1
|
Qin G, Sun Z, Jin Y, Ren X, Zhang Z, Wang S, Zhou G, Huang K, Zhao H, Jiang X. The association between the triglyceride-glucose index and prognosis in postoperative renal cell carcinoma patients: a retrospective cohort study. Front Endocrinol (Lausanne) 2024; 15:1301703. [PMID: 38476671 PMCID: PMC10927751 DOI: 10.3389/fendo.2024.1301703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Background Insulin resistance has been proven to be associated with renal cell carcinoma (RCC). However, the prognostic value of the triglyceride-glucose (TyG) index, as a marker for insulin resistance (IR), is still unclear. Therefore, we conducted research to explore the prognostic value and the predictive performance of the TyG index in postoperative RCC patients. Methods A total of 651 postoperative RCC patients from January 2016 to June 2018 were enrolled in the final study. Their clinical and laboratory parameters were collected from medical records and through follow-up by phone. The triglyceride-glucose (TyG) index was calculated as follows: TyG = Ln[TG (mg/dl) × FBG (mg/dL)/2]. The overall survival (OS) and disease-free survival (DFS) were identified as the main outcomes. Results The TyG index is an independent prognostic factor for OS (HR = 2.340, 95% CI = 1.506 to 3.64, P < 0.001) and DFS (HR = 2.027, 95% CI = 1.347 to 3.051, P < 0.001) in postoperative RCC patients. Kaplan-Meier survival curves of the different TyG index levels showed statistically significant differences in terms of OS and DFS (log-rank test, P < 0.0001). Furthermore, the TyG index was significantly associated with RCC risk factors. Conclusion The TyG index is significantly associated with RCC survival. The mechanisms responsible for these results may contribute toward the improvement of RCC prognosis and immunotherapy efficacy and the development of new immunotherapeutic targets.
Collapse
Affiliation(s)
- Guoliang Qin
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhuang Sun
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuxiang Jin
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiangguo Ren
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhaocun Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shuo Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Guanwen Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Kun Huang
- Department of Urology, Changle County People’s Hospital, Weifang, China
| | - Haifeng Zhao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xianzhou Jiang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Association of Diabetes Severity and Mortality with Lung Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14102553. [PMID: 35626156 PMCID: PMC9139965 DOI: 10.3390/cancers14102553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The survival impact of diabetes severity on lung cancer survival remains unclear. We performed head-to-head propensity score matching to estimate the survival impact of various adapted diabetes complications severity index (aDCSI) scores in patients with both diabetes and lung squamous cell carcinoma (SqCLC). The results indicated that diabetes severity (aDCSI ≥ 2) is an independent prognostic factor for the overall survival of patients with both diabetes and lung SqCLC who receive standard treatments. Prevention of diabetes progression is necessary for patients with diabetes; it affects not only diabetes control but also improves survival for patients with lung SqCLC. Abstract Purpose: The survival impact of diabetes severity on lung cancer remains unclear. We performed head-to-head propensity score matching to estimate the survival impact of various adapted diabetes complications severity index (aDCSI) scores in patients with both diabetes and lung squamous cell carcinoma (SqCLC). Patients and Methods: We enrolled patients with both diabetes and lung SqCLC and categorized them into the mild (aDCSI = 0–1) and moderate-to-severe (aDCSI ≥ 2) diabetes groups. The patients in both groups were matched at a 1:1 ratio. Results: the matching process yielded a final cohort of 5742 patients with both diabetes and lung SqCLC (2871 patients in the mild diabetes group and 2871 patients in the moderate-to-severe diabetes groups) who were eligible for further analysis. A multivariate Cox regression analysis revealed that the adjusted hazard ratio (aHR; 95% confidence interval) of all-cause death for the mild diabetes group relative to the moderate-to-severe diabetes group was 1.17 (1.08–1.28; p = 0.0005). Conclusion: severe diabetes (aDCSI ≥ 2) is an independent prognostic factor for OS among patients with both diabetes and lung SqCLC who receive standard treatments. Preventing diabetes progression is necessary for patients with diabetes because it not only supports diabetes control but also improves survival for patients with lung SqCLC.
Collapse
|
3
|
Renal Cell Cancer and Obesity. Int J Mol Sci 2022; 23:ijms23063404. [PMID: 35328822 PMCID: PMC8951303 DOI: 10.3390/ijms23063404] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
Cancers are a frequent cause of morbidity and mortality. There are many risk factors for tumours, including advanced age, personal or family history of cancer, some types of viral infections, exposure to radiation and some chemicals, smoking and alcohol consumption, as well as obesity. Increasing evidence suggest the role of obesity in the initiation and progression of various cancers, including renal cell carcinoma. Since tumours require energy for their uncontrollable growth, it appears plausible that their initiation and development is associated with the dysregulation of cells metabolism. Thus, any state characterised by an intake of excessive energy and nutrients may favour the development of various cancers. There are many factors that promote the development of renal cell carcinoma, including hypoxia, inflammation, insulin resistance, excessive adipose tissue and adipokines and others. There are also many obesity-related alterations in genes expression, including DNA methylation, single nucleotide polymorphisms, histone modification and miRNAs that can promote renal carcinogenesis. This review focuses on the impact of obesity on the risk of renal cancers development, their aggressiveness and patients’ survival.
Collapse
|
4
|
Nakhjavani M, Smith E, Palethorpe HM, Tomita Y, Yeo K, Price TJ, Townsend AR, Hardingham JE. Anti-Cancer Effects of an Optimised Combination of Ginsenoside Rg3 Epimers on Triple Negative Breast Cancer Models. Pharmaceuticals (Basel) 2021; 14:ph14070633. [PMID: 34208799 PMCID: PMC8308773 DOI: 10.3390/ph14070633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/26/2022] Open
Abstract
Key problems of chemotherapies, as the mainstay of treatment for triple-negative breast cancer (TNBC), are toxicity and development of tumour resistance. Using response surface methodology, we previously optimised the combination of epimers of ginsenoside Rg3 (Rg3) for anti-angiogenic action. Here, we show that the optimised combination of 50 µM SRg3 and 25 µM RRg3 (C3), derived from an RSM model of migration of TNBC cell line MDA-MB-231, inhibited migration of MDA-MB-231 and HCC1143, in 2D and 3D migration assays (p < 0.0001). C3 inhibited mammosphere formation efficiency in both cell lines and decreased the CD44+ stem cell marker in the mammospheres. Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. C3 affected the signalling of AKT in MDA-MB-231 and HCC1143 mammospheres. In a mouse model of metastatic TNBC, an equivalent dose of C3 (23 mg/kg SRg3 + 11 mg/kg RRg3) or an escalated dose of 46 mg/kg SRg3 + 23 mg/kg RRg3 was administered to NSG mice bearing MDA-MB-231-Luc cells. Calliper and IVIS spectrum measurement of the primary and secondary tumour showed that the treatment shrunk the primary tumour and decreased the load of metastasis in mice. In conclusion, this combination of Rg3 epimers showed promising results as a potential treatment option for TNBC patients.
Collapse
Affiliation(s)
- Maryam Nakhjavani
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (Y.T.); (K.Y.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| | - Eric Smith
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (Y.T.); (K.Y.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Correspondence: ; Tel.: +61-8-8222-6142
| | - Helen M. Palethorpe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia;
| | - Yoko Tomita
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (Y.T.); (K.Y.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Kenny Yeo
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (Y.T.); (K.Y.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| | - Tim J. Price
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Amanda R. Townsend
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Jennifer E. Hardingham
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (Y.T.); (K.Y.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| |
Collapse
|
5
|
Ogawa H, Fujibayashi Y, Nishikubo M, Nishioka Y, Tane S, Kitamura Y, Nishio W. Prognostic significance of preoperative haemoglobin A1c level in patients with lung adenocarcinoma. Interact Cardiovasc Thorac Surg 2021; 33:534-540. [PMID: 34115869 DOI: 10.1093/icvts/ivab140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/31/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES We investigated the influence of the preoperative haemoglobin A1c (HbA1c) value on the prognosis and pathology of patients with lung adenocarcinoma who underwent surgery. METHODS We reviewed the medical records of 400 lung adenocarcinoma patients who underwent lobectomy with mediastinal lymph node dissection between 2009 and 2013 using a prospectively maintained database. We stratified 400 patients into 4 groups according to the preoperative HbA1c value as follows: HbA1c ≤ 5.9 (n = 296), 6.0 ≤ HbA1c ≤ 6.9 (n = 70), 7.0 ≤ HbA1c ≤ 7.9 (n = 21) and HbA1c ≥ 8.0 (n = 12). We compared the recurrence-free survival and overall survival (OS) among these 4 groups. Univariate and multivariate analyses were performed to identify the risk factors for recurrence. RESULTS The median follow-up period was 61.2 months. On comparing the recurrence-free survival and OS rates among these 4 groups, we found that these rates among patients in the HbA1c ≥ 8.0 group were significantly poorer compared with the other 3 groups (5-year recurrence-free survival: HbA1c ≤ 5.9, 70.4%; 6.0 ≤ HbA1c ≤ 6.9, 69.7%; 7.0 ≤ HbA1c ≤ 7.9, 70.7%; ≥8.0 HbA1c, 18.8%; P = 0.002; and 5-year OS: HbA1c ≤ 5.9, 88.7%; 6.0 ≤ HbA1c ≤ 6.9, 80.6%; 7.0 ≤ HbA1c ≤ 7.9, 90.2%; ≥8.0 HbA1c, 66.7%; P = 0.046). Patients in the HbA1c ≥ 8.0 group had significantly more tumours with vascular invasion (P = 0.041) and experienced distant metastasis significantly more often (P = 0.028) than those with other values. A multivariate analysis revealed that preoperative HbA1c ≥ 8.0 [hazard ratio (HR) 2.33; P = 0.026] and lymph node metastasis (HR 3.94; P < 0.001) were significant independent prognostic factors for recurrence. CONCLUSIONS Our results revealed that preoperative HbA1c ≥ 8.0 is associated to poor prognosis due to the occurrence of distant metastasis and we should carefully follow these patients after surgery. CLINICAL REGISTRATION NUMBER Hyogo Cancer Center, G-57.
Collapse
Affiliation(s)
- Hiroyuki Ogawa
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Japan
| | | | - Megumi Nishikubo
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Japan
| | - Yuki Nishioka
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Japan
| | - Shinya Tane
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Japan
| | | | - Wataru Nishio
- Department of Thoracic Surgery, Hyogo Cancer Center, Akashi, Japan
| |
Collapse
|
6
|
Hu Y, Yang Z, Bao D, Ni JS, Lou J. miR-455-5p suppresses hepatocellular carcinoma cell growth and invasion via IGF-1R/AKT/GLUT1 pathway by targeting IGF-1R. Pathol Res Pract 2019; 215:152674. [DOI: 10.1016/j.prp.2019.152674] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/16/2019] [Accepted: 09/27/2019] [Indexed: 12/18/2022]
|
7
|
Yang H, Yin K, Wang Y, Xia M, Zhang R, Wang W, Chen J, Wang C, Shuang W. Pre-existing type 2 diabetes is an adverse prognostic factor in patients with renal cell carcinoma. J Diabetes 2019; 11:993-1001. [PMID: 31141620 PMCID: PMC7379251 DOI: 10.1111/1753-0407.12957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/13/2019] [Accepted: 05/26/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Diabetes is a risk factor for various cancers, but its prognostic role in renal cell carcinoma (RCC) is controversial and understudied. This study investigated the prognostic value of type 2 diabetes (T2D) in RCC patients. METHODS The clinicopathological and follow-up data of 451 RCC patients undergoing radical or partial nephrectomy at the First Hospital of Shanxi Medical University from 2013 to 2018 were reviewed. Associations of T2D with clinicopathological parameters of RCC were evaluated using the Kaplan-Meier method for survival estimates and Cox regression univariate and multivariate analyses. RESULTS Of 451 patients, 74 (16.4%) had T2D. These patients were older, had a higher body mass index, higher incidence rates of hypertension and cardiovascular morbidity, a higher rate of laparoscopic surgery, and smaller neoplasms (all P < .05). Patients with T2D exhibited shorter overall survival (OS; P = .009), cancer-specific survival (CSS; P = .043), and recurrence-free survival (RFS; P = .008) than patients without T2D. Fuhrman grade (hazard ratio [HR] 2.542, 95% confidence interval [CI] 1.115-5.795, P = .026) and T2D (HR 3.391, CI 1.458-7.886, P = .005) were independent predictors of OS; T2D was an independent predictor of CSS (HR = 4.637, 95% CI 1.420-15.139, P = .011) and RFS (HR 3.492, 95% CI 1.516-8.044, P = .003). CONCLUSIONS Renal cell carcinoma patients with T2D have a shorter OS and higher recurrence rate and mortality risk than those without T2D.
Collapse
Affiliation(s)
- Haosen Yang
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Keqiang Yin
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Yusheng Wang
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Mancheng Xia
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Ruiqin Zhang
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Wenzhan Wang
- Department of GynecologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Jiawei Chen
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Chaoqi Wang
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Weibing Shuang
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
8
|
The tumor suppressor NDRG2 cooperates with an mTORC1 inhibitor to suppress the Warburg effect in renal cell carcinoma. Invest New Drugs 2019; 38:956-966. [DOI: 10.1007/s10637-019-00839-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/12/2019] [Indexed: 10/26/2022]
|
9
|
Ren L, Yao Y, Wang Y, Wang S. MiR-505 suppressed the growth of hepatocellular carcinoma cells via targeting IGF-1R. Biosci Rep 2019; 39:BSR20182442. [PMID: 31160483 PMCID: PMC6603277 DOI: 10.1042/bsr20182442] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/14/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers globally. An increasing body of evidence has demonstrated the critical function of microRNAs (miRNAs) in the initiation and progression of human cancers. Here, we showed that miR-505 was down-regulated in HCC tissues and cell lines. Reduced expression of miR-505 was significantly correlated with the worse prognosis of HCC patients. Overexpression of miR-505 suppressed the proliferation, colony formation and induced apoptosis of both HepG2 and Huh7 cells. Further mechanism study uncovered that miR-505 bound the 3'-untranslated region (3'-UTR) of the insulin growth factor receptor (IGF-1R) and inhibited the expression of IGF-1R in HCC cells. The down-regulation of IGF-1R by miR-505 further suppressed the phosphorylation of AKT at the amino acid S473. Consistently, the abundance of glucose transporter (GLUT) 1 (GLUT1) was reduced with the overexpression of miR-505. Down-regulation of GLUT1 by miR-505 consequently attenuated the glucose uptake, lactate production and ATP generation of HCC cells. Collectively, our results demonstrated the tumor suppressive function of miR-505 possibly via inhibiting the glycolysis of HCC cells. These findings suggested miR-505 as an interesting target for designing anti-cancer strategy in HCC.
Collapse
Affiliation(s)
- Liang Ren
- Department of Ultrasound and Imaging, Yichang Yiling Hospital, Yichang city 443100, Hubei province, China
| | - Yongshan Yao
- Emergency and Trauma Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang city 443100, Hubei province, China
| | - Yang Wang
- Department of Ultrasound and Imaging, Yichang Yiling Hospital, Yichang city 443100, Hubei province, China
| | - Shengqiang Wang
- Department of Pediatrics, Yichang Yiling Hospital, No.32 of Dong Hu street, Yiling district, Yichang city 443100, Hubei province, China
| |
Collapse
|
10
|
Solarek W, Koper M, Lewicki S, Szczylik C, Czarnecka AM. Insulin and insulin-like growth factors act as renal cell cancer intratumoral regulators. J Cell Commun Signal 2019; 13:381-394. [PMID: 30929166 PMCID: PMC6732138 DOI: 10.1007/s12079-019-00512-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
The risk of renal cell carcinoma development is correlated with obesity and type II diabetes. Since insulin and insulin-like growth factors play a key role during development of both metabolic diseases, these molecules may be important in RCC pathophysiology We investigated the effect of insulin and IGFs on RCC cells using in vitro model with 786-O, 769-P, Caki-1, Caki-2, ACHN cancer cell lines. Cancer cells were compared with normal kidney cells - PCS-400-010 and HEK293. The growth, viability of cells as well as migration rate were assessed upon hormonal stimulation. The insulin receptor and Insulin-like growth factor 1 receptor presence were evaluated and the expression of 84 genes related to insulin signaling pathway. In all RCC cell lines IGF-1R expression was confirmed in contrast to IR, which was expressed only in control HEK293 cell line. Insulin and IGFs stimulated RCC cells growth and migration rate. Insulin, IGF-1 and IGF-2 triggered both IR and IGF-1R phosphorylation. Analyzed RCC did not secret insulin, IGF-1 or IGF-2 and were not activated in autocrine-paracrine signaling loop. Insulin and IGFs stimulations triggered down-regulation of PI3K-Akt-mTOR and Ras-MAPK pathway gens, as well as DOK2-3, INS, FRS3, IRS1-2, IGF1R - genes encoding insulin receptor-associated proteins. In conclusion, we showed that IGFs and insulin may play a stimulatory role for renal cancer cells, thus they can possibly affect renal cancer tumorigenesis and progression on cellular level.
Collapse
Affiliation(s)
- Wojciech Solarek
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Michal Koper
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Slawomir Lewicki
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of Oncology, European Health Centre, Otwock, Poland.,Medical Center for Postgraduate Education, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, Warsaw, 04-141, Poland.
| |
Collapse
|
11
|
Sun L, Gao Z, Luo L, Tan H, Zhang G. Estrogen affects cell growth and IGF-1 receptor expression in renal cell carcinoma. Onco Targets Ther 2018; 11:5873-5878. [PMID: 30271170 PMCID: PMC6149902 DOI: 10.2147/ott.s172149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Both obesity and gender are important etiological factors in renal cell carcinoma (RCC) development, suggesting a pivotal role of sex hormone signaling pathway and insulin-like growth factor (IGF) family in RCC carcinogenesis. Here, we aimed to investigate the effect of estrogen on RCC growth and the possible interaction between estrogen/estrogen receptor (ER) signaling pathway and the IGF axis. Methods ER-α and ER-β were detected in four human RCC cell lines. Cells were treated with 17β-estradiol (E2), and cell proliferation was determined using the cell counting kit-8 assay. Using siRNA, ER-β was downregulated in RCC cells and the effect of E2 on cell growth and IGF-1 receptor (IGF-1R) expression was examined. Results E2 inhibited 786-O cell but not A498 cell growth significantly. After the downregulation of ER-β, E2 showed no obvious inhibitory role in 786-O cells. E2 stimulation increased the expression of IGF-1R in 786-O cells. Downregulation of ER-β, as well as fulvestrant, attenuated the stimulatory effect of E2 on IGF-1R expression. Conclusion Our results revealed that estrogen induced RCC growth inhibition via an ER-β-dependent pathway. Estrogen also upregulated the expression of IGF-1R, suggesting a link between estrogen/ER and IGF axis.
Collapse
Affiliation(s)
- Lijiang Sun
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China,
| | - Zhemin Gao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China,
| | - Lei Luo
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China,
| | - Hailin Tan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China,
| | - Guiming Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China,
| |
Collapse
|
12
|
Lai Y, Zhao Z, Zeng T, Liang X, Chen D, Duan X, Zeng G, Wu W. Crosstalk between VEGFR and other receptor tyrosine kinases for TKI therapy of metastatic renal cell carcinoma. Cancer Cell Int 2018. [PMID: 29527128 PMCID: PMC5838927 DOI: 10.1186/s12935-018-0530-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC), and is frequently accompanied by the genetic features of von Hippel–Lindau (VHL) loss. VHL loss increases the expression of hypoxia-inducible factors (HIFs) and their targets, including epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF). The primary treatment for metastatic RCC (mRCC) is molecular-targeted therapy, especially anti-angiogenic therapy. VEGF monoclonal antibodies and VEGF receptor (VEGFR) tyrosine kinase inhibitors (TKIs) are the main drugs used in anti-angiogenic therapy. However, crosstalk between VEGFR and other tyrosine kinase or downstream pathways produce resistance to TKI treatment, and the multi-target inhibitors, HIF inhibitors or combination strategies are promising strategies for mRCC. HIFs are upstream of the crosstalk between the growth factors, and these factors may regulate the expression of VEGR, EGF, PDGF and other growth factors. The frequent VHL loss in ccRCC increases HIF expression, and HIFs may be an ideal candidate to overcome the TKI resistance. The combination of HIF inhibitors and immune checkpoint inhibitors is also anticipated. Various clinical trials of programmed cell death protein 1 inhibitors are planned. The present study reviews the effects of current and potential TKIs on mRCC, with a focus on VEGF/VEGFR and other targets for mRCC therapy.
Collapse
Affiliation(s)
- Yongchang Lai
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Zhijian Zhao
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Tao Zeng
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Xiongfa Liang
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Dong Chen
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Wenqi Wu
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| |
Collapse
|
13
|
Pearson ADJ, Federico SM, Aerts I, Hargrave DR, DuBois SG, Iannone R, Geschwindt RD, Wang R, Haluska FG, Trippett TM, Geoerger B. A phase 1 study of oral ridaforolimus in pediatric patients with advanced solid tumors. Oncotarget 2018; 7:84736-84747. [PMID: 27713169 PMCID: PMC5356695 DOI: 10.18632/oncotarget.12450] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/16/2016] [Indexed: 11/25/2022] Open
Abstract
Purpose Ridaforolimus is an investigational, potent, selective mTOR inhibitor. This study was conducted to determine the recommended phase 2 dose (RP2D), maximum tolerated dose, safety, pharmacokinetics, and antitumor activity of oral ridaforolimus in children with advanced solid tumors. Experimental Design In this phase 1, multicenter, open-label study in children aged 6 to <18 years with advanced solid tumors, ridaforolimus was administered orally for 5 consecutive days/week in 28-day cycles until progression, unacceptable toxicity, or consent withdrawal. Dose started at 22 mg/m2 and increased to 28 mg/m2 and 33 mg/m2, followed by expansion at the RP2D. Results Twenty patients were treated; 18 were evaluable for dose-limiting toxicities. One dose-limiting toxicity (grade 3 increased alanine aminotransferase) occurred in 1 patient at 33 mg/m2. Dose escalation concluded at 33 mg/m2; the maximum tolerated dose was not determined. The most common treatment-related adverse events (frequency ≥40%) were manageable grade 1–2 stomatitis, thrombocytopenia, hypertriglyceridemia, increased alanine aminotransferase, fatigue, hypercholesterolemia, anemia, and increased aspartate aminotransferase. Ridaforolimus exposure at 28 mg/m2 and 33 mg/m2 exceeded adult target levels. The RP2D for oral ridaforolimus in children was defined as 33 mg/m2. Four patients received at least 4 cycles; 2 with pineoblastoma and diffuse intrinsic pontine glioma had stable disease for 12 and 46 cycles, respectively. Conclusions Ridaforolimus is orally bioavailable and well tolerated in children with advanced solid tumors. The RP2D (33 mg/m2, 5 days/week) exceeds the adult RP2D. The favorable toxicity and pharmacokinetic profiles may allow for combination therapy, a promising therapeutic option in pediatric malignancies.
Collapse
Affiliation(s)
- Andrew D J Pearson
- Paediatric Drug Development Unit, Children and Young People's Unit, Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Sara M Federico
- Department of Pediatric Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Isabelle Aerts
- Department of Pediatric, Adolescent and Young Adult Oncology, Institut Curie, Paris, France
| | - Darren R Hargrave
- Haematology and Oncology Department, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Steven G DuBois
- Department of Pediatrics, University of California San Francisco School of Medicine, and Benioff Children's Hospital, San Francisco, CA, USA.,Current affiliation: Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Robert Iannone
- Clinical Research, Merck & Co., Inc., North Wales, PA, USA
| | | | - Ruixue Wang
- BARDS, MSD R&D (China) Co. Ltd., Beijing, China
| | - Frank G Haluska
- Clinical Research & Development, ARIAD Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Tanya M Trippett
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Birgit Geoerger
- Department of Childhood and Adolescent Oncology, Gustave Roussy, University Paris-Sud, Villejuif, France
| |
Collapse
|
14
|
Simpson A, Petnga W, Macaulay VM, Weyer-Czernilofsky U, Bogenrieder T. Insulin-Like Growth Factor (IGF) Pathway Targeting in Cancer: Role of the IGF Axis and Opportunities for Future Combination Studies. Target Oncol 2017; 12:571-597. [PMID: 28815409 PMCID: PMC5610669 DOI: 10.1007/s11523-017-0514-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite a strong preclinical rationale for targeting the insulin-like growth factor (IGF) axis in cancer, clinical studies of IGF-1 receptor (IGF-1R)-targeted monotherapies have been largely disappointing, and any potential success has been limited by the lack of validated predictive biomarkers for patient enrichment. A large body of preclinical evidence suggests that the key role of the IGF axis in cancer is in driving treatment resistance, via general proliferative/survival mechanisms, interactions with other mitogenic signaling networks, and class-specific mechanisms such as DNA damage repair. Consequently, combining IGF-targeted agents with standard cytotoxic agents, other targeted agents, endocrine therapies, or immunotherapies represents an attractive therapeutic approach. Anti-IGF-1R monoclonal antibodies (mAbs) do not inhibit IGF ligand 2 (IGF-2) activation of the insulin receptor isoform-A (INSR-A), which may limit their anti-proliferative activity. In addition, due to their lack of specificity, IGF-1R tyrosine kinase inhibitors are associated with hyperglycemia as a result of interference with signaling through the classical metabolic INSR-B isoform; this may preclude their use at clinically effective doses. Conversely, IGF-1/IGF-2 ligand-neutralizing mAbs inhibit proliferative/anti-apoptotic signaling via IGF-1R and INSR-A, without compromising the metabolic function of INSR-B. Therefore, combination regimens that include these agents may be more efficacious and tolerable versus IGF-1R-targeted combinations. Herein, we review the preclinical and clinical experience with IGF-targeted therapies to-date, and discuss the rationale for future combination approaches as a means to overcome treatment resistance.
Collapse
Affiliation(s)
- Aaron Simpson
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV, Dr. Boehringer Gasse 5-11, 1121, Vienna, Austria.
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
15
|
Gao S, Li S, Duan X, Gu Z, Ma Z, Yuan X, Feng X, Wang H. Inhibition of glycogen synthase kinase 3 beta (GSK3β) suppresses the progression of esophageal squamous cell carcinoma by modifying STAT3 activity. Mol Carcinog 2017; 56:2301-2316. [PMID: 28574599 DOI: 10.1002/mc.22685] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/13/2017] [Accepted: 06/01/2017] [Indexed: 12/26/2022]
Abstract
Although GSK3β has been reported to have contrasting effects on the progression of different tumors, it's possible functions in esophageal squamous cell carcinoma (ESCC) and the related molecular mechanisms remain unknown. Here, we investigated the expression, function, and molecular mechanism of GSK3β in the development of ESCC in vitro and in vivo. Though the expression of total GSK3β was significantly increased, the phosphorylated (inactivated) form of GSK3β (Ser9) was concurrently decreased in the cancerous tissues of patients with ESCC compared with controls, suggesting that GSK3β activity was enhanced in cancerous tissues. Further pathological data analysis revealed that higher GSK3β expression was associated with poorer differentiation, higher metastasis rates, and worse prognosis of ESCC. These results were confirmed in different ESCC cell lines using a pharmacological inhibitor and specific siRNA to block GSK3β. Using a cancer phospho-antibody array, we found that STAT3 is a target of GSK3β. GSK3 inhibition reduced STAT3 phosphorylation, and overexpression of constitutively active GSK3β had the opposite effect. Moreover, STAT3 inhibition mimicked the effects of GSK3β inhibition on ESCC cell migration and viability, while overexpression of a plasmid encoding mutant STAT3 (Y705F) abrogated these effects, and these results were further substantiated by clinicopathological data. In addition, a GSK3 inhibitor (LiCl) and/or STAT3 inhibitor (WP-1066) efficiently suppressed the growth of ESCC cells in a xenograft tumor model. Altogether, these results reveal that higher GSK3β expression promotes ESCC progression through STAT3 in vitro and in vivo, and GSK3β-STAT3 signaling could be a potential therapeutic target for ESCC treatment.
Collapse
Affiliation(s)
- Shegan Gao
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical college of Henan University of Science and Technology, Luoyang, China
| | - Shuoguo Li
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical college of Henan University of Science and Technology, Luoyang, China
| | - Xiaoxian Duan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky
| | - Zhen Gu
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky
| | - Zhikun Ma
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical college of Henan University of Science and Technology, Luoyang, China
| | - Xiang Yuan
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical college of Henan University of Science and Technology, Luoyang, China
| | - Xiaoshan Feng
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical college of Henan University of Science and Technology, Luoyang, China
| | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky
| |
Collapse
|
16
|
Duff D, Long A. Roles for RACK1 in cancer cell migration and invasion. Cell Signal 2017; 35:250-255. [PMID: 28336233 DOI: 10.1016/j.cellsig.2017.03.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/16/2023]
Abstract
Migration and invasion of cancer cells into surrounding tissue and vasculature is an important initial step in cancer metastasis. Metastasis is the leading cause of cancer related death and thus it is crucial that we improve our understanding of the mechanisms that promote this life-threatening phenomenon. Cell migration involves a complex, multistep process that leads to the actin-driven movement of cells on or through the tissues of the body. The multifunctional scaffolding protein RACK1 plays important roles in nucleating cell signalling hubs, anchoring proteins at specific subcellular locations and regulating protein activity. It is essential for cell migration and accumulating evidence now demonstrates multiple roles for RACK1 in regulating migration and invasion of tumour cells. The possibility of designing drugs that block the migratory and invasive capabilities of cancer cells represents an attractive therapeutic strategy for treating malignant disease with RACK1 being a potential target. In this review we summarize this evidence and examine the mechanisms that underlie the contribution of RACK1 to the various stages of cell migration and invasion.
Collapse
Affiliation(s)
- Deirdre Duff
- Trinity Translational Medicine Institute, Trinity College Dublin, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
17
|
Tracz AF, Szczylik C, Porta C, Czarnecka AM. Insulin-like growth factor-1 signaling in renal cell carcinoma. BMC Cancer 2016; 16:453. [PMID: 27405474 PMCID: PMC4942928 DOI: 10.1186/s12885-016-2437-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) incidence is highest in highly developed countries and it is the seventh most common neoplasm diagnosed. RCC management include nephrectomy and targeted therapies. Type 1 insulin-like growth factor (IGF-1) pathway plays an important role in cell proliferation and apoptosis resistance. IGF-1 and insulin share overlapping downstream signaling pathways in normal and cancer cells. IGF-1 receptor (IGF1R) stimulation may promote malignant transformation promoting cell proliferation, dedifferentiation and inhibiting apoptosis. Clear cell renal cell carcinoma (ccRCC) patients with IGF1R overexpression have 70 % increased risk of death compared to patients who had tumors without IGF1R expression. IGF1R signaling deregulation may results in p53, WT, BRCA1, VHL loss of function. RCC cells with high expression of IGF1R are more resistant to chemotherapy than cells with low expression. Silencing of IGF1R increase the chemosensitivity of ccRCC cells and the effect is greater in VHL mutated cells. Understanding the role of IGF-1 signaling pathway in RCC may result in development of new targeted therapeutic interventions. First preclinical attempts with anti-IGF-1R monoclonal antibodies or fragment antigen-binding (Fab) fragments alone or in combination with an mTOR inhibitor were shown to inhibit in vitro growth and reduced the number of colonies formed by of RCC cells.
Collapse
Affiliation(s)
- Adam F Tracz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,First Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Camillo Porta
- Department of Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| |
Collapse
|
18
|
Kim JH, Hwang KH, Lkhagvadorj S, Jung JH, Chung HC, Park KS, Kong ID, Eom M, Cha SK. Klotho plays a critical role in clear cell renal cell carcinoma progression and clinical outcome. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:297-304. [PMID: 27162484 PMCID: PMC4860372 DOI: 10.4196/kjpp.2016.20.3.297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/04/2022]
Abstract
Klotho functions as a tumor suppressor predominantly expressed in renal tubular cells, the origin of clear cell renal cell carcinoma (ccRCC). Altered expression and/or activity of growth factor receptor have been implicated in ccRCC development. Although Klotho suppresses a tumor progression through growth factor receptor signaling including insulin-like growth factor-1 receptor (IGF-1R), the role of Klotho acting on IGF-1R in ccRCC and its clinical relevance remains obscure. Here, we show that Klotho is favorable prognostic factor for ccRCC and exerts tumor suppressive role for ccRCC through inhibiting IGF-1R signaling. Our data shows the following key findings. First, in tumor tissues, the level of Klotho and IGF-1R expression are low or high, respectively, compared to that of adjacent non-neoplastic parenchyma. Second, the Klotho expression is clearly low in higher grade of ccRCC and is closely associated with clinical outcomes in tumor progression. Third, Klotho suppresses IGF-1-stimulated cell proliferation and migration by inhibiting PI3K/Akt pathway. These results provide compelling evidence supporting that Klotho acting on IGF-1R signaling functions as tumor suppressor in ccRCC and suggest that Klotho is a potential carcinostatis substance for ccRCC.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Department of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Kyu-Hee Hwang
- Department of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Sayamaa Lkhagvadorj
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Jae Hung Jung
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Hyun Chul Chung
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Kyu-Sang Park
- Department of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Korea.; Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - In Deok Kong
- Department of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Korea.; Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Seung-Kuy Cha
- Department of Physiology and Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Korea.; Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| |
Collapse
|
19
|
Zhu L, Cao H, Zhang T, Shen H, Dong W, Wang L, Du J. The Effect of Diabetes Mellitus on Lung Cancer Prognosis: A PRISMA-compliant Meta-analysis of Cohort Studies. Medicine (Baltimore) 2016; 95:e3528. [PMID: 27124062 PMCID: PMC4998725 DOI: 10.1097/md.0000000000003528] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Previous studies suggested that diabetes mellitus (DM) was associated with risk and mortality of cancer, but studies investigating the correlation between DM and lung cancer prognosis remain controversial. Herein, a meta-analysis was performed to derive a more precise estimate of the prognostic role of DM in lung cancer.Medline and Embase were searched for eligible articles from inception to October 25, 2015. The pooled hazard ratio (HR) with its 95% confidence interval (95% CI) was calculated to evaluate the correlation between DM and lung cancer prognosis. Subgroup meta-analysis was performed based on the histology and the treatment methods.A total of 20 cohort studies from 12 articles were included in the meta-analysis. Also, 16 studies investigated the overall survival (OS) and 4 studies investigated the progression-free survival (PFS). DM was significantly associated with the inferior OS of lung cancer with the pooled HR 1.28 (95% CI: 1.10-1.49, P = 0.001). The association was prominent in the nonsmall cell lung cancer (NSCLC) subgroup (HR 1.35, 95%CI: 1.14-1.60, P = 0.002), whereas the association was not significant in the small cell lung cancer (SCLC) subgroup (HR 1.33, 95% CI: 0.87-2.03, P = 0.18). When NSCLC patients were further stratified by treatment methods, DM had more influence on the surgically treated subgroup than the nonsurgically treated subgroup. There was no obvious evidence for publication bias by Begg's and Egger's test.The results of this meta-analysis exhibit an association of DM with inferior prognosis amongst lung cancer patients, especially the surgically treated NSCLC patients. Given the small number of studies included in this meta-analysis, the present conclusion should be consolidated with more high-quality prospective cohort studies or randomized controlled trials.
Collapse
Affiliation(s)
- Linhai Zhu
- From the Institute of Oncology (LZ, HC, TZ, LW), Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, P.R. China; Department of Oncology (HS), Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, P.R. China; Department of thoracic surgery (WD, JD), Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, P.R. China
| | | | | | | | | | | | | |
Collapse
|
20
|
Solarek W, Czarnecka AM, Escudier B, Bielecka ZF, Lian F, Szczylik C. Insulin and IGFs in renal cancer risk and progression. Endocr Relat Cancer 2015; 22:R253-64. [PMID: 26330483 DOI: 10.1530/erc-15-0135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin and IGFs play a significant role in cancer development and progression, including renal cell carcinoma (RCC). RCC is the most frequent type of kidney cancer in adults and the tenth most common malignancy worldwide. Insulin is normally associated with metabolism control, whereas IGFs are defined as proliferation regulators. Today, there is convincing evidence of an association between obesity and the risk of RCC. Indicated risk factors together with type 2 diabetes are irreversibly connected with circulating insulin and IGF levels. The interplay between these molecules, their receptors, and IGF-binding proteins might be crucial for RCC cell biology and RCC progression. Given the potent activity IGF/IGF receptor 1 (IGF1R) inhibitors demonstrate against RCC in basic research, some type of combination therapy may prove to be beneficial clinically in the management of RCC. This review addresses not only molecular but also clinical associations between insulin and IGF1 signaling pathways and both RCC biology and clinical course. Revealing these interactions may improve our understanding of basic molecular oncology processes in RCC and improve treatment strategies.
Collapse
Affiliation(s)
- W Solarek
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| | - A M Czarnecka
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| | - B Escudier
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| | - Z F Bielecka
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| | - F Lian
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| | - C Szczylik
- Laboratory of Molecular Oncology Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland School of Molecular Medicine Medical University of Warsaw, Warsaw, Poland Institut Gustave Roussy 114 rue Edouard Vaillant, 94805 Villejuif, France Emory University School of Medicine Atlanta, Georgia, USA
| |
Collapse
|
21
|
Chen L, Li H, Gu L, Ma X, Li X, Gao Y, Zhang Y, Shen D, Fan Y, Wang B, Bao X, Zhang X. The Impact of Diabetes Mellitus on Renal Cell Carcinoma Prognosis: A Meta-Analysis of Cohort Studies. Medicine (Baltimore) 2015; 94:e1055. [PMID: 26131819 PMCID: PMC4504607 DOI: 10.1097/md.0000000000001055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Previous studies that investigated the relationship between DM and survival in renal cell carcinoma (RCC) patients reported inconsistent findings. Hence, we conducted a meta-analysis to obtain a more precise evaluation of the prognostic significance of DM in RCC. A systematic review was conducted with PubMed, Embase, and Web of Science to identify relevant articles that evaluated the effect of DM on RCC patients. Based on the inclusion and quality assessment criteria, 18 studies were eligible for the meta-analysis. Pooled hazard ratios (HR) and corresponding 95% confidence intervals (CI) for overall survival (OS), cancer-specific survival (CSS), and recurrence-free survival (RFS) were calculated by standard meta-analysis techniques. The results suggested that DM was associated with poor OS (HR 1.56, 95% CI, 1.35-1.81, P < 0.001), poor CSS (HR 2.03, 95% CI, 1.37-3.01, P < 0.001), and poor RFS (HR 1.73, 95% CI, 1.25-2.39, P = 0.012). In addition, for patients with localized RCC, patients with clear cell RCC, or patients receiving nephrectomy, DM was associated with both poor OS and CSS by subgroup analyses. Our study revealed that there was a significant negative impact of DM on OS, CSS, and RFS in RCC patients. Therefore, more attention should be paid to RCC patients with preexisting DM because of their poor prognosis.
Collapse
Affiliation(s)
- Luyao Chen
- From the State Key Laboratory of Kidney Diseases, Department of Urology, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing (LC, HL, LG, XM, XL, YG, YZ, DS, YF, BW, XZ); and Medical School, Nankai University, Tianjin, People's Republic of China (XB)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhao Q, Tran H, Dimitrov DS, Cheung NKV. A dual-specific anti-IGF-1/IGF-2 human monoclonal antibody alone and in combination with temsirolimus for therapy of neuroblastoma. Int J Cancer 2015; 137:2243-52. [PMID: 25924852 DOI: 10.1002/ijc.29588] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/14/2015] [Indexed: 12/16/2022]
Abstract
The insulin-like growth factors (IGFs), IGF-1 and IGF-2, have been implicated in the growth, survival and metastasis of a broad range of malignancies including pediatric tumors. They bind to the IGF receptor type 1 (IGF-1R) and the insulin receptor (IR) which are overexpressed in many types of solid malignancies. Activation of the IR by IGF-2 results in increased survival of tumor cells. We have previously identified a novel human monoclonal antibody, m708.5, which binds with high (pM) affinity to both human IGF-1 and IGF-2, and potently inhibits phosphorylation of the IGF-1R and the IR in tumor cells. m708.5 exhibited strong antitumor activity as a single agent against most cell lines derived from neuroblastoma, Ewing family of tumor, rhabdomyosarcoma and osteosarcoma. When tested in neuroblastoma cell lines, it showed strong synergy with temsirolimus and synergy with chemotherapeutic agents in vitro. In xenograft models, the combination of m708.5 and temsirolimus significantly inhibited neuroblastoma growth and prolonged mouse survival. Taken together, these results support the clinical development of m708.5 for pediatric solid tumors with potential for synergy with chemotherapy and mTOR inhibitors.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY.,Laboratory of Fully Human Antibody Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Guangdong, China
| | - Hoa Tran
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Dimiter S Dimitrov
- Protein Interaction Section, Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, MD, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
23
|
Low Expression of miR-126 Is a Prognostic Marker for Metastatic Clear Cell Renal Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:693-703. [DOI: 10.1016/j.ajpath.2014.11.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 11/07/2014] [Accepted: 11/13/2014] [Indexed: 01/18/2023]
|
24
|
Czarnecka AM, Kornakiewicz A, Lian F, Szczylik C. Future perspectives for mTOR inhibitors in renal cell cancer treatment. Future Oncol 2015; 11:801-17. [DOI: 10.2217/fon.14.303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
ABSTRACT Everolimus is a mTOR inhibitor that demonstrates antitumor and antiangiogenic activities. In a randomized Phase III trial, patients with metastatic renal cell carcinoma who progressed on sunitinib/sorafenib were treated with everolimus and showed significant improvement in progression-free survival compared with best supportive care. Novel approaches in treatment are expected to ensure less toxic therapies and increase efficacy of everolimus. To provide a new perspective for mTOR inhibitor research and therapy, we discuss renal cell carcinoma cancer stem cells as a potential target for mTOR inhibitors and present new concepts on emerging antiangiogenic therapies. Finally, we point why systems biology approach with reverse molecular engineering may also contribute to the field of drug discovery in renal cell carcinoma.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Fei Lian
- Emory School of Medicine Atlanta, GA 30322, USA
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| |
Collapse
|
25
|
Kornakiewicz A, Solarek W, Bielecka ZF, Lian F, Szczylik C, Czarnecka AM. Mammalian Target of Rapamycin Inhibitors Resistance Mechanisms in Clear Cell Renal Cell Carcinoma. CURRENT SIGNAL TRANSDUCTION THERAPY 2014; 8:210-218. [PMID: 25152703 PMCID: PMC4141323 DOI: 10.2174/1574362409666140206222746] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/21/2014] [Accepted: 01/29/2014] [Indexed: 11/22/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a kinase protein involved in PI3K/AKT signaling with a central role in the processes of cell growth, survival and angiogenesis. Frequent mutations of this pathway make upstream and downstream components novel targets for tailored therapy design. Two mTOR inhibitors - everolimus and temsirolimus - enable an increase in overall survival (OS) or progression-free survival (PFS) time in a treatment of renal cancer. Despite recent advances in renal cancer treatment, resistance to targeted therapy is common. Understanding of molecular mechanisms is the basis of drug resistance which can facilitate prediction of success or failure in combinational or sequential targeted therapy. The article provides current knowledge on the mTOR signaling network and gives insight into the mechanisms of resistance to mTOR inhibitors from the complex perspective of RCC biology. The mechanisms of resistance developed not only by cancer cells, but also by interactions with tumor microenvironment are analyzed to emphasize the role of angiogenesis in ccRCC pathogenesis. As recent studies have shown the role of PI3K/AKT-mTOR pathway in proliferation and differentiation of cancer stem cells, we discuss cancer stem cell hypothesis and its possible contribution to ccRCC resistance. In the context of drug resistance, we also elaborate on a new approach considering ccRCC as a metabolic disease. In conclusion we speculate on future developments in agents targeting the mTOR pathway taking into consideration the singular biology of ccRCC.
Collapse
Affiliation(s)
- Anna Kornakiewicz
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- I Faculty of Medicine, Medical University of Warsaw,Poland
- Collegium Invisibile, Warsaw,Poland
| | - Wojciech Solarek
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw,Poland
| | - Zofia F. Bielecka
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw,Poland
| | - Fei Lian
- Department of Urology, Emory School of Medicine, Atlanta, GA ,USA
| | - Cezary Szczylik
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
| | - Anna M. Czarnecka
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
| |
Collapse
|
26
|
Subramani R, Lopez-Valdez R, Arumugam A, Nandy S, Boopalan T, Lakshmanaswamy R. Targeting insulin-like growth factor 1 receptor inhibits pancreatic cancer growth and metastasis. PLoS One 2014; 9:e97016. [PMID: 24809702 PMCID: PMC4014591 DOI: 10.1371/journal.pone.0097016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 04/15/2014] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers. Increasing incidence and mortality indicates that there is still much lacking in detection and management of the disease. This is partly due to a lack of specific symptoms during early stages of the disease. Several growth factor receptors have been associated with pancreatic cancer. Here, we have investigated if an RNA interference approach targeted to IGF-IR could be effective and efficient against pancreatic cancer growth and metastasis. For that, we evaluated the effects of IGF-1R inhibition using small interfering RNA (siRNAs) on tumor growth and metastasis in HPAC and PANC-1 pancreatic cancer cell lines. We found that silencing IGF-1R inhibits pancreatic cancer growth and metastasis by blocking key signaling pathways such AKT/PI3K, MAPK, JAK/STAT and EMT. Silencing IGF-1R resulted in an anti-proliferative effect in PANC-1 and HPAC pancreatic cancer cell lines. Matrigel invasion, transwell migration and wound healing assays also revealed a role for IGF-1R in metastatic properties of pancreatic cancer. These results were further confirmed using Western blotting analysis of key intermediates involved in proliferation, epithelial mesenchymal transition, migration, and invasion. In addition, soft agar assays showed that silencing IGF-1R also blocks the colony forming capabilities of pancreatic cancer cells in vitro. Western blots, as well as, flow cytometric analysis revealed the induction of apoptosis in IGF-1R silenced cells. Interestingly, silencing IGF-1R also suppressed the expression of insulin receptor β. All these effects together significantly control pancreatic cancer cell growth and metastasis. To conclude, our results demonstrate the significance of IGF-1R in pancreatic cancer.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Rebecca Lopez-Valdez
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Arunkumar Arumugam
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Sushmita Nandy
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Thiyagarajan Boopalan
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Rajkumar Lakshmanaswamy
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
- * E-mail:
| |
Collapse
|