1
|
AlBarakat MM, Ahmed YB, Alshwayyat S, Ellaithy A, Y. Al-Shammari Y, Soliman Y, Rezq H, Abdelazeem B, Kunadi A. The efficacy and safety of cabozantinib in patients with metastatic or advanced renal cell carcinoma: a systematic review and meta-analysis. Proc AMIA Symp 2024; 37:822-830. [PMID: 39165809 PMCID: PMC11332639 DOI: 10.1080/08998280.2024.2363616] [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: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 08/22/2024] Open
Abstract
Background Cabozantinib, a new first-line treatment for advanced renal cell carcinoma (aRCC), targets essential tyrosine kinases and outperforms the established comparator (sunitinib) in various efficacy outcomes. This systematic review and meta-analysis aimed to assess the efficacy and safety of cabozantinib compared to other aRCC treatments. Methods Following PRISMA and Cochrane guidelines, our protocol was registered in PROSPERO. A systematic search, without date limits, was conducted on PubMed, Cochrane, Web of Science, and EMBASE until October 8, 2023. Data extraction encompassed study details, baseline information, and outcomes. Hazard ratios (HR) and risk ratios (RR) with 95% confidence intervals were employed for each outcome, and a random-effects model was applied to account for expected heterogeneity. Results Three studies, encompassing 967 patients, were included in our analysis. In terms of efficacy, the pooled rate for overall survival significantly favored cabozantinib. However, in subgroup analyses, cabozantinib was only statistically superior to everolimus. For progression-free survival and tumor objective response rate, cabozantinib outperformed both everolimus and sunitinib. In adverse events, compared to sunitinib, cabozantinib exhibited inferiority in nearly all evaluated aspects, except for nausea and stomatitis, which showed no difference between the two groups. Conversely, it demonstrated a comparable risk profile with everolimus across various side effects. Conclusion Cabozantinib shows significant efficacy in extending overall survival, progression-free survival, and tumor objective response rate despite a potentially higher risk of adverse events compared to sunitinib. These findings support cabozantinib as a first-line therapy for aRCC, either as an initial treatment or after prior VEGFR-targeted therapies.
Collapse
Affiliation(s)
- Majd M. AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Yaman B. Ahmed
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Sakhr Alshwayyat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Asmaa Ellaithy
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | | | - Hazem Rezq
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Arvind Kunadi
- Internal Medicine and Nephrology Departments, McLaren Health Care, Flint, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
2
|
Zhang X, Zhang B, Zhang Y, Zhang F. Association analysis of hepatocellular carcinoma-related hub proteins and hub genes. Proteomics Clin Appl 2023; 17:e2200090. [PMID: 37050894 DOI: 10.1002/prca.202200090] [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: 10/26/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. The occurrence and development of HCC are closely related to epigenetic modifications. Epigenetic modifications can regulate gene expression and related functions through DNA methylation. This paper presents an association analysis method of HCC-related hub proteins and hub genes. EXPERIMENTAL DESIGN Bioinformatics analysis of HCC-related DNA methylation data is carried out to clarify the molecular mechanism of HCC-related genes and to find hub genes (genes with more connections in the network) by constructing in the gene interaction network. This paper proposes an accurate prediction method of protein-protein interaction (PPI) based on deep learning model DeepSG2PPI. The trained DeepSG2PPI model predicts the interaction relationship between the synthetic proteins regulated by HCC-related genes. RESULTS This paper finds that four genes are the intersection of hub genes and hub proteins. The four genes are: FBL, CCNB2, ALDH18A1, and RPLP0. The association of RPLP0 gene with HCC is a new finding of this study. RPLP0 is expected to become a new biomarker for the treatment, diagnosis, and prognosis of HCC. The four proteins corresponding to the four genes are: ENSP00000221801, ENSP00000288207, ENSP00000360268, and ENSP00000449328. CONCLUSIONS AND CLINICAL RELEVANCE The association between the hub genes with the hub proteins is analyzed. The mutual verification of the hub genes and the hub proteins can obtain more credible HCC-related genes and proteins, which is helpful for the diagnosis, treatment, and drug development of HCC.
Collapse
Affiliation(s)
- Xinhong Zhang
- School of Software, Henan University, Kaifeng, China
| | - Boyan Zhang
- School of Software, Henan University, Kaifeng, China
| | - Yawei Zhang
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| | - Fan Zhang
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| |
Collapse
|
3
|
Nixon AB, Halabi S, Liu Y, Starr MD, Brady JC, Shterev I, Luo B, Hurwitz HI, Febbo PG, Rini BI, Beltran H, Small EJ, Morris MJ, George DJ. Predictive Biomarkers of Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with IFNα ± Bevacizumab: Results from CALGB 90206 (Alliance). Clin Cancer Res 2022; 28:2771-2778. [PMID: 34965953 PMCID: PMC9240110 DOI: 10.1158/1078-0432.ccr-21-2386] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/25/2021] [Accepted: 12/21/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE CALGB 90206 was a phase III trial of 732 patients with metastatic renal cell carcinoma (mRCC) comparing bevacizumab plus IFNα (BEV + IFN) with IFNα alone (IFN). No difference in overall survival (OS) was observed. Baseline samples were analyzed to identify predictive biomarkers for survival benefit. PATIENTS AND METHODS A total of 32 biomarkers were assessed in 498 consenting patients randomly assigned into training (n = 279) and testing (n = 219) sets. The proportional hazards model was used to test for treatment arm and biomarker interactions of OS. The estimated coefficients from the training set were used to compute a risk score for each patient and to classify patients by risk in the testing set. The resulting model was assessed for predictive accuracy using the time-dependent area under the ROC curve (tAUROC). RESULTS A statistically significant three-way interaction between IL6, hepatocyte growth factor (HGF), and bevacizumab treatment was observed in the training set and confirmed in the testing set (P < 0.0001). The model based on IL6, HGF, and bevacizumab treatment was predictive of OS (P < 0.001), with the high- and low-risk groups having a median OS of 10.2 [95% confidence interval (CI), 8.0-13.8] and 34.3 (95% CI, 28.5-40.5) months, respectively. The average tAUROC for the final model of OS based on 100 randomly split testing sets was 0.78 (first, third quartiles = 0.77, 0.79). CONCLUSIONS IL6 and HGF are potential predictive biomarkers of OS benefit from BEV + IFN in patients with mRCC. The model based on key biological and clinical factors demonstrated predictive efficacy for OS. These markers warrant further validation in future anti-VEGF and immunotherapy in mRCC trials. See related commentaries by Mishkin and Kohn, p. 2722 and George and Bertagnolli, p. 2725.
Collapse
Affiliation(s)
- Andrew B. Nixon
- Department of Medical Oncology, Duke Cancer Institute, Duke University Medical Center; Durham, NC
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics and Alliance Statistics and Data Center, Duke University Medical Center; Durham, NC
| | - Yingmiao Liu
- Department of Medical Oncology, Duke Cancer Institute, Duke University Medical Center; Durham, NC
| | - Mark D. Starr
- Department of Medical Oncology, Duke Cancer Institute, Duke University Medical Center; Durham, NC
| | - John C. Brady
- Department of Medical Oncology, Duke Cancer Institute, Duke University Medical Center; Durham, NC
| | - Ivo Shterev
- Department of Biostatistics and Bioinformatics and Alliance Statistics and Data Center, Duke University Medical Center; Durham, NC
- Current address: Illumina, Redwood City, CA
| | - Bin Luo
- Department of Biostatistics and Bioinformatics and Alliance Statistics and Data Center, Duke University Medical Center; Durham, NC
| | | | | | - Brian I. Rini
- Department of Medicine, Cleveland Clinic Taussig Cancer Institute, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
| | - Himisha Beltran
- Department of Medicine, Dana-Farber/Partners Cancer Care, Harvard Cancer Center; Boston, MA
| | - Eric J. Small
- Department of Medicine, University of California, San Francisco; San Francisco, CA
| | - Michael J. Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel J. George
- Department of Medical Oncology, Duke Cancer Institute, Duke University Medical Center; Durham, NC
| |
Collapse
|
4
|
c-Met and EPHA7 Receptor Tyrosine Kinases Are Related to Prognosis in Clear Cell Renal Cell Carcinoma: Focusing on the Association with Myoferlin Expression. Cancers (Basel) 2022; 14:cancers14041095. [PMID: 35205843 PMCID: PMC8870418 DOI: 10.3390/cancers14041095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are important targets for clear cell renal cell carcinoma (ccRCC) treatment. Myoferlin is a strong regulator of RTKs. To identify myoferlin-associated RTKs and their prognostic implications in ccRCC, we investigated the expression of RTKs and myoferlin using proteome-based evaluation and immunohistochemical staining in tissue microarray. Multivariate Cox analysis adjusted for TNM stage and WHO grade was performed (n = 410 and 506). Proteomic analysis suggested c-Met and EPHA7 as novel candidates for myoferlin-associated RTKs. We immunohistochemically validated the positive association between c-Met and myoferlin expression. High c-Met expression was independently associated with overall (hazard ratio (HR) = 1.153-2.919) and cancer-specific survival (HR = 1.150-3.389). The prognostic effect of high c-Met expression was also determined in an independent cohort (overall survival, HR = 1.503-3.771). Although expression of EPHA7 and myoferlin was not correlated, EPHA7 expression was independently associated with progression-free (HR = 1.237-4.319) and cancer-specific survival (HR = 1.214-4.558). In addition, network-based prioritization showed co-functional enrichment of c-Met and myoferlin, suggesting a novel regulatory function of myoferlin in c-Met signaling. This study indicates that c-Met and EPHA7 might be useful prognostic biomarkers, and the presumed myoferlin/c-Met pathway could be a novel therapeutic target in ccRCC.
Collapse
|
5
|
Yang X, Liao HY, Zhang HH. Roles of MET in human cancer. Clin Chim Acta 2021; 525:69-83. [PMID: 34951962 DOI: 10.1016/j.cca.2021.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/18/2023]
Abstract
The MET proto-oncogene was first identified in osteosarcoma cells exposed to carcinogens. Although expressed in many normal cells, MET is overexpressed in many human cancers. MET is involved in the initiation and development of various human cancers and mediates proliferation, migration and invasion. Accordingly, MET has been successfully used as a biomarker for diagnosis and prognosis, survival, post-operative recurrence, risk assessment and pathologic grading, as well as a therapeutic target. In addition, recent work indicates that inhibition of MET expression and function has potential clinical benefit. This review summarizes the role, mechanism, and clinical significance of MET in the formation and development of human cancer.
Collapse
Affiliation(s)
- Xin Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
6
|
Brisset M, Grandin M, Bernet A, Mehlen P, Hollande F. Dependence receptors: new targets for cancer therapy. EMBO Mol Med 2021; 13:e14495. [PMID: 34542930 PMCID: PMC8573599 DOI: 10.15252/emmm.202114495] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 12/22/2022] Open
Abstract
Dependence receptors are known to promote survival and positive signaling such as proliferation, migration, and differentiation when activated, but to actively trigger apoptosis when unbound to their ligand. Their abnormal regulation was shown to be an important feature of tumorigenesis, allowing cancer cells to escape apoptosis triggered by these receptors while promoting in parallel major aspects of tumorigenesis such as proliferation, angiogenesis, invasiveness, and chemoresistance. This involvement in multiple cancer hallmarks has raised interest in dependence receptors as targets for cancer therapy. Although additional studies remain necessary to fully understand the complexity of signaling pathways activated by these receptors and to target them efficiently, it is now clear that dependence receptors represent very exciting targets for future cancer treatment. This manuscript reviews current knowledge on the contribution of dependence receptors to cancer and highlights the potential for therapies that activate pro-apoptotic functions of these proteins.
Collapse
Affiliation(s)
- Morgan Brisset
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| | - Mélodie Grandin
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| | - Agnès Bernet
- Apoptosis, Cancer and Development LaboratoryCentre de Recherche en Cancérologie de Lyon, INSERM U1052‐CNRS UMR5286Centre Léon BérardUniversité de LyonLyonFrance
| | - Patrick Mehlen
- Apoptosis, Cancer and Development LaboratoryCentre de Recherche en Cancérologie de Lyon, INSERM U1052‐CNRS UMR5286Centre Léon BérardUniversité de LyonLyonFrance
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| |
Collapse
|
7
|
Naung NY, Duncan WJ, De Silva RK, Coates DE. HGF/MET in osteogenic differentiation of primary human palatal periosteum-derived mesenchymal stem cells. J Oral Sci 2021; 63:341-346. [PMID: 34526445 DOI: 10.2334/josnusd.21-0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
PURPOSE This study aimed to determine expressions of hepatocyte growth factor (HGF) and MET proto-oncogene receptor tyrosine kinase (MET) in palatal periosteum (PP) and to examine the effect of HGF/MET on osteogenic differentiation of human palatal periosteum-derived mesenchymal stem cells (PD-MSCs). METHODS HGF/MET proteins in human palatal periosteum (n = 3) were localized using immunohistochemistry. PD-MSCs (n = 3) were cultured in serum-free Essential 8 (E8) medium or osteogenic medium with and without Capmatinib, a selective ATP-inhibitor of MET. HGF concentration in vitro was measured with ELISA. Relative gene expression was quantified from PD-MSCs by quantitative reverse transcription real-time polymerase chain reaction. RESULTS Immunohistochemistry detected co-localization of HGF and MET protein in PP. HGF protein levels were significantly higher (P < 0.05) in osteogenic media (day 21: 12.19 ± 8.36 ng/mL) than in E8 medium (day 21: 0.42 ± 0.72 ng/mL). MET inhibitor had a limited feedback effect on the expression profile of the osteogenic genes tested. Gene expression levels for all but three genes were comparable in serum-free and osteogenic media at all time points. CONCLUSION HGF/MET present in human PP and HGF is upregulated in vitro during osteogenesis; however the targeted pathways controlled by MET may not involve osteoblast maturation.
Collapse
Affiliation(s)
- Noel Ye Naung
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago.,Pun Hlaing Hospitals
| | - Warwick J Duncan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| | - Rohana K De Silva
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago
| |
Collapse
|
8
|
Chen JX, Xu D, Cao JW, Zuo L, Han ZT, Tian YJ, Chu CM, Zhou W, Pan XW, Cui XG. TRIM47 promotes malignant progression of renal cell carcinoma by degrading P53 through ubiquitination. Cancer Cell Int 2021; 21:129. [PMID: 33622324 PMCID: PMC7903798 DOI: 10.1186/s12935-021-01831-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is one of the most common malignant tumors originating from the renal parenchymal urinary epithelial system. Tripartite motif 47 (TRIM47) is a member of the TRIM family proteins, which has E3 ligase activity and has been demonstrated to be involved in the occurrence and prognosis of many tumors. The main purpose of this study is to explore the role and potential mechanism of TRIM47 in promoting malignant biological behavior of RCC. Materials and methods TRIM47 mRNA and protein levels in human renal cancer and paired normal adjacent tissues were detected by qRT-PCR and Western blot. The effects of TRIM47 knockdown and overexpression in renal cell carcinoma cells on cell proliferation, invasion and xenograft tumor growth in nude mice were analyzed. The molecular mechanism was explored by mass spectrometric exploration,Western blot and immunoprecipitation assays. Results TRIM47 promoted RCC cell proliferation in vitro and in vivo as an oncogene. Mechanistically, TRIM47 exerted an E3 ligase activity by interacting with P53 protein to increase its ubiquitination and degradation, which further promoted the malignant biological behavior of RCC. Conclusions Our study demonstrated that the TRIM47-P53 axis played a functional role in RCC progression and suggested a potential therapeutic target for RCC.
Collapse
Affiliation(s)
- Jia-Xin Chen
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai
| | - Da Xu
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai
| | - Jian-Wei Cao
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai
| | - Li Zuo
- Department of Urology, Changzhou Second People's Hospital, Changzhou, 213000, China
| | - Zhi-Tao Han
- Nanjing University of Traditional Chinese Medicine School of Medical and Life Sciences, Nanjing, 210023, China
| | - Yi-Jun Tian
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai
| | - Chuan-Min Chu
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai
| | - Wang Zhou
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai.
| | - Xiu-Wu Pan
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai.
| | - Xin-Gang Cui
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, 200433, China, Shanghai.
| |
Collapse
|
9
|
Chen S, Wang Y, Chen L, Xia Y, Cui J, Wang W, Jiang X, Wang J, Zhu Y, Sun S, Zou Y, Gong Y, Shi B. CUL4B promotes aggressive phenotypes of renal cell carcinoma via upregulating c-Met expression. Int J Biochem Cell Biol 2020; 130:105887. [PMID: 33227394 DOI: 10.1016/j.biocel.2020.105887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022]
Abstract
Cullin 4B (CUL4B), encoding a scaffold protein in Cullin RING ubiquitin-ligase complexes (CRL4B), is overexpressed and serves as an oncogene in various solid tumors. However, the roles and the underlying mechanisms of CUL4B in renal cell carcinoma (RCC) are still unknown. In this study, we demonstrated that CUL4B was significantly upregulated in RCC cells and clinical specimens, and its overexpression was correlated with poor survival of RCC patients. Knockdown of CUL4B resulted in the inhibition of proliferation, migration and invasion of RCC cells. Furthermore, we found that the expression of CUL4B is positively correlated with c-Met expression in RCC cells and tissues. Konckdown of c-Met or treatment with c-Met inhibitor, SU11274, could block the increase in cell proliferation, migration and invasion induced by CUL4B-overexpression. We also showed that CUL4B overexpression significantly accelerated xenograft tumor growth, and administration of SU11274 could also abrogate the accelerated tumor growth induced by CUL4B overexpression in vivo. These findings shed light on the contribution of CUL4B to tumorigenesis in RCC via activating c-Met signaling and its therapeutic implications in RCC patients.
Collapse
Affiliation(s)
- Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University, School of Basic Medical Sciences, Jinan, Shandong, 250012, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, Shandong, 250012, China
| | - Yong Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University, School of Basic Medical Sciences, Jinan, Shandong, 250012, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, Shandong, 250012, China
| | - Lipeng Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yangyang Xia
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jianfeng Cui
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Wenfu Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xuewen Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jian Wang
- Department of Urology, The People's Hospital of Laoling City, Dezhou, Shandong, 253600, China
| | - Yaofeng Zhu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shuna Sun
- Department of Dermatology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University, School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University, School of Basic Medical Sciences, Jinan, Shandong, 250012, China.
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
10
|
Fang P, Zhou L, Lim LY, Fu H, Yuan ZX, Lin J. Targeting Strategies for Renal Cancer Stem Cell Therapy. Curr Pharm Des 2020; 26:1964-1978. [PMID: 32188377 DOI: 10.2174/1381612826666200318153106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/27/2020] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) is an intractable genitourinary malignancy that accounts for approximately 4% of adult malignancies. Currently, there is no approved targeted therapy for RCC that has yielded durable remissions, and they remain palliative in intent. Emerging evidence has indicated that renal tumorigenesis and RCC treatment-resistance may originate from renal cancer stem cells (CSCs) with tumor-initiating capacity (CSC hypothesis). A better understanding of the mechanism underlying renal CSCs will help to dissect RCC heterogeneity and drug treatment efficiency, to promote more personalized and targeted therapies. In this review, we summarized the stem cell characteristics of renal CSCs. We outlined the targeting strategies and challenges associated with developing therapies that target renal CSCs angiogenesis, immunosuppression, signaling pathways, surface biomarkers, microRNAs and nanomedicine. In conclusion, CSCs are an important role in renal carcinogenesis and represent a valid target for treatment of RCC patients.
Collapse
Affiliation(s)
- Pengchao Fang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liuting Zhou
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lee Y Lim
- Department of Pharmacy, School of Medicine and Pharmacology, The University of Western Australia, Crawley WA 6009, Perth, Australia
| | - Hualin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Juchun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
11
|
Mukai S, Yamasaki K, Fujii M, Nagai T, Terada N, Kataoka H, Kamoto T. Dysregulation of Type II Transmembrane Serine Proteases and Ligand-Dependent Activation of MET in Urological Cancers. Int J Mol Sci 2020; 21:ijms21082663. [PMID: 32290402 PMCID: PMC7215454 DOI: 10.3390/ijms21082663] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 01/09/2023] Open
Abstract
Unlike in normal epithelium, dysregulated overactivation of various proteases have been reported in cancers. Degradation of pericancerous extracellular matrix leading to cancer cell invasion by matrix metalloproteases is well known evidence. On the other hand, several cell-surface proteases, including type II transmembrane serine proteases (TTSPs), also induce progression through activation of growth factors, protease activating receptors and other proteases. Hepatocyte growth factor (HGF) known as a multifunctional growth factor that upregulates cancer cell motility, invasiveness, proliferative, and anti-apoptotic activities through phosphorylation of MET (a specific receptor of HGF). HGF secreted as inactive zymogen (pro-HGF) from cancer associated stromal fibroblasts, and the proteolytic activation by several TTSPs including matriptase and hepsin is required. The activation is strictly regulated by HGF activator inhibitors (HAIs) in physiological condition. However, downregulation is frequently observed in cancers. Indeed, overactivation of MET by upregulation of matriptase and hepsin accompanied by the downregulation of HAIs in urological cancers (prostate cancer, renal cell carcinoma, and bladder cancer) are also reported, a phenomenon observed in cancer cells with malignant phenotype, and correlated with poor prognosis. In this review, we summarized current reports focusing on TTSPs, HAIs, and MET signaling axis in urological cancers.
Collapse
Affiliation(s)
- Shoichiro Mukai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
- Correspondence: ; Tel.: +81-985-85-2968
| | - Koji Yamasaki
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
| | - Masato Fujii
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
| | - Takahiro Nagai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
| | - Naoki Terada
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
| | - Hiroaki Kataoka
- Oncopathology and Regenerative Biology Section, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Toshiyuki Kamoto
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan; (K.Y.); (M.F.); (T.N.); (N.T.); (T.K.)
| |
Collapse
|
12
|
Prisciandaro M, Ratta R, Massari F, Fornarini G, Caponnetto S, Iacovelli R, De Giorgi U, Facchini G, Scagliarini S, Sabbatini R, Caserta C, Peverelli G, Mennitto A, Verzoni E, Procopio G. Safety and Efficacy of Cabozantinib for Metastatic Nonclear Renal Cell Carcinoma: Real-world Data From an Italian Managed Access Program. Am J Clin Oncol 2019; 42:42-45. [PMID: 30204614 DOI: 10.1097/coc.0000000000000478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The activity of cabozantinib in nonclear cell histologies has not been evaluated. MATERIALS AND METHODS Data were collected across 24 Italian hospitals. Patients were aged 18 years and older with advanced nonclear cell renal cell carcinoma (RCC), with an Eastern Cooperative Oncology Group Performance Status 0 to 2, who had relapsed after previous systemic treatments for metastatic disease. Cabozantinib was administered orally at 60 mg once a day in 28 days cycles. Dose reductions to 40 or 20 mg were made due to toxicity. Adverse events (AEs) were monitored using CTCAE version 4.0. RESULTS Seventeen patients were enrolled. Three (18%) patients were diagnosed type I papillary RCC, 9 (53%) type II papillary, 3 (18%) chromophobe, and 2 (11%) with Bellini duct carcinoma. In total, 11 patients started with 60 mg. Six patients started a lower dose of 40 mg. Median progression-free survival was 7.83 months (0.4 to 13.4 mo), while median overall survival was not reached but 1-year overall survival was about 60%. Six patients (35%) experienced a partial response to treatment and 6 patients (35%) showed a stable disease. In the remaining 5 (30%), we observed a progressive disease. Grade 3 and 4 AEs were observed in 41% of patients. Among 20 patients, only 1 (6%) discontinued treatment due to AEs. Asthenia (41%), diarrhea (35%), aminotransferase increasing (35%), mucosal inflammation (35%), hand and foot syndrome (24%), and hypothyroidism (24%) were the most frequently AEs. CONCLUSIONS Our data showed that, cabozantinib is a active and feasible treatment in patient with nonclear cell RCC.
Collapse
Affiliation(s)
- Michele Prisciandaro
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - Raffaele Ratta
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | | | - Giuseppe Fornarini
- Medical Oncology Department, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genova
| | - Salvatore Caponnetto
- Department of Medical Oncology B, Policlinico Umberto I "Sapienza" University of Rome, Rome
| | - Roberto Iacovelli
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Integrata (AOUI), University of Verona, Verona
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola
| | - Gaetano Facchini
- Department of Uro-Gynaecological Oncology, Division of Medical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale (IRCCS)
| | | | - Roberto Sabbatini
- Department of Oncology and Haematology and Respiratory Disease, University Hospital, Modena
| | | | - Giorgia Peverelli
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - Alessia Mennitto
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - Elena Verzoni
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - Giuseppe Procopio
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| |
Collapse
|
13
|
Huaqi Y, Caipeng Q, Qiang W, Yiqing D, Xiang D, Xu T, Xiaowei Z, Qing L, Shijun L, Tao X. Transcription Factor SOX18 Promotes Clear Cell Renal Cell Carcinoma Progression and Alleviates Cabozantinib-Mediated Inhibitory Effects. Mol Cancer Ther 2019; 18:2433-2445. [PMID: 31527225 DOI: 10.1158/1535-7163.mct-19-0043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/12/2019] [Accepted: 09/09/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Yin Huaqi
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Qin Caipeng
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Wang Qiang
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Du Yiqing
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Dai Xiang
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Tang Xu
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Zhang Xiaowei
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Li Qing
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Liu Shijun
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China
| | - Xu Tao
- Department of Urology, Peking University People's Hospital, Peking University Second School of Clinical Medicine, Beijing, China.
| |
Collapse
|
14
|
Du M, Wang J, Chen H, Wang S, Chen L, Xu Y, Su F, Lu X. MicroRNA‑200a suppresses migration and invasion and enhances the radiosensitivity of NSCLC cells by inhibiting the HGF/c‑Met signaling pathway. Oncol Rep 2018; 41:1497-1508. [PMID: 30569179 PMCID: PMC6365696 DOI: 10.3892/or.2018.6925] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Hepatocyte growth factor (HGF), an activator of the c‑Met signaling pathway, is involved in tumor invasiveness, metastasis and radiotherapy resistance. In the present study, a novel HGF regulatory pathway in lung cancer involving micro-RNAs (miRNAs/miR) is described. Immunohistochemical staining and western blot analyses demonstrated that HGF was upregulated and associated with miR‑200a downregulation in non‑small cell lung cancer (NSCLC) samples compared with normal lung tissues. The association between HGF and miR‑200a was associated with the degree of tumor malignancy and cell migration and invasion. miR‑200a negatively regulated HGF expression by targeting the 3'‑untranslated region of the HGF mRNA. miR‑200a overexpression induced HGF downregulation, decreased NSCLC cell migration and invasion, promoted apoptosis, and decreased cell survival in A549 and H1299 cells in response to ionizing radiation. The present results revealed a previously uncharacterized role of miRNA‑200a in regulating tumor malignancy and radiosensitivity by suppressing HGF expression, a key factor in the HGF/c‑Met pathway.
Collapse
Affiliation(s)
- Menghua Du
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Jin Wang
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Huan Chen
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Shouli Wang
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Liesong Chen
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Yichang Xu
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Fengtao Su
- Cancer Institute, Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Xueguan Lu
- Department of Oncology and Radiotherapy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
15
|
Bersanelli M, Leonardi F, Buti S. Spotlight on cabozantinib for previously untreated advanced renal cell carcinoma: evidence to date. Cancer Manag Res 2018; 10:3773-3780. [PMID: 30288108 PMCID: PMC6159801 DOI: 10.2147/cmar.s160485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background In the past few years, new drugs made their appearance in the first-line setting of treatment for metastatic renal cell carcinoma (mRCC), and cabozantinib is one among them. The present systematic review aims to point out any evidence published to date about first-line treatment with cabozantinib for mRCC patients, describing their outcome in all end points explored by the literature. Methods PRISMA guidelines were followed. A systematic assessment of literature and peer- reviewed presentations was performed by searching PubMed and major oncology meeting resources, from the database inception until June 25, 2018. The following keywords were used: "cabozantinib or cabozantinib-s-malate or XL184" and "renal cell carcinoma or kidney cancer or clear cell renal carcinoma or renal cancer" and "first-line or untreated or treatment-naïve or primary treatment". All types of original clinical studies were included, evaluating either cabozantinib monotherapy or any systemic drug combination containing cabozantinib for previously untreated patients with mRCC. Results From potential 75 titles and abstracts, seven publications were selected. One was the main report of a randomized clinical trial (the CABOSUN study); four papers reported updated results, secondary or subgroup analyses from the same study population; and further two reports consisted of network meta-analyses. From the additional search for ongoing clinical trials, six studies currently in progress were reported. Conclusion According to the reported evidence, cabozantinib may be a viable first-line option in mRCC patients with intermediate or poor risk according to International Metastatic Renal Cell Carcinoma Database Consortium model. It offers an undoubtful advantage in terms of progression-free survival, despite quite high rates of G3-4 toxicity, modest objective response rate, and no survival advantage. Nevertheless, given the availability of an immunotherapy combination that significantly improved overall survival for the same population in a Phase III trial and the indisputable efficacy of cabozantinib as second-line treatment, this drug may be devoted as a rescue option in patients progressive to primary therapy.
Collapse
Affiliation(s)
| | | | - Sebastiano Buti
- University Hospital of Parma, Medical Oncology Unit, Parma, Italy,
| |
Collapse
|
16
|
Procopio G, Prisciandaro M, Iacovelli R, Cortesi E, Fornarini G, Facchini G, Cartenì G, Sabbatini R, Del Bene G, Galli L, Caserta C, Multari AG, Bregni M, Massari F, Buti S, De Giorgi U, Zustovich F, Milella M, Calabrò F, Mancini ML, Tortora G, Vernieri C, Santini D, Sorarù M, Ricotta R, Masini C, Tucci M, Fedeli SL, Ortega C, Mecozzi A, Ratta R, Sternberg CN, Verzoni E. Safety and Efficacy of Cabozantinib in Metastatic Renal-Cell Carcinoma: Real-World Data From an Italian Managed Access Program. Clin Genitourin Cancer 2018; 16:e945-e951. [DOI: 10.1016/j.clgc.2018.03.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/20/2018] [Accepted: 03/23/2018] [Indexed: 12/15/2022]
|
17
|
Correlation of c-MET Expression with PD-L1 Expression in Metastatic Clear Cell Renal Cell Carcinoma Treated by Sunitinib First-Line Therapy. Target Oncol 2018; 12:487-494. [PMID: 28550387 DOI: 10.1007/s11523-017-0498-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is highly metastatic. Cabozantinib, an anti-angiogenic tyrosine kinase inhibitor that targets c-MET, provided interesting results in metastatic ccRCC treatment. OBJECTIVE To understand better the role of c-MET in ccRCC, we assessed its status in a population of patients with metastatic ccRCC. PATIENTS AND METHODS For this purpose, tumor samples were analyzed for c-MET expression by immunohistochemistry (IHC), for c-MET copy number alterations by fluorescence in situ hybridization (FISH), and for c-MET mutations by next generation sequencing (NGS) in a retrospective cohort of 90 primary ccRCC of patients with metastases treated by first-line sunitinib. The expression of c-MET was correlated with pathological, immunohistochemical (VEGFA, CAIX, PD-L1), clinical, and molecular criteria (VHL status) by univariate and multivariate analyses and to clinical outcome using Kaplan-Meier curves compared by log-rank test. RESULTS Of ccRCC, 31.1% had low c-MET expression (absent to weak intensity by IHC) versus 68.9% with high expression (moderate to strong intensity). High expression of c-MET was associated with a gain in FISH analysis (p=0.0284) without amplification. No mutations were detected in NGS. Moreover, high c-MET expression was associated with lymph node metastases (p=0.004), sarcomatoid component (p=0.029), VEGFA (p=0.037), and PD-L1 (p=0.001) overexpression, the only factor that remained independently associated (p<0.001) after logistic regression. No difference was observed in clinical outcomes. CONCLUSION This study is the first to analyse c-MET status in metastatic ccRCC. The high expression of c-MET in the majority of ccRCC and its independent association with PD-L1 expression, may suggest a potential benefit from combining c-MET inhibitors and targeted immunotherapy.
Collapse
|
18
|
Bhandari A, Xia E, Zhou Y, Guan Y, Xiang J, Kong L, Wang Y, Yang F, Wang O, Zhang X. ITGA7 functions as a tumor suppressor and regulates migration and invasion in breast cancer. Cancer Manag Res 2018; 10:969-976. [PMID: 29760566 PMCID: PMC5937492 DOI: 10.2147/cmar.s160379] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Breast cancer is the most common malignancy in women and the underlying mechanism of breast cancer cell metastasis is still far from uncover. Integrin subunit alpha 7 (ITGA7) is a functioning protein. It has been detected in many malignancies. But the function of ITGA7 in breast cancer is not clear. Our aim is to explore ITGA7 expression and its role in breast cancer. Methods Real-time PCR was performed to determine ITGA7 expression in BC tissues and normal adjacent tissues. The specific functions of ITGA7 in breast cancer cell lines (MDA-MB-231 and BT-549) transfected with small interfering RNA were determined through migration, invasion assays. Western blot assays were performed to determine the expression of c-met and vimentin. Results ITGA7 was down-regulated in breast cancer tissues compared to the adjacent normal tissues (T:N =7.68±27.38: 41.01± 31.47, P<0.001) and this observation was consistent with the TCGA cohort (T:N =4.51±0.45:5.40±0.61, P<0.0001). In vitro experiments showed that knocking down ITGA7 significantly inhibited the migration and invasion of the breast cancer cell lines (MDA-MB-231 and BT-549). Meanwhile, knockdown of ITGA7 promoted c-met and vimentin expression, which may induce invasion and migration. Conclusion ITGA7 plays an important tumorigenic function and acts as a suppress gene in breast cancer. Our findings indicate that ITGA7 was the gene associated with breast cancer.
Collapse
Affiliation(s)
- Adheesh Bhandari
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Erjie Xia
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yuying Zhou
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yaoyao Guan
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Jingjing Xiang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Lingguo Kong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yinghao Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Fan Yang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Ouchen Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaohua Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Large-scale genomic profiling has shed new light on the molecular underpinnings of renal cell carcinoma (RCC), spurring a much needed refinement of RCC subclassification based on an integrative assessment of histopathologic features and molecular alterations. At the same time, renal mass biopsies have become increasingly commonplace, necessitating ancillary tools to help guide clinical management. Herein, we briefly review our current understanding of RCC genomics, highlighting areas of possible clinical utility, as well as potential limitations, for renal mass biopsies. RECENT FINDINGS Distinct RCC subtypes harbor characteristic molecular features, including somatic mutations, copy number alterations, and genomic rearrangements. Existing ancillary tools, including fluorescent in-situ hybridization and immunohistochemistry, may be useful for diagnostic subclassification. Recurrent secondary molecular alterations in clear cell RCC (BAP1, SETD2, PBRM1, and TP53) and papillary RCC (CDKN2A) may be associated with poor prognosis; however, intratumoral genomic heterogeneity may limit the clinical utility of these molecular biomarkers in renal mass biopsies. SUMMARY Recent technological advances have the potential to fundamentally alter the clinical management of RCC by leveraging our increasing understanding of RCC genomics to assess hundreds of molecular biomarkers simultaneously. Additional focused molecular analyses of renal mass biopsy cohorts are needed prior to widespread implementation of molecular biomarker assays.
Collapse
|
20
|
Guo T, Zhao S, Wang P, Xue X, Zhang Y, Yang M, Li N, Li Z, Xu L, Jiang L, Zhao L, Ma PC, Rosell R, Li J, Gu C. YB-1 regulates tumor growth by promoting MACC1/c-Met pathway in human lung adenocarcinoma. Oncotarget 2018. [PMID: 28624808 PMCID: PMC5564630 DOI: 10.18632/oncotarget.18262] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aberrant overexpression of the transcription/translation factor Y-box-binding protein (YB-1) is associated with poor prognosis of lung adenocarcinoma, however the underlying mechanism by which YB-1 acts has not been fully elucidated. Here, we reported that inhibition of YB-1 diminished proliferation, migration and invasion of lung adenocarcinoma cells. Interestingly, we identified metastasis associated in colon cancer-1 (MACC1) as a target of YB-1. Depletion of YB-1 markedly decreased MACC1 promoter activity and suppressed the MACC1/c-Met signaling pathway in lung adenocarcinoma cells. Additionally, chromatin immunoprecipitation (ChIP) assay demonstrated that YB-1 bound to the MACC1 promoter. Moreover, YB-1 was positively correlated with MACC1, and both proteins were over-expressed in lung adenocarcinoma tissues. The Cox-regression analysis indicated that high YB-1 expression was an independent risk factor for prognosis in enrolled patients. Furthermore, depletion of YB-1 attenuated tumorigenesis in a xenograft mouse model and reduced MACC1 expression in tumor tissues. Collectively, our data suggested that targeting YB-1 suppressed lung adenocarcinoma progression through the MACC1/c-Met pathway and that the high expression of YB-1/MACC1 is a potential prognostic marker in lung adenocarcinoma.
Collapse
Affiliation(s)
- Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Peng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoyuan Xue
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yan Zhang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Mengying Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Nan Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lingzhi Xu
- The Second Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Lei Jiang
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei 230000, China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Patrick C Ma
- Aerodigestive Oncology Translational Research THOR, Department of Solid Tumor Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rafael Rosell
- Breakthrough Cancer Research Unit, Pangaea Biotech, Dexeus University Institute, Catalan Institute of Oncology, Badalona 08916, Spain
| | - Jinxiu Li
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
21
|
Cella D, Escudier B, Tannir NM, Powles T, Donskov F, Peltola K, Schmidinger M, Heng DY, Mainwaring PN, Hammers HJ, Lee JL, Roth BJ, Marteau F, Williams P, Baer J, Mangeshkar M, Scheffold C, Hutson TE, Pal S, Motzer RJ, Choueiri TK. Quality of Life Outcomes for Cabozantinib Versus Everolimus in Patients With Metastatic Renal Cell Carcinoma: METEOR Phase III Randomized Trial. J Clin Oncol 2018; 36:757-764. [PMID: 29377755 PMCID: PMC6804841 DOI: 10.1200/jco.2017.75.2170] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose In the phase III METEOR trial ( ClinicalTrials.gov identifier: NCT01865747), 658 previously treated patients with advanced renal cell carcinoma were randomly assigned 1:1 to receive cabozantinib or everolimus. The cabozantinib arm had improved progression-free survival, overall survival, and objective response rate compared with everolimus. Changes in quality of life (QoL), an exploratory end point, are reported here. Patients and Methods Patients completed the 19-item Functional Assessment of Cancer Therapy-Kidney Symptom Index (FKSI-19) and the five-level EuroQol (EQ-5D-5L) questionnaires at baseline and throughout the study. The nine-item FKSI-Disease-Related Symptoms (FKSI-DRS), a subset of FKSI-19, was also investigated. Data were summarized descriptively and by repeated-measures analysis (for which a clinically relevant difference was an effect size ≥ 0.3). Time to deterioration (TTD) was defined as the earlier of date of death, radiographic progressive disease, or ≥ 4-point decrease from baseline in FKSI-DRS. Results The QoL questionnaire completion rates remained ≥ 75% through week 48 in each arm. There was no difference over time for FKSI-19 Total, FKSI-DRS, or EQ-5D data between the cabozantinib and everolimus arms. Among the individual FKSI-19 items, cabozantinib was associated with worse diarrhea and nausea; everolimus was associated with worse shortness of breath. These differences are consistent with the adverse event profile of each drug. Cabozantinib improved TTD overall, with a marked improvement in patients with bone metastases at baseline. Conclusion In patients with advanced renal cell carcinoma, relative to everolimus, cabozantinib generally maintained QoL to a similar extent. Compared with everolimus, cabozantinib extended TTD overall and markedly improved TTD in patients with bone metastases.
Collapse
Affiliation(s)
- David Cella
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Bernard Escudier
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Nizar M. Tannir
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Powles
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Frede Donskov
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Katriina Peltola
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Manuela Schmidinger
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel Y.C. Heng
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Paul N. Mainwaring
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Hans J. Hammers
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Jae Lyun Lee
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Bruce J. Roth
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Florence Marteau
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Paul Williams
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - John Baer
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Milan Mangeshkar
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Christian Scheffold
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas E. Hutson
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Sumanta Pal
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J. Motzer
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Toni K. Choueiri
- David Cella, Northwestern University Feinberg School of Medicine, Chicago, IL; Bernard Escudier, Institut Gustave Roussy, Villejuif; Florence Marteau, Ipsen Pharma, Boulogne-Billancourt; Paul Williams, Mapi Group, Lyon, France; Nizar M. Tannir, University of Texas MD Anderson Cancer Center Hospital, Houston; Hans J. Hammers, University of Texas Southwestern Medical Center; Thomas E. Hutson, Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX; Thomas Powles, Queen Mary University of London, London, United Kingdom; Frede Donskov, Aarhus University Hospital, Aarhus, Denmark; Katriina Peltola, Helsinki University Hospital Cancer Center, Helsinki, Finland; Manuela Schmidinger, Medical University of Vienna, Vienna, Austria; Daniel Y.C. Heng, University of Calgary, Calgary, Alberta, Canada; Paul N. Mainwaring, Icon Cancer Care, Brisbane, Queensland, Australia; Jae Lyun Lee, University of Ulsan College of Medicine, Seoul, South Korea; Bruce J. Roth, Washington University in St Louis, St Louis, MO; John Baer, Milan Mangeshkar, and Christian Scheffold, Exelixis, South San Francisco; Sumanta Pal, City of Hope National Medical Center, Duarte, CA; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
22
|
Lalani AKA, Gray KP, Albiges L, Callea M, Pignon JC, Pal S, Gupta M, Bhatt RS, McDermott DF, Atkins MB, Woude GFV, Harshman LC, Choueiri TK, Signoretti S. Differential expression of c-Met between primary and metastatic sites in clear-cell renal cell carcinoma and its association with PD-L1 expression. Oncotarget 2017; 8:103428-103436. [PMID: 29262573 PMCID: PMC5732739 DOI: 10.18632/oncotarget.21952] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
In preclinical models, c-Met promotes survival of renal cancer cells through the regulation of programmed death-ligand 1 (PD-L1). However, this relationship in human clear cell renal cell carcinoma (ccRCC) is not well characterized. We evaluated c-Met expression in ccRCC patients using paired primary and metastatic samples and assessed the association with PD-L1 expression and other clinical features. Areas with predominant and highest Fuhrman nuclear grade (FNG) were selected. c-Met expression was evaluated by IHC using an anti-Met monoclonal antibody (MET4 Ab) and calculated by a combined score (CS, 0-300): intensity of c-Met staining (0-3) x % of positive cells (0-100). PD-L1 expression in tumor cells was previously assessed by IHC and PD-L1+ was defined as PD-L1 > 0% positive cells. Our cohort consisted of 45 pairs of primary and metastatic ccRCC samples. Overall, c-Met expression was higher in metastatic sites compared to primary sites (average c-Met CS: 55 vs. 28, p = 0.0003). Higher c-Met expression was associated with higher FNG (4 vs. 3) in primary tumors (average c-Met CS: 52 vs. 20, p = 0.04). c-Met expression was numerically greater in PD-L1+ vs. PD-L1- tumors. Higher c-Met expression in metastatic sites compared to primary tumors suggests that testing for biomarkers of response to c-Met inhibitors should be conducted in metastases. While higher c-Met expression in PD-L1+ tumors requires further investigation, it supports exploring these targets in combination clinical trials.
Collapse
Affiliation(s)
- Aly-Khan A Lalani
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathryn P Gray
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Marcella Callea
- Department of Pathology, Ospedale San Raffaele, Milan, Italy
| | | | - Soumitro Pal
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
| | - Mamta Gupta
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rupal S Bhatt
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David F McDermott
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington D.C., USA
| | | | - Lauren C Harshman
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
23
|
Abstract
The influx of multiple novel therapeutic options in the mRCC field has brought a challenge for treatment sequencing in this disease. In the past few years, cabozantinib, nivolumab and the combination of lenvatinib and everolimus have been approved in the second-line setting. As there is no direct comparison between these agents and the studies have failed to show improved benefit among a biomarker-selected patient population, appropriate patient selection based on clinical factors for individualized therapy is critical. Herein we provide a comprehensive overview of current data from each agent through the discussion of disease biology, clinical trials, potential biomarkers and distilling future perspectives in the field.
Collapse
Affiliation(s)
- Manuel Caitano Maia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Nazli Dizman
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Meghan Salgia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Sumanta Kumar Pal
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
24
|
Chen S, Zhu Y, Cui J, Wang Y, Xia Y, Song J, Cheng S, Zhou C, Zhang D, Zhang B, Shi B. The role of c-Met in prognosis and clinicopathology of renal cell carcinoma: Results from a single-centre study and systematic review. Urol Oncol 2017; 35:532.e15-532.e23. [PMID: 28427859 DOI: 10.1016/j.urolonc.2017.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 03/12/2017] [Accepted: 03/21/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND OBJECTIVES The c-Met proto-oncogene pathway plays an important role in the progression of various cancers. However, the effect of the c-Met pathway on renal cell carcinoma (RCC) remains controversial. We decided to clarify the role of c-Met in prognosis and clinicopathology of RCC. METHODS A total of 10 pairs of tumour and adjacent tissues were obtained from patients with primary RCC between 2013 and 2014 and tissue microarrays to assess c-Met expression in tumour tissues from 90 patients with RCC by Western blot and immunohistochemical staining. We also presented a meta-analysis to explore the correlation between c-Met and pathological grade and stage of RCC. The two-tailed Pearson's χ2 and Fischer exact tests were used to compare categorical variables. Multivariate analysis was performed using the multivariate Cox proportional hazards model. RESULTS C-Met protein levels were increased in 8 of 10 RCC tissue samples compared with their adjacent normal tissue and c-Met expression levels were positively associated with a high nuclear grade (P = 0.008) and pT stage (P = 0.002). Multivariate analysis showed that a high expression of c-Met was an independent predictor of disease-specific survival (P = 0.017). A meta-analysis found that increased c-Met expression in RCC tissues was closely correlated with high tumour grade (P<0.001) and high pT stage (P = 0.001). Most importantly, c-Met expression was significantly correlated with disease-specific survival (P<0.001). CONCLUSIONS Because c-Met is strongly associated with pathological grade, stage and disease-specific survival, c-Met levels may have potential to predict patient prognosis and to guide clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Shouzhen Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yaofeng Zhu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Jianfeng Cui
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yong Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yangyang Xia
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Jing Song
- Laboratory of Experimental Teratology, Ministry of Education and Institute of Experimental Nuclear Medicine, School of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Shanshan Cheng
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Changkuo Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Dongqing Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Bing Zhang
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, People's Republic of China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
25
|
Sustained inhibition of cMET-VEGFR2 signaling using liposome-mediated delivery increases efficacy and reduces toxicity in kidney cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1853-1861. [DOI: 10.1016/j.nano.2016.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 03/17/2016] [Accepted: 04/04/2016] [Indexed: 11/20/2022]
|
26
|
Grassi P, Verzoni E, Ratta R, Mennitto A, de Braud F, Procopio G. Cabozantinib in the treatment of advanced renal cell carcinoma: design, development, and potential place in the therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2167-72. [PMID: 27462141 PMCID: PMC4939993 DOI: 10.2147/dddt.s104225] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The treatment of metastatic renal cell carcinoma (mRCC) has markedly improved over the last few years with the introduction of several targeted agents in clinical practice. Nevertheless, either primary or secondary resistance to inhibition of VEGF and mTOR pathways has limited the clinical benefit of these systemic treatments. Recently, a better understanding of the involvement of MET and its ligand HGF in many biological processes made this signaling pathway an attractive therapeutic target in oncology, particularly in mRCC. Herein, we review the development of cabozantinib, a recently approved inhibitor of multiple tyrosine kinase receptors, including MET, VEGFRs, and AXL, which has proven to increase progression-free survival and overall survival when compared to everolimus in mRCC patients who had progressed after VEGFR-targeted therapy. Finally, we discuss the potential role of cabozantinib within the current treatment landscape for mRCC.
Collapse
Affiliation(s)
- Paolo Grassi
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| | - Raffaele Ratta
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| | - Alessia Mennitto
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Genitourinary Cancer Unit, Fondazione IRCCS Istituto nazionale Tumori, Milan, Italy
| |
Collapse
|
27
|
Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, Lee JL, Heng DYC, Schmidinger M, Agarwal N, Sternberg CN, McDermott DF, Aftab DT, Hessel C, Scheffold C, Schwab G, Hutson TE, Pal S, Motzer RJ. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol 2016; 17:917-927. [PMID: 27279544 DOI: 10.1016/s1470-2045(16)30107-3] [Citation(s) in RCA: 695] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/18/2016] [Accepted: 04/21/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cabozantinib is an oral inhibitor of tyrosine kinases including MET, VEGFR, and AXL. The randomised phase 3 METEOR trial compared the efficacy and safety of cabozantinib versus the mTOR inhibitor everolimus in patients with advanced renal cell carcinoma who progressed after previous VEGFR tyrosine-kinase inhibitor treatment. Here, we report the final overall survival results from this study based on an unplanned second interim analysis. METHODS In this open-label, randomised phase 3 trial, we randomly assigned (1:1) patients aged 18 years and older with advanced or metastatic clear-cell renal cell carcinoma, measurable disease, and previous treatment with one or more VEGFR tyrosine-kinase inhibitors to receive 60 mg cabozantinib once a day or 10 mg everolimus once a day. Randomisation was done with an interactive voice and web response system. Stratification factors were Memorial Sloan Kettering Cancer Center risk group and the number of previous treatments with VEGFR tyrosine-kinase inhibitors. The primary endpoint was progression-free survival as assessed by an independent radiology review committee in the first 375 randomly assigned patients and has been previously reported. Secondary endpoints were overall survival and objective response in all randomly assigned patients assessed by intention-to-treat. Safety was assessed per protocol in all patients who received at least one dose of study drug. The study is closed for enrolment but treatment and follow-up of patients is ongoing for long-term safety evaluation. This trial is registered with ClinicalTrials.gov, number NCT01865747. FINDINGS Between Aug 8, 2013, and Nov 24, 2014, 658 patients were randomly assigned to receive cabozantinib (n=330) or everolimus (n=328). The median duration of follow-up for overall survival and safety was 18·7 months (IQR 16·1-21·1) in the cabozantinib group and 18·8 months (16·0-21·2) in the everolimus group. Median overall survival was 21·4 months (95% CI 18·7-not estimable) with cabozantinib and 16·5 months (14·7-18·8) with everolimus (hazard ratio [HR] 0·66 [95% CI 0·53-0·83]; p=0·00026). Cabozantinib treatment also resulted in improved progression-free survival (HR 0·51 [95% CI 0·41-0·62]; p<0·0001) and objective response (17% [13-22] with cabozantinib vs 3% [2-6] with everolimus; p<0·0001) per independent radiology review among all randomised patients. The most common grade 3 or 4 adverse events were hypertension (49 [15%] in the cabozantinib group vs 12 [4%] in the everolimus group), diarrhoea (43 [13%] vs 7 [2%]), fatigue (36 [11%] vs 24 [7%]), palmar-plantar erythrodysaesthesia syndrome (27 [8%] vs 3 [1%]), anaemia (19 [6%] vs 53 [17%]), hyperglycaemia (3 [1%] vs 16 [5%]), and hypomagnesaemia (16 [5%] vs none). Serious adverse events grade 3 or worse occurred in 130 (39%) patients in the cabozantinib group and in 129 (40%) in the everolimus group. One treatment-related death occurred in the cabozantinib group (death; not otherwise specified) and two occurred in the everolimus group (one aspergillus infection and one pneumonia aspiration). INTERPRETATION Treatment with cabozantinib increased overall survival, delayed disease progression, and improved the objective response compared with everolimus. Based on these results, cabozantinib should be considered as a new standard-of-care treatment option for previously treated patients with advanced renal cell carcinoma. Patients should be monitored for adverse events that might require dose modifications. FUNDING Exelixis Inc.
Collapse
Affiliation(s)
| | | | - Thomas Powles
- Barts Cancer Institute, Cancer Research UK Experimental Cancer Medicine Centre, Queen Mary University of London, Royal Free NHS Trust, London, UK
| | - Nizar M Tannir
- University of Texas, MD Anderson Cancer Center Hospital, Houston, TX, USA
| | | | | | - Hans J Hammers
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | | | - Bruce J Roth
- Washington University in St Louis, St Louis, MO, USA
| | - Katriina Peltola
- Helsinki University Central Hospital Cancer Center, Helsinki, Finland
| | - Jae Lyun Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Daniel Y C Heng
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute at The University of Utah, Salt Lake City, Utah
| | | | | | | | | | | | | | - Thomas E Hutson
- Texas Oncology-Baylor Sammons Cancer Center, Dallas, TX, USA
| | - Sumanta Pal
- City of Hope National Medical Center, Duarte, CA, USA
| | | | | |
Collapse
|
28
|
Zhang T, Boominathan R, Foulk B, Rao C, Kemeny G, Strickler JH, Abbruzzese JL, Harrison MR, Hsu DS, Healy P, Li J, Pi C, Prendergast KM, Hobbs C, Gemberling S, George DJ, Hurwitz HI, Connelly M, Garcia-Blanco MA, Armstrong AJ. Development of a Novel c-MET-Based CTC Detection Platform. Mol Cancer Res 2016; 14:539-47. [PMID: 26951228 DOI: 10.1158/1541-7786.mcr-16-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 02/25/2016] [Indexed: 11/16/2022]
Abstract
UNLABELLED Amplification of the MET oncogene is associated with poor prognosis, metastatic dissemination, and drug resistance in many malignancies. We developed a method to capture and characterize circulating tumor cells (CTC) expressing c-MET using a ferromagnetic antibody. Immunofluorescence was used to characterize cells for c-MET, DAPI, and pan-CK, excluding CD45(+) leukocytes. The assay was validated using appropriate cell line controls spiked into peripheral blood collected from healthy volunteers (HV). In addition, peripheral blood was analyzed from patients with metastatic gastric, pancreatic, colorectal, bladder, renal, or prostate cancers. CTCs captured by c-MET were enumerated, and DNA FISH for MET amplification was performed. The approach was highly sensitive (80%) for MET-amplified cells, sensitive (40%-80%) for c-MET-overexpressed cells, and specific (100%) for both c-MET-negative cells and in 20 HVs. Of 52 patients with metastatic carcinomas tested, c-MET CTCs were captured in replicate samples from 3 patients [gastric, colorectal, and renal cell carcinoma (RCC)] with 6% prevalence. CTC FISH demonstrated that MET amplification in both gastric and colorectal cancer patients and trisomy 7 with gain of MET gene copies in the RCC patient. The c-MET CTC assay is a rapid, noninvasive, sensitive, and specific method for detecting MET-amplified tumor cells. CTCs with MET amplification can be detected in patients with gastric, colorectal, and renal cancers. IMPLICATIONS This study developed a novel c-MET CTC assay for detecting c-MET CTCs in patients with MET amplification and warrants further investigation to determine its clinical applicability. Mol Cancer Res; 14(6); 539-47. ©2016 AACR.
Collapse
Affiliation(s)
- Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Rengasamy Boominathan
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Brad Foulk
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Chandra Rao
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Gabor Kemeny
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - John H Strickler
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - James L Abbruzzese
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - David S Hsu
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Patrick Healy
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Jing Li
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Cinthia Pi
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Katherine M Prendergast
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Carey Hobbs
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Sarah Gemberling
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Herbert I Hurwitz
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Mark Connelly
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Mariano A Garcia-Blanco
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina. Department of Pharmacology and Cancer Biology, Duke University, Durham, NC.
| |
Collapse
|
29
|
Volz B, Schmidt M, Heinrich K, Kapp K, Schroff M, Wittig B. Design and characterization of the tumor vaccine MGN1601, allogeneic fourfold gene-modified vaccine cells combined with a TLR-9 agonist. Mol Ther Oncolytics 2016; 3:15023. [PMID: 27119114 PMCID: PMC4824560 DOI: 10.1038/mto.2015.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 11/20/2022] Open
Abstract
The tumor vaccine MGN1601 was designed and developed for treatment of metastatic renal cell carcinoma (mRCC). MGN1601 consists of a combination of fourfold gene-modified cells with the toll-like receptor 9 agonist dSLIM, a powerful connector of innate and adaptive immunity. Vaccine cells originate from a renal cell carcinoma cell line (grown from renal cell carcinoma tissue), express a variety of known tumor-associated antigens (TAA), and are gene modified to transiently express two co-stimulatory molecules, CD80 and CD154, and two cytokines, GM-CSF and IL-7, aimed to support immune response. Proof of concept of the designed vaccine was shown in mice: The murine homologue of the vaccine efficiently (100%) prevented tumor growth when used as prophylactic vaccine in a syngeneic setting. Use of the vaccine in a therapeutic setting showed complete response in 92% of mice as well as synergistic action and necessity of the components. In addition, specific cellular and humoral immune responses in mice were found when used in an allogeneic setting. Immune response to the vaccine was also shown in mRCC patients treated with MGN1601: Peptide array analysis revealed humoral CD4-based immune response to TAA expressed on vaccine cells, including survivin, cyclin D1, and stromelysin.
Collapse
Affiliation(s)
- Barbara Volz
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
- Mologen AG, Berlin, Germany
| | | | - Kerstin Heinrich
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
- Mologen AG, Berlin, Germany
| | | | | | - Burghardt Wittig
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
30
|
Raval SH, Singh RD, Joshi DV, Patel HB, Mody SK. Recent developments in receptor tyrosine kinases targeted anticancer therapy. Vet World 2016; 9:80-90. [PMID: 27051190 PMCID: PMC4819356 DOI: 10.14202/vetworld.2016.80-90] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 12/22/2022] Open
Abstract
Novel concepts and understanding of receptors lead to discoveries and optimization of many small molecules and antibodies as anti-cancerous drugs. Receptor tyrosine kinases (RTKs) are such a promising class of receptors under the investigation in past three decades. RTKs are one of the essential mediators of cell signaling mechanism for various cellular processes. Transformations such as overexpression, dysregulation, or mutations of RTKs may result into malignancy, and thus are an important target for anticancer therapy. Numerous subfamilies of RTKs, such as epidermal growth factor receptor, vascular endothelial growth factor receptor, fibroblast growth factor receptors, insulin-like growth factor receptor, and hepatocyte growth factor receptor, have been being investigated in recent years as target for anticancer therapy. The present review focuses several small molecules drugs as well as monoclonal antibodies targeting aforesaid subfamilies either approved or under investigation to treat the various cancers.
Collapse
Affiliation(s)
- Samir H. Raval
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Banaskantha - 385 506, Gujarat, India
| | - Ratn D. Singh
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Banaskantha - 385 506, Gujarat, India
| | - Dilip V. Joshi
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Banaskantha - 385 506, Gujarat, India
| | - Hitesh B. Patel
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Banaskantha - 385 506, Gujarat, India
| | - Shailesh K. Mody
- Department of Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Banaskantha - 385 506, Gujarat, India
| |
Collapse
|
31
|
Hakimi AA, Ostrovnaya I, Jacobsen A, Susztak K, Coleman JA, Russo P, Winer AG, Mano R, Sankin AI, Motzer RJ, Voss MH, Offit K, Purdue M, Pomerantz M, Freedman M, Choueiri TK, Hsieh JJ, Klein RJ. Validation and genomic interrogation of the MET variant rs11762213 as a predictor of adverse outcomes in clear cell renal cell carcinoma. Cancer 2015; 122:402-10. [PMID: 26505625 DOI: 10.1002/cncr.29765] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/02/2015] [Accepted: 09/15/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND The exonic single-nucleotide variant rs11762213 located in the MET oncogene has recently been identified as a prognostic marker in clear cell renal cell carcinoma (ccRCC). This finding was validated with The Cancer Genome Atlas (TCGA) cohort, and the biologic implications were explored. METHODS The genotype status for rs11762213 was available for 272 patients. Paired tumor-normal data, genomic data, and clinical information were acquired from ccRCC TCGA data sets. Cancer-specific survival (CSS) was analyzed with the competing risk method, and Cox proportional hazards regression was used for the analysis of the time to recurrence (TTR). Multivariate competing risk models were fitted to adjust for the validated Mayo Clinic Stage, Size, Grade, and Necrosis (SSIGN) score. RESULTS The variant allele of rs11762213 was detected in 10.3% of the cohort. After adjustments for the SSIGN score, the risk allele remained a significant predictor for adverse CSS (hazard ratio [HR], 3.88; 95% confidence interval [CI], 1.99-7.56; P < .0001) and for TTR (OR, 2.97; 95% CI, 1.43-6.2; P = .003). The mapping of rs11762213 to regulatory regions within the genome suggested that it might affect a DNA enhancer region. RNA and protein sequencing data for MET did not reveal differences in steady-state expression with stratification by risk allele. CONCLUSIONS The exonic MET variant rs11762213 is an independent predictor of adverse CSS and TTR in ccRCC and should be integrated into clinical practice for prognostic stratification. Genomic analysis suggests that the single-nucleotide polymorphism may affect an enhancer region located in the coding region of MET. Further biological mechanistic interrogation is currently underway.
Collapse
Affiliation(s)
- A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | - Paul Russo
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew G Winer
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roy Mano
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Martin H Voss
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark Purdue
- National Cancer Institute, Bethesda, Maryland
| | - Mark Pomerantz
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Matthew Freedman
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - James J Hsieh
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
32
|
Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene 2015; 35:2687-97. [PMID: 26364599 DOI: 10.1038/onc.2015.343] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/14/2022]
Abstract
Antiangiogenic therapy resistance occurs frequently in patients with metastatic renal cell carcinoma (RCC). The purpose of this study was to understand the mechanism of resistance to sunitinib, an antiangiogenic small molecule, and to exploit this mechanism therapeutically. We hypothesized that sunitinib-induced upregulation of the prometastatic MET and AXL receptors is associated with resistance to sunitinib and with more aggressive tumor behavior. In the present study, tissue microarrays containing sunitinib-treated and untreated RCC tissues were stained with MET and AXL antibodies. The low malignant RCC cell line 786-O was chronically treated with sunitinib and assayed for AXL, MET, epithelial-mesenchymal transition (EMT) protein expression and activation. Co-culture experiments were used to examine the effect of sunitinib pretreatment on endothelial cell growth. The effects of AXL and MET were evaluated in various cell-based models by short hairpin RNA or inhibition by cabozantinib, the multi-tyrosine kinases inhibitor that targets vascular endothelial growth factor receptor, MET and AXL. Xenograft mouse models tested the ability of cabozantinib to rescue sunitinib resistance. We demonstrated that increased AXL and MET expression was associated with inferior clinical outcome in patients. Chronic sunitinib treatment of RCC cell lines activated both AXL and MET, induced EMT-associated gene expression changes, including upregulation of Snail and β-catenin, and increased cell migration and invasion. Pretreatment with sunitinib enhanced angiogenesis in 786-0/human umbilical vein endothelial cell co-culture models. The suppression of AXL or MET expression and the inhibition of AXL and MET activation using cabozantinib both impaired chronic sunitinib treatment-induced prometastatic behavior in cell culture and rescued acquired resistance to sunitinib in xenograft models. In summary, chronic sunitinib treatment induces the activation of AXL and MET signaling and promotes prometastatic behavior and angiogenesis. The inhibition of AXL and MET activity may overcome resistance induced by prolonged sunitinib therapy in metastatic RCC.
Collapse
|
33
|
Kim HL, Halabi S, Li P, Mayhew G, Simko J, Nixon AB, Small EJ, Rini B, Morris MJ, Taplin ME, George D. A Molecular Model for Predicting Overall Survival in Patients with Metastatic Clear Cell Renal Carcinoma: Results from CALGB 90206 (Alliance). EBioMedicine 2015; 2:1814-20. [PMID: 26870806 PMCID: PMC4740313 DOI: 10.1016/j.ebiom.2015.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/06/2015] [Accepted: 09/07/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Prognosis associated with metastatic renal cell carcinoma (mRCC) can vary widely. METHODS This study used pretreatment nephrectomy specimens from a randomized phase III trial. Expression levels of candidate genes were determined from archival tumors using the OpenArray® platform for TaqMan® RT-qPCR. The dataset was randomly divided at 2:1 ratio into training (n = 221) and testing (n = 103) sets to develop a multigene prognostic signature. FINDINGS Gene expressions were measured in 324 patients. In the training set, multiple models testing 424 candidate genes identified a prognostic signature containing 8 genes plus MSKCC clinical risk factors. In the testing set, the time dependent (td) AUC for a prognostic model containing the 8 genes with and without MSKCC risk factors were 0.72 and 0.69, respectively. The tdAUC for the clinical risk factors alone was 0.61. Additional primary mRCCs from patients with mRCC (n = 12) were sampled in multiple sites and standard deviations of gene expressions within a tumor were used as a measure of heterogeneity. All 8 genes in the final prognostic model met our criteria for minimal heterogeneity. CONCLUSIONS A molecular prognostic signature based on 8 genes was developed and is ready for external validation in this patient population and other related settings such as nonmetastatic RCC.
Collapse
Affiliation(s)
- Hyung L Kim
- Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, and Alliance Statistics and Data Center, Duke University, Durham, NC, United States
| | - Ping Li
- Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Greg Mayhew
- GeneCentric Diagnostics, Durham, NC, United States
| | - Jeff Simko
- University of California at San Francisco, San Francisco, CA, United States
| | | | - Eric J Small
- University of California at San Francisco, San Francisco, CA, United States
| | - Brian Rini
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, United States
| | - Michael J Morris
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Daniel George
- Department of Biostatistics and Bioinformatics, and Alliance Statistics and Data Center, Duke University, Durham, NC, United States
| | | |
Collapse
|
34
|
Abstract
The MET receptor tyrosine kinase and its ligand hepatocyte growth factor/scatter factor (HGF/SF) are potential therapeutic targets in many human malignancies, making this pathway an important focus of molecular and cancer research. MET mutations have been detected in various tumours. In addition, many tumour types demonstrate MET and HGF/SF overexpression and amplification. The MET signal transduction cascade is complex, and manifests in a broad spectrum of mitogenic and morphogenic functions, affecting cell proliferation, migration, differentiation, morphology and survival. Cancer cells commandeer the physiological functions of this signalling axis to facilitate invasion and metastasis. Significant progress has been made in the development of agents that inhibit MET-HGF/SF signalling. In this article, we outline the key features of the MET gene, its protein product and the ligand HGF/SF, to provide an overview of this important signalling pathway and offer a summary of the relevant pathological and clinical directions of research.
Collapse
Affiliation(s)
- Garret Skead
- Division of Anatomical Pathology, University of Cape Town and National Health Laboratory Service, Groote Schuur hospital, Cape Town, South Africa
| | - Dhirendra Govender
- Division of Anatomical Pathology, University of Cape Town and National Health Laboratory Service, Groote Schuur hospital, Cape Town, South Africa
| |
Collapse
|
35
|
Leiser D, Medová M, Mikami K, Nisa L, Stroka D, Blaukat A, Bladt F, Aebersold DM, Zimmer Y. KRAS and HRAS mutations confer resistance to MET targeting in preclinical models of MET-expressing tumor cells. Mol Oncol 2015; 9:1434-46. [PMID: 25933688 DOI: 10.1016/j.molonc.2015.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/23/2015] [Accepted: 04/05/2015] [Indexed: 12/16/2022] Open
Abstract
The MET receptor tyrosine kinase is often deregulated in human cancers and several MET inhibitors are evaluated in clinical trials. Similarly to EGFR, MET signals through the RAS-RAF-ERK/MAPK pathway which plays key roles in cell proliferation and survival. Mutations of genes encoding for RAS proteins, particularly in KRAS, are commonly found in various tumors and are associated with constitutive activation of the MAPK pathway. It was shown for EGFR, that KRAS mutations render upstream EGFR inhibition ineffective in EGFR-positive colorectal cancers. Currently, there are no clinical studies evaluating MET inhibition impairment due to RAS mutations. To test the impact of RAS mutations on MET targeting, we generated tumor cells responsive to the MET inhibitor EMD1214063 that express KRAS G12V, G12D, G13D and HRAS G12V variants. We demonstrate that these MAPK-activating RAS mutations differentially interfere with MET-mediated biological effects of MET inhibition. We report increased residual ERK1/2 phosphorylation indicating that the downstream pathway remains active in presence of MET inhibition. Consequently, RAS variants counteracted MET inhibition-induced morphological changes as well as anti-proliferative and anchorage-independent growth effects. The effect of RAS mutants was reversed when MET inhibition was combined with MEK inhibitors AZD6244 and UO126. In an in vivo mouse xenograft model, MET-driven tumors harboring mutated RAS displayed resistance to MET inhibition. Taken together, our results demonstrate for the first time in details the role of KRAS and HRAS mutations in resistance to MET inhibition and suggest targeting both MET and MEK as an effective strategy when both oncogenic drivers are expressed.
Collapse
Affiliation(s)
- Dominic Leiser
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Kei Mikami
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Lluís Nisa
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Andree Blaukat
- Merck Serono an Affiliate of Merck Serono Research & Development, Merck KGaA, 64271 Darmstadt, Germany
| | - Friedhelm Bladt
- Merck Serono an Affiliate of Merck Serono Research & Development, Merck KGaA, 64271 Darmstadt, Germany
| | - Daniel M Aebersold
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Switzerland.
| |
Collapse
|
36
|
Golovine K, Makhov P, Naito S, Raiyani H, Tomaszewski J, Mehrazin R, Tulin A, Kutikov A, Uzzo RG, Kolenko VM. Piperlongumine and its analogs down-regulate expression of c-Met in renal cell carcinoma. Cancer Biol Ther 2015; 16:743-9. [PMID: 25801713 PMCID: PMC4623021 DOI: 10.1080/15384047.2015.1026511] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/01/2015] [Indexed: 12/29/2022] Open
Abstract
The c-Met protein, a transmembrane receptor tyrosine kinase, is the product of a proto-oncogene. Its only known ligand, hepatocyte growth factor (HGF), regulates cell growth, motility, migration, invasion, proliferation, and angiogenesis. The aberrant expression of c-Met is often associated with poor prognosis in multiple cancers, including renal cell carcinoma (RCC). Silencing or inactivation of c-Met leads to decreased viability of cancer cells, thereby making ablation of c-Met signaling an attractive concept for developing novel strategies for the treatment of renal tumors. Naturally-occurring products or substances are the most consistent source of drug development. As such, we investigated the functional impact of piperlongumine (PL), a naturally occurring alkaloid present in the Long pepper (Piper longum) on c-Met expression in RCC cells and demonstrated that PL and its analogs rapidly reduce c-Met protein and RNA levels in RCC cells via ROS-dependent mechanism. PL-mediated c-Met depletion coincided with the inhibition of downstream c-Met signaling; namely Erk/MAPK, STAT3, NF-κB and Akt/mTOR. As such, PL and PL analogs hold promise as potential therapeutic agents for the treatment of metastatic RCC and the prevention of postoperative RCC recurrence.
Collapse
Key Words
- Erk, Extracellular signal-regulated kinase
- FAK, Focal adhesion kinase
- HGF, Hepatocyte growth factor
- MAPK, Mitogen-activated protein kinase
- NF-kB, Nuclear factor kappaB
- PL, Piperlongumine
- PL-Di, PL-Dimer
- PL-FPh, PL-fluorophenyl
- RCC, Renal cell carcinoma
- RECIST, Response evaluation criteria in solid tumors
- RNA, Ribonucleic acid
- ROS
- ROS, Reactive oxygen species
- STAT, Signal transducer and activator of transcription
- TKIs, Tyrosine kinase inhibitors
- VEGFR, Vascular endothelial growth factor receptor
- c-Met
- cancer
- mTOR, Mammalian target of rapamycin
- piperlongumine
- renal
Collapse
Affiliation(s)
| | - Peter Makhov
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Sei Naito
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Henish Raiyani
- Cancer Biology Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Jeffrey Tomaszewski
- Division of Urologic Oncology; Department of Surgery; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Reza Mehrazin
- Division of Urologic Oncology; Department of Surgery; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Alexei Tulin
- Cancer Epigenetics Program; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Alexander Kutikov
- Division of Urologic Oncology; Department of Surgery; Fox Chase Cancer Center; Philadelphia, PA, USA
| | - Robert G Uzzo
- Division of Urologic Oncology; Department of Surgery; Fox Chase Cancer Center; Philadelphia, PA, USA
| | | |
Collapse
|
37
|
Gelsomino F, Rossi G, Tiseo M. MET and Small-Cell Lung Cancer. Cancers (Basel) 2014; 6:2100-15. [PMID: 25314153 PMCID: PMC4276958 DOI: 10.3390/cancers6042100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/21/2014] [Accepted: 09/22/2014] [Indexed: 01/21/2023] Open
Abstract
Small-cell lung cancer (SCLC) is one of the most aggressive lung tumors. The majority of patients with SCLC are diagnosed at an advanced stage. This tumor type is highly sensitive to chemo-radiation treatment, with very high response rates, but invariably relapses. At this time, treatment options are still limited and the prognosis of these patients is poor. A better knowledge of the molecular biology of SCLC allowed us to identify potential druggable targets. Among these, the MET/HGF axis seems to be one of the most aberrant signaling pathways involved in SCLC invasiveness and progression. In this review, we describe briefly all recent literature on the different molecular profiling in SCLC; in particular, we discuss the specific alterations involving c-MET gene and their implications as a potential target in SCLC.
Collapse
Affiliation(s)
- Francesco Gelsomino
- Medical Oncology Unit 1, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milano, Italy.
| | - Giulio Rossi
- Operative Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, Via del Pozzo 71, 41124 Modena, Italy.
| | - Marcello Tiseo
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria, Viale A. Gramsci 14, 43126 Parma, Italy.
| |
Collapse
|
38
|
Hong H, Chen F, Zhang Y, Cai W. New radiotracers for imaging of vascular targets in angiogenesis-related diseases. Adv Drug Deliv Rev 2014; 76:2-20. [PMID: 25086372 DOI: 10.1016/j.addr.2014.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/03/2023]
Abstract
Tremendous advances over the last several decades in positron emission tomography (PET) and single photon emission computed tomography (SPECT) allow for targeted imaging of molecular and cellular events in the living systems. Angiogenesis, a multistep process regulated by the network of different angiogenic factors, has attracted world-wide interests, due to its pivotal role in the formation and progression of different diseases including cancer, cardiovascular diseases (CVD), and inflammation. In this review article, we will summarize the recent progress in PET or SPECT imaging of a wide variety of vascular targets in three major angiogenesis-related diseases: cancer, cardiovascular diseases, and inflammation. Faster drug development and patient stratification for a specific therapy will become possible with the facilitation of PET or SPECT imaging and it will be critical for the maximum benefit of patients.
Collapse
|
39
|
Choueiri TK, Fay AP, Gray KP, Callea M, Ho TH, Albiges L, Bellmunt J, Song J, Carvo I, Lampron M, Stanton ML, Hodi FS, McDermott DF, Atkins MB, Freeman GJ, Hirsch MS, Signoretti S. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann Oncol 2014; 25:2178-2184. [PMID: 25193987 DOI: 10.1093/annonc/mdu445] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) expression in nonclear-cell RCC (non-ccRCC) and its association with clinical outcomes are unknown. METHODS Formalin-fixed paraffin-embedded (FFPE) specimens were obtained from 101 patients with non-ccRCC. PD-L1 expression was evaluated by immunohistochemistry in both tumor cell membrane and tumor-infiltrating mononuclear cells (TIMC). PD-L1 tumor positivity was defined as ≥5% tumor cell membrane staining. For PD-L1 expression in TIMC, a combined score based on the extent of infiltrate and percentage of positive cells was used. Baseline clinico-pathological characteristics and outcome data [time to recurrence (TTR) and overall survival (OS)] were correlated with PD-L1 staining. RESULTS Among 101 patients, 11 (10.9%) were considered PD-L1+ in tumor cells: 2/36 (5.6%) of chromophobe RCC, 5/50 (10%) of papillary RCC, 3/10 (30%) of Xp11.2 translocation RCC and 1/5 (20%) of collecting duct carcinoma. PD-L1 positivity (PD-L1+) in tumor cells was significantly associated with higher stage (P = 0.01) and grade (P = 0.03), as well as shorter OS (P < 0.001). On the other hand, PD-L1 positivity by TIMC was observed in 57 (56.4%) patients: 13/36 (36.1%) of chromophobe RCC, 30/50 (60%) of papillary RCC, 9/10 (90%) of Xp11.2 translocation RCC and 5/5 (100%) of collecting duct carcinoma. A trend toward shorter OS was observed in patients with PD-L1+ in TIMC (P = 0.08). PD-L1+ in both tumor cell membrane and TIMC cells were associated with shorter TTR (P = 0.02 and P = 0.03, respectively). CONCLUSION In non-ccRCC, patients with PD-L1+ tumors appear to have worse clinical outcomes, although only PD-L1 positivity in tumor cells is associated with higher tumor stage and grade.
Collapse
Affiliation(s)
- T K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Department of Medical Oncology, Brigham and Women's Hospital, Boston; Harvard Medical School, Boston.
| | - A P Fay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - K P Gray
- Biostatistics and Computational Biology, Harvard School of Public Health, Boston
| | - M Callea
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - T H Ho
- Department of Medical Oncology, Mayo Clinic, Scottsdale
| | - L Albiges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - J Bellmunt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Department of Medical Oncology, Brigham and Women's Hospital, Boston; Harvard Medical School, Boston
| | - J Song
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - I Carvo
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - M Lampron
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - M L Stanton
- Department of Medical Oncology, Mayo Clinic, Scottsdale
| | - F S Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston; Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston
| | - D F McDermott
- Harvard Medical School, Boston; Department of Medical Oncology, Beth-Israel Deaconess Medical Center, Boston
| | - M B Atkins
- Department of Medical Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, USA
| | - G J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston
| | - M S Hirsch
- Harvard Medical School, Boston; Department of Pathology, Brigham and Women's Hospital, Boston
| | - S Signoretti
- Harvard Medical School, Boston; Department of Pathology, Brigham and Women's Hospital, Boston
| |
Collapse
|
40
|
Direct regulation of GAS6/AXL signaling by HIF promotes renal metastasis through SRC and MET. Proc Natl Acad Sci U S A 2014; 111:13373-8. [PMID: 25187556 DOI: 10.1073/pnas.1404848111] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.
Collapse
|
41
|
Bhatt JR, Finelli A. Landmarks in the diagnosis and treatment of renal cell carcinoma. Nat Rev Urol 2014; 11:517-25. [PMID: 25112856 DOI: 10.1038/nrurol.2014.194] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common renal cancer is renal cell carcinoma (RCC), which arises from the renal parenchyma. The global incidence of RCC has increased over the past two decades by 2% per year. RCC is the most lethal of the common urological cancers: despite diagnostic advances, 20-30% of patients present with metastatic disease. A clearer understanding of the genetic basis of RCC has led to immune-based and targeted treatments for this chemoresistant cancer. Despite promising results in advanced disease, overall response rates and durable complete responses are rare. Surgery remains the main treatment modality, especially for organ-confined disease, with a selective role in advanced and metastatic disease. Smaller tumours are increasingly managed with biopsy, minimally invasive interventions and surveillance. The future promises multimodal, integrated and personalized care, with further understanding of the disease leading to new treatment options.
Collapse
Affiliation(s)
- Jaimin R Bhatt
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| | - Antonio Finelli
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
42
|
Cerbone L, Recine F, Sternberg CN. Pharmacotherapy options for advanced renal cell carcinoma. Expert Opin Orphan Drugs 2014. [DOI: 10.1517/21678707.2014.911079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Albiges L, Guegan J, Le Formal A, Verkarre V, Rioux-Leclercq N, Sibony M, Bernhard JC, Camparo P, Merabet Z, Molinie V, Allory Y, Orear C, Couvé S, Gad S, Patard JJ, Escudier B. MET is a potential target across all papillary renal cell carcinomas: result from a large molecular study of pRCC with CGH array and matching gene expression array. Clin Cancer Res 2014; 20:3411-21. [PMID: 24658158 DOI: 10.1158/1078-0432.ccr-13-2173] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Papillary renal cell carcinomas (pRCC) are the most common nonclear cell RCC subtype. Germline mutations of the MET oncogene at 7q31 have been detected in patients with hereditary type I pRCC and in 13% of sporadic type I pRCC. Recent report of MET inhibition strengthened the role of c-Met inhibition across pRCC. EXPERIMENTAL DESIGN We collected 220 frozen samples of sporadic pRCC through the French RCC Network and quality controlled for percentage of malignant cells >70%. Gene expression was assessed on 98 pRCC using human whole-genome Agilent 8 × 60K arrays. Copy number alterations were analyzed using Agilent Human 2 × 400K and 4× 180K array for type II pRCC and comparative genomic microarray analysis method for type I pRCC. MET gene sequencing was performed on type I pRCC. RESULTS MET expression level was high across all pRCC. We identified copy number alterations (gain) in 46% of type II pRCC and in 81% of type I pRCC. Correlation between DNA copy number alterations and mRNA expression level was highly significant. Eleven somatic mutations of MET gene were identified amongst 51 type I pRCC (21.6%), including 4 new mutations. We validated LRRK2 cokinase as highly correlated to MET expression. CONCLUSION The present report expands the role of MET activation as a potential target across all pRCC subtypes. These data support investigating MET inhibitors in pRCC in correlation with MET activation status.
Collapse
Affiliation(s)
- Laurence Albiges
- Authors' Affiliations: Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France; INSERM U753, IGR, Villejuif, France;
| | | | | | - Virginie Verkarre
- Department of Pathology, Necker-Enfants Malades Hospital, AP-HP, Université Paris Descartes
| | - Nathalie Rioux-Leclercq
- Department of Pathology, CHU Rennes, Faculté de Médecine, Université de Rennes1, Rennes; and
| | - Mathilde Sibony
- Department of Pathology, Tenon Hospital, AP-HP, Université Paris Pierre et Marie Curie
| | | | | | - Zahira Merabet
- Department of Pathology, Institut Gustave Roussy, Villejuif, France
| | | | - Yves Allory
- Department of Pathology, Hopital Mondor, Faculté Paris Sud, Creteil
| | | | - Sophie Couvé
- INSERM U753, IGR, Villejuif, France; Laboratoire de Génétique Oncologique EPHE, Institut Gustave Roussy
| | - Sophie Gad
- INSERM U753, IGR, Villejuif, France; Laboratoire de Génétique Oncologique EPHE, Institut Gustave Roussy
| | - Jean-Jacques Patard
- INSERM U753, IGR, Villejuif, France; Department of Urology, Kremlin Bicetre Hospital, Université Paris Sud, Kremlin Bicêtre, Paris
| | - Bernard Escudier
- Authors' Affiliations: Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France; INSERM U753, IGR, Villejuif, France
| |
Collapse
|
44
|
Fay AP, Signoretti S, Choueiri TK. MET as a Target in Papillary Renal Cell Carcinoma. Clin Cancer Res 2014; 20:3361-3. [DOI: 10.1158/1078-0432.ccr-14-0690] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Satcher RL, Pan T, Cheng CJ, Lee YC, Lin SC, Yu G, Li X, Hoang AG, Tamboli P, Jonasch E, Gallick GE, Lin SH. Cadherin-11 in renal cell carcinoma bone metastasis. PLoS One 2014; 9:e89880. [PMID: 24587095 PMCID: PMC3933681 DOI: 10.1371/journal.pone.0089880] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 12/13/2022] Open
Abstract
Bone is one of the common sites of metastases from renal cell carcinoma (RCC), however the mechanism by which RCC preferentially metastasize to bone is poorly understood. Homing/retention of RCC cells to bone and subsequent proliferation are necessary steps for RCC cells to colonize bone. To explore possible mechanisms by which these processes occur, we used an in vivo metastasis model in which 786-O RCC cells were injected into SCID mice intracardially, and organotropic cell lines from bone, liver, and lymph node were selected. The expression of molecules affecting cell adhesion, angiogenesis, and osteolysis were then examined in these selected cells. Cadherin-11, a mesenchymal cadherin mainly expressed in osteoblasts, was significantly increased on the cell surface in bone metastasis-derived 786-O cells (Bo-786-O) compared to parental, liver, or lymph node-derived cells. In contrast, the homing receptor CXCR4 was equivalently expressed in cells derived from all organs. No significant difference was observed in the expression of angiogenic factors, including HIF-1α, VEGF, angiopoeitin-1, Tie2, c-MET, and osteolytic factors, including PTHrP, IL-6 and RANKL. While the parental and Bo-786-O cells have similar proliferation rates, Bo-786-O cells showed an increase in migration compared to the parental 786-O cells. Knockdown of Cadherin-11 using shRNA reduced the rate of migration in Bo-786-O cells, suggesting that Cadherin-11 contributes to the increased migration observed in bone-derived cells. Immunohistochemical analysis of cadherin-11 expression in a human renal carcinoma tissue array showed that the number of human specimens with positive cadherin-11 activity was significantly higher in tumors that metastasized to bone than that in primary tumors. Together, these results suggest that Cadherin-11 may play a role in RCC bone metastasis.
Collapse
Affiliation(s)
- Robert L. Satcher
- Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Tianhong Pan
- Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Chien-Jui Cheng
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Chen Lee
- Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Song-Chang Lin
- Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Guoyu Yu
- Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiaoxia Li
- Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Anh G. Hoang
- Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Pheroze Tamboli
- Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Eric Jonasch
- Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gary E. Gallick
- Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sue-Hwa Lin
- Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
- Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|