemotherapy are the current standard protocol for GBM patients, there is still a poor prognosis in these patients. Temozolomide (TMZ) as a first-line therapeutic agent in GBM can easily cross from the blood-brain barrier to inhibit tumor cell proliferation. However, there is a high rate of TMZ resistance in GBM patients. Since, there are limited therapeutic choices for GBM patients who develop TMZ resistance; it is required to clarify the molecular mechanisms of chemo resistance to introduce the novel therapeutic targets. MicroRNAs (miRNAs) regulate chemo resistance through regulation of drug metabolism, absorption, DNA repair, apoptosis, and cell cycle. In the present review we discussed the role of miRNAs in TMZ response of GBM cells. It has been reported that miRNAs mainly induced TMZ sensitivity by regulation of signaling pathways and autophagy in GBM cells. Therefore, miRNAs can be used as the reliable diagnostic/prognostic markers in GBM patients. They can also be used as the therapeutic targets to improve the TMZ response in GBM cells.
Collapse
Affiliation(s)
- Mahsa Palizkaran Yazdi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Barjasteh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Onyije FM, Dolatkhah R, Olsson A, Bouaoun L, Deltour I, Erdmann F, Bonaventure A, Scheurer ME, Clavel J, Schüz J. Risk factors for childhood brain tumours: A systematic review and meta-analysis of observational studies from 1976 to 2022. Cancer Epidemiol 2024; 88:102510. [PMID: 38056243 PMCID: PMC10835339 DOI: 10.1016/j.canep.2023.102510] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Childhood brain tumours (CBTs) are the leading cause of cancer death in children under the age of 20 years globally. Though the aetiology of CBT remains poorly understood, it is thought to be multifactorial. We aimed to synthesize potential risk factors for CBT to inform primary prevention. METHODS We conducted a systematic review and meta-analysis of epidemiological studies indexed in the PubMed, Web of Science, and Embase databases from the start of those resources through 27 July 2023. We included data from case-control or cohort studies that reported effect estimates for each risk factor around the time of conception, during pregnancy and/or during post-natal period. Random effects meta-analysis was used to estimate summary effect sizes (ES) and 95% confidence intervals (CIs). We also quantified heterogeneity (I2) across studies. FINDINGS A total of 4040 studies were identified, of which 181 studies (85 case-control and 96 cohort studies) met our criteria for inclusion. Of all eligible studies, 50% (n = 91) were conducted in Europe, 32% (n = 57) in North America, 9% (n = 16) in Australia, 8% (n = 15) in Asia, 1% (n = 2) in South America, and none in Africa. We found associations for some modifiable risk factors including childhood domestic exposures to insecticides (ES 1.44, 95% CI 1.20-1.73) and herbicides (ES 2.38, 95% CI 1.31-4.33). Maternal domestic exposure to insecticides (ES 1.45, 95% CI 1.09-1.94), maternal consumption of cured meat (ES 1.51, 95% CI 1.05-2.17) and coffee ≥ 2 cups/day (ES 1.45, 95% 95% CI 1.07-1.95) during pregnancy, and maternal exposure to benzene (ES 2.22; 95% CI 1.01-4.88) before conception were associated with CBTs in case-control studies. Also, paternal occupational exposure to pesticides (ES 1.48, 95% CI 1.23-1.77) and benzene (ES 1.74, 95% CI 1.10-2.76) before conception and during pregnancy were associated in case-control studies and in combined analysis. On the other hand, assisted reproductive technology (ART) (ES 1.32, 95% CI 1.05-1.67), caesarean section (CS) (ES 1.12, 95% CI 1.01-1.25), paternal occupational exposure to paint before conception (ES 1.56, 95% CI 1.02-2.40) and maternal smoking > 10 cigarettes per day during pregnancy (ES 1.18, 95% CI 1.00-1.40) were associated with CBT in cohort studies. Maternal intake of vitamins and folic acid during pregnancy was inversely associated in cohort studies. Hormonal/infertility treatment, breastfeeding, child day-care attendance, maternal exposure to electric heated waterbed, tea and alcohol consumption during pregnancy were among those not associated with CBT in both case-control and cohort studies. CONCLUSION Our results should be interpreted with caution, especially as most associations between risk factors and CBT were discordant between cohort and case-control studies. At present, it is premature for any CBT to define specific primary prevention guidelines.
Collapse
Affiliation(s)
- Felix M Onyije
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France.
| | - Roya Dolatkhah
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Ann Olsson
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Liacine Bouaoun
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Isabelle Deltour
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Friederike Erdmann
- Research Group Aetiology and Inequalities in Childhood Cancer, Division of Childhood Cancer Epidemiology Institute of Medical Biostatistics, Epidemiology, and Informatics (IMBEI), University Medical Center Mainz, Langenbeckstraβe 1, 55131 Mainz, Germany
| | - Audrey Bonaventure
- Epidemiology of Childhood and Adolescent Cancers Team, Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), Villejuif, France
| | - Michael E Scheurer
- Department of Pediatrics, Hematology-Oncology, Baylor College of Medicine and Texas Children's Hospital Cancer Center, Houston, TX, United States
| | - Jacqueline Clavel
- Epidemiology of Childhood and Adolescent Cancers Team, Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), Villejuif, France; National Registry of Childhood Cancers, Hôpital Paul Brousse, Groupe Hospitalier Universitaire Paris-Sud, Assistance Publique Hôpitaux de Paris (AP-HP), Villejuif, France; Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre-lès-Nancy, France
| | - Joachim Schüz
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| |
Collapse
|
3
|
Lu J, Wang Z, He Z, Hu Y, Duan H, Liu Z, Li D, Zhong S, Ren J, Zhao G, Mou Y, Yao M. Oligomer-Aβ42 suppress glioma progression via potentiating phagocytosis of microglia. CNS Neurosci Ther 2024; 30:e14495. [PMID: 37849438 PMCID: PMC10805446 DOI: 10.1111/cns.14495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/12/2023] [Accepted: 10/01/2023] [Indexed: 10/19/2023] Open
Abstract
AIMS Glioma is characterized by an immunosuppressed environment and a poor prognosis. The accumulation of Amyloid β (Aβ) leads to an active environment during the early stages of Alzheimer's disease (AD). Aβ is also present in glioma tissues; however, the biological and translational implications of Aβ in glioma are elusive. METHODS Immunohistochemical (IHC) staining, Kaplan-Meier (KM) survival analysis and Cox regression analysis on a cohort of 79 patients from our institution were performed to investigate the association between Aβ and the malignancy of glioma. Subsequently, the potential of oligomer-Aβ42 (OAβ42) to inhibit glioma growth was investigated in vivo and in vitro. Immunofluorescence staining and phagocytosis assays were performed to evaluate the activation of microglia. Finally, RNA-seq was utilized to identify the predominant signaling involved in this process and in vitro studies were performed to validate them. RESULTS A positive correlation between Aβ and a favorable prognosis was observed in glioma. Furthermore, OAβ42 suppressed glioma growth by enhancing the phagocytic activity of microglia. Insulin-like growth factor 1 (IGF-1) secreted by OAβ42-activated microglia was essential in the engulfment process. CONCLUSION Our study proved an anti-glioma effect of Aβ, and microglia could serve as a cellular target for treating glioma with OAβ42.
Collapse
Affiliation(s)
- Jie Lu
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Zhenning Wang
- Department of Neurosurgery, Dongguan People's Hospital (Affiliated Dongguan Hospital)Southern Medical UniversityDongguanChina
| | - Zhenqiang He
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yang Hu
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Hao Duan
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Zihao Liu
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Depei Li
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Sheng Zhong
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jiaoyan Ren
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhouChina
| | - Guojun Zhao
- Laboratory Animal CenterThe Sixth Affiliated Hospital of Guangzhou Medical UniversityQingyuanChina
| | - Yonggao Mou
- Department of Neurosurgery/Neuro‐oncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Maojin Yao
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhouChina
| |
Collapse
|
4
|
Trojan A, Lone YC, Briceno I, Trojan J. Anti-Gene IGF-I Vaccines in Cancer Gene Therapy: A Review of a Case of Glioblastoma. Curr Med Chem 2024; 31:1983-2002. [PMID: 38031775 DOI: 10.2174/0109298673237968231106095141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE Vaccines for the deadliest brain tumor - glioblastoma (GBM) - are generally based on targeting growth factors or their receptors, often using antibodies. The vaccines described in the review were prepared to suppress the principal cancer growth factor - IGF-I, using anti-gene approaches either of antisense (AS) or of triple helix (TH) type. Our objective was to increase the median survival of patients treated with AS and TH cell vaccines. METHODOLOGY The cells were transfected in vitro by both constructed IGF-I AS and IGF-I TH expression episomal vectors; part of these cells was co-cultured with plant phytochemicals, modulating IGF-I expression. Both AS and TH approaches completely suppressed IGF-I expression and induced MHC-1 / B7 immunogenicity related to the IGF-I receptor signal. RESULTS This immunogenicity proved to be stronger in IGF-I TH than in IGF-I AS-prepared cell vaccines, especially in TH / phytochemical cells. The AS and TH vaccines generated an important TCD8+ and TCD8+CD11b- immune response in treated GBM patients and increased the median survival of patients up to 17-18 months, particularly using TH vaccines; in some cases, 2- and 3-year survival was reported. These clinical results were compared with those obtained in therapies targeting other growth factors. CONCLUSION The anti-gene IGF-I vaccines continue to be applied in current GBM personalized medicine. Technical improvements in the preparation of AS and TH vaccines to increase MHC-1 and B7 immunogenicity have, in parallel, allowed to increase in the median survival of patients.
Collapse
Affiliation(s)
- Annabelle Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- Faculty of Medicine, University of Cartagena, PO Box: 130014 Cartagena de Indias, Colombia
| | - Yu-Chun Lone
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
| | - Ignacio Briceno
- Faculty of Medicine, University of La Sabana, PO Box: 250008 Chia, Colombia
| | - Jerzy Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
- National Academy of Medicine - ANM, PO Box: 75272 Paris, France
| |
Collapse
|
5
|
Zhang W, Peng Y, Kang X, Wang C, Chen F, He Y, Li W. Healthy and Unhealthy Plant-Based Diets and Glioma in the Chinese Population. Brain Sci 2023; 13:1401. [PMID: 37891770 PMCID: PMC10605677 DOI: 10.3390/brainsci13101401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Plant-based diets have been suggested to help prevent various chronic diseases, including cancer. However, there are few reports on central nervous system tumors, and data on dose-response relationships are lacking. This individual-matched case-control study included 506 cases and 506 controls. The overall plant-based diet index (PDI), the healthy plant-based diet index (hPDI), and the unhealthy plant-based diet index (uPDI) were calculated using dietary information collected through a food frequency questionnaire, with higher scores indicating better adherence. We analyzed the relationship of plant-based diets with glioma. After adequate adjustment for confounders, PDI was associated with a reduced glioma risk (OR = 0.42, 95% CI: 0.24-0.72). Conversely, uPDI was associated with an elevated glioma risk (OR = 8.04, 95% CI: 4.15-15.60). However, hPDI was not significantly associated with glioma risk (OR = 0.83, 95% CI: 0.48-1.45). For subgroups, PDI was not significant in analyzing young age, BMI, or any pathological subtypes. The restricted cubic spline function showed a significant dose-response relationship between PDI (p-nonlinearity< 0.0001) and uPDI (p-nonlinearity= 0.0711) and glioma. Further analysis found that refined grains had the greatest effect on gliomas in the less healthy plant-based food group. Therefore, following a plant-based diet was linked to a lower risk of glioma, especially when consuming fewer unhealthy plant-based foods.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (W.Z.); (Y.P.); (X.K.); (C.W.); (F.C.); (Y.H.)
| |
Collapse
|
6
|
Rabah N, Ait Mohand FE, Kravchenko-Balasha N. Understanding Glioblastoma Signaling, Heterogeneity, Invasiveness, and Drug Delivery Barriers. Int J Mol Sci 2023; 24:14256. [PMID: 37762559 PMCID: PMC10532387 DOI: 10.3390/ijms241814256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The most prevalent and aggressive type of brain cancer, namely, glioblastoma (GBM), is characterized by intra- and inter-tumor heterogeneity and strong spreading capacity, which makes treatment ineffective. A true therapeutic answer is still in its infancy despite various studies that have made significant progress toward understanding the mechanisms behind GBM recurrence and its resistance. The primary causes of GBM recurrence are attributed to the heterogeneity and diffusive nature; therefore, monitoring the tumor's heterogeneity and spreading may offer a set of therapeutic targets that could improve the clinical management of GBM and prevent tumor relapse. Additionally, the blood-brain barrier (BBB)-related poor drug delivery that prevents effective drug concentrations within the tumor is discussed. With a primary emphasis on signaling heterogeneity, tumor infiltration, and computational modeling of GBM, this review covers typical therapeutic difficulties and factors contributing to drug resistance development and discusses potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem 91120, Israel; (N.R.); (F.-E.A.M.)
| |
Collapse
|
7
|
Huang X, Hansen J, Lee PC, Wu CK, Federman N, Arah OA, Li CY, Olsen J, Ritz B, Heck JE. Maternal diabetes and childhood cancer risks in offspring: two population-based studies. Br J Cancer 2022; 127:1837-1842. [PMID: 36088507 PMCID: PMC9643384 DOI: 10.1038/s41416-022-01961-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The effect of maternal diabetes on childhood cancer has not been widely studied. METHODS We examined this in two population-based studies in Denmark (N = 6420 cancer cases, 160,484 controls) and Taiwan (N = 2160 cancer cases, 2,076,877 non-cases) using logistic regression and Cox proportional hazard regression adjusted for birth year, child's sex, maternal age and birth order. RESULTS Gestational diabetes in Denmark [odds ratio (OR) = 0.98, 95% confidence interval (CI): 0.71-1.35] or type II and gestational diabetes in Taiwan (type II: hazard ratio (HR) = 0.81, 95% CI: 0.63-1.05; gestational diabetes: HR = 1.06, 95% CI: 0.92-1.22) were not associated with cancer (all types combined). In Denmark, maternal type I diabetes was associated with the risk of glioma (OR = 2.33, 95% CI: 1.04-5.22), while in Taiwan, the risks of glioma (HR = 1.59, 95% CI: 1.01-2.50) were elevated among children whose mothers had gestational diabetes. There was a twofold increased risk for hepatoblastoma with maternal type II diabetes (HR = 2.02, 95% CI: 1.02-4.00). CONCLUSIONS Our results suggest that maternal diabetes is an important risk factor for certain types of childhood cancers, emphasising the need for effective interventions targeting maternal diabetes to prevent serious health effects in offspring.
Collapse
Affiliation(s)
- Xiwen Huang
- Department of Epidemiology, Fielding School of Public Health, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095-1772, USA
| | - Johnni Hansen
- Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Pei-Chen Lee
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, 89 Nei-Chiang St, Wan-Hua Dist, Taipei, 10845, Taiwan.
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm U1018, Team "Exposome, heredity, cancer and health", CESP, 94807, Villejuif, France.
- Department of Public Health, National Cheng Kung University, #1, University Road, Tainan, 70101, Taiwan.
| | - Chia-Kai Wu
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm U1018, Team "Exposome, heredity, cancer and health", CESP, 94807, Villejuif, France
| | - Noah Federman
- Department of Pediatrics, Geffen School of Medicine, UCLA, Los Angeles, CA, 90095-1752, USA
| | - Onyebuchi A Arah
- Department of Epidemiology, Fielding School of Public Health, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095-1772, USA
- Department of Statistics, UCLA College of Letters and Science, Los Angeles, CA, USA
- Section for Epidemiology, Department of Public Health, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Chung-Yi Li
- Department of Public Health, National Cheng Kung University, #1, University Road, Tainan, 70101, Taiwan
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Jorn Olsen
- Department of Clinical Epidemiology, Aarhus University, Olof Palmes Allé 43-45 8200 Aarhus N, Aarhus, Denmark
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095-1772, USA
| | - Julia E Heck
- Department of Epidemiology, Fielding School of Public Health, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095-1772, USA.
- College of Health and Public Service, University of North Texas, 1155 Union Circle #305250, Denton, TX, 76203-5017, USA.
| |
Collapse
|
8
|
Marley AR, Domingues A, Ghosh T, Turcotte LM, Spector LG. Maternal BMI, Diabetes, and Gestational Weight Gain and Risk for Pediatric Cancer in Offspring: A Systematic Review and Meta-Analysis. JNCI Cancer Spectr 2022; 6:6537542. [PMID: 35603850 PMCID: PMC8982388 DOI: 10.1093/jncics/pkac020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background Pediatric cancer incidence has steadily increased concurrent with rising adult obesity, but associations between maternal obesity and associated comorbidities and pediatric cancer risk remain understudied. We aimed to quantitatively characterize associations of pediatric cancer risk with maternal prepregnancy body mass index (BMI), gestational weight gain, and maternal diabetes. Methods We performed a comprehensive and systematic literature search in Ovid and EMBASE from their inception to March 15, 2021. Eligible studies reported risk estimates and sample sizes and provided sufficient description of outcome and exposure ascertainment. Random effects models were used to estimate pooled effects. Results Thirty-four studies were included in the analysis. Prepregnancy BMI was positively associated with leukemia risk in offspring (odds ratio [OR] per 5-unit BMI increase =1.07, 95% confidence intervals [CI] = 1.04 to 1.11; I2 = 0.0%). Any maternal diabetes was positively associated with acute lymphoblastic leukemia risk (OR = 1.46, 95% CI = 1.28 to 1.67; I2 = 0.0%), even after restricting to birthweight-adjusted analyses (OR = 1.74, 95% CI = 1.29 to 2.34; I2 = 0.0%), and inversely associated with risk of central nervous system tumors (OR = 0.73, 95% CI = 0.55 to 0.97; I2 = 0.0%). Pregestational diabetes (OR = 1.57, 95% CI = 1.11 to 2.24; I2 = 26.8%) and gestational diabetes (OR = 1.40, 95% CI = 1.12 to 1.75; I2 = 0.0%) were also positively associated with acute lymphoblastic leukemia risk. No statistically significant associations were observed for gestational weight gain. Conclusions Maternal obesity and diabetes may be etiologically linked to pediatric cancer, particularly leukemia and central nervous system tumors. Our findings support weight management and glycemic control as important components of maternal and offspring health. Further validation is warranted.
Collapse
Affiliation(s)
- Andrew R Marley
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Allison Domingues
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Taumoha Ghosh
- Division of Hematology/Oncology, Department of Pediatrics, University of Miami, Miami, FL, USA
| | - Lucie M Turcotte
- Division of Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Logan G Spector
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
9
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
10
|
Tilak M, Holborn J, New LA, Lalonde J, Jones N. Receptor Tyrosine Kinase Signaling and Targeting in Glioblastoma Multiforme. Int J Mol Sci 2021; 22:1831. [PMID: 33673213 PMCID: PMC7918566 DOI: 10.3390/ijms22041831] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is amongst the deadliest of human cancers, with a median survival rate of just over one year following diagnosis. Characterized by rapid proliferation and diffuse infiltration into the brain, GBM is notoriously difficult to treat, with tumor cells showing limited response to existing therapies and eventually developing resistance to these interventions. As such, there is intense interest in better understanding the molecular alterations in GBM to guide the development of more efficient targeted therapies. GBM tumors can be classified into several molecular subtypes which have distinct genetic signatures, and they show aberrant activation of numerous signal transduction pathways, particularly those connected to receptor tyrosine kinases (RTKs) which control glioma cell growth, survival, migration, invasion, and angiogenesis. There are also non-canonical modes of RTK signaling found in GBM, which involve G-protein-coupled receptors and calcium channels. This review uses The Cancer Genome Atlas (TCGA) GBM dataset in combination with a data-mining approach to summarize disease characteristics, with a focus on select molecular pathways that drive GBM pathogenesis. We also present a unique genomic survey of RTKs that are frequently altered in GBM subtypes, as well as catalog the GBM disease association scores for all RTKs. Lastly, we discuss current RTK targeted therapies and highlight emerging directions in GBM research.
Collapse
Affiliation(s)
| | | | | | | | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.T.); (J.H.); (L.A.N.); (J.L.)
| |
Collapse
|
11
|
Tirrò E, Massimino M, Romano C, Martorana F, Pennisi MS, Stella S, Pavone G, Di Gregorio S, Puma A, Tomarchio C, Vitale SR, Manzella L, Vigneri P. Prognostic and Therapeutic Roles of the Insulin Growth Factor System in Glioblastoma. Front Oncol 2021; 10:612385. [PMID: 33604294 PMCID: PMC7885861 DOI: 10.3389/fonc.2020.612385] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain malignancy and is often resistant to conventional treatments due to its extensive cellular heterogeneity. Thus, the overall survival of GBM patients remains extremely poor. Insulin-like growth factor (IGF) signaling entails a complex system that is a key regulator of cell transformation, growth and cell-cycle progression. Hence, its deregulation is frequently involved in the development of several cancers, including brain malignancies. In GBM, differential expression of several IGF system components and alterations of this signaling axis are linked to significantly worse prognosis and reduced responsiveness to temozolomide, the most commonly used pharmacological agent for the treatment of the disease. In the present review we summarize the biological role of the IGF system in the pathogenesis of GBM and comprehensively discuss its clinical significance and contribution to the development of resistance to standard chemotherapy and experimental treatments.
Collapse
Affiliation(s)
- Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy.,Medical Oncology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Giuliana Pavone
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy.,Medical Oncology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Sandra Di Gregorio
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Cristina Tomarchio
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Silvia Rita Vitale
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy.,Medical Oncology, A.O.U. Policlinico "G. Rodolico-San Marco", Catania, Italy
| |
Collapse
|
12
|
Saini M, Jha AN, Tangri R, Qudratullah M, Ali S. MN1 overexpression with varying tumor grade is a promising predictor of survival of glioma patients. Hum Mol Genet 2021; 29:3532-3545. [PMID: 33105486 PMCID: PMC7788295 DOI: 10.1093/hmg/ddaa231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/31/2022] Open
Abstract
Gliomas have substantial mortality to incidence rate ratio and a dismal clinical course. Newer molecular insights, therefore, are imperative to refine glioma diagnosis, prognosis and therapy. Meningioma 1 (MN1) gene is a transcriptional co-regulator implicated in other malignancies, albeit its significance in glioma pathology remains to be explored. IGFBP5 is regulated transcriptionally by MN1 and IGF1 and is associated with higher glioma grade and shorter survival time, prompting us to ascertain their correlation in these tumors. We quantified the expression of MN1, IGFBP5 and IGF1 in 40 glioma samples and examined their interrelatedness. MN1 mRNA-protein inter-correlation and the gene's copy number were evaluated in these tumors. Publicly available TCGA datasets were used to examine the association of MN1 expression levels with patient survival and for validating our findings. We observed MN1 overexpression correlated with low-grade (LGGs) and not high-grade gliomas and is not determined by the copy number alteration of the gene. Notably, gliomas with upregulated MN1 have better overall survival (OS) and progression-free survival (PFS). IGFBP5 expression associated inversely with MN1 expression levels in gliomas but correlated positively with IGF1 expression in only LGGs. This suggests a potential grade-specific interplay between repressive and activating roles of MN1 and IGF1, respectively, in the regulation of IGFBP5. Thus, MN1 overexpression, a promising predictor of OS and PFS in gliomas, may serve as a prognostic biomarker in clinical practice to categorize patients with survival advantage.
Collapse
Affiliation(s)
- Masum Saini
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Ajaya Nand Jha
- Max Super Specialty Hospital, 1, Press Enclave Road, Saket, New Delhi 110017, India
| | - Rajiv Tangri
- Max Super Specialty Hospital, 1, Press Enclave Road, Saket, New Delhi 110017, India
- Dr. Lal PathLabs, National Reference Laboratory, Sector 18, Rohini, New Delhi 110085, India
| | - Md Qudratullah
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sher Ali
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Department of Life Sciences, SBSR, Sharda University, KP-III, Greater Noida 201310, India
| |
Collapse
|
13
|
Aloizou AM, Pateraki G, Siokas V, Mentis AFA, Liampas I, Lazopoulos G, Kovatsi L, Mitsias PD, Bogdanos DP, Paterakis K, Dardiotis E. The role of MiRNA-21 in gliomas: Hope for a novel therapeutic intervention? Toxicol Rep 2020; 7:1514-1530. [PMID: 33251119 PMCID: PMC7677650 DOI: 10.1016/j.toxrep.2020.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Gliomas are the most common primary brain tumors in adults. They are generally very resistant to treatment and are therefore associated with negative outcomes. MicroRNAs (miRNAs) are small, non-coding RNA molecules that affect many cellular processes by regulating gene expression and, post-transcriptionally, the translation of mRNAs. MiRNA-21 has been consistently shown to be upregulated in glioma and research has shown that it is involved in a wide variety of biological pathways, promoting tumor cell survival and invasiveness. Furthermore, it has been implicated in resistance to treatment, both against chemotherapy and radiotherapy. In this review, we gathered the existent data on miRNA-21 and gliomas, in terms of its expression levels, association with grade and prognosis, the pathways it involves and its targets in glioma, and finally how it leads to treatment resistance. Furthermore, we discuss how this knowledge could be applied in clinical practice in the years to come. To our knowledge, this is the first review to assess in extent and depth the role of miRNA-21 in gliomas.
Collapse
Affiliation(s)
- Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Georgia Pateraki
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Alexios-Fotios A Mentis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece.,Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
| | - Ioannis Liampas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - George Lazopoulos
- Department of Cardiothoracic Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Panayiotis D Mitsias
- Department of Neurology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, University General Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis 40500, Larissa, Greece
| | - Konstantinos Paterakis
- Department of Neurosurgery, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| |
Collapse
|
14
|
Mollinedo F, Gajate C. Lipid rafts as signaling hubs in cancer cell survival/death and invasion: implications in tumor progression and therapy: Thematic Review Series: Biology of Lipid Rafts. J Lipid Res 2020; 61:611-635. [PMID: 33715811 PMCID: PMC7193951 DOI: 10.1194/jlr.tr119000439] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cholesterol/sphingolipid-rich membrane domains, known as lipid rafts or membrane rafts, play a critical role in the compartmentalization of signaling pathways. Physical segregation of proteins in lipid rafts may modulate the accessibility of proteins to regulatory or effector molecules. Thus, lipid rafts serve as sorting platforms and hubs for signal transduction proteins. Cancer cells contain higher levels of intracellular cholesterol and lipid rafts than their normal non-tumorigenic counterparts. Many signal transduction processes involved in cancer development (insulin-like growth factor system and phosphatidylinositol 3-kinase-AKT) and metastasis [cluster of differentiation (CD)44] are dependent on or modulated by lipid rafts. Additional proteins playing an important role in several malignant cancers (e.g., transmembrane glycoprotein mucin 1) are also being detected in association with lipid rafts, suggesting a major role of lipid rafts in tumor progression. Conversely, lipid rafts also serve as scaffolds for the recruitment and clustering of Fas/CD95 death receptors and downstream signaling molecules leading to cell death-promoting raft platforms. The partition of death receptors and downstream signaling molecules in aggregated lipid rafts has led to the formation of the so-called cluster of apoptotic signaling molecule-enriched rafts, or CASMER, which leads to apoptosis amplification and can be pharmacologically modulated. These death-promoting rafts can be viewed as a linchpin from which apoptotic signals are launched. In this review, we discuss the involvement of lipid rafts in major signaling processes in cancer cells, including cell survival, cell death, and metastasis, and we consider the potential of lipid raft modulation as a promising target in cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain. mailto:
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain
| |
Collapse
|
15
|
Park AK, Kim P, Ballester LY, Esquenazi Y, Zhao Z. Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma. Neuro Oncol 2020; 21:59-70. [PMID: 30053126 DOI: 10.1093/neuonc/noy120] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background A high heterogeneity and activation of multiple oncogenic pathways have been implicated in failure of targeted therapies in glioblastoma (GBM). Methods Using The Cancer Genome Atlas data, we identified subtype-specific prognostic core genes by a combined approach of genome-wide Cox regression and Gene Set Enrichment Analysis. The results were validated with 8 combined public datasets containing 608 GBMs. We further examined prognostic chromosome aberrations and mutations. Results In classical and mesenchymal subtypes, 2 receptor tyrosine kinases (RTKs) (MET and IGF1R), and the genes in RTK downstream pathways such as phosphatidylinositol-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and nuclear factor-kappaB (NF-kB), were commonly detected as prognostic core genes. Classical subtype-specific prognostic core genes included those in cell cycle, DNA repair, and the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. Immune-related genes were enriched in the prognostic genes showing negative promoter cytosine-phosphate-guanine (CpG) methylation/expression correlations. Mesenchymal subtype-specific prognostic genes were those related to mesenchymal cell movement, PI3K/Akt, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), Wnt/β-catenin, and Wnt/Ca2+ pathways. In copy number alterations and mutations, 6p loss and TP53 mutation were associated with poor and good survival, respectively, in the classical subtype. In the mesenchymal subtype, patients with PIK3R1 or PCLO mutations showed poor prognosis. In the glioma CpG island methylator phenotype (G-CIMP) subtype, patients harboring 10q loss, 12p gain, or 14q loss exhibited poor survival. Furthermore, 10q loss was significantly associated with the recently recognized G-CIMP subclass showing relatively low CpG methylation and poor prognosis. Conclusion These subtype-specific alterations have promising potentials as new prognostic biomarkers and therapeutic targets combined with surrogate markers of GBM subtypes. However, considering the small number of events, the results of copy number alterations and mutations require further validations.
Collapse
Affiliation(s)
- Ae Kyung Park
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea.,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Pora Kim
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
16
|
Chen KC, Chen PH, Ho KH, Shih CM, Chou CM, Cheng CH, Lee CC. IGF-1-enhanced miR-513a-5p signaling desensitizes glioma cells to temozolomide by targeting the NEDD4L-inhibited Wnt/β-catenin pathway. PLoS One 2019; 14:e0225913. [PMID: 31805126 PMCID: PMC6894868 DOI: 10.1371/journal.pone.0225913] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022] Open
Abstract
Temozolomide (TMZ) is a first-line alkylating agent for glioblastoma multiforme (GBM). Clarifying the mechanisms inducing TMZ insensitivity may be helpful in improving its therapeutic effectiveness against GBM. Insulin-like growth factor (IGF)-1 signaling and micro (mi)RNAs are relevant in mediating GBM progression. However, their roles in desensitizing GBM cells to TMZ are still unclear. We aimed to identify IGF-1-mediated miRNA regulatory networks that elicit TMZ insensitivity for GBM. IGF-1 treatment attenuated TMZ cytotoxicity via WNT/β-catenin signaling, but did not influence glioma cell growth. By miRNA array analyses, 93 upregulated and 148 downregulated miRNAs were identified in IGF-1-treated glioma cells. miR-513a-5p from the miR-513a-2 gene locus was upregulated by IGF-1-mediated phosphoinositide 3-kinase (PI3K) signaling. Its elevated levels were also observed in gliomas versus normal cells, in array data of The Cancer Genome Atlas (TCGA), and the GSE61710, GSE37366, and GSE41032 datasets. In addition, lower levels of neural precursor cell-expressed developmentally downregulated 4-like (NEDD4L), an E3 ubiquitin protein ligase that inhibits WNT signaling, were found in gliomas by analyzing cells, arrays, and RNA sequencing data of TCGA glioma patients. Furthermore, a negative correlation was identified between miR-513a-5p and NEDD4L in glioma. NEDD4L was also validated as a direct target gene of miR-513a-5p, and it was reduced by IGF-1 treatment. Overexpression of NEDD4L inhibited glioma cell viability and reversed IGF-1-repressed TMZ cytotoxicity. In contrast, miR-513a-5p significantly affected NEDD4L-inhibited WNT signaling and reduced TMZ cytotoxicity. These findings demonstrate a distinct role of IGF-1 signaling through miR-513a-5p-inhibited NEDD4L networks in influencing GBM's drug sensitivity to TMZ.
Collapse
Affiliation(s)
- Ku-Chung Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Peng-Hsu Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chwen-Ming Shih
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Chou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Hsiung Cheng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Cheng Lee
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Xi JJ, He RY, Zhang JK, Cai ZB, Zhuang RX, Zhao YM, Shao YD, Pan XW, Shi TT, Dong ZJ, Liu SR, Kong LM. Design, synthesis, and biological evaluation of novel 3-(thiophen-2-ylthio)pyridine derivatives as potential multitarget anticancer agents. Arch Pharm (Weinheim) 2019; 352:e1900024. [PMID: 31338897 DOI: 10.1002/ardp.201900024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022]
Abstract
A series of novel 3-(thiophen-2-ylthio)pyridine derivatives as insulin-like growth factor 1 receptor (IGF-1R) inhibitors was designed and synthesized. IGF-1R kinase inhibitory activities and cytotoxicities against HepG2 and WSU-DLCL2 cell lines were tested. For all of these compounds, potent cancer cell proliferation inhibitory activities were observed, but not through the inhibition of IGR-1R. Selected compounds were further screened against various kinases. Typical compound 22 (50% inhibitory concentration [IC50 ] values, HepG2: 2.98 ± 1.11 μM and WSU-DLCL2: 4.34 ± 0.84 μM) exhibited good inhibitory activities against fibroblast growth factor receptor-2 (FGFR2), FGFR3, epidermal growth factor receptor, Janus kinase, and RON (receptor originated from Nantes), with IC50 values ranging from 2.14 to 12.20 μM. Additionally, the cell-cycle analysis showed that compound 22 could arrest HepG2 cells in the G1/G0 phase. Taken together, all the experiments confirmed that the compounds in this series were multitarget anticancer agents worth further optimizing.
Collapse
Affiliation(s)
- Jian-Jun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Ruo-Yu He
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Jian-Kang Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Zhao-Bin Cai
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Rang-Xiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Yan-Mei Zhao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Yi-Dan Shao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Xu-Wang Pan
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Ting-Ting Shi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Zuo-Jun Dong
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang Province, China
| | - Shou-Rong Liu
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, China
| | - Li-Min Kong
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
18
|
Zhang J, Xi J, He R, Zhuang R, Kong L, Fu L, Zhao Y, Zhang C, Zeng L, Lu J, Tao R, Liu Z, Zhu H, Liu S. Discovery of 3-(thiophen/thiazole-2-ylthio)pyridine derivatives as multitarget anticancer agents. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02400-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
19
|
Seliger C, Genbrugge E, Gorlia T, Chinot O, Stupp R, Nabors B, Weller M, Hau P. Use of metformin and outcome of patients with newly diagnosed glioblastoma: Pooled analysis. Int J Cancer 2019; 146:803-809. [PMID: 30980539 DOI: 10.1002/ijc.32337] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/11/2019] [Accepted: 03/18/2019] [Indexed: 01/03/2023]
Abstract
Metformin has been linked to improve survival of patients with various cancers. There is little information on survival of glioblastoma patients after use of metformin. We assessed the association between metformin use and survival in a pooled analysis of patient data from 1,731 individuals from the randomized AVAglio, CENTRIC and CORE trials. We performed multivariate Cox analyses for overall survival (OS) and progression-free survival (PFS) comparing patients' use of metformin at baseline and/or during concomitant radiochemotherapy (TMZ/RT). Further exploratory analyses investigated the effect of metformin with a history of diabetes and nonfasting glucose levels in relation to OS or PFS of glioblastoma patients. Metformin alone or in any combination was not significantly associated with OS or PFS (at baseline, hazard ratio [HR] for OS = 0.87; 95% confidence interval [CI] = 0.65-1.16; HR for PFS = 0.84; 95% CI = 0.64-1.10; during TMZ/RT HR for OS = 0.97; 95% CI = 0.68-1.38; HR for PFS = 1.02; 95% CI = 0.74-1.41). We found a statistically nonsignificant association of metformin monotherapy with glioblastoma survival at baseline (HR for OS = 0.68; 95% CI = 0.42-1.10; HR for PFS = 0.57; 95% CI = 0.36-0.91), but not during the TMZ/RT period (HR for OS = 0.90; 95% CI = 0.51-1.56; HR for PFS = 1.05; 95% CI = 0.64-1.73). Diabetes mellitus or increased nonfasting glucose levels were not associated with a difference in OS or PFS in our selected study population. Metformin did not prolong survival of patients with newly diagnosed glioblastoma in our analysis. Additional studies may identify patients with specific tumor characteristics that are associated with potential benefit from treatment with metformin, possibly due to metabolic vulnerabilities.
Collapse
Affiliation(s)
- Corinna Seliger
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | | | | | - Olivier Chinot
- Aix-Marseille University, APHM, CNRS, INP, Institute of Neurophysiopathology, CHU Timone, Service de Neuro-Oncologie, Marseille, France
| | - Roger Stupp
- Malnati Brain Tumor Institute of the Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Burt Nabors
- Department of Neurology and Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | | |
Collapse
|
20
|
Wang Y, Sun Y, Tang J, Zhou W, Liu X, Bi Y, Zhang ZJ. Does diabetes decrease the risk of glioma? A systematic review and meta-analysis of observational studies. Ann Epidemiol 2019; 30:22-29.e3. [PMID: 30545764 DOI: 10.1016/j.annepidem.2018.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/02/2018] [Accepted: 11/18/2018] [Indexed: 01/11/2023]
Abstract
PURPOSE Increasing epidemiologic evidence suggests that diabetes mellitus (DM) may be associated with a decreased risk of glioma. This systematic review assessed whether DM was associated with glioma risk. METHODS Electronic searches were performed in PubMed, Web of Science, EMBASE, and Cochrane Library databases up to August 30, 2018. A random-effects model was performed to calculate summary effect size with corresponding 95% confidence intervals (CIs). RESULTS In total, 10 studies (eight case-control studies and two cohort studies) matched the inclusion criteria. Meta-analyses of case-control studies showed that DM decreased the risk of glioma by 23% (odds ratio: 0.77, 95% CI: 0.61-0.96; P = .02, I2 = 82.0%). However, no such effect was observed in cohort studies (relative risk: 0.71, 95% CI: 0.10-4.80; P = .72, I2 = 61.6%). In the subgroup analyses, DM was associated with a decreased risk of glioma in Caucasians but not in Asians; the inverse association was slightly higher in males than in females. CONCLUSIONS Our results indicate that DM decreases the risk of glioma, but the inverse association may vary in subgroups. The present conclusions should be confirmed with further studies.
Collapse
Affiliation(s)
- Yongbo Wang
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Yi Sun
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Juan Tang
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Wei Zhou
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Xiaoxue Liu
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Yongyi Bi
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China
| | - Zhi-Jiang Zhang
- Department of Epidemiology and Biostatistics, School of Health Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Abstract
The important role of insulin-like growth factor 1 receptor (IGF-1R) in malignant tumors has been well established. Increased IGF-1R activity promotes cancer cell proliferation, migration, and invasion and is associated with tumor metastasis, treatment resistance, poor prognosis, and shortened survival in patients with cancer. However, while IGF-1R has become a promising target for cancer therapy, IGF-1R-targeted therapy is ineffective in unselected patients. It is therefore essential to evaluate IGF-1R expression before treatment in order to identify responsive patients, monitor therapy efficacy, and estimate prognosis. Insulin-like growth factor 1 receptor molecular imaging is an optimal method for assessing the expression of IGF-1R in vivo accurately and noninvasively. In this review, we will summarize the current status of IGF-1R molecular imaging in cancer, in which 5 major classes of ligands that have been developed for noninvasive IGF-1R molecular imaging will be discussed: natural ligands, monoclonal antibodies, antibody fragments, affibodies, and small molecules. For decades, IGF-1R molecular imaging is studied in full swing and more effort is needed in the future.
Collapse
Affiliation(s)
- Yingying Sun
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xilin Sun
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,3 Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Baozhong Shen
- 1 Molecular Imaging Research Center, Harbin Medical University, Harbin, Heilongjiang, China.,2 TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
22
|
Dahlhaus A, Prengel P, Spector L, Pieper D. Birth weight and subsequent risk of childhood primary brain tumors: An updated meta-analysis. Pediatr Blood Cancer 2017; 64. [PMID: 27804208 DOI: 10.1002/pbc.26299] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/04/2016] [Accepted: 09/08/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND Primary brain tumors are common in childhood, but the etiology is largely unclear. As studies on birth weight as a risk factor for the occurrence of histologically specified tumors have been inconclusive, we decided to update a 2008 meta-analysis on the subject. METHODS A search strategy was performed in Medline and EMBASE for the period 2007-2016. We included six new studies and performed further subgroup analyses for medulloblastoma and primitive neuroectodermal tumors (PNETs). Dichotomous analyses were performed for low (2,500 g) and high birth weight (4,000 g cutoff point). RESULTS Our results confirmed that high birth weight increases the risk of astrocytoma (odds ratio [OR] = 1.60, 95% confidence interval [CI]: 1.23-2.09) and medulloblastoma/PNET. However, subgroup analysis revealed an increased risk of medulloblastoma (OR = 1.31, 95% CI: 1.08-1.58) but not of PNET (OR = 1.16, 95% CI: 0.92-1.46). Low birth weight was associated with an increased risk of medulloblastoma/PNET. Subgroup analysis for medulloblastoma and PNET revealed increased risk but CIs included zero. Neither low nor high birth weight was associated with the risk of ependymoma. CONCLUSIONS While an association between high birth weight and astrocytoma was confirmed, more studies are needed to investigate medulloblastoma and PNET risk in children with high and low birth weight.
Collapse
Affiliation(s)
- Anne Dahlhaus
- Institute of General Practice, Goethe-University Frankfurt, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peggy Prengel
- Institute for Research in Operative Medicine, Witten/Herdecke University, Cologne, Germany
| | - Logan Spector
- Division of Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Dawid Pieper
- Institute for Research in Operative Medicine, Witten/Herdecke University, Cologne, Germany
| |
Collapse
|
23
|
The microRNA-302b-inhibited insulin-like growth factor-binding protein 2 signaling pathway induces glioma cell apoptosis by targeting nuclear factor IA. PLoS One 2017; 12:e0173890. [PMID: 28323865 PMCID: PMC5360322 DOI: 10.1371/journal.pone.0173890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/28/2017] [Indexed: 02/02/2023] Open
Abstract
MicroRNAs are small noncoding RNAs that post-transcriptionally control the expression of genes involved in glioblastoma multiforme (GBM) development. Although miR-302b functions as a tumor suppressor, its role in GBM is still unclear. Therefore, this study comprehensively explored the roles of miR-302b-mediated gene networks in GBM cell death. We found that miR-302b levels were significantly higher in primary astrocytes than in GBM cell lines. miR-302b overexpression dose dependently reduced U87-MG cell viability and induced apoptosis through caspase-3 activation and poly(ADP ribose) polymerase degradation. A transcriptome microarray revealed 150 downregulated genes and 380 upregulated genes in miR-302b-overexpressing cells. Nuclear factor IA (NFIA), higher levels of which were significantly related to poor survival, was identified as a direct target gene of miR-302b and was involved in miR-302b-induced glioma cell death. Higher NFIA levels were observed in GBM cell lines and human tumor sections compared with astrocytes and non-tumor tissues, respectively. NFIA knockdown significantly enhanced apoptosis. We found high levels of insulin-like growth factor-binding protein 2 (IGFBP2), another miR-302b-downregulated gene, in patients with poor survival. We verified that NFIA binds to the IGFBP2 promoter and transcriptionally enhances IGFBP2 expression levels. We identified that NFIA-mediated IGFBP2 signaling pathways are involved in miR-302b-induced glioma cell death. The identification of a regulatory loop whereby miR-302b inhibits NFIA, leading to a decrease in expression of IGFBP-2, may provide novel directions for developing therapies to target glioblastoma tumorigenesis.
Collapse
|
24
|
Abdolhoseinpour H, Mehrabi F, Shahraki K, Khoshnood RJ, Masoumi B, Yahaghi E, Goudarzi PK. Investigation of serum levels and tissue expression of two genes IGFBP-2 and IGFBP-3 act as potential biomarker for predicting the progression and survival in patients with glioblastoma multiforme. J Neurol Sci 2016; 366:202-206. [PMID: 27288807 DOI: 10.1016/j.jns.2016.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Identification of genetic copy number changes in glial tumors is of importance in the context of improved/refined diagnostic, prognostic procedures and therapeutic decision-making. Blood-derived biomarkers, therefore, would be useful as minimally invasive markers that could support diagnosis and enable monitoring of tumour growth and response to treatment. OBJECTIVE The aim of this study was to evaluate the clinical significance of IGFBP-2/3 in glioblastoma multiforme (GBM) and their value as predictors of survival. METHODS We examined the plasma levels of IGFBP-2 and IGFBP-3 using ELISA in patient suffering from GBM and controls groups. Furthermore, immunohistochemistry method was used to evaluate the expression levels of these markers. RESULTS Preoperative plasma levels of IGFBP-2 and IGFBP-3 were markedly higher in glioblastoma patients (mean±SD: 521.5±164.2ng/ml; 402.4±126ng/ml) when compared with healthy controls (301.28±73.12; 244±89.5ng/ml; p<0.001). Immunohistochemical results indicated that the median H score for glioblastoma tissues was higher when compared with normal tissues. The mean scores for IGFBP-2 expression in glioblastoma was higher than normal tissues (p<0.001). Our result showed that the median H score for glioblastoma tissues was higher when compared with normal tissue for IGFBP-3 expression. The mean scores for glioblastoma tissues was higher than normal tissues (p<0.001). We also evaluated whether plasma IGFBP-2 and IGFBP-3 levels were related to clinical features. The plasma IGFBP-2 level was strongly linked to the patient's age (R=0.769, P=0.001) that were strongly increased in patients with older age (>65), (mean±SD: 594.36±33.3ng/ml). On the other hand, plasma IGFBP-3 level was not correlated with age (P=0.462), sex (P=0.532), and tumor size (P=0.245). Our findings indicated that the tissue IGFBP-2 level was also markedly correlated with the patient's age (R=0.612, P=0.015). On the other hand, tissue IGFBP-3 expression level was not correlated with age (P=0.472), sex (P=0.512), and tumor size (P=0.241). Kaplan-Meier survival and log-rank analysis suggested that patients with high plasma level of IGFBP-2 and tissue expression of IGFBP-2 had shorter overall survival than those with low levels (log-rank test P=0.027; P<0.001). Kaplan-Meier survival and log-rank analysis suggested that patients with high plasma level of IGFBP-3 and tissue expression of IGFBP-3 had shorter overall survival than those with low levels groups (log-rank test P=0.018; P<0.001). CONCLUSION These data suggest that plasma levels and tissue levels of IGFBP-2 and IGFBP-3 may be as potential biomarkers for predicting the progression and survival in patients with GBM.
Collapse
Affiliation(s)
- Hesam Abdolhoseinpour
- Department of Neurosurgery, Bou Ali Hospital, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Farzad Mehrabi
- Department of Neurology, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Shahraki
- Department of Ophthalmology, Alzahra Eye Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Reza Jalili Khoshnood
- Department of Neurosurgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Masoumi
- Department of Emergency Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Emad Yahaghi
- Department of Molecular Biology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
25
|
Birth Size Characteristics and Risk of Brain Tumors in Early Adulthood: Results from a Swedish Cohort Study. Cancer Epidemiol Biomarkers Prev 2016; 25:678-85. [DOI: 10.1158/1055-9965.epi-15-1096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/13/2016] [Indexed: 11/16/2022] Open
|
26
|
Dai ZF, Huang QL, Liu HP. Different body mass index grade on the risk of developing glioma: a meta-analysis. Chin Neurosurg J 2015. [DOI: 10.1186/s41016-015-0008-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
27
|
Yang CH, Li K, Pfeffer SR, Pfeffer LM. The Type I IFN-Induced miRNA, miR-21. Pharmaceuticals (Basel) 2015; 8:836-47. [PMID: 26610525 PMCID: PMC4695812 DOI: 10.3390/ph8040836] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/10/2015] [Accepted: 11/20/2015] [Indexed: 01/21/2023] Open
Abstract
The interferon (IFN) family of cytokines not only has antiviral properties at various steps in the viral replication cycle, but also anticancer activity through multiple pathways that include inhibiting cell proliferation, regulating cellular responses to inducers of apoptosis and modulating angiogenesis and the immune system. IFNs are known to induce their biological activity through the induction of protein encoding IFN-stimulated genes. However, recent studies have established that IFNs also induce the expression of microRNAs (miRNAs), which are small endogenous non-coding RNAs that suppress gene expression at the post-transcriptional level. MiRNAs play critical roles in tumorigenesis and have been implicated to act as either oncogenes or tumor suppressors in various human cancers. Therefore, IFN-induced miRNAs play an important role, not only in the host response to innate immune response to cancer, but also in the tumorigenic process itself. Furthermore, IFN-induced miRNAs may participate in and/or orchestrate antiviral defense in certain viral infections. In this review, we describe our recent studies on the induction of miR-21 by type I IFN, the role of the STAT3 and NFκB signaling pathways in IFN-induced miR-21 expression, the role of miR-21 in different cancers and the role of miR-21 in regulating the antiviral response.
Collapse
Affiliation(s)
- Chuan He Yang
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
- Center for Cancer Research, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
| | - Kui Li
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, 858 Madison Avenue, Memphis, TN 38163, USA.
| | - Susan R Pfeffer
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
- Center for Cancer Research, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
- Center for Cancer Research, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
| |
Collapse
|
28
|
Maris C, D'Haene N, Trépant AL, Le Mercier M, Sauvage S, Allard J, Rorive S, Demetter P, Decaestecker C, Salmon I. IGF-IR: a new prognostic biomarker for human glioblastoma. Br J Cancer 2015; 113:729-37. [PMID: 26291053 PMCID: PMC4559821 DOI: 10.1038/bjc.2015.242] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/02/2015] [Accepted: 06/11/2015] [Indexed: 01/18/2023] Open
Abstract
Background: Glioblastomas (GBMs) are the most common malignant primary brain tumours in adults and are refractory to conventional therapy, including surgical resection, radiotherapy and chemotherapy. The insulin-like growth factor (IGF) system is a complex network that includes ligands (IGFI and IGFII), receptors (IGF-IR and IGF-IIR) and high-affinity binding proteins (IGFBP-1 to IGFBP-6). Many studies have reported a role for the IGF system in the regulation of tumour cell biology. However, the role of this system remains unclear in GBMs. Methods: We investigate the prognostic value of both the IGF ligands' and receptors' expression in a cohort of human GBMs. Tissue microarray and image analysis were conducted to quantitatively analyse the immunohistochemical expression of these proteins in 218 human GBMs. Results: Both IGF-IR and IGF-IIR were overexpressed in GBMs compared with normal brain (P<10−4 and P=0.002, respectively). Moreover, with regard to standard clinical factors, IGF-IR positivity was identified as an independent prognostic factor associated with shorter survival (P=0.016) and was associated with a less favourable response to temozolomide. Conclusions: This study suggests that IGF-IR could be an interesting target for GBM therapy.
Collapse
Affiliation(s)
- C Maris
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - N D'Haene
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - A-L Trépant
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - M Le Mercier
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - S Sauvage
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), Académie Universitaire Wallonie-Bruxelles, Gosselies 6041, Belgium
| | - J Allard
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - S Rorive
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging (CMMI), Académie Universitaire Wallonie-Bruxelles, Gosselies 6041, Belgium
| | - P Demetter
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - C Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), Académie Universitaire Wallonie-Bruxelles, Gosselies 6041, Belgium.,Laboratories of Image, Signal processing and Acoustics (LISA), Brussels School of Engineering/Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Brussels 1050, Belgium
| | - I Salmon
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging (CMMI), Académie Universitaire Wallonie-Bruxelles, Gosselies 6041, Belgium
| |
Collapse
|
29
|
Zhou X, Shen F, Ma P, Hui H, Pei S, Chen M, Wang Z, Zhou W, Jin B. GSK1838705A, an IGF-1R inhibitor, inhibits glioma cell proliferation and suppresses tumor growth in vivo. Mol Med Rep 2015; 12:5641-6. [PMID: 26238593 PMCID: PMC4581800 DOI: 10.3892/mmr.2015.4129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 05/08/2015] [Indexed: 12/14/2022] Open
Abstract
Glioma is a type of primary malignant tumor of the central nervous system in humans. At present, standard treatment involves surgical resection, followed by radiation therapy and chemotherapy. However, the prognosis is poor and the long-term survival rate remains low. An improved understanding of the molecular basis for glioma tumorigenesis is in urgently required. The pro-survival effect of the insulin-like growth factor (IGF) signaling pathway has been implicated in progression of the glioma disease state. GSK1838705A is a novel, small molecule kinase inhibitor of IGF-IR, which inhibits IGF signal transduction and downstream target activation. Its anti-proliferative activity has been demonstrated in various tumor cell lines. The present study investigated the potential use of GSK1838705A for the treatment of glioma. Human U87MG glioma cells were used to examine the inhibitory activity of GSK1838705A in cell proliferation, migration and apoptosis. The antitumor activity of GSK1838705A was assessed in a xenograft mouse model. GSK1838705A inhibited the growth and induced the apoptosis of the U87MG glioma cells in a dose-dependent manner. The GSK1838705A-treated cells exhibited reduced migratory activity in response to chemoattractants. The present study further demonstrated the antitumor activity of GSK1838705A in vivo. The administration of GSK1838705A significantly inhibited the growth of glioma tumors by inducing the apoptosis of tumor cells. These results suggested that targeting IGF signaling with GSK1838705A may be a promising therapeutic strategy for the treatment of patients with glioma.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Fazheng Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Pengju Ma
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Hongyan Hui
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Sujuan Pei
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Ming Chen
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Zhongwei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Wenke Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Baozhe Jin
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| |
Collapse
|
30
|
Seliger C, Ricci C, Meier CR, Bodmer M, Jick SS, Bogdahn U, Hau P, Leitzmann MF. Diabetes, use of antidiabetic drugs, and the risk of glioma. Neuro Oncol 2015; 18:340-9. [PMID: 26093337 DOI: 10.1093/neuonc/nov100] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/07/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Prior epidemiologic studies suggest inverse relations between diabetes and glioma risk, but the underlying mechanisms, including use of antidiabetic drugs, are unknown. METHODS We therefore performed a matched case-control analysis using the Clinical Practice Research Datalink (CPRD). We identified incident glioma cases diagnosed between 1995 and 2012 and matched each case with 10 controls on age, gender, calendar time, general practice, and years of active history in the CPRD. We performed conditional logistic regression to estimate odds ratios (ORs) with 95% CIs, adjusted for body mass index and smoking. RESULTS We identified 2005 cases and 20 050 controls. Diabetes was associated with decreased risk of glioma (OR = 0.74; 95% CI = 0.60-0.93), particularly glioblastoma (OR = 0.69; 95% CI = 0.51-0.94). Glioblastoma risk reduction was markedly pronounced among diabetic men (OR = 0.60; 95% CI = 0.40-0.90), most apparently for those with diabetes of long-term duration (OR for >5 vs 0 y = 0.46; 95% CI = 0.26-0.82) or poor glycemic control (OR for HbA1c ≥ 8 vs <6.5% = 0.20; 95% CI = 0.06-0.70). In contrast, the effect of diabetes on glioblastoma risk was absent among women (OR = 0.85; 95% CI = 0.53-1.36). No significant associations with glioma were found for use of metformin (OR for ≥ 30 vs 0 prescriptions = 0.72; 95% CI = 0.38-1.39), sulfonylureas (OR = 0.71; 95% CI = 0.39-1.30), or insulin (OR = 0.79; 95% CI = 0.37-1.69). CONCLUSIONS Antidiabetic treatment appears to be unrelated to glioma, but long-term diabetes duration and increased HbA1c both show decreased glioma risk. Stronger findings in men than women suggest low androgen levels concurrent with diabetes as a biologic mechanism.
Collapse
Affiliation(s)
- Corinna Seliger
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Cristian Ricci
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Christoph R Meier
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Michael Bodmer
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Susan S Jick
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Ulrich Bogdahn
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| | - Michael F Leitzmann
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany (C.S., U.B., P.H.); Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany (C.R., M.F.L.); Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland (C.R.M., M.B.); Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, Boston, Massachusetts, (C.R.M., S.S.J.); Hospital Pharmacy, University Hospital Basel, Basel, Switzerland (C.R.M.); Department of General Internal Medicine, University of Bern, Inselspital/Universitätsspital, Bern, Switzerland (M.B.)
| |
Collapse
|
31
|
Pfeffer SR, Yang CH, Pfeffer LM. The Role of miR-21 in Cancer. Drug Dev Res 2015; 76:270-7. [PMID: 26082192 DOI: 10.1002/ddr.21257] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are small endogenous noncoding RNAs that suppress gene expression at the post-transcriptional level. In the past decade, miRNAs have been extensively studied in a number of different human cancers. MiRNAs have been identified to act both as oncogenes and as tumor suppressors. In addition, miRNAs are associated with the intrinsic resistance of cancer to various forms of therapy, and they are implicated in both tumor progression and metastasis. The characterization of the specific alterations in the patterns of miRNA expression in cancer has great potential for identifying biomarkers for early cancer detection, as well as for potential therapeutic intervention in cancer treatment. In this chapter, we describe the ever-expanding role of miR-21 and its target genes in different cancers, and provide insight into how this oncogenic miRNA regulates cancer cell proliferation, migration, and apoptosis by suppressing the expression of tumor suppressors.
Collapse
Affiliation(s)
- Susan R Pfeffer
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Chuan He Yang
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
32
|
Zhou Q, Zhang J, Cui Q, Li X, Gao G, Wang Y, Xu Y, Gao X. GSK1904529A, an insulin-like growth factor-1 receptor inhibitor, inhibits glioma tumor growth, induces apoptosis and inhibits migration. Mol Med Rep 2015; 12:3381-3385. [PMID: 26035416 PMCID: PMC4526077 DOI: 10.3892/mmr.2015.3869] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 03/26/2015] [Indexed: 12/27/2022] Open
Abstract
Malignant gliomas are the most common type of primary malignancy of the central nervous system, with a poor prognosis. The therapeutic options for malignant gliomas are limited and far from satisfactory, and novel treatment strategies are urgently required to improve the outcome of the disease. Insulin-like growth factor (IGF)/IGF-1 receptor (IGF-1R) signaling pathway regulates cell proliferation, motility and survival. The dysregulation of this signaling pathway has been implicated in the development of malignant gliomas. In the present study, GSK1904529A, a small molecule inhibitor of IGF-1R, suppressed glioma cell viability, induced glioma cell apoptosis and inhibited glioma cell migration in vitro. In addition, GSK1904529A inhibited glioma tumor growth and induced tumor cell apoptosis in vivo. In conclusion, the results of the present study suggested GSK1904529A as a promising agent for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Pathology, Children's Hospital of Zhengzhou, Zhengzhou, Henan 450052, P.R. China
| | - Junxia Zhang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Qinying Cui
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaodong Li
- Department of Ophthalmology, People's Hospital of Zhengzhou, Zhengzhou, Henan 450012, P.R. China
| | - Ge Gao
- Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yanfen Wang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuping Xu
- Department of Dermatology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xiaoqun Gao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
33
|
Lin KW, Liao A, Qutub AA. Simulation predicts IGFBP2-HIF1α interaction drives glioblastoma growth. PLoS Comput Biol 2015; 11:e1004169. [PMID: 25884993 PMCID: PMC4401766 DOI: 10.1371/journal.pcbi.1004169] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/02/2015] [Indexed: 12/21/2022] Open
Abstract
Tremendous strides have been made in improving patients’ survival from cancer with one glaring exception: brain cancer. Glioblastoma is the most common, aggressive and highly malignant type of primary brain tumor. The average overall survival remains less than 1 year. Notably, cancer patients with obesity and diabetes have worse outcomes and accelerated progression of glioblastoma. The root cause of this accelerated progression has been hypothesized to involve the insulin signaling pathway. However, while the process of invasive glioblastoma progression has been extensively studied macroscopically, it has not yet been well characterized with regards to intracellular insulin signaling. In this study we connect for the first time microscale insulin signaling activity with macroscale glioblastoma growth through the use of computational modeling. Results of the model suggest a novel observation: feedback from IGFBP2 to HIF1α is integral to the sustained growth of glioblastoma. Our study suggests that downstream signaling from IGFI to HIF1α, which has been the target of many insulin signaling drugs in clinical trials, plays a smaller role in overall tumor growth. These predictions strongly suggest redirecting the focus of glioma drug candidates on controlling the feedback between IGFBP2 and HIF1α. Current treatment for glioblastoma patients is limited to nonspecific methods: surgery followed by a combination of radio- and chemotherapy. With these methods, glioma patient survival is less than one year post-diagnosis. Targeting specific protein signaling pathways offers potentially more potent therapies. One promising potential target is the insulin signaling pathway, which is known to contribute to glioblastoma progression. However, drugs targeting this pathway have shown mixed results in clinical trials, and the detailed mechanisms of how the insulin signaling pathway promotes glioblastoma growth remain to be elucidated. Here, we developed a computational model of insulin signaling in glioblastoma in order to study this pathway’s role in tumor progression. Using the model, we systematically test contributions of different insulin signaling protein interactions on glioblastoma growth. Our model highlights a key driver for the growth of glioblastoma: IGFBP2-HIF1α feedback. This interaction provides a target that could open the door for new therapies in glioma and other solid tumors.
Collapse
Affiliation(s)
- Ka Wai Lin
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Angela Liao
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Amina A. Qutub
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
34
|
Abstract
Objective: To review the mechanisms of anti-cancer activity of fenofibrate (FF) and other Peroxisome Proliferator Activator Receptor α (PPARα) agonists based on evidences reported in the published literature.Methods: We extensively reviewed the literature concerning FF as an off target anti-cancer drug. Controversies regarding conflicting findings were also addressed.Results: The main mechanism involved in anti-cancer activity is anti-angiogenesis through down-regulation of Vascular Endothelial Growth Factor (VEGF), Vascular Endothelial Growth Factor Receptor (VEGFR) and Hypoxia Inducible factor-1 α (HIF-1α), inhibition of endothelial cell migration, up-regulation of endostatin and thrombospondin-1, but there are many other contributing mechanisms like apoptosis and cell cycle arrest, down-regulation of Nuclear Factor Kappa B (NF-kB) and Protein kinase B (Akt) and decrease of cellular energy by impairing mitochondrial function. Growth impairment is related to down-regulation of Phospho-Inositol 3 Kinase (PI3K)/Akt axis and down-regulation of the p38 map kinase (MAPK) cascade. A possible role should be assigned to FF stimulated over-expression of Tribbles Homolog-3 (TRIB3) which inhibits Akt phosphorylation. Important anti-cancer and anti-metastatic activities are due to down-regulation of MCP-1 (monocyte chemotactic protein-1), decreased Metalloprotease-9 (MMP-9) production, weak down-regulation of adhesion molecules like E selectin, intercellular adhesion molecules (ICAM) and Vascular Endothelial Adhesion Molecules (VCAM), and decreased secretion of chemokines like Interleukin-6 (IL-6), and down-regulation of cyclin D-1. There is no direct link between FF activity in lipid metabolism and anticancer activity, except for the fact that many anticancer actions are dependent from PPARα agonism. FF exhibits also PPARα independent anti-cancer activities.Conclusions: There are strong evidences indicating that FF can disrupt growth-related activities in many different cancers, due to anti-angiogenesis and anti-inflammatory effects. Therefore FF may be useful as a complementary adjunct treatment of cancer, particularly included in anti-angiogenic protocols like those currently increasingly used in glioblastoma. There are sound reasons to initiate well planned phase II clinical trials for FF as a complementary adjunct treatment of cancer.
Collapse
|
35
|
Zhang D, Jiang S, Meng H. Role of the Insulin-Like Growth Factor Type 1 Receptor in the Pathogenesis of Diabetic Encephalopathy. Int J Endocrinol 2015; 2015:626019. [PMID: 26089889 PMCID: PMC4451562 DOI: 10.1155/2015/626019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
Defective cognitive function is common in patients with diabetes, suggesting that insulin normally exerts anabolic actions in neuron, namely, diabetic encephalopathy. However, because insulin can cross-activate the insulin-like growth factor type 1 receptor (IGF-1R), which also functions in most of tissues, such as muscle and bone, it has been difficult to establish the direct (IGF-1-independent) actions of insulin in the pathogenesis of diabetic encephalopathy. To overcome this problem, we examined insulin signaling and action in primary PC-12 cells engineered for conditional disruption of the IGF-1 receptor (ΔIGF-1R). The results showed that the lower glucose metabolism and high expression of IGF-1R occurred in the brain of the DE rat model. The results also showed the defect of IGF-1R could significantly improve the ability of glucose consumption and enhance sensitivity to insulin-induced IR and Akt phosphorylation in PC12 cells. And meanwhile, IGF-1R allele gene knockout (IGF-1R(neo)) mice treated with HFD/STZ had better cognitive abilities than those of wild mice. Those results indicate that insulin exerts direct anabolic actions in neuron-like cells by activation of its cognate receptor and prove that IGF-1R plays an important role in the pathogenesis of diabetic encephalopathy.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Radiology, Affiliated Hospital of BeiHua University, JiLin 132011, China
| | - Shuang Jiang
- College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Heng Meng
- Department of Radiology, Affiliated Hospital of BeiHua University, JiLin 132011, China
- *Heng Meng:
| |
Collapse
|
36
|
Zamykal M, Martens T, Matschke J, Günther HS, Kathagen A, Schulte A, Peters R, Westphal M, Lamszus K. Inhibition of intracerebral glioblastoma growth by targeting the insulin-like growth factor 1 receptor involves different context-dependent mechanisms. Neuro Oncol 2014; 17:1076-85. [PMID: 25543125 DOI: 10.1093/neuonc/nou344] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/24/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Signaling by insulin-like growth factor 1 receptor (IGF-1R) can contribute to the formation and progression of many diverse tumor types, including glioblastoma. We investigated the effect of the IGF-1R blocking antibody IMC-A12 on glioblastoma growth in different in vivo models. METHODS U87 cells were chosen to establish rapidly growing, angiogenesis-dependent tumors in the brains of nude mice, and the GS-12 cell line was used to generate highly invasive tumors. IMC-A12 was administered using convection-enhanced local delivery. Tumor parameters were quantified histologically, and the functional relevance of IGF-1R activation was analyzed in vitro. RESULTS IMC-A12 treatment inhibited the growth of U87 and GS-12 tumors by 75% and 50%, respectively. In GS-12 tumors, the invasive tumor extension and proliferation rate were significantly reduced by IMC-A12 treatment, while apoptosis was increased. In IMC-A12-treated U87 tumors, intratumoral vascularization was markedly decreased, and tumor cell proliferation was moderately reduced. Flow cytometry showed that <2% of U87 cells but >85% of GS-12 cells expressed IGF-1R. Activation of IGF-1R by IGF-1 and IGF-2 in GS-12 cells was blocked by IMC-A12. Both ligands stimulated GS-12 cell proliferation, and IGF-2 also stimulated migration. IMC-A12 inhibited these stimulatory effects and increased apoptosis. In U87 cells, stimulation with either ligand had no functional effect. CONCLUSIONS IGF-1R blockade can inhibit glioblastoma growth by different mechanisms, including direct effects on the tumor cells as well as indirect anti-angiogenic effects. Hence, blocking IGF-1R may be useful to target both the highly proliferative, angiogenesis-dependent glioblastoma core component as well as the infiltrative periphery.
Collapse
Affiliation(s)
- Martin Zamykal
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Tobias Martens
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Jakob Matschke
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Hauke S Günther
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Annegret Kathagen
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Alexander Schulte
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Regina Peters
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Manfred Westphal
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| | - Katrin Lamszus
- Department of Neurosurgery (M.Z., T.M., H.S.G., A.K., A.S., R.P., M.W., K.L.) and Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.M.)
| |
Collapse
|
37
|
Crump C, Sundquist J, Sieh W, Winkleby MA, Sundquist K. Perinatal and familial risk factors for brain tumors in childhood through young adulthood. Cancer Res 2014; 75:576-83. [PMID: 25511376 DOI: 10.1158/0008-5472.can-14-2285] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Perinatal factors, including high birth weight, have been associated with childhood brain tumors in case-control studies. However, the specific contributions of gestational age and fetal growth remain unknown, and these issues have never been examined in large cohort studies with follow-up into adulthood. We conducted a national cohort study of 3,571,574 persons born in Sweden in 1973-2008, followed up for brain tumor incidence through 2010 (maximum age 38 years) to examine perinatal and familial risk factors. There were 2,809 brain tumors in 69.7 million person-years of follow-up. After adjusting for potential confounders, significant risk factors for brain tumors included high fetal growth [incidence rate ratio (IRR) per additional 1 SD, 1.04; 95% confidence interval (CI), 1.01-1.08, P = 0.02], first-degree family history of a brain tumor (IRR, 2.43; 95% CI, 1.86-3.18, P < 0.001), parental country of birth (IRR for both parents born in Sweden vs. other countries, 1.21; 95% CI, 1.09-1.35, P < 0.001), and high maternal education level (Ptrend = 0.01). These risk factors did not vary by age at diagnosis. The association with high fetal growth appeared to involve pilocytic astrocytomas, but not other astrocytomas, medulloblastomas, or ependymomas. Gestational age at birth, birth order, multiple birth, and parental age were not associated with brain tumors. In this large cohort study, high fetal growth was associated with an increased risk of brain tumors (particularly pilocytic astrocytomas) independently of gestational age, not only in childhood but also into young adulthood, suggesting that growth factor pathways may play an important long-term role in the etiology of certain brain tumor subtypes.
Collapse
Affiliation(s)
- Casey Crump
- Department of Medicine, Stanford University, Stanford, California.
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden. Stanford Prevention Research Center, Stanford University, Stanford, California
| | - Weiva Sieh
- Department of Health Research and Policy, Stanford University, Stanford, California
| | - Marilyn A Winkleby
- Stanford Prevention Research Center, Stanford University, Stanford, California
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden. Stanford Prevention Research Center, Stanford University, Stanford, California
| |
Collapse
|
38
|
Azevedo H, Fujita A, Bando SY, Iamashita P, Moreira-Filho CA. Transcriptional network analysis reveals that AT1 and AT2 angiotensin II receptors are both involved in the regulation of genes essential for glioma progression. PLoS One 2014; 9:e110934. [PMID: 25365520 PMCID: PMC4217762 DOI: 10.1371/journal.pone.0110934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/26/2014] [Indexed: 01/25/2023] Open
Abstract
Gliomas are aggressive primary brain tumors with high infiltrative potential. The expression of Angiotensin II (Ang II) receptors has been associated with poor prognosis in human astrocytomas, the most common type of glioma. In this study, we investigated the role of Angiotensin II in glioma malignancy through transcriptional profiling and network analysis of cultured C6 rat glioma cells exposed to Ang II and to inhibitors of its membrane receptor subtypes. C6 cells were treated with Ang II and specific antagonists of AT1 and AT2 receptors. Total RNA was isolated after three and six hours of Ang II treatment and analyzed by oligonucleotide microarray technology. Gene expression data was evaluated through transcriptional network modeling to identify how differentially expressed (DE) genes are connected to each other. Moreover, other genes co-expressing with the DE genes were considered in these analyses in order to support the identification of enriched functions and pathways. A hub-based network analysis showed that the most connected nodes in Ang II-related networks exert functions associated with cell proliferation, migration and invasion, key aspects for glioma progression. The subsequent functional enrichment analysis of these central genes highlighted their participation in signaling pathways that are frequently deregulated in gliomas such as ErbB, MAPK and p53. Noteworthy, either AT1 or AT2 inhibitions were able to down-regulate different sets of hub genes involved in protumoral functions, suggesting that both Ang II receptors could be therapeutic targets for intervention in glioma. Taken together, our results point out multiple actions of Ang II in glioma pathogenesis and reveal the participation of both Ang II receptors in the regulation of genes relevant for glioma progression. This study is the first one to provide systems-level molecular data for better understanding the protumoral effects of Ang II in the proliferative and infiltrative behavior of gliomas.
Collapse
Affiliation(s)
- Hátylas Azevedo
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - André Fujita
- Department of Computer Science, Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvia Yumi Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Priscila Iamashita
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Carlos Alberto Moreira-Filho
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
39
|
Yang CH, Yue J, Pfeffer SR, Fan M, Paulus E, Hosni-Ahmed A, Sims M, Qayyum S, Davidoff AM, Handorf CR, Pfeffer LM. MicroRNA-21 promotes glioblastoma tumorigenesis by down-regulating insulin-like growth factor-binding protein-3 (IGFBP3). J Biol Chem 2014; 289:25079-87. [PMID: 25059666 DOI: 10.1074/jbc.m114.593863] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite advances in surgery, imaging, chemotherapy, and radiation, patients with glioblastoma multiforme (GBM), the most common histological subtype of glioma, have an especially dismal prognosis; >70% of GBM patients die within 2 years of diagnosis. In many human cancers, the microRNA miR-21 is overexpressed, and accumulating evidence indicates that it functions as an oncogene. Here, we report that miR-21 is overexpressed in human GBM cell lines and tumor tissue. Moreover, miR-21 expression in GBM patient samples is inversely correlated with patient survival. Knockdown of miR-21 in GBM cells inhibited cell proliferation in vitro and markedly inhibited tumor formation in vivo. A number of known miR-21 targets have been identified previously. By microarray analysis, we identified and validated insulin-like growth factor (IGF)-binding protein-3 (IGFBP3) as a novel miR-21 target gene. Overexpression of IGFBP3 in glioma cells inhibited cell proliferation in vitro and inhibited tumor formation of glioma xenografts in vivo. The critical role that IGFBP3 plays in miR-21-mediated actions was demonstrated by a rescue experiment, in which IGFBP3 knockdown in miR-21KD glioblastoma cells restored tumorigenesis. Examination of tumors from GBM patients showed that there was an inverse relationship between IGFBP3 and miR-21 expression and that increased IGFBP3 expression correlated with better patient survival. Our results identify IGFBP3 as a novel miR-21 target gene in glioblastoma and suggest that the oncogenic miRNA miR-21 down-regulates the expression of IGFBP3, which acts as a tumor suppressor in human glioblastoma.
Collapse
Affiliation(s)
- Chuan He Yang
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Junming Yue
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Susan R Pfeffer
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Meiyun Fan
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | | | - Amira Hosni-Ahmed
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Michelle Sims
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Sohail Qayyum
- From the Departments of Pathology and Laboratory Medicine and
| | - Andrew M Davidoff
- From the Departments of Pathology and Laboratory Medicine and Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee 38103
| | - Charles R Handorf
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Lawrence M Pfeffer
- From the Departments of Pathology and Laboratory Medicine and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| |
Collapse
|
40
|
Thota B, Arimappamagan A, Kandavel T, Shastry AH, Pandey P, Chandramouli BA, Hegde AS, Kondaiah P, Santosh V. STAT-1 expression is regulated by IGFBP-3 in malignant glioma cells and is a strong predictor of poor survival in patients with glioblastoma. J Neurosurg 2014; 121:374-83. [PMID: 24878287 DOI: 10.3171/2014.4.jns131198] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECT Insulin-like growth factor binding proteins (IGFBPs) have been implicated in the pathogenesis of glioma. In a previous study the authors demonstrated that IGFBP-3 is a novel glioblastoma biomarker associated with poor survival. Since signal transducer and activator of transcription 1 (STAT-1) has been shown to be regulated by IGFBP-3 during chondrogenesis and is a prosurvival and radioresistant molecule in different tumors, the aim in the present study was to explore the functional significance of IGFBP-3 in malignant glioma cells, to determine if STAT-1 is indeed regulated by IGFBP-3, and to study the potential of STAT-1 as a biomarker in glioblastoma. METHODS The functional significance of IGFBP-3 was investigated using the short hairpin (sh)RNA gene knockdown approach on U251MG cells. STAT-1 regulation by IGFBP-3 was tested on U251MG and U87MG cells by shRNA gene knockdown and exogenous treatment with recombinant IGFBP-3 protein. Subsequently, the expression of STAT-1 was analyzed with real-time reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) in glioblastoma and control brain tissues. Survival analyses were done on a uniformly treated prospective cohort of adults with newly diagnosed glioblastoma (136 patients) using Kaplan-Meier and Cox regression models. RESULTS IGFBP-3 knockdown significantly impaired proliferation, motility, migration, and invasive capacity of U251MG cells in vitro (p < 0.005). Exogenous overexpression of IGFBP-3 in U251MG and U87MG cells demonstrated STAT-1 regulation. The mean transcript levels (by real-time RT-PCR) and the mean labeling index of STAT-1 (by IHC) were significantly higher in glioblastoma than in control brain tissues (p = 0.0239 and p < 0.001, respectively). Multivariate survival analysis revealed that STAT-1 protein expression (HR 1.015, p = 0.033, 95% CI 1.001-1.029) along with patient age (HR 1.025, p = 0.005, 95% CI 1.008-1.042) were significant predictors of shorter survival in patients with glioblastoma. CONCLUSIONS IGFBP-3 influences tumor cell proliferation, migration, and invasion and regulates STAT-1 expression in malignant glioma cells. STAT-1 is overexpressed in human glioblastoma tissues and emerges as a novel prognostic biomarker.
Collapse
|
41
|
WILLIAMS PAULT. Reduced Risk of Brain Cancer Mortality from Walking and Running. Med Sci Sports Exerc 2014; 46:927-32. [DOI: 10.1249/mss.0000000000000176] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
42
|
Hsieh WT, Yeh WL, Cheng RY, Lin C, Tsai CF, Huang BR, Wu CYJ, Lin HY, Huang SS, Lu DY. Exogenous endothelin-1 induces cell migration and matrix metalloproteinase expression in U251 human glioblastoma multiforme. J Neurooncol 2014; 118:257-269. [PMID: 24756349 DOI: 10.1007/s11060-014-1442-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 04/09/2014] [Indexed: 10/25/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal type of primary brain tumor characterized by its rapid infiltration to surrounding tissues during the early stages. The fast spreading of GBM obscures the initiation of the tumor mass making the treatment outcome undesirable. Endothelin-1 is known as a secretory protein presented in various types of brain cells, which has been indicated as a factor for cancer pathology. The aim of the present study was to investigate the molecular mechanism of cell migration in GBM. We found that various malignant glioma cells expressed higher amounts of endothelin-1, ETA, and ETB receptors than nonmalignant human astrocytes. The application of endothelin-1 enhanced the migratory activity in human U251 glioma cells corresponding to increased expression of matrix metalloproteinase (MMP)-9 and MMP-13. The endothelin-1-induced cell migration was attenuated by MMP-9 and MMP-13 inhibitors and inhibitors of mitogen-activated protein (MAP) kinase and PI3 kinase/Akt. Furthermore, the elevated levels of phosphate c-Jun accumulation in the nucleus and activator protein-1 (AP-1)-DNA binding activity were also found in endothelin-1 treated glioma cells. In migration-prone sublines, cells with greater migration ability showed higher endothelin-1, ETB receptor, and MMP expressions. These results indicate that endothelin-1 activates MAP kinase and AP-1 signaling, resulting in enhanced MMP-9 and MMP-13 expressions and cell migration in GBM.
Collapse
Affiliation(s)
- Wen-Tsong Hsieh
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering and Department of Medical Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Ruo-Yuo Cheng
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Caren Yu-Ju Wu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No. 91 Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
43
|
Visani M, de Biase D, Marucci G, Cerasoli S, Nigrisoli E, Bacchi Reggiani ML, Albani F, Baruzzi A, Pession A. Expression of 19 microRNAs in glioblastoma and comparison with other brain neoplasia of grades I-III. Mol Oncol 2014; 8:417-30. [PMID: 24412053 PMCID: PMC5528554 DOI: 10.1016/j.molonc.2013.12.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/09/2013] [Accepted: 12/16/2013] [Indexed: 02/08/2023] Open
Abstract
Several biomarkers have been proposed as useful parameters to better specify the prognosis or to delineate new target therapy strategies for glioblastoma patients. MicroRNAs could represent putative target molecules, considering their role in tumorigenesis, cancer progression and their specific tissue expression. Although several studies have tried to identify microRNA signature for glioblastoma, a microRNA profile is still far from being well-defined. In this work the expression of 19 microRNAs (miR-7, miR-9, miR-9∗, miR-10a, miR-10b, miR-17, miR-20a, miR-21, miR-26a, miR-27a, miR-31, miR-34a, miR-101, miR-137, miR-182, miR-221, miR-222, miR-330, miR-519d) was evaluated in sixty formalin-fixed and paraffin-embedded glioblastoma samples using a locked nucleic acid real-time PCR. Moreover, a comparison of miRNA expressions was performed between primary brain neoplasias of different grades (grades IV-I). The analysis of 14 validated miRNA expression in the 60 glioblastomas, using three different non-neoplastic references as controls, revealed a putative miRNA signature: mir-10b and miR-21 were up-regulated, while miR-7, miR-31, miR-101, miR-137, miR-222 and miR-330 were down-regulated in glioblastomas. Comparing miRNA expression between glioblastoma group and gliomas of grades I-III, 3 miRNAs (miR-10b, mir-34a and miR-101) showed different regulation statuses between high-grade and low-grade tumors. miR-10b was up-regulated in high grade and significantly down-regulated in low-grade gliomas, suggesting that could be a candidate for a GBM target therapy. This study provides further data for the identification of a miRNA profile for glioblastoma and suggests that different-grade neoplasia could be characterized by different expression of specific miRNAs.
Collapse
Affiliation(s)
- Michela Visani
- Department of Pharmacy and Biotechnology, FaBiT, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, FaBiT, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy; Department of Experimental, Diagnostic and Specialty Medicine, DIMES, Anatomic Pathology at Bellaria Hospital, University of Bologna, via Altura 3, 40139 Bologna, Italy.
| | - Gianluca Marucci
- Department of Biomedical Science and Neuromotor Sciences, DIBINEM, Anatomic Pathology, Bellaria Hospital, University of Bologna, via Altura 3, 40139 Bologna, Italy
| | - Serenella Cerasoli
- Anatomic Pathology, M. Bufalini Hospital, Viale Ghirotti 286, 47023 Cesena (FC), Italy
| | - Evandro Nigrisoli
- Anatomic Pathology, M. Bufalini Hospital, Viale Ghirotti 286, 47023 Cesena (FC), Italy
| | - Maria Letizia Bacchi Reggiani
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Via Massarenti 9, 40100 Bologna, Italy
| | - Fiorenzo Albani
- IRCCS Istituto delle Scienze Neurologiche, via Altura 3, 40139 Bologna, Italy; Department of Biomedical & Neuromotor Sciences, University of Bologna, via Altura 3, 40139 Bologna, Italy
| | - Agostino Baruzzi
- IRCCS Istituto delle Scienze Neurologiche, via Altura 3, 40139 Bologna, Italy; Department of Biomedical & Neuromotor Sciences, University of Bologna, via Altura 3, 40139 Bologna, Italy
| | - Annalisa Pession
- Department of Pharmacy and Biotechnology, FaBiT, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | | |
Collapse
|
44
|
Hervouet E, Staehlin O, Pouliquen D, Debien E, Cartron PF, Menanteau J, Vallette FM, Olivier C. Antioxidants delay clinical signs and systemic effects of ENU induced brain tumors in rats. Nutr Cancer 2014; 65:686-94. [PMID: 23859036 DOI: 10.1080/01635581.2013.789541] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
According to our previous study suggesting that antioxidant properties of phytochemicals in the diet decrease glioma aggressiveness, we used a SUVIMAX-like diet ("Supplementation en VItamines et Minéraux AntioXydants") (enriched with alpha-tocopherol, beta carotene, vitamin C, zinc, and sodium selenite), adapted to rats. The present results showed that each of the antioxidants inhibited growth of glioma cells in vitro. When used in combination for in vivo studies, we showed a highly significant delay in the clinical signs of the disease, but not a statistical significant difference in the incidence of glioma in an Ethyl-nitrosourea (ENU)-model. The SUVIMAX-like diet decreased candidate markers of tumoral aggressiveness and gliomagenesis progression. The mRNA expressions of 2 common markers in human glioma: Mn-SOD (Manganese Superoxide Dismutase) and IGFBP5 (insulin growth factor binding protein) were reduced in the tumors of rats fed the antioxidant diet. In addition, the transcripts of two markers linked to brain tumor proliferation, PDGFRb (platelet-derived growth factor receptor beta) and Ki-67, were also significantly decreased. On the whole, our results suggest a protective role for antioxidants to limit aggressiveness and to some extent, progression of gliomas, in a rat model.
Collapse
Affiliation(s)
- E Hervouet
- UMR INSERM 892-CNRS 6299, Centre de Recherche en Cancérologie Nantes-Angers, Nantes, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Patel VN, Gokulrangan G, Chowdhury SA, Chen Y, Sloan AE, Koyutürk M, Barnholtz-Sloan J, Chance MR. Network signatures of survival in glioblastoma multiforme. PLoS Comput Biol 2013; 9:e1003237. [PMID: 24068912 PMCID: PMC3777929 DOI: 10.1371/journal.pcbi.1003237] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 08/08/2013] [Indexed: 12/02/2022] Open
Abstract
To determine a molecular basis for prognostic differences in glioblastoma multiforme (GBM), we employed a combinatorial network analysis framework to exhaustively search for molecular patterns in protein-protein interaction (PPI) networks. We identified a dysregulated molecular signature distinguishing short-term (survival<225 days) from long-term (survival>635 days) survivors of GBM using whole genome expression data from The Cancer Genome Atlas (TCGA). A 50-gene subnetwork signature achieved 80% prediction accuracy when tested against an independent gene expression dataset. Functional annotations for the subnetwork signature included “protein kinase cascade,” “IκB kinase/NFκB cascade,” and “regulation of programmed cell death” – all of which were not significant in signatures of existing subtypes. Finally, we used label-free proteomics to examine how our subnetwork signature predicted protein level expression differences in an independent GBM cohort of 16 patients. We found that the genes discovered using network biology had a higher probability of dysregulated protein expression than either genes exhibiting individual differential expression or genes derived from known GBM subtypes. In particular, the long-term survivor subtype was characterized by increased protein expression of DNM1 and MAPK1 and decreased expression of HSPA9, PSMD3, and CANX. Overall, we demonstrate that the combinatorial analysis of gene expression data constrained by PPIs outlines an approach for the discovery of robust and translatable molecular signatures in GBM. Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor in adults, and, while the median survival time for treated patients is approximately one year, subgroups of patients respond differently to the same treatments, with some patients showing little improvement and other patients living far longer than expected. These differences in treatment response indicate that the tumors may show molecular differences that we can harness to tailor cancer therapy. To this end, we sought to identify biomarkers of patient survival in GBM. To improve the applicability of our molecular markers to other patient groups, we constrained our markers using maps of protein-protein interactions, and we also employed a unique computational strategy that incorporates patient-to-patient molecular variability into the results. We identified a set of 50 genes comprising a subnetwork signature that successfully separated GBM patients by their survival times. Our approach to identifying this subnetwork signature also improved our ability to identify its protein products in an independent cohort of patients. In the ongoing search to improve cancer detection and treatment, our work represents a successful strategy for identifying reproducible biomarkers that can more efficiently lead to the discovery of druggable protein targets.
Collapse
Affiliation(s)
- Vishal N. Patel
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| | - Giridharan Gokulrangan
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Salim A. Chowdhury
- School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Yanwen Chen
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Andrew E. Sloan
- Brain Tumor & Neuro-Oncology Center, University Hospital-Case Medical Center, Cleveland, Ohio, United States of America
| | - Mehmet Koyutürk
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Electrical Engineering & Computer Science, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jill Barnholtz-Sloan
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mark R. Chance
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
46
|
Vlachostergios PJ, Voutsadakis IA, Papandreou CN. The role of ubiquitin-proteasome system in glioma survival and growth. Growth Factors 2013; 31:106-13. [PMID: 23688106 DOI: 10.3109/08977194.2013.799156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
High-grade gliomas represent a group of aggressive brain tumors with poor prognosis due to an inherent capacity of persistent cell growth and survival. The ubiquitin-proteasome system (UPS) is an intracellular machinery responsible for protein turnover. Emerging evidence implicates various proteins targeted for degradation by the UPS in key survival and proliferation signaling pathways of these tumors. In this review, we discuss the involvement of UPS in the regulation of several mediators and effectors of these pathways in malignant gliomas.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, University Hospital of Larissa, Larissa, Greece.
| | | | | |
Collapse
|
47
|
|
48
|
Wilk A, Urbanska K, Grabacka M, Mullinax J, Marcinkiewicz C, Impastato D, Estrada JJ, Reiss K. Fenofibrate-induced nuclear translocation of FoxO3A triggers Bim-mediated apoptosis in glioblastoma cells in vitro. Cell Cycle 2012; 11:2660-71. [PMID: 22732497 DOI: 10.4161/cc.21015] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Anti-neoplastic potential of calorie restriction or ligand-induced activation of peroxisome proliferator activated receptors (PPARs) has been demonstrated in multiple studies; however, mechanism(s) by which tumor cells respond to these stimuli remain to be elucidated. One of the potent agonists of PPARα, fenofibrate, is a commonly used lipid-lowering drug with low systemic toxicity. Fenofibrate-induced PPARα transcriptional activity is expected to shift energy metabolism from glycolysis to fatty acid β-oxidation, which in the long-term, could target weak metabolic points of glycolysis-dependent glioblastoma cells. The results of this study demonstrate that 25 μM fenofibrate can effectively repress malignant growth of primary glial tumor cells and glioblastoma cell lines. This cytostatic action involves G(1) arrest accompanied by only a marginal level of apoptotic cell death. Although the cells treated with 25 μM fenofibrate remain arrested, the cells treated with 50 μM fenofibrate undergo massive apoptosis, which starts after 72 h of the treatment. This delayed apoptotic event was preceded by FoxO3A nuclear accumulation, FoxO3A phosphorylation on serine residue 413, its elevated transcriptional activity and expression of FoxO-dependent apoptotic protein, Bim. siRNA-mediated inhibition of FoxO3A attenuated fenofibrate-induced apoptosis, indicating a direct involvement of this transcription factor in the fenofibrate action against glioblastoma. These properties of fenofibrate, coupled with its low systemic toxicity, make it a good candidate in support of conventional therapies against glial tumors.
Collapse
Affiliation(s)
- Anna Wilk
- Neurological Cancer Research, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kulkarni A, Thota B, Srividya MR, Thennarasu K, Arivazhagan A, Santosh V, Chandramouli BA. Expression pattern and prognostic significance of IGFBP isoforms in anaplastic astrocytoma. Pathol Oncol Res 2012; 18:961-7. [PMID: 22547392 DOI: 10.1007/s12253-012-9526-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 03/27/2012] [Indexed: 11/28/2022]
Abstract
The role of insulin-like growth factors and their regulatory proteins (IGFBP isoforms) in gliomas, particularly glioblastoma, has been a subject of active research in recent years. There is paucity of literature on their expression and impact on clinical outcome in anaplastic astrocytomas. To evaluate the expression patterns of IGFBP isoforms in anaplastic astrocytoma and correlate with clinical outcome, a retrospective study of 53 adult patients operated for supratentorial lobar anaplastic astrocytoma was performed. The protein expression of IGFBP isoforms (IGFBP-2, -3, -5 and -7), was studied by immunohistochemistry on all samples. The patients were followed up and outcome was documented. The median age at presentation in the present study was 35 years. The pattern of staining was intra cytoplasmic, homogenous and diffuse for IGFBP-2, -3 and -5 and granular for IGFBP-7. IGFBP-2 expression was significantly low in anaplastic astrocytoma as compared to other isoforms (P < 0.001). IGFBP-3 expression was higher than the other isoforms. However, its' expression correlated with favorable overall survival and demonstrated a trend towards significance on univariate analysis. The present study is the first of its kind to describe comprehensively the pattern of expression of IGFBP isoforms (IGFBP-2, -3, -5 and -7) in anaplastic astrocytomas. IGFBP-2 and IGFBP-3 expression patterns and correlation to prognosis were distinct in anaplastic astrocytoma patients, contradictory to what has been reported in glioblastoma, thus giving further evidence that anaplastic astrocytomas are molecularly distinct from glioblastoma.
Collapse
Affiliation(s)
- A Kulkarni
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | | | | | | | | | | |
Collapse
|
50
|
Qian XM, Shi ZD, Ren Y, Liu CY, Ji YR, Long LX, Pu P, Sheng J, Yuan XB, Kang CS. Synergistic inhibition of human glioma cell line by temozolomide and PAMAM-mediated miR-21i. J Appl Polym Sci 2012. [DOI: 10.1002/app.37823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|