1
|
Dolan M, Shi Y, Mastri M, Long MD, McKenery A, Hill JW, Vaghi C, Benzekry S, Barbi J, Ebos JM. A senescence-mimicking (senomimetic) VEGFR TKI side-effect primes tumor immune responses via IFN/STING signaling. Mol Cancer Ther 2024; 23:745113. [PMID: 38690835 PMCID: PMC11527799 DOI: 10.1158/1535-7163.mct-24-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) that block the vascular endothelial growth factor receptors (VEGFRs) disrupt tumor angiogenesis but also have many unexpected side-effects that impact tumor cells directly. This includes the induction of molecular markers associated with senescence, a form of cellular aging that typically involves growth arrest. We have shown that VEGFR TKIs can hijack these aging programs by transiently inducting senescence-markers (SMs) in tumor cells to activate senescence-associated secretory programs that fuel drug resistance. Here we show that these same senescence-mimicking ('senomimetic') VEGFR TKI effects drive an enhanced immunogenic signaling that, in turn, can alter tumor response to immunotherapy. Using a live-cell sorting method to detect beta-galactosidase, a commonly used SM, we found that subpopulations of SM-expressing (SM+) tumor cells have heightened interferon (IFN) signaling and increased expression of IFN-stimulated genes (ISGs). These ISG increases were under the control of the STimulator of INterferon Gene (STING) signaling pathway, which we found could be directly activated by several VEGFR TKIs. TKI-induced SM+ cells could stimulate or suppress CD8 T-cell activation depending on host:tumor cell contact while tumors grown from SM+ cells were more sensitive to PD-L1 inhibition in vivo, suggesting that offsetting immune-suppressive functions of SM+ cells can improve TKI efficacy overall. Our findings may explain why some (but not all) VEGFR TKIs improve outcomes when combined with immunotherapy and suggest that exploiting senomimetic drug side-effects may help identify TKIs that uniquely 'prime' tumors for enhanced sensitivity to PD-L1 targeted agents.
Collapse
Affiliation(s)
- Melissa Dolan
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center Buffalo, NY, 14263. USA
| | - Yuhao Shi
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center Buffalo, NY, 14263. USA
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263. USA
| | - Mark D. Long
- Department of Bioinformatics and Statistics, Roswell Park Comprehensive Cancer Center Buffalo, NY, 14263. USA
| | - Amber McKenery
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263. USA
| | - James W. Hill
- Jacobs School of Medicine and Biomedical Sciences, SUNY at Buffalo, Buffalo, New York, 14263. USA
| | - Cristina Vaghi
- Inria Team MONC, Inria Bordeaux Sud-Ouest, Talence, France
- Computational Pharmacology and Clinical Oncology (COMPO), Inria Sophia Antipolis–Méditerranée, Cancer Research Center of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, 13385 Marseille, France
| | - Sebastien Benzekry
- Inria Team MONC, Inria Bordeaux Sud-Ouest, Talence, France
- Computational Pharmacology and Clinical Oncology (COMPO), Inria Sophia Antipolis–Méditerranée, Cancer Research Center of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, 13385 Marseille, France
| | - Joseph Barbi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263. USA
| | - John M.L. Ebos
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center Buffalo, NY, 14263. USA
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263. USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center Buffalo, NY, 14263. USA
- Lead Contact
| |
Collapse
|
2
|
Benzekry S, Mastri M, Nicolò C, Ebos JML. Machine-learning and mechanistic modeling of metastatic breast cancer after neoadjuvant treatment. PLoS Comput Biol 2024; 20:e1012088. [PMID: 38701089 PMCID: PMC11095706 DOI: 10.1371/journal.pcbi.1012088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/15/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Clinical trials involving systemic neoadjuvant treatments in breast cancer aim to shrink tumors before surgery while simultaneously allowing for controlled evaluation of biomarkers, toxicity, and suppression of distant (occult) metastatic disease. Yet neoadjuvant clinical trials are rarely preceded by preclinical testing involving neoadjuvant treatment, surgery, and post-surgery monitoring of the disease. Here we used a mouse model of spontaneous metastasis occurring after surgical removal of orthotopically implanted primary tumors to develop a predictive mathematical model of neoadjuvant treatment response to sunitinib, a receptor tyrosine kinase inhibitor (RTKI). Treatment outcomes were used to validate a novel mathematical kinetics-pharmacodynamics model predictive of perioperative disease progression. Longitudinal measurements of presurgical primary tumor size and postsurgical metastatic burden were compiled using 128 mice receiving variable neoadjuvant treatment doses and schedules (released publicly at https://zenodo.org/records/10607753). A non-linear mixed-effects modeling approach quantified inter-animal variabilities in metastatic dynamics and survival, and machine-learning algorithms were applied to investigate the significance of several biomarkers at resection as predictors of individual kinetics. Biomarkers included circulating tumor- and immune-based cells (circulating tumor cells and myeloid-derived suppressor cells) as well as immunohistochemical tumor proteins (CD31 and Ki67). Our computational simulations show that neoadjuvant RTKI treatment inhibits primary tumor growth but has little efficacy in preventing (micro)-metastatic disease progression after surgery and treatment cessation. Machine learning algorithms that included support vector machines, random forests, and artificial neural networks, confirmed a lack of definitive biomarkers, which shows the value of preclinical modeling studies to identify potential failures that should be avoided clinically.
Collapse
Affiliation(s)
- Sebastien Benzekry
- Computational Pharmacology and Clinical Oncology (COMPO), Inria Sophia Antipolis–Méditerranée, Cancer Research Center of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, Marseille, France
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Chiara Nicolò
- InSilicoTrials Technologies S.P.A, Riva Grumula, Trieste, Italy
| | - John M. L. Ebos
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| |
Collapse
|
3
|
Tzetzo SL, Kramer ED, Mohammadpour H, Kim M, Rosario SR, Yu H, Dolan MR, Oturkar CC, Morreale BG, Bogner PN, Stablewski AB, Benavides FJ, Brackett CM, Ebos JM, Das GM, Opyrchal M, Nemeth MJ, Evans SS, Abrams SI. Downregulation of IRF8 in alveolar macrophages by G-CSF promotes metastatic tumor progression. iScience 2024; 27:109187. [PMID: 38420590 PMCID: PMC10901102 DOI: 10.1016/j.isci.2024.109187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Tissue-resident macrophages (TRMs) are abundant immune cells within pre-metastatic sites, yet their functional contributions to metastasis remain incompletely understood. Here, we show that alveolar macrophages (AMs), the main TRMs of the lung, are susceptible to downregulation of the immune stimulatory transcription factor IRF8, impairing anti-metastatic activity in models of metastatic breast cancer. G-CSF is a key tumor-associated factor (TAF) that acts upon AMs to reduce IRF8 levels and facilitate metastasis. Translational relevance of IRF8 downregulation was observed among macrophage precursors in breast cancer and a CD68hiIRF8loG-CSFhi gene signature suggests poorer prognosis in triple-negative breast cancer (TNBC), a G-CSF-expressing subtype. Our data highlight the underappreciated, pro-metastatic roles of AMs in response to G-CSF and identify the contribution of IRF8-deficient AMs to metastatic burden. AMs are an attractive target of local neoadjuvant G-CSF blockade to recover anti-metastatic activity.
Collapse
Affiliation(s)
- Stephanie L. Tzetzo
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elliot D. Kramer
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hemn Mohammadpour
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Minhyung Kim
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Melissa R. Dolan
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C. Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Brian G. Morreale
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Paul N. Bogner
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aimee B. Stablewski
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Fernando J. Benavides
- Department of Epigenetics and Molecular Carcinogenesis, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - John M.L. Ebos
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Gokul M. Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mateusz Opyrchal
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Michael J. Nemeth
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sharon S. Evans
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
4
|
Zarychta E, Bielawski K, Wrzeszcz K, Rhone P, Ruszkowska-Ciastek B. Unraveling the Angiogenic Puzzle: Pre-Treatment sVEGFR1 and sVEGFR2 Levels as Promising Prognostic Indicators in Early-Stage Breast Cancer Patients. Int J Mol Sci 2023; 24:13508. [PMID: 37686312 PMCID: PMC10487545 DOI: 10.3390/ijms241713508] [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: 08/01/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Despite the advancements in breast cancer (BrC) diagnosis and treatment, a considerable proportion of patients with early-stage disease still experience local recurrence or metastasis. This study aimed to assess the levels of specific angiogenic parameters in the EDTA plasma of BrC patients before and after treatment and to explore their clinical and prognostic significance. The levels of vascular endothelial growth factor A (VEGF-A), soluble form of vascular endothelial growth factor receptor type 1 (sVEGFR1), and soluble form of vascular endothelial growth factor receptor type 2 (sVEGFR2) were measured in 84 early BrC patients, both prior to surgery and within a median time of nine months post-treatment. Prognostic significance was evaluated using Kaplan-Meier survival and Cox regression analyses. Linear regression models were employed to examine the independent impact of selected angiogenic factors on DFS in breast cancer patients. The results of uni- and multivariate analyses indicated that a pre-treatment concentration of sVEGFR1 above 30.99 pg/mL was associated with improved disease-free survival (DFS) (p < 0.0001 for both analyses), while a pre-treatment concentration of sVEGFR2 above 9475.67 pg/mL was associated with an increased risk of BrC relapse (p < 0.0001 for both analyses). Additionally, a post-treatment concentration of sVEGFR2 above 7361.71 pg/mL was associated with better overall survival (OS) based on the Kaplan-Meier survival analysis (p = 0.0141). Furthermore, linear regression models revealed a significant inverse association between pre-treatment levels of sVEGFR1 and the risk of relapse (standardized β -0.2578, p = 0.0499) and a significant positive association of VEGF-A levels with the risk of recurrence (standardized β 0.2958, p = 0.0308). In conclusion, the findings suggest that both pre- and post-treatment levels of sVEGFR1 and sVEGFR2 may hold promise as potential prognostic markers for BrC patients.
Collapse
Affiliation(s)
- Elżbieta Zarychta
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
- Invicta Fertility Clinic, 6 Zlota Street, 00-019 Warsaw, Poland
| | - Kornel Bielawski
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| | - Katarzyna Wrzeszcz
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| | - Piotr Rhone
- Clinical Ward of Breast Cancer and Reconstructive Surgery, Oncology Centre Prof. F. Łukaszczyk Memorial Hospital, 2 Romanowska Street, 85-796 Bydgoszcz, Poland;
| | - Barbara Ruszkowska-Ciastek
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 9 Curie Sklodowska Street, 85-094 Bydgoszcz, Poland; (K.B.); (K.W.); (B.R.-C.)
| |
Collapse
|
5
|
Zheng J, Wang X, Yu J, Zhan Z, Guo Z. IL-6, TNF-α and IL-12p70 levels in patients with colorectal cancer and their predictive value in anti-vascular therapy. Front Oncol 2022; 12:997665. [PMID: 36226059 PMCID: PMC9549173 DOI: 10.3389/fonc.2022.997665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/30/2022] [Indexed: 01/19/2023] Open
Abstract
We aimed to analyze the levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and interleukin-12 (IL-12p70) in colorectal cancer and evaluate the predictive significance of clinical efficacy of patients with colorectal cancer treated with anti-vascular therapy combined with chemotherapy. A retrospective study of 162 patients with colorectal cancer in Fujian Medical University Hospital was conducted from January 2019 to December 2020. A comparative analysis of the levels of IL-6, TNF-α and IL-12p70 between the two groups were studied. The relationship between the levels and the clinical characteristics of patients was observed; the factors affecting the levels of IL-6, TNF-α, and IL-12p70 in colorectal cancer patients were analyzed, and the predictive validity of the efficacy of anti-vascular therapy was evaluated. We observed that the individual expression levels of IL-6, TNF-α and IL-12p70 in the patients with colorectal cancer are related to lymph node metastasis, TNM staging, and degree of differentiation (P<0.05); however, they are irrelevant to the age, sex, and tumor location of patients with colorectal cancer (P>0.05). The multiple stepwise regression analysis indicates that lymph node metastasis and TNM staging are independent risk factors that correlate with IL-6 and IL-12p70 levels in colorectal cancer patients (P<0.01). The degree of differentiation was found to be an independent risk factor connected to TNF- α levels of patients with colorectal cancer. The change of IL-12p70 level could predict the validity of anti-vascular treatment for advanced colorectal cancer. When evaluated for combined expression, IL-6 and IL-12p70 in patients with colorectal cancer closely related to lymph node metastasis and TNM staging. IL-12p70 can be used as a predictor of anti-vascular therapy with colorectal cancer.
Collapse
Affiliation(s)
- Jingxian Zheng
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaojie Wang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
| | - Jiami Yu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
| | - Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
| | - Zengqing Guo
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, The Affiliated Cancer Hospital of Fujian Medical University, Fuzhou, China
- *Correspondence: Zengqing Guo,
| |
Collapse
|
6
|
Wang B, Zhang X, Chen H, Koh A, Zhao C, Chen Y. A Review of Intraocular Biomolecules in Retinal Vein Occlusion: Toward Potential Biomarkers for Companion Diagnostics. Front Pharmacol 2022; 13:859951. [PMID: 35559255 PMCID: PMC9086509 DOI: 10.3389/fphar.2022.859951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/25/2022] [Indexed: 12/04/2022] Open
Abstract
Retinal vein occlusion (RVO) is one of the most common retinal vascular diseases. The pathogenesis of RVO is multifactorial and involves a complex interplay among a variety of vascular and inflammatory mediators. Many cytokines, chemokines, growth factors, and cell adhesion molecules have been reported to be implicated. Treatments for RVO are directed at the management of underlying risk factors and vision-threatening complications, including macula edema (ME) and neovascularization. Intravitreal anti-VEGF agents are currently considered as the first-line treatment for ME secondary to RVO (RVO-ME), but a substantial proportion of patients responded insufficiently to anti-VEGF agents. Since RVO-ME refractory to anti-VEGF agents generally responds to corticosteroids and its visual outcome is negatively correlated to disease duration, prediction of treatment response at baseline in RVO-ME may significantly improve both cost-effectiveness and visual prognosis. Several bioactive molecules in the aqueous humor were found to be associated with disease status in RVO. This review aims to present a comprehensive review of intraocular biomolecules reported in RVO, including VEGF, IL-6, IL-8, MCP-1, sICAM-1, IL-12, IL-13, sVEGFR-1, sVEGFR-2, PDGF-AA, etc., highlighting their association with disease severity and/or phenotype, and their potential roles in prognostic prediction and treatment selection. Some of these molecules may serve as biomarkers for aqueous humor-based companion diagnostics for the treatment of RVO in the future.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Xiao Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Adrian Koh
- Eye & Retina Surgeons, Camden Medical Centre, Singapore, Singapore
| | - Chan Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youxin Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Kong JH, Kim YD, Park SP, Kim Y. Aqueous Humor Cytokine Levels and Choroidal Thicknesses of Patients with Age‐related Macular Degeneration and Pachychoroid Neovasculopathy. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2022. [DOI: 10.3341/jkos.2022.63.4.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Purpose: We sought correlations between the subfoveal choroidal thickness (SCT) and changes in the levels of aqueous humor cytokines before and after anti‐vascular endothelial growth factor (anti‐VEGF) treatment of patients with neovascular age‐related macular degeneration (nAMD) and pachychoroid neovasculopathy.Methods: We measured changes in the SCT and levels of aqueous humor cytokines (VEGF, soluble VEGF receptor‐2 [sVEGFR‐ 2], platelet‐derived growth factor [PDGF]‐AA, monocyte chemoattractant protein 1 [MCP‐1], interleukin [IL]‐6, and IL‐8) after anti‐ VEGF treatment of 11 eyes of 11 nAMD patients and nine eyes of nine pachychoroid neovasculopathy patients. The aqueous humor cytokine levels were compared between the two groups.Results: After anti‐VEGF treatment, the aqueous levels of VEGF and PDGF‐AA decreased significantly, whereas that of sVEGFR‐2 increased. The amount of change in sVEGFR‐2 concentration before and after anti‐VEGF treatment correlated with the SCT and its change after treatment. nAMD patients with relatively thin SCTs and smaller SCT changes after anti‐VEGF treatment showed greater increases in sVEGFR‐2 levels following treatment. We found significant correlations among the MCP‐1, IL‐6, and IL‐8 levels in the nAMD group, and between the sVEGFR‐2 and MCP‐1, and MCP‐1 and PDGF‐AA, levels in the pachychoroid neovasculopathy group.Conclusions: Patients with nAMD exhibited significant increases in aqueous sVEGFR‐2 levels following anti‐VEGF treatment and significant correlations among the levels of the inflammatory cytokines MCP‐1, IL‐6, and IL‐8, suggesting that angiogenic factors and inflammatory cytokines may affect the pathophysiologies of the two diseases differently.
Collapse
|
8
|
Özgürtaş T, Yücel Ç, Sertoğlu E, Hayran Y, Çolak S, Tekgöz E, Omma A, Durukan AH. Evaluation of the relationship of lymphangiogenesis markers with disease pathogenesis in patients with Behçet's uveitis. Acta Clin Belg 2022; 77:387-395. [PMID: 33629934 DOI: 10.1080/17843286.2021.1890451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The present study aims to evaluate the relationship between Behçet's uveitis and lymphangiogenesis by determining levels of Vascular endothelial growth factor-C (VEGF-C, its receptors sVEGFR-2, sVEGFR-3 and lymphangiogenesis markers podoplanin (PDPN) and lymphatic vessel endothelial hyaluronan receptor 1(LYVE-1), and C-type lectin domain family 1 member B (CLEC2). MATERIALS AND METHODS 55 patients with BD uveitis and 31 healthy control subjects were enrolled in the study. RESULTS sVEGFR-2, sVEGFR-3, VEGF-C/sVEGFR-2 ratio, PDPN and LYVE-1 levels were higher in the patient group. A positive correlation was found between LYVE-1 and hsCRP levels. PDPN had a strong predictive value for progression with a cut-off value of 2 pg/mL, with 69% sensitivity and 68% specificity (p = 0.001). CONCLUSION LYVE-1 and PDPN can be good representatives of the ongoing inflammatory processes in BD uveitis and point out that the disease can be related to lymphangiogenesis.
Collapse
Affiliation(s)
- Taner Özgürtaş
- Department of Clinical Biochemistry, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Çiğdem Yücel
- Department of Clinical Biochemistry, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Erdim Sertoğlu
- Department of Clinical Biochemistry, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Yıldız Hayran
- Department of Dermatology, Ankara City Hospital, Ankara, Turkey
| | - Seda Çolak
- Department of Rheumatology, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Emre Tekgöz
- Department of Rheumatology, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Ahmet Omma
- Department of Rheumatology, Ankara City Hospital, Ankara, Turkey
| | - Ali Hakan Durukan
- Department of Ophthalmology, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
9
|
Schirò G, Balistreri CR. The close link between brain vascular pathological conditions and neurodegenerative diseases: Focus on some examples and potential treatments. Vascul Pharmacol 2021; 142:106951. [PMID: 34942382 DOI: 10.1016/j.vph.2021.106951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Abstract
A close relationship is emerging among the age-related neurodegenerative decline, and the age-related typical alterations, dysfunctions, and related diseases of the cerobro-and/or cardiovascular system, which contributes in a significative manner to the triggering and progressing of neurodegenerative diseases (NeuroDegD). Specifically, macroinfarcts, microinfarcts, micro-hemorrhages (and particularly their number), atherosclerosis, arteriolosclerosis and cerebral amyloid angiopathy have been documented to be significantly associated with the onset of the cognitive impairment. In addition, vascular alterations and dysfunctions resulting in a reduced cerebral blood flow, and anomalies in the brain blood barrier (BBB), have been also demonstrated to contribute to NeuroDegD pathophysiologic processes. At the same time, such vascular alterations are also observed in cognitively unimpaired subjects. Here, some of these aspects are described with a particular focus on some NeuroDegD, as well as potential strategies for delaying or stopping their onset and progression.
Collapse
Affiliation(s)
- Giuseppe Schirò
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90134 Palermo, Italy
| | - Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90134 Palermo, Italy.
| |
Collapse
|
10
|
Ambari AM, Lilihata G, Zuhri E, Ekawati E, Wijaya SA, Dwiputra B, Sukmawan R, Radi B, Haryana SM, Adiarto S, Hanafy DA, Zamroni D, Elen E, Mangkuanom AS, Santoso A. External Counterpulsation Improves Angiogenesis by Preserving Vascular Endothelial Growth Factor-A and Vascular Endothelial Growth Factor Receptor-2 but Not Regulating MicroRNA-92a Expression in Patients With Refractory Angina. Front Cardiovasc Med 2021; 8:761112. [PMID: 34760951 PMCID: PMC8573065 DOI: 10.3389/fcvm.2021.761112] [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: 08/19/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Objective: External counterpulsation (ECP) provides long-term benefits of improved anginal frequency and exercise tolerance in patients with refractory angina (RA). This is postulated as a result of improved angiogenesis and endothelial function through an increase in shear stress. Angiogenesis is mainly represented by vascular endothelial growth factor-A (VEGF-A) and its receptor, vascular endothelial growth factor receptor-2 (VEGFR-2). The microRNA-92a (miR-92a) is a flow-sensitive miRNA that regulates atherosclerosis and angiogenesis in response to shear stress. Thus, ECP beneficial effect might be achieved through interaction between VEGF-A, VEGFR-2, and miR-92a. This study aims to evaluate the ECP effect on VEGF-A, VEGFR-2, and miR-92a in patients with RA in a sham-controlled manner. Methods: This was a randomized sham-controlled trial, enrolling 50 patients with RA who have coronary artery disease (CAD). Participants were randomized (1:1 ratio) to 35 sessions of either ECP (n = 25) or sham (n = 25), each session lasting for 1 h. Plasma levels of VEGF-A and VEGFR-2 were assayed by the ELISA technique. The quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to measure miR-92a circulating levels in plasma. Result: External counterpulsation significantly preserved VEGF-A and VEGFR-2 level compared to sham [ΔVEGF-A: 1 (-139 to 160) vs.-136 (-237 to 67) pg/ml, p = 0.026; ΔVEGFR-2: -171(-844 to +1,166) vs. -517(-1,549 to +1,407) pg/ml, p = 0.021, respectively]. Circulating miR-92a increased significantly in ECP [5.1 (4.2-6.4) to 5.9 (4.8-6.4), p < 0.001] and sham [5.2 (4.1-9.4) to 5.6 (4.8-6.3), p = 0.008] post-intervention. The fold changes tended to be higher in ECP group, although was not statistically different from sham [fold changes ECP = 4.6 (0.3-36.5) vs. sham 2.8 (0-15), p = 0.33)]. Conclusion: External counterpulsation improved angiogenesis by preserving VEGF-A and VEGFR-2 levels. Both ECP and sham increased miR-92a significantly, yet the changes were not different between the two groups. (Study registered on www.clinicaltrials.gov, no: NCT03991871, August 8, 2019, and received a grant from the National Health Research and Development of Ministry of Health of Indonesia, No: HK.02.02/I/27/2020).
Collapse
Affiliation(s)
- Ade Meidian Ambari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Gracia Lilihata
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Ervan Zuhri
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Elok Ekawati
- Division of Cardiovascular Research and Development, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Shoma Adhi Wijaya
- Division of Cardiovascular Research and Development, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Bambang Dwiputra
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Renan Sukmawan
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Basuni Radi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Sofia Mubarika Haryana
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Suko Adiarto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Dicky A. Hanafy
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Dian Zamroni
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Elen Elen
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Arwin S. Mangkuanom
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Anwar Santoso
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia - National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| |
Collapse
|
11
|
Iguchi M, Wada H, Shinozaki T, Suzuki M, Ajiro Y, Matsuda M, Koike A, Koizumi T, Shimizu M, Ono Y, Takenaka T, Sakagami S, Morita Y, Fujimoto K, Yonezawa K, Yoshida K, Ninomiya A, Nakamura T, Funada J, Kajikawa Y, Oishi Y, Kato T, Kotani K, Abe M, Akao M, Hasegawa K. Soluble vascular endothelial growth factor receptor 2 and prognosis in patients with chronic heart failure. ESC Heart Fail 2021; 8:4187-4198. [PMID: 34387398 PMCID: PMC8497334 DOI: 10.1002/ehf2.13555] [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: 03/22/2021] [Revised: 07/09/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Aims Endothelial cell vascular endothelial growth factor receptor 2 (VEGFR‐2) plays a pivotal role in angiogenesis, which induces physiological cardiomyocyte hypertrophy via paracrine signalling between endothelial cells and cardiomyocytes. We investigated whether a decrease in circulating soluble VEGFR‐2 (sVEGFR‐2) levels is associated with poor prognosis in patients with chronic heart failure (HF). Methods and results We performed a multicentre prospective cohort study of 1024 consecutive patients with HF, who were admitted to hospitals due to acute decompensated HF and were stabilized after initial management. Serum levels of sVEGFR‐2 were measured at discharge. Patients were followed up over 2 years. The outcomes were cardiovascular death, all‐cause death, major adverse cardiovascular events (MACE) defined as a composite of cardiovascular death and HF hospitalization, and HF hospitalization. The mean age of the patients was 75.5 (standard deviation, 12.6) years, and 57% were male. Patients with lower sVEGFR‐2 levels were older and more likely to be female, and had greater proportions of atrial fibrillation and anaemia, and lower proportions of diabetes, dyslipidaemia, and HF with reduced ejection fraction (<40%). During the follow‐up, 113 cardiovascular deaths, 211 all‐cause deaths, 350 MACE, and 309 HF hospitalizations occurred. After adjustment for potential clinical confounders and established biomarkers [N‐terminal B‐type natriuretic peptide (NT‐proBNP), high‐sensitivity cardiac troponin I, and high‐sensitivity C‐reactive protein], a low sVEGFR‐2 level below the 25th percentile was significantly associated with cardiovascular death [hazard ratio (HR), 1.79; 95% confidence interval (CI), 1.16–2.74] and all‐cause death (HR, 1.43; 95% CI, 1.04–1.94), but not with MACE (HR, 1.11; 95% CI, 0.86–1.43) or HF hospitalization (HR, 1.03; 95% CI, 0.78–1.35). The stratified analyses revealed that a low sVEGFR‐2 level below the 25th percentile was significantly associated with cardiovascular death (HR, 1.76; 95% CI, 1.07–2.85) and all‐cause death (HR, 1.49; 95% CI, 1.03–2.15) in the high‐NT‐proBNP group (above the median), but not in the low‐NT‐proBNP group. Notably, the patients with high‐NT‐proBNP and low‐sVEGFR‐2 (below the 25th percentile) had a 2.96‐fold higher risk (95% CI, 1.56–5.85) for cardiovascular death and a 2.40‐fold higher risk (95% CI, 1.52–3.83) for all‐cause death compared with those with low‐NT‐proBNP and high‐sVEGFR‐2. Conclusions A low sVEGFR‐2 value was independently associated with cardiovascular death and all‐cause death in patients with chronic HF. These associations were pronounced in those with high NT‐proBNP levels.
Collapse
Affiliation(s)
- Moritake Iguchi
- Department of Cardiology, National Hospital Organization Kyoto Medical Center, 1-1, Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan
| | - Hiromichi Wada
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tsuyoshi Shinozaki
- Department of Cardiology, National Hospital Organization Sendai Medical Center, Sendai, Japan
| | - Masahiro Suzuki
- Department of Clinical Research, National Hospital Organization Saitama Hospital, Wako, Japan
| | - Yoichi Ajiro
- Division of Clinical Research, National Hospital Organization Yokohama Medical Center, Yokohama, Japan
| | - Morihiro Matsuda
- Division of Preventive Medicine Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Akihiro Koike
- Department of Cardiology, National Hospital Organization Fukuokahigashi Medical Center, Koga, Japan
| | - Tomomi Koizumi
- Department of Cardiology, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Masatoshi Shimizu
- Department of Cardiology, National Hospital Organization Kobe Medical Center, Kobe, Japan
| | - Yujiro Ono
- Department of Cardiology, National Hospital Organization Higashihiroshima Medical Center, Hiroshima, Japan
| | - Takashi Takenaka
- Department of Cardiology, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| | - Satoru Sakagami
- Institute for Clinical Research, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Yukiko Morita
- Department of Cardiology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan
| | - Kazuteru Fujimoto
- Department of Cardiology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Kazuya Yonezawa
- Division of Clinical Research, National Hospital Organization Hakodate Hospital, Hakodate, Japan
| | - Kazuro Yoshida
- Department of Cardiology, National Hospital Organization Nagasakikawadana Medical Center, Higashisonogi, Japan.,Department of Cardiology, National Hospital Organization Nagasaki Hospital, Nagasaki, Japan
| | - Akiyo Ninomiya
- Department of Cardiology, National Hospital Organization Nagasakikawadana Medical Center, Higashisonogi, Japan
| | - Toshihiro Nakamura
- Department of Cardiology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Junichi Funada
- Department of Cardiology, National Hospital Organization Ehime Medical Center, Toon, Japan
| | - Yutaka Kajikawa
- Department of Cardiology, National Hospital Organization Fukuyama Medical Center, Fukuyama, Japan
| | - Yoshifumi Oishi
- Department of Cardiology, National Hospital Organization Higashitokushima Medical Center, Itano, Japan
| | - Toru Kato
- Department of Clinical Research, National Hospital Organization Tochigi Medical Center, Utsunomiya, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Mitsuru Abe
- Department of Cardiology, National Hospital Organization Kyoto Medical Center, 1-1, Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan
| | - Masaharu Akao
- Department of Cardiology, National Hospital Organization Kyoto Medical Center, 1-1, Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan
| | - Koji Hasegawa
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | | |
Collapse
|
12
|
Divergent Impact of Breast Cancer Laterality on Clinicopathological, Angiogenic, and Hemostatic Profiles: A Potential Role of Tumor Localization in Future Outcomes. J Clin Med 2020; 9:jcm9061708. [PMID: 32498398 PMCID: PMC7356400 DOI: 10.3390/jcm9061708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 01/07/2023] Open
Abstract
To date, lateral differences of invasive breast cancer (IBrC) with respect to the angiogenic and hemostatic profiles were never studied. Here, we aimed to determine the relationship of tumor laterality with various clinical and pathological parameters including angiogenic and hemostatic profiles. A total of 92 women that were initially non-metastatic and treated by surgery were included in this single-center prospective study. Patients were grouped according to tumor localization. A four-year follow-up was accomplished in all patients with a 15.22% recurrence rate. An immunoassay of selected angiogenic and hemostatic parameters, as well as immunohistochemistry of estrogen and progesterone receptors, human epidermal growth factor receptor 2 (HER2), and Ki67, was comparatively performed in groups with right- and left-sided IBrC. The same analysis was carried out in a subgroup of patients with luminal A molecular subtype of cancer. Patients with right-sided tumors free of nodal involvement had a significantly longer overall survival compared to their left-sided counterparts (p = 0.0491). Additionally, right-sided tumors had a higher predisposition to be a luminal-A subtype of IBrC (p = 0.0016). Furthermore, 10% of left-sided tumors exhibited an overexpression of HER2, while only 2% patients suffering right-sided tumors displayed a positive score (p = 0.0357). Our findings revealed a significantly higher concentration of vascular endothelial growth factor (VEGF)-A (p = 0.0136), lower anti-angiogenic ratios (sVEGFR1/VEGF-A (p = 0.0208) and sVEGFR2/VEGF-A (p = 0.0068)), and elevated plasminogen activator inhibitor type 1 (PAI-1) (p = 0.0229) in patients with breast cancer localized in the left breast, regardless of the molecular subtype of IBrC. Our study showed that left-sided breast tumors without lymph node metastases demonstrate worse overall survival. Laterality of IBrC is associated with pro-angiogenic and pro-thrombotic conditions. We propose to consider laterality as a prognostic factor of IBrC.
Collapse
|
13
|
Okamoto M, Watanabe M, Inoue N, Ogawa K, Hidaka Y, Iwatani Y. Gene polymorphisms of VEGF and VEGFR2 are associated with the severity of Hashimoto's disease and the intractability of Graves' disease, respectively. Endocr J 2020; 67:545-559. [PMID: 32074519 DOI: 10.1507/endocrj.ej19-0480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is one of main regulators of angiogenesis that functions by binding to its receptors, including VEGF receptor (VEGFR) 2. There are few data available regarding the association between VEGF and VEGFR polymorphisms and the susceptibility to and prognosis of autoimmune thyroid diseases (AITDs). To elucidate this association, we genotyped four functional VEGF and two VEGFR2 polymorphisms and measured serum VEGF levels. In the four functional VEGF polymorphisms, the frequencies of the I carrier and I allele of VEGF -2549 I/D, which has lower activity, were higher in patients with severe HD than in those with mild HD. In the two functional VEGFR2 polymorphisms, the frequency of the rs2071559 CC genotype, which has higher activity, was higher in patients with intractable GD than in controls, and the proportion of GD patients with larger goiters was higher in those with the CC genotype. Moreover, the frequency of the rs1870377 TT genotype with higher activity was higher in patients with intractable GD than in those with GD in remission. Combinations of VEGF and VEGFR2 polymorphisms with stronger interactions were associated with the intractability of GD. Serum VEGF levels were higher in HD and AITD patients than those in controls. In conclusion, VEGF polymorphisms with lower activity were associated with the severity of HD, while VEGFR2 polymorphisms and the combinations of VEGF and VEGFR2 polymorphisms, which have stronger interactions, were associated with the intractability of GD. VEGF and VEGFR2 polymorphisms were associated with HD severity and GD intractability, respectively.
Collapse
Affiliation(s)
- Mami Okamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Mikio Watanabe
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Naoya Inoue
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Laboratory for Clinical Investigation, Osaka University Hospital, Osaka 565-0871, Japan
| | - Kazane Ogawa
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yoh Hidaka
- Laboratory for Clinical Investigation, Osaka University Hospital, Osaka 565-0871, Japan
| | - Yoshinori Iwatani
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| |
Collapse
|
14
|
A Novel 4-Gene Score to Predict Survival, Distant Metastasis and Response to Neoadjuvant Therapy in Breast Cancer. Cancers (Basel) 2020; 12:cancers12051148. [PMID: 32370309 PMCID: PMC7281399 DOI: 10.3390/cancers12051148] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
We generated a 4-gene score with genes upregulated in LM2-4, a metastatic variant of MDA-MB-231 (DOK 4, HCCS, PGF, and SHCBP1) that was strongly associated with disease-free survival (DFS) in TCGA cohort (hazard ratio [HR]>1.2, p < 0.02). The 4-gene score correlated with overall survival of TCGA (HR = 1.44, p < 0.001), which was validated with DFS and disease-specific survival of METABRIC cohort. The 4-gene score was able to predict worse survival or clinically aggressive tumors, such as high Nottingham pathological grade and advanced cancer staging. High score was associated with worse survival in the hormonal receptor (HR)-positive/Her2-negative subtype. High score enriched cell proliferation-related gene sets in GSEA. The score was high in primary tumors that originated, in and metastasized to, brain and lung, and it predicted worse progression-free survival for metastatic tumors. Good tumor response to neoadjuvant chemotherapy or hormonal therapy was accompanied by score reduction. High scores were also predictive of response to neoadjuvant chemotherapy for HR-positive/Her2-negative subtype. High score tumors had increased expression of T cell exhaustion marker genes, suggesting that the score may also be a biomarker for immunotherapy response. Our novel 4-gene score with both prognostic and predictive values may, therefore, be clinically useful particularly in HR-positive breast cancer.
Collapse
|
15
|
Auvray M, Tougeron D, Auclin E, Moulin V, Artru P, Hautefeuille V, Hammel P, Lecomte T, Locher C, Sickersen G, Coriat R, Lecaille C, Vernerey D, Taieb J, Pernot S. Efficacy and Safety of Aflibercept in Combination With Chemotherapy Beyond Second-Line Therapy in Metastatic Colorectal Carcinoma Patients: An AGEO Multicenter Study. Clin Colorectal Cancer 2020; 19:39-47.e5. [PMID: 31648924 DOI: 10.1016/j.clcc.2019.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/10/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Although no data have been reported beyond second-line therapy, aflibercept is approved in this setting in many countries. We conducted a multicenter study to analyze the efficacy and safety of a aflibercept-chemotherapy regimen beyond second-line therapy in patients with metastatic colorectal cancer. PATIENTS AND METHODS Metastatic colorectal cancer patients treated with aflibercept beyond second-line therapy were included. Objective response rate, overall survival (OS), and progression-free survival (PFS) were assessed. RESULTS A total of 130 patients were included. Median OS and PFS were 7.6 months (95% confidence interval, 6.2-9.3) and 3.3 months (95% confidence interval, 2.7-3.8), respectively. The best response rates were partial response 6.9%, stable disease 38.5%, progressive disease 42.5%, and not evaluable 12%. According to whether patients received previous FOLFIRI (leucovorin, 5-fluorouracil, irinotecan, and oxaliplatin)-bevacizumab or not, OS was 7.7 and 8.1 months (P = .31), and PFS was 2.9 and 3.9 months (P = .02), respectively. Interestingly, PFS and OS were both significantly improved by 4% and 5% per month, respectively, without antiangiogenic treatment before the initiation of the aflibercept regimen. The negative effect of prior FOLFIRI-bevacizumab or shorter time since last bevacizumab was maintained in multivariate analysis for both OS and PFS. CONCLUSION The aflibercept-chemotherapy regimen is a therapeutic option in patients with chemorefractory disease beyond second-line therapy, in particular in patients with an antiangiogenic-free interval.
Collapse
Affiliation(s)
- Marie Auvray
- Department of Digestive Oncology, European Georges Pompidou Hospital, René Descartes University, Paris, France
| | - David Tougeron
- Department of Hepato-Gastroenterology, Center Hospitalo-Universitaire de Poitiers, Poitiers, France
| | - Edouard Auclin
- Methodology and quality of life unit in oncology, University hospital of Besançon, Besançon, France; University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Valérie Moulin
- Department of Digestive Oncology Center Hospitalier de La Rochelle, La Rochelle, France
| | - Pascal Artru
- Department of Hepato-Gastroenterology, Jean Mermoz Hospital, Lyon, France
| | | | - Pascal Hammel
- Department of Digestive Oncology, Beaujon University Hospital, AP-HP and University Paris 7, Denis Diderot, Clichy, France
| | - Thierry Lecomte
- Department of Hepato-Gastroenterology and Digestive Oncology, CHU de Tours, Hôpital Trousseau, Tours, France
| | - Christophe Locher
- Department of Hepato-Gastroenterology, Center Hospitalier de Meaux, Meaux, France
| | - Gaëlle Sickersen
- Department of Hepato-Gastroenterology, CHU Saint-Louis, Paris, France
| | - Romain Coriat
- Department of Hepato-Gastroenterology and Digestive Oncology, CHU Cochin, Paris, France
| | - Cédric Lecaille
- Department of Gastroenterology and Digestive Oncology, Polyclinique de Bordeaux Nord, Bordeaux, France
| | - Dewi Vernerey
- Methodology and quality of life unit in oncology, University hospital of Besançon, Besançon, France
| | - Julien Taieb
- Department of Digestive Oncology, European Georges Pompidou Hospital, René Descartes University, Paris, France
| | - Simon Pernot
- Department of Digestive Oncology, European Georges Pompidou Hospital, René Descartes University, Paris, France.
| |
Collapse
|
16
|
Hait NC, Maiti A, Xu P, Qi Q, Kawaguchi T, Okano M, Takabe K, Yan L, Luo C. Regulation of hypoxia-inducible factor functions in the nucleus by sphingosine-1-phosphate. FASEB J 2020; 34:4293-4310. [PMID: 32017264 PMCID: PMC10112293 DOI: 10.1096/fj.201901734rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
Sphingosine kinase 2 (SphK2) is known to phosphorylate the nuclear sphingolipid metabolite to generate sphingosine-1-phosphate (S1P). Nuclear S1P is involved in epigenetic regulation of gene expression; however, the underlying mechanisms are not well understood. In this work, we have identified the role of nuclear S1P and SphK2 in regulating hypoxia-responsive master transcription factors hypoxia-inducible factor (HIF)-1α/2α, and their functions in breast cancer, with a focus on triple-negative breast cancer (TNBC). We have shown SphK2 is associated with HIF-1α in protein complexes, and is enriched at the promoters of HIF target genes, including vascular endothelial growth factor (VEGF), where it enhances local histone H3 acetylation and transcription. S1P specifically binds to the PAS domains of HIF-1α. SphK2, and HIF-1α expression levels are elevated in metastatic estrogen receptor-positive (ER+) and TNBC clinical tissue specimens compared to healthy breast tissue samples. To determine if S1P formation in the nucleus by SphK2 is a key regulator of HIF functions, we found using a preclinical TNBC xenograft mouse model, and an existing selective SphK2 inhibitor K-145, that nuclear S1P, histone acetylation, HIF-1α expression, and TNBC tumor growth were all reduced in vivo. Our results suggest that S1P and SphK2 in the nucleus are linked to the regulation of HIF-1α/2α functions associated with breast cancer progression, and may provide potential therapeutic targets.
Collapse
Affiliation(s)
- Nitai C Hait
- Division of Breast Surgery and Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Aparna Maiti
- Division of Breast Surgery and Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Pan Xu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Qianya Qi
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tsutomu Kawaguchi
- Division of Breast Surgery and Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Maiko Okano
- Division of Breast Surgery and Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kazuaki Takabe
- Division of Breast Surgery and Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Vaghi C, Rodallec A, Fanciullino R, Ciccolini J, Mochel JP, Mastri M, Poignard C, Ebos JML, Benzekry S. Population modeling of tumor growth curves and the reduced Gompertz model improve prediction of the age of experimental tumors. PLoS Comput Biol 2020; 16:e1007178. [PMID: 32097421 PMCID: PMC7059968 DOI: 10.1371/journal.pcbi.1007178] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 03/06/2020] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor growth curves are classically modeled by means of ordinary differential equations. In analyzing the Gompertz model several studies have reported a striking correlation between the two parameters of the model, which could be used to reduce the dimensionality and improve predictive power. We analyzed tumor growth kinetics within the statistical framework of nonlinear mixed-effects (population approach). This allowed the simultaneous modeling of tumor dynamics and inter-animal variability. Experimental data comprised three animal models of breast and lung cancers, with 833 measurements in 94 animals. Candidate models of tumor growth included the exponential, logistic and Gompertz models. The exponential and-more notably-logistic models failed to describe the experimental data whereas the Gompertz model generated very good fits. The previously reported population-level correlation between the Gompertz parameters was further confirmed in our analysis (R2 > 0.92 in all groups). Combining this structural correlation with rigorous population parameter estimation, we propose a reduced Gompertz function consisting of a single individual parameter (and one population parameter). Leveraging the population approach using Bayesian inference, we estimated times of tumor initiation using three late measurement timepoints. The reduced Gompertz model was found to exhibit the best results, with drastic improvements when using Bayesian inference as compared to likelihood maximization alone, for both accuracy and precision. Specifically, mean accuracy (prediction error) was 12.2% versus 78% and mean precision (width of the 95% prediction interval) was 15.6 days versus 210 days, for the breast cancer cell line. These results demonstrate the superior predictive power of the reduced Gompertz model, especially when combined with Bayesian estimation. They offer possible clinical perspectives for personalized prediction of the age of a tumor from limited data at diagnosis. The code and data used in our analysis are publicly available at https://github.com/cristinavaghi/plumky.
Collapse
Affiliation(s)
- Cristina Vaghi
- MONC team, Inria Bordeaux Sud-Ouest, Talence, France
- Institut de Mathématiques de Bordeaux, CNRS UMR 5251, Bordeaux University, Talence, France
| | - Anne Rodallec
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Aix Marseille Université, Marseille, France; Laboratoire de Pharmacocinétique et Toxicologie, La Timone University Hospital of Marseille, Marseille, France
| | - Raphaëlle Fanciullino
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Aix Marseille Université, Marseille, France; Laboratoire de Pharmacocinétique et Toxicologie, La Timone University Hospital of Marseille, Marseille, France
| | - Joseph Ciccolini
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Aix Marseille Université, Marseille, France; Laboratoire de Pharmacocinétique et Toxicologie, La Timone University Hospital of Marseille, Marseille, France
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Clair Poignard
- MONC team, Inria Bordeaux Sud-Ouest, Talence, France
- Institut de Mathématiques de Bordeaux, CNRS UMR 5251, Bordeaux University, Talence, France
| | - John M. L. Ebos
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
- Departments of Medicine and Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Sébastien Benzekry
- MONC team, Inria Bordeaux Sud-Ouest, Talence, France
- Institut de Mathématiques de Bordeaux, CNRS UMR 5251, Bordeaux University, Talence, France
| |
Collapse
|
18
|
Zarychta E, Rhone P, Bielawski K, Michalska M, Rość D, Ruszkowska-Ciastek B. Anti-angiogenic efficacy in invasive breast carcinoma patients depends on clinicopathological determinants. Adv Med Sci 2019; 64:216-223. [PMID: 30818220 DOI: 10.1016/j.advms.2019.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/19/2018] [Accepted: 02/08/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE The biggest problem with the occurrence of breast cancer is late diagnosis, which is associated with high mortality rates. The aim of the study was to appraise the number of circulating endothelial precursors and the concentration of vascular endothelial growth factor A (VEGF-A) and the soluble forms of its receptors, sVEGFR1 and sVEGFR2, in breast cancer patients with respect to clinicopathological features. MATERIAL AND METHODS The study involved 85 women of Caucasian ethnicity aged 45-66 with primary breast cancer without distant metastases (M0). Inclusion criteria were as follows: histopathological examination confirming the diagnosis of primary breast cancer, without previous radiotherapy and chemotherapy. Immunohistochemistry evaluation of oestrogen and progesterone receptors, human epidermal growth factor receptor 2, Ki67 expression was made in all cases. In the EDTA-plasma, the concentrations of VEGF-A and its soluble receptors, sVEGFR1 and sVEGFR2, were measured applying immunoassay techniques. Circulating endothelial progenitor cells (EPCs) were identified with the immune-phenotype CD45-, CD34+, CD133+, CD31+ using flow cytometry. RESULTS Older women with breast cancer had significantly higher concentrations of VEGF-A as well as sVEGFR2 compared with their younger counterparts. A significantly higher concentration of the soluble form of VEGF receptor type 1 in patients with T1 breast cancer in relation to T2 cases was noted. Also, negative correlations between circulating EPCs and histological grading as well as a soluble form of VEGFR2 with histological grading of breast cancer according to the Elston-Ellis classification were observed. CONCLUSIONS Anti-angiogenic potential is divergent in relation to the clinicopathological determinants.
Collapse
|
19
|
Enhanced efficacy of sitravatinib in metastatic models of antiangiogenic therapy resistance. PLoS One 2019; 14:e0220101. [PMID: 31369645 PMCID: PMC6675057 DOI: 10.1371/journal.pone.0220101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/08/2019] [Indexed: 01/09/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) that primarily target angiogenesis are approved to treat several cancers in the metastatic setting; however, resistance is common. Sequential treatment or 'switching' from one TKI to another following failure can be effective, but predicting which drugs will have cross-over sensitivity remains a challenge. Here we examined sitravatinib (MGCD516), a spectrum-selective TKI able to block MET, TAM (TYRO3, AXL, MerTK) and multiple receptor families (including PDGFRs, VEGFRs, and Ephs). Transcriptomic analysis of several mouse and human cell lines revealed diverse molecular changes after resistance to two TKIs (sunitinib and axitinib) with multiple sitravatinib targets found to be upregulated. Sitravatinib treatment in vitro resulted in enhanced anti-proliferative effects in resistant cells and was improved compared to TKIs with similar target profiles. In vivo, primary tumor growth inhibition after sitravatinib treatment in mice was enhanced in resistant tumors and metastasis suppression improved when tumors were surgically removed. Together, these results suggest that the diverse and often inconsistent compensatory signaling mechanisms found to contribute to TKI resistance may paradoxically improve the tumor-inhibiting effects of broad-spectrum TKIs such as sitravatinib that are able to block multiple signaling pathways. Sitravatinib in the second-line setting following antiangiogenic TKI treatment may have enhanced inhibitory effects in local and disseminated disease, and improve outcomes in patients with refractory disease.
Collapse
|
20
|
Deyell RJ, Wu B, Rassekh SR, Tu D, Samson Y, Fleming A, Bouffet E, Sun X, Powers J, Seymour L, Baruchel S, Morgenstern DA. Phase I study of vinblastine and temsirolimus in pediatric patients with recurrent or refractory solid tumors: Canadian Cancer Trials Group Study IND.218. Pediatr Blood Cancer 2019; 66:e27540. [PMID: 30393943 DOI: 10.1002/pbc.27540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/11/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022]
Abstract
UNLABELLED Combining mammalian target of rapamycin (mTOR) inhibitors and vinca alkaloids has shown therapeutic synergy in xenograft models of pediatric cancers. This phase I study assessed safety and toxicity of temsirolimus in combination with vinblastine in children. PROCEDURE Patients ≥ 1 and ≤ 18 years with recurrent/refractory solid or CNS tumors were eligible. Vinblastine (4 mg/m2 ) and temsirolimus (15 mg/m2 ) were administered i.v. weekly, with planned dose escalation of vinblastine using a rolling six phase I design. Pharmacokinetic and pharmacodynamic data were collected. RESULTS Seven patients with median age 12 years (range, 8-18 years) were enrolled; all were evaluable for toxicity and six for response. At dose level 1, four of six patients developed grade 3 mucositis, of which one met duration criteria for dose-limiting toxicity (DLT). Four patients required dose omissions for grade 3 or 4 hematologic toxicity, including one prolonged neutropenia DLT. A subsequent patient was enrolled on dose level -2 (temsirolimus 10 mg/m2 , vinblastine 4 mg/m2 ) with no protocol-related toxicity > grade 1, except grade 2 neutropenia. Two serious adverse events (SAE) occurred-an allergic reaction to temsirolimus (grade 2) and an intracranial hemorrhage in a CNS tumor patient (grade 3)-unlikely related to study therapy. Soluble VEGFR2 was reduced at cycle 1, day 36 in keeping with inhibition of angiogenesis. Four patients achieved prolonged stable disease for a median of 5.0 months (range, 3.1-8.3 months). CONCLUSION The combination of weekly temsirolimus (15 mg/m2 ) and vinblastine (4 mg/m2 ) exceeds the maximum tolerated dose in children, with frequent oral mucositis and hematologic toxicity.
Collapse
Affiliation(s)
- Rebecca J Deyell
- Division of Pediatric Hematology/Oncology/BMT, University of British Columbia, British Columbia Children's Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Bing Wu
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Rod Rassekh
- Division of Pediatric Hematology/Oncology/BMT, University of British Columbia, British Columbia Children's Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Yvan Samson
- Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Adam Fleming
- McMaster Children's Hospital at Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Eric Bouffet
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqun Sun
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Jean Powers
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Lesley Seymour
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Sylvain Baruchel
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniel A Morgenstern
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Abstract
Alzheimer’s disease (AD) is a common disorder of progressive cognitive decline among elderly subjects. Angiogenesis-related factors including vascular endothelial growth factor (VEGF) might be involved in the pathogenesis of AD. Soluble form of the VEGF receptor is likely to be an intrinsic negative counterpart of VEGF. We measured the plasma levels of VEGF and its two soluble receptors (sVEGFR1 and sVEGFR2) in 120 control subjects, 75 patients with mild cognitive impairment, and 76 patients with AD using ELISA. Plasma levels of VEGF in patients with AD were higher than those in healthy control subjects. However, plasma levels of sVEGFR1 and sVEGFR2 were lower in patients with AD than in healthy control subjects. Levels of VEGFR2 mRNA were significantly decreased in human umbilical vein endothelial cells after amyloid-beta treatment. Further, protein levels of VEGFR2 were also decreased in the brains of AD model mice. In addition, we show that the expression of sVEGFR2 and VEGFR2 was also decreased by the transfection with the Notch intracellular domain. These results indicate that the alterations of VEGF and its two receptors levels might be associated with those at risk for Alzheimer’s disease.
Collapse
|
22
|
Komatsu H, Oishi T, Itamochi H, Shimada M, Sato S, Chikumi J, Sato S, Nonaka M, Sawada M, Wakahara M, Umekita Y, Harada T. Serum Vascular Endothelial Growth Factor-A as a Prognostic Biomarker for Epithelial Ovarian Cancer. Int J Gynecol Cancer 2017; 27:1325-1332. [PMID: 28557832 DOI: 10.1097/igc.0000000000001027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Bevacizumab, which targets vascular endothelial growth factor (VEGF)-A, has recently been proven to be effective for the treatment of epithelial ovarian cancer (EOC). Thus, interest in VEGF-A has increased. There are few reports on concomitant detection of both ligands and its soluble receptors in serum samples, and the significance of serum VEGF-A in EOC is unclear, unlike the situation with tissue samples. We conducted the present study to explore the levels of serum VEGF family and its receptors and to evaluate their utility as prognostic biomarkers. METHODS A total of 128 patients with EOC, who were consecutively treated at Tottori University Hospital between 2006 and 2012, were included. Blood samples were collected before initial surgery. Serum concentrations of VEGF-A, VEGF-C, VEGFR-1, and VEGFR-2 were analyzed by enzyme-linked immunosorbent assay. We also examined the mRNA and protein expression of VEGF-A in tumor tissue from 30 cases by real-time reverse transcription polymerase chain reaction and immunohistochemistry. RESULTS The levels of VEGF-A in patients with stage III/IV disease were significantly higher than those with stage I/II disease (P = 0.0036). On the other hand, the level of VEGFR-2 in stage III/IV was significantly lower than that in stage I/II (P = 0.0026). With the cutoff value of VEGF/VEGFRs at the median level, the overall survival (OS) for patients with high VEGF-A levels was significantly lower than those with low levels (P = 0.015). Patients with high VEGFR-2 levels showed better prognosis than those with low VEGFR-2 levels (P = 0.023). Multivariate analysis revealed that International Federation of Gynecology and Obstetrics stage and serum VEGF-A were independent prognostic factors for OS [hazard ratio 2.01, 95% confidence interval (1.13-3.63), P = 0.017]. There was no significant correlation between mRNA or protein expression and serum levels of VEGF-A. CONCLUSIONS Serum VEGF-A is an independent prognostic factor for OS in patients with EOC, implying that serum VEGF-A is a prognostic biomarker for EOC. Further study to validate the data is needed.
Collapse
Affiliation(s)
- Hiroaki Komatsu
- *Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago; †Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka; and ‡Department of Pathology, Tottori University School of Medicine, Yonago, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Schindler E, Amantea MA, Karlsson MO, Friberg LE. A Pharmacometric Framework for Axitinib Exposure, Efficacy, and Safety in Metastatic Renal Cell Carcinoma Patients. CPT Pharmacometrics Syst Pharmacol 2017; 6:373-382. [PMID: 28378918 PMCID: PMC5488123 DOI: 10.1002/psp4.12193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/15/2023] Open
Abstract
The relationships between exposure, biomarkers (vascular endothelial growth factor (VEGF), soluble VEGF receptors (sVEGFR)-1, -2, -3, and soluble stem cell factor receptor (sKIT)), tumor sum of longest diameters (SLD), diastolic blood pressure (dBP), and overall survival (OS) were investigated in a modeling framework. The dataset included 64 metastatic renal cell carcinoma patients (mRCC) treated with oral axitinib. Biomarker timecourses were described by indirect response (IDR) models where axitinib inhibits sVEGFR-1, -2, and -3 production, and VEGF degradation. No effect was identified on sKIT. A tumor model using sVEGFR-3 dynamics as driver predicted SLD data well. An IDR model, with axitinib exposure stimulating the response, characterized dBP increase. In a time-to-event model the SLD timecourse predicted OS better than exposure, biomarker- or dBP-related metrics. This type of framework can be used to relate pharmacokinetics, efficacy, and safety to long-term clinical outcome in mRCC patients treated with VEGFR inhibitors. (ClinicalTrial.gov identifier NCT00569946.).
Collapse
Affiliation(s)
- E Schindler
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| | | | - MO Karlsson
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| | - LE Friberg
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
24
|
Roskoski R. Vascular endothelial growth factor (VEGF) and VEGF receptor inhibitors in the treatment of renal cell carcinomas. Pharmacol Res 2017; 120:116-132. [PMID: 28330784 DOI: 10.1016/j.phrs.2017.03.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 12/12/2022]
Abstract
One Von Hippel-Lindau (VHL) tumor suppressor gene is lost in most renal cell carcinomas while the nondeleted allele exhibits hypermethylation-induced inactivation or inactivating somatic mutations. As a result of these genetic modifications, there is an increased production of VEGF-A and pro-angiogenic growth factors in this disorder. The important role of angiogenesis in the pathogenesis of renal cell carcinomas and other tumors has focused the attention of investigators on the biology of VEGFs and VEGFR1-3 and to the development of inhibitors of the intricate and multifaceted angiogenic pathways. VEGFR1-3 contain an extracellular segment with seven immunoglobulin-like domains, a transmembrane segment, a juxtamembrane segment, a protein kinase domain with an insert of about 70 amino acid residues, and a C-terminal tail. VEGF-A stimulates the activation of preformed VEGFR2 dimers by the auto-phosphorylation of activation segment tyrosines followed by the phosphorylation of additional protein-tyrosines that recruit phosphotyrosine binding proteins thereby leading to signalling by the ERK1/2, AKT, Src, and p38 MAP kinase pathways. VEGFR1 modulates the activity of VEGFR2, which is the chief pathway in vasculogenesis and angiogenesis. VEGFR3 and its ligands (VEGF-C and VEGF-D) are involved primarily in lymphangiogenesis. Small molecule VEGFR1/2/3 inhibitors including axitinib, cabozantinib, lenvatinib, sorafenib, sunitinib, and pazopanib are approved by the FDA for the treatment of renal cell carcinomas. Most of these agents are type II inhibitors of VEGFR2 and inhibit the so-called DFG-Aspout inactive enzyme conformation. These drugs are steady-state competitive inhibitors with respect to ATP and like ATP they form hydrogen bonds with the hinge residues that connect the small and large protein kinase lobes. Bevacizumab, a monoclonal antibody that binds to VEGF-A, is also approved for the treatment of renal cell carcinomas. Resistance to these agents invariably occurs within one year of treatment and clinical studies are underway to determine the optimal sequence of treatment with these anti-angiogenic agents. The nivolumab immune checkpoint inhibitor is also approved for the second-line treatment of renal cell carcinomas. Owing to the resistance of renal cell carcinomas to cytotoxic drugs and radiation therapy, the development of these agents has greatly improved the therapeutic options in the treatment of these malignancies.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116, Box 19, Horse Shoe, NC 28742-8814, United States.
| |
Collapse
|
25
|
Iwona BS. Growth Factors in the Pathogenesis of Retinal Neurodegeneration in Diabetes Mellitus. Curr Neuropharmacol 2017; 14:792-804. [PMID: 27528260 PMCID: PMC5333593 DOI: 10.2174/1570159x14666160813182009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/12/2015] [Accepted: 01/06/2015] [Indexed: 12/13/2022] Open
Abstract
Neurodegeneration is an initial process in the development of diabetic retinopathy (DR). High quantities of glutamate, oxidative stress, induction of the renin-angiotensin system (RAS) and elevated levels of RAGE are crucial elements in the retinal neurodegeneration caused by diabetes mellitus. At least, there is emerging proof to indicate that the equilibrium between the neurotoxic and neuroprotective components will affect the state of the retinal neurons. Somatostatin (SST), pigment epithelium-derived factor (PEDF), and erythropoietin (Epo) are endogenous neuroprotective peptides that are decreased in the eye of diabetic persons and play an essential role in retinal homeostasis. On the other hand, insulin-like growth factor 1 (IGF-1), and vascular endothelial growth factor (VEGF) are pivotal proteins which participate in the development of new capillaries and finally cause damage to the retinal neurons. During recent years, our knowledge about the function of growth factors in the pathogenesis of retinal neurodegeneration has increased. However, intensive investigations are needed to clarify the basic processes that contribute to retinal neurodegeneration and its association with damage to the capillary blood vessels. The objective of this review article is to show new insights on the role of neurotransmitters and growth factors in the pathogenesis of diabetic retinopathy. The information contained in this manuscript may provide the basis for novel strategies based on the factors of neurodegeneration to diagnose, prevent and treat DR in its earliest phases.
Collapse
Affiliation(s)
- Ben-Skowronek Iwona
- Department Pediatric Endocrinology and Diabetology, Medical University of Lublin, ul. Prof. A. Gebali 6, 20-093 Lublin, Poland
| |
Collapse
|
26
|
Maiti A, Takabe K, Hait NC. Metastatic triple-negative breast cancer is dependent on SphKs/S1P signaling for growth and survival. Cell Signal 2017; 32:85-92. [PMID: 28108260 DOI: 10.1016/j.cellsig.2017.01.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/16/2022]
Abstract
About 40,000 American women die from metastatic breast cancer each year despite advancements in treatment. Approximately, 15% of breast cancers are triple-negative for estrogen receptor, progesterone receptor, and HER2. Triple-negative cancer is characterized by more aggressive, harder to treat with conventional approaches and having a greater possibility of recurrence. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid signaling mediator has emerged as a key regulatory molecule in breast cancer progression. Therefore, we investigated whether cytosolic sphingosine kinase type 1 (SphK1) and nuclear sphingosine kinase type 2 (SphK2), the enzymes that make S1P are critical for growth and PI3K/AKT, ERK-MAP kinase mediated survival signaling of lung metastatic variant LM2-4 breast cancer cells, generated from the parental triple-negative MDA-MB-231 human breast cancer cell line. Similar with previous report, SphKs/S1P signaling is critical for the growth and survival of estrogen receptor positive MCF-7 human breast cancer cells, was used as our study control. MDA-MB-231 did not show a significant effect of SphKs/S1P signaling on AKT, ERK, and p38 pathways. In contrast, LM2-4 cells that gained lung metastatic phenotype from primary MDA-MB-231 cells show a significant effect of SphKs/S1P signaling requirement on cell growth, survival, and cell motility. PF-543, a selective potent inhibitor of SphK1, attenuated epidermal growth factor (EGF)-mediated cell growth and survival signaling through inhibition of AKT, ERK, and p38 MAP kinase pathways mainly in LM2-4 cells but not in parental MDA-MB-231 human breast cancer cells. Moreover, K-145, a selective inhibitor of SphK2, markedly attenuated EGF-mediated cell growth and survival of LM2-4 cells. We believe this study highlights the importance of SphKs/S1P signaling in metastatic triple-negative breast cancers and targeted therapies.
Collapse
Affiliation(s)
- Aparna Maiti
- Roswell Park Cancer Institute, Division of Breast Surgery, Department of Surgical Oncology, Department of Molecular and Cellular Biology, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Kazuaki Takabe
- Roswell Park Cancer Institute, Division of Breast Surgery, Department of Surgical Oncology, Department of Molecular and Cellular Biology, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Nitai C Hait
- Roswell Park Cancer Institute, Division of Breast Surgery, Department of Surgical Oncology, Department of Molecular and Cellular Biology, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
27
|
Mailankody S, Devlin SM, Korde N, Lendvai N, Lesokhin A, Landau H, Hassoun H, Ballagi A, Ekman D, Chung DJ, Patel M, Koehne G, Giralt S, Landgren O. Proteomic profiling in plasma cell disorders: a feasibility study. Leuk Lymphoma 2016; 58:1757-1759. [PMID: 27908223 DOI: 10.1080/10428194.2016.1258699] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Sham Mailankody
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Sean M Devlin
- c Department of Biostatistics and Epidemiology , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Neha Korde
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Nikoletta Lendvai
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Alexander Lesokhin
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Heather Landau
- b Department of Medicine , Weill Cornell Medical College , New York , NY , USA.,d Department of Medicine, Adult Bone Marrow Transplant Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Hani Hassoun
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | | | | | - David J Chung
- b Department of Medicine , Weill Cornell Medical College , New York , NY , USA.,d Department of Medicine, Adult Bone Marrow Transplant Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Minal Patel
- f Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Guenther Koehne
- b Department of Medicine , Weill Cornell Medical College , New York , NY , USA.,d Department of Medicine, Adult Bone Marrow Transplant Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Sergio Giralt
- b Department of Medicine , Weill Cornell Medical College , New York , NY , USA.,d Department of Medicine, Adult Bone Marrow Transplant Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Ola Landgren
- a Department of Medicine, Myeloma Service , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,b Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| |
Collapse
|
28
|
Ashley SL, Xia M, Murray S, O’Dwyer DN, Grant E, White ES, Flaherty KR, Martinez FJ, Moore BB. Six-SOMAmer Index Relating to Immune, Protease and Angiogenic Functions Predicts Progression in IPF. PLoS One 2016; 11:e0159878. [PMID: 27490795 PMCID: PMC4973878 DOI: 10.1371/journal.pone.0159878] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/08/2016] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Biomarkers in easily accessible compartments like peripheral blood that can predict disease progression in idiopathic pulmonary fibrosis (IPF) would be clinically useful regarding clinical trial participation or treatment decisions for patients. In this study, we used unbiased proteomics to identify relevant disease progression biomarkers in IPF. METHODS Plasma from IPF patients was measured using an 1129 analyte slow off-rate modified aptamer (SOMAmer) array, and patient outcomes were followed over the next 80 weeks. Receiver operating characteristic (ROC) curves evaluated sensitivity and specificity for levels of each biomarker and estimated area under the curve (AUC) when prognostic biomarker thresholds were used to predict disease progression. Both logistic and Cox regression models advised biomarker selection for a composite disease progression index; index biomarkers were weighted via expected progression-free days lost during follow-up with a biomarker on the unfavorable side of the threshold. RESULTS A six-analyte index, scaled 0 to 11, composed of markers of immune function, proteolysis and angiogenesis [high levels of ficolin-2 (FCN2), cathepsin-S (Cath-S), legumain (LGMN) and soluble vascular endothelial growth factor receptor 2 (VEGFsR2), but low levels of inducible T cell costimulator (ICOS) or trypsin 3 (TRY3)] predicted better progression-free survival in IPF with a ROC AUC of 0.91. An index score ≥ 3 (group ≥ 2) was strongly associated with IPF progression after adjustment for age, gender, smoking status, immunomodulation, forced vital capacity % predicted and diffusing capacity for carbon monoxide % predicted (HR 16.8, 95% CI 2.2-126.7, P = 0.006). CONCLUSION This index, derived from the largest proteomic analysis of IPF plasma samples to date, could be useful for clinical decision making in IPF, and the identified analytes suggest biological processes that may promote disease progression.
Collapse
Affiliation(s)
- Shanna L. Ashley
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - Meng Xia
- Biostatistics Department, University of Michigan School of Public Health, Ann Arbor, MI, United States of America
| | - Susan Murray
- Biostatistics Department, University of Michigan School of Public Health, Ann Arbor, MI, United States of America
| | - David N. O’Dwyer
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Ethan Grant
- MedImmune, Gaithersburg, MD, United States of America
| | - Eric S. White
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Kevin R. Flaherty
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Fernando J. Martinez
- Department of Internal Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Bethany B. Moore
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
29
|
Scartozzi M, Vincent L, Chiron M, Cascinu S. Aflibercept, a New Way to Target Angiogenesis in the Second Line Treatment of Metastatic Colorectal Cancer (mCRC). Target Oncol 2016; 11:489-500. [DOI: 10.1007/s11523-016-0447-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
30
|
Aghagolzadeh P, Radpour R. New trends in molecular and cellular biomarker discovery for colorectal cancer. World J Gastroenterol 2016; 22:5678-5693. [PMID: 27433083 PMCID: PMC4932205 DOI: 10.3748/wjg.v22.i25.5678] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/16/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death worldwide, which is consequence of multistep tumorigenesis of several genetic and epigenetic events. Since CRC is mostly asymptomatic until it progresses to advanced stages, the early detection using effective screening approaches, selection of appropriate therapeutic strategies and efficient follow-up programs are essential to reduce CRC mortalities. Biomarker discovery for CRC based on the personalized genotype and clinical information could facilitate the classification of patients with certain types and stages of cancer to tailor preventive and therapeutic approaches. These cancer-related biomarkers should be highly sensitive and specific in a wide range of specimen(s) (including tumor tissues, patients’ fluids or stool). Reliable biomarkers which enable the early detection of CRC, can improve early diagnosis, prognosis, treatment response prediction, and recurrence risk. Advances in our understanding of the natural history of CRC have led to the development of different CRC associated molecular and cellular biomarkers. This review highlights the new trends and approaches in CRC biomarker discovery, which could be potentially used for early diagnosis, development of new therapeutic approaches and follow-up of patients.
Collapse
|
31
|
Ratnasari N, Nurdjanah S, Sadewa AH, Hakimi M. The role of vascular endothelial growth factor -634 G/C and its soluble receptor on chronic liver disease and hepatocellular carcinoma. Arab J Gastroenterol 2016; 17:61-66. [PMID: 27426958 DOI: 10.1016/j.ajg.2016.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 02/14/2016] [Accepted: 06/12/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND STUDY AIMS The single nucleotide polymorphism (SNP) of the vascular endothelial growth factor (VEGF) gene -634 G/C (rs2010963) influences the progression of hepatocellular carcinoma (HCC). There have been no studies on the role of VEGF SNP -634 G/C in chronic liver disease (CLD). The aim of the present study was to analyse the correlation between VEGF SNP -634 and the clinical severity of CLD and HCC. PATIENTS AND METHODS A cross sectional study was conducted on 182 subjects (46 HCC, 39 liver cirrhotic/LC, 38 chronic hepatitis/CH; and 57 healthy subjects). The study was conducted from 2010 to 2014 at the Dr. Sardjito Hospital Yogyakarta, Indonesia. All subjects submitted blood serum for DNA sequencing examination using primer. The clinical data of CLD and HCC were assessed, and sVEGFR-2 was examined in 149 subjects. All data were analysed using STATA programme 11.0. RESULTS Significant differences were observed in genotypic frequency (GG/GC/CC) between HCC, LC, CH and healthy subjects (p=0.004), but though no significant differences were observed between the G>G and C>G genotypic frequencies (p=0.337). The frequency of genotype GG was significantly higher than genotype GC or CC in HCC and was associated with declining of clinical conditions (p<0.05). No significant difference in the distribution genotypes was observed with respect to the level of sVEGFR-2 in the serum. However, we observed a significant correlation between sVEGFR-2 and clinical characteristics in LC and CH (p<0.05). CONCLUSION Genotype GG of the VEGF SNP -634 is the dominant genotype in severe CLD and HCC. sVEGFR-2 correlates with the disease severity but is not directly associated with the SNP -634 genotype.
Collapse
Affiliation(s)
- Neneng Ratnasari
- Internal Medicine Department, Faculty of Medicine, Gadjah Mada University/Dr. Sardjito General Hospital, Yogyakarta, Indonesia.
| | - Siti Nurdjanah
- Internal Medicine Department, Faculty of Medicine, Gadjah Mada University/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Ahmad H Sadewa
- Biochemistry Department, Faculty of Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| | - Mohammad Hakimi
- Public Health Department, Faculty of Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| |
Collapse
|
32
|
Lam SW, Nota NM, Jager A, Bos MMEM, van den Bosch J, van der Velden AMT, Portielje JEA, Honkoop AH, van Tinteren H, Boven E. Angiogenesis- and Hypoxia-Associated Proteins as Early Indicators of the Outcome in Patients with Metastatic Breast Cancer Given First-Line Bevacizumab-Based Therapy. Clin Cancer Res 2016; 22:1611-20. [PMID: 26823602 DOI: 10.1158/1078-0432.ccr-15-1005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 10/07/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE We examined whether pretreatment levels of angiogenesis- or hypoxia-related proteins and their changes after one cycle of first-line bevacizumab-based therapy were associated with response, PFS, or OS in patients with metastatic breast cancer. EXPERIMENTAL DESIGN We included 181 patients enrolled in the phase II ATX trial evaluating first-line paclitaxel and bevacizumab without or with capecitabine (NTR1348). Plasma samples were analyzed for VEGF-A, soluble VEGFR2 (sVEGFR2), angiopoietin 2 (ANG2), soluble TIE2 (sTIE2), IL6, IL8, and carbonic anhydrase 9 (CA9). Baseline serum CA15-3 was documented. HR was adjusted for confounding factors. Where appropriate, an optimal cut-off value defining a high and a low group was determined with Martingale residuals. RESULTS At baseline, multiple proteins were significantly associated with PFS (ANG2, IL6, IL8, CA9, CA15-3) and OS (ANG2, sTIE2, IL6, IL8, CA9, CA15-3). After one cycle, VEGF-A, ANG2, sTIE2, and IL8 significantly decreased, while sVEGFR2 and CA9 significantly increased. The relative change in sVEGFR2 (P= 0.01) and IL8 (P= 0.001) was associated with response. Defining optimal cut-off, patients with a high CA9 rise (>2.9%) had better PFS (HR 0.45) and OS (HR 0.54) than those with low/no rise. CONCLUSIONS Multiple angiogenesis- or hypoxia-related proteins were prognostic for PFS and OS. Molecular agents targeting these proteins might be beneficial in patients with high levels. Changes in IL8 or sVEGFR2 levels at second cycle appear predictive for response. Changes in CA9 levels during bevacizumab-based therapy for prediction of PFS and OS merit further study.
Collapse
Affiliation(s)
- Siu W Lam
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Nienke M Nota
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Agnes Jager
- Erasmus Medical Center-Daniel den Hoed Cancer Center, Rotterdam, the Netherlands
| | | | | | | | | | | | - Harm van Tinteren
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| | | |
Collapse
|
33
|
Metastatic breast cancer patients treated with low-dose metronomic chemotherapy with cyclophosphamide and celecoxib: clinical outcomes and biomarkers of response. Cancer Chemother Pharmacol 2015; 77:365-74. [PMID: 26721701 DOI: 10.1007/s00280-015-2947-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/11/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Preclinical results showing therapeutic effect and low toxicity of metronomic chemotherapy with cyclophosphamide (Cy) + celecoxib (Cel) for mammary tumors encouraged its translation to the clinic for treating advanced breast cancer patients (ABCP). PATIENTS AND METHODS A single-arm, mono-institutional, non-randomized, phase II, two-step clinical trial (approved by Bioethics Committee and Argentine Regulatory Authority) was designed. Patients received Cy (50 mg po.d) + Cel (200 mg p.o.bid). Patient eligibility criteria included: ABCP who progressed to anthracyclines, taxanes and capecitabine, ≤4 chemotherapy schemes, with good performance status. Several pro- and anti-angiogenic molecules and cells were determined as biomarkers. Informed consent was signed by all patients. Primary endpoint was clinical benefit (CB). RESULTS Twenty patients were enrolled. Main clinical outcomes were prolonged disease stabilization and partial remission in 10/20 and 1/20 patients, respectively. CB was 55 %, and time to progression (TTP) was 21.1 weeks. Median TTP in patients who achieved CB was 35.6 weeks, and mean overall survival was 44.20 weeks. There were no grade 3/4 toxicities associated with treatment. Circulating endothelial cells (CECs) increased at the time of progression in patients who showed CB (P = 0.014). Baseline CECs and circulating endothelial progenitor cells showed marginal associations with TTP. Serum VEGF decreased (P = 0.050), sVEGFR-2 increased (P = 0.005) and VEGF/sVEGFR-2 ratio decreased during treatment (P = 0.041); baseline VEGF and VEGF/sVEGFR-2 were associated with TTP (P = 0.035 and P = 0.030, respectively), while sVEGFR-2 did not. CONCLUSIONS Treatment was effective, showing low toxicity profile and excellent tolerability. The combination had anti-angiogenic effect. Increased levels of CEC could be useful for detecting progression. Baseline VEGF and VEGF/sVEGFR-2 values could be useful as early predictors of response. TRIAL REGISTRATION ANMAT#4596/09.
Collapse
|
34
|
Benzekry S, Tracz A, Mastri M, Corbelli R, Barbolosi D, Ebos JML. Modeling Spontaneous Metastasis following Surgery: An In Vivo-In Silico Approach. Cancer Res 2015; 76:535-47. [PMID: 26511632 DOI: 10.1158/0008-5472.can-15-1389] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022]
Abstract
Rapid improvements in the detection and tracking of early-stage tumor progression aim to guide decisions regarding cancer treatments as well as predict metastatic recurrence in patients following surgery. Mathematical models may have the potential to further assist in estimating metastatic risk, particularly when paired with in vivo tumor data that faithfully represent all stages of disease progression. Herein, we describe mathematical analysis that uses data from mouse models of spontaneous metastasis developing after surgical removal of orthotopically implanted primary tumors. Both presurgical (primary tumor) growth and postsurgical (metastatic) growth were quantified using bioluminescence and were then used to generate a mathematical formalism based on general laws of the disease (i.e., dissemination and growth). The model was able to fit and predict pre/postsurgical data at the level of the individual as well as the population. Our approach also enabled retrospective analysis of clinical data describing the probability of metastatic relapse as a function of primary tumor size. In these data-based models, interindividual variability was quantified by a key parameter of intrinsic metastatic potential. Critically, our analysis identified a highly nonlinear relationship between primary tumor size and postsurgical survival, suggesting possible threshold limits for the utility of tumor size as a predictor of metastatic recurrence. These findings represent a novel use of clinically relevant models to assess the impact of surgery on metastatic potential and may guide optimal timing of treatments in neoadjuvant (presurgical) and adjuvant (postsurgical) settings to maximize patient benefit.
Collapse
Affiliation(s)
- Sebastien Benzekry
- Inria Bordeaux Sud-Ouest, Team MONC, Institut de Mathematiques de Bordeaux, Bordeaux, France.
| | - Amanda Tracz
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Michalis Mastri
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Ryan Corbelli
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Dominique Barbolosi
- SMARTc Pharmacokinetics Unit, Inserm S 911 CRO2, Aix Marseille University, Marseille, France
| | - John M L Ebos
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York. Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
35
|
Mousa L, Salem ME, Mikhail S. Biomarkers of Angiogenesis in Colorectal Cancer. BIOMARKERS IN CANCER 2015; 7:13-9. [PMID: 26543385 PMCID: PMC4624093 DOI: 10.4137/bic.s25250] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/17/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and accounts for 10% of all new cancer diagnoses. Angiogenesis is a tightly regulated process that is mediated by a group of angiogenic factors such as vascular endothelial growth factor and its receptors. Given the widespread use of antiangiogenic agents in CRC, there has been considerable interest in the development of methods to identify novel markers that can predict outcome in the treatment of this disease with angiogenesis inhibitors. Multiple biomarkers are in various phases of development and include tissue, serum, and imaging biomarkers. The complexity of the angiogenesis pathway and the overlap between the various angiogenic factors present a significant challenge to biomarker discovery. In our review, we discuss the angiogenesis pathway and the most promising evolving concepts in biomarker discovery, as well as highlight the landmark studies that identify subgroups of patients with CRC who may preferentially benefit from angiogenesis inhibitors.
Collapse
Affiliation(s)
- Luay Mousa
- The Medstar Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Mohamed E Salem
- Medstar Georgetown University Hospital, Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Sameh Mikhail
- The Medstar Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| |
Collapse
|
36
|
Bouattour M, Payancé A, Wassermann J. Evaluation of antiangiogenic efficacy in advanced hepatocellular carcinoma: Biomarkers and functional imaging. World J Hepatol 2015; 7:2245-2263. [PMID: 26380650 PMCID: PMC4568486 DOI: 10.4254/wjh.v7.i20.2245] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/16/2015] [Accepted: 08/30/2015] [Indexed: 02/06/2023] Open
Abstract
Many years after therapeutic wilderness, sorafenib finally showed a clinical benefit in patients with advanced hepatocellular carcinoma. After the primary general enthusiasm worldwide, some disappointments emerged particularly since no new treatment could exceed or at least match sorafenib in this setting. Without these new drugs, research focused on optimizing care of patients treated with sorafenib. One challenging research approach deals with identifying prognostic and predictive biomarkers of sorafenib in this population. The task still seems difficult; however appropriate investigations could resolve this dilemma, as observed for some malignancies where other drugs were used.
Collapse
Affiliation(s)
- Mohamed Bouattour
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| | - Audrey Payancé
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| | - Johanna Wassermann
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| |
Collapse
|
37
|
Bilen MA, Zurita AJ, Ilias-Khan NA, Chen HC, Wang X, Kearney AY, Hodges S, Jonasch E, Huang S, Khakoo AY, Tannir NM. Hypertension and Circulating Cytokines and Angiogenic Factors in Patients With Advanced Non-Clear Cell Renal Cell Carcinoma Treated With Sunitinib: Results From a Phase II Trial. Oncologist 2015; 20:1140-8. [PMID: 26306901 DOI: 10.1634/theoncologist.2015-0143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND We evaluated the significance of hypertension developing during vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor (VEGFR-TKI) treatment and a group of cytokines and angiogenic factors (CAFs) in advanced non-clear cell renal cell carcinoma (nccRCC) patients treated with sunitinib in a phase II study. MATERIALS AND METHODS Using multiplex assays, we analyzed the levels of 38 CAFs in plasma at baseline and after 4 weeks of sunitinib therapy. Sunitinib benefit was defined as a partial response or stable disease using the Response Evaluation Criteria in Solid Tumors lasting ≥4 months. Cox proportional hazards regression models were used to assess the associations among hypertension, CAFs, and progression-free (PFS) and overall survival (OS). RESULTS Fifty-seven patients were evaluable; 53 had baseline CAF levels available. The median PFS and OS were 2.9 months (95% confidence interval [CI], 1.4-5.5) and 16.8 months (95% CI, 10.7-27.4), respectively. Sunitinib benefit was observed in 21 patients (37%). However, 33 patients (60%) developed hypertension during treatment, although no association was found with survival or response. Elevated baseline soluble tumor necrosis factor (TNF) receptor I, interleukin-8, growth-regulated oncogene, transforming growth factor-α, and VEGFR-2 levels were associated with an increased risk of death on multivariate analysis. CONCLUSION We found no association between the development of hypertension and survival or sunitinib benefit in advanced nccRCC. TNF and angiogenic/immunomodulatory mediators were identified for evaluation as markers of prognosis and VEGFR-TKI benefit in future studies.
Collapse
Affiliation(s)
- Mehmet Asim Bilen
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Amado J Zurita
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Nasreen A Ilias-Khan
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Hsiang-Chun Chen
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Xuemei Wang
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Alper Y Kearney
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Sherie Hodges
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Eric Jonasch
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Shixia Huang
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Aarif Yusuf Khakoo
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| | - Nizar M Tannir
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, Department of Cardiology, and Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Amgen, Inc., San Francisco, California, USA
| |
Collapse
|
38
|
Varkaris A, Corn PG, Parikh NU, Efstathiou E, Song JH, Lee YC, Aparicio A, Hoang AG, Gaur S, Thorpe L, Maity SN, Bar Eli M, Czerniak BA, Shao Y, Alauddin M, Lin SH, Logothetis CJ, Gallick GE. Integrating Murine and Clinical Trials with Cabozantinib to Understand Roles of MET and VEGFR2 as Targets for Growth Inhibition of Prostate Cancer. Clin Cancer Res 2015; 22:107-21. [PMID: 26272062 DOI: 10.1158/1078-0432.ccr-15-0235] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/26/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE We performed parallel investigations in cabozantinib-treated patients in a phase II trial and simultaneously in patient-derived xenograft (PDX) models to better understand the roles of MET and VEGFR2 as targets for prostate cancer therapy. EXPERIMENTAL DESIGN In the clinical trial, radiographic imaging and serum markers were examined, as well as molecular markers in tumors from bone biopsies. In mice harboring PDX intrafemurally or subcutaneously, cabozantinib effects on tumor growth, MET, PDX in which MET was silenced, VEGFR2, bone turnover, angiogenesis, and resistance were examined. RESULTS In responsive patients and PDX, islets of viable pMET-positive tumor cells persisted, which rapidly regrew after drug withdrawal. Knockdown of MET in PDX did not affect tumor growth in mice nor did it affect cabozantinib-induced growth inhibition but did lead to induction of FGFR1. Inhibition of VEGFR2 and MET in endothelial cells reduced the vasculature, leading to necrosis. However, each islet of viable cells surrounded a VEGFR2-negative vessel. Reduction of bone turnover was observed in both cohorts. CONCLUSIONS Our studies demonstrate that MET in tumor cells is not a persistent therapeutic target for metastatic castrate-resistant prostate cancer (CRPC), but inhibition of VEGFR2 and MET in endothelial cells and direct effects on osteoblasts are responsible for cabozantinib-induced tumor inhibition. However, vascular heterogeneity represents one source of primary therapy resistance, whereas induction of FGFR1 in tumor cells suggests a potential mechanism of acquired resistance. Thus, integrated cross-species investigations demonstrate the power of combining preclinical models with clinical trials to understand mechanisms of activity and resistance of investigational agents.
Collapse
Affiliation(s)
- Andreas Varkaris
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nila U Parikh
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jian H Song
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu-Chen Lee
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anh G Hoang
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanchaika Gaur
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas. Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Lynnelle Thorpe
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas. Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Sankar N Maity
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Menashe Bar Eli
- Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bogdan A Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yiping Shao
- Department of Imaging Physics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mian Alauddin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sue-Hwa Lin
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas. Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
| |
Collapse
|
39
|
Ebos JML, Mastri M, Lee CR, Tracz A, Hudson JM, Attwood K, Cruz-Munoz WR, Jedeszko C, Burns P, Kerbel RS. Neoadjuvant antiangiogenic therapy reveals contrasts in primary and metastatic tumor efficacy. EMBO Mol Med 2015; 6:1561-76. [PMID: 25361689 PMCID: PMC4287975 DOI: 10.15252/emmm.201403989] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Thousands of cancer patients are currently in clinical trials evaluating antiangiogenic therapy
in the neoadjuvant setting, which is the treatment of localized primary tumors prior to surgical
intervention. The rationale is that shrinking a tumor will improve surgical outcomes and minimize
growth of occult micrometastatic disease—thus delaying post-surgical recurrence and improving
survival. But approved VEGF pathway inhibitors have not been tested in clinically relevant
neoadjuvant models that compare pre- and post-surgical treatment effects. Using mouse models of
breast, kidney, and melanoma metastasis, we demonstrate that primary tumor responses to neoadjuvant
VEGFR TKI treatment do not consistently correlate with improved post-surgical survival, with
survival worsened in certain settings. Similar negative effects did not extend to protein-based VEGF
pathway inhibitors and could be reversed with altered dose, surgical timing, and treatment duration,
or when VEGFR TKIs are combined with metronomic ‘anti-metastatic’ chemotherapy
regimens. These studies represent the first attempt to recapitulate the complex clinical parameters
of neoadjuvant therapy in mice and identify a novel tool to compare systemic antiangiogenic
treatment effects on localized and disseminated disease.
Collapse
Affiliation(s)
- John M L Ebos
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Michalis Mastri
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Christina R Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Amanda Tracz
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - John M Hudson
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - William R Cruz-Munoz
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Christopher Jedeszko
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Peter Burns
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Harrison IP, Selemidis S. Understanding the biology of reactive oxygen species and their link to cancer: NADPH oxidases as novel pharmacological targets. Clin Exp Pharmacol Physiol 2015; 41:533-42. [PMID: 24738947 DOI: 10.1111/1440-1681.12238] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS), the cellular products of myriad physiological processes, have long been understood to lead to cellular damage if produced in excess and to be a causative factor in cancer through the oxidation and nitration of various macromolecules. Reactive oxygen species influence various hallmarks of cancer, such as cellular proliferation and angiogenesis, through the promotion of cell signalling pathways intrinsic to these processes and can also regulate the function of key immune cells, such as macrophages and regulatory T cells, which promote angiogenesis in the tumour environment. Herein we emphasize the family of NADPH oxidase enzymes as the most likely source of ROS, which promote angiogenesis and tumourigenesis through signalling pathways within endothelial, immune and tumour cells. In this review we focus on the pharmacological inhibitors of NADPH oxidases and suggest that, compared with traditional anti-oxidants, they are likely to offer better alternatives for suppression of tumour angiogenesis. Despite the emerging enthusiasm towards the use of NADPH oxidase inhibitors for cancer therapy, this field is still in its infancy; in particular, there is a glaring lack of knowledge of the roles of NADPH oxidases in in vivo animal models and in human cancers. Certainly a clearer understanding of the relevant signalling pathways influenced by NADPH oxidases during angiogenesis in cancer is likely to yield novel therapeutic approaches.
Collapse
Affiliation(s)
- Ian P Harrison
- Department of Pharmacology, Monash University, Melbourne, Vic., Australia
| | | |
Collapse
|
41
|
Zurita AJ, Khajavi M, Wu HK, Tye L, Huang X, Kulke MH, Lenz HJ, Meropol NJ, Carley W, DePrimo SE, Lin E, Wang X, Harmon CS, Heymach JV. Circulating cytokines and monocyte subpopulations as biomarkers of outcome and biological activity in sunitinib-treated patients with advanced neuroendocrine tumours. Br J Cancer 2015; 112:1199-205. [PMID: 25756398 PMCID: PMC4385961 DOI: 10.1038/bjc.2015.73] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/23/2014] [Accepted: 01/29/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sunitinib is approved worldwide for treatment of advanced pancreatic neuroendocrine tumours (pNET), but no validated markers exist to predict response. This analysis explored biomarkers associated with sunitinib activity and clinical benefit in patients with pNET and carcinoid tumours in a phase II study. METHODS Plasma was assessed for vascular endothelial growth factor (VEGF)-A, soluble VEGF receptor (sVEGFR)-2, sVEGFR-3, interleukin (IL)-8 (n=105), and stromal cell-derived factor (SDF)-1α (n=28). Pre-treatment levels were compared between tumour types and correlated with response, progression-free (PFS), and overall survival (OS). Changes in circulating myelomonocytic and endothelial cells were also analysed. RESULTS Stromal cell-derived factor-1α and sVEGFR-2 levels were higher in pNET than in carcinoid (P=0.003 and 0.041, respectively). High (above-median) baseline SDF-1α was associated with worse PFS, OS, and response in pNET, and high sVEGFR-2 with longer OS (P⩽0.05). For carcinoid, high IL-8, sVEGFR-3, and SDF-1α were associated with shorter PFS and OS, and high IL-8 and SDF-1α with worse response (P⩽0.05). Among circulating cell types, monocytes showed the largest on-treatment decrease, particularly CD14+ monocytes co-expressing VEGFR-1 or CXCR4. CONCLUSIONS Interleukin-8, sVEGFR-3, and SDF-1α were identified as predictors of sunitinib clinical outcome. Putative pro-tumorigenic CXCR4+ and VEGFR-1+ monocytes represent novel candidate markers and biologically relevant targets explaining the activity of sunitinib.
Collapse
Affiliation(s)
- A J Zurita
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| | - M Khajavi
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| | - H-K Wu
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| | - L Tye
- Pfizer Inc., La Jolla, CA, USA
| | - X Huang
- Pfizer Inc., La Jolla, CA, USA
| | - M H Kulke
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - H-J Lenz
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - N J Meropol
- University Hospitals Case Medical Center Seidman Cancer Center, Case Comprehensive Cancer Center and Case Western Reserve University, Cleveland, OH, USA
| | | | | | - E Lin
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| | - X Wang
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| | | | - J V Heymach
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374 Houston, TX, USA
| |
Collapse
|
42
|
Sallinen H, Heikura T, Koponen J, Kosma VM, Heinonen S, Ylä-Herttuala S, Anttila M. Serum angiopoietin-2 and soluble VEGFR-2 levels predict malignancy of ovarian neoplasm and poor prognosis in epithelial ovarian cancer. BMC Cancer 2014; 14:696. [PMID: 25245329 PMCID: PMC4179851 DOI: 10.1186/1471-2407-14-696] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/19/2014] [Indexed: 01/18/2023] Open
Abstract
Background The aim of the study was to explore the serum levels of eight angiogenesis biomarkers in patients with benign, borderline or malignant epithelial ovarian neoplasms and to compare them to those of healthy controls. In addition, we aimed to study how those biomarkers predict the clinical course and survival of patients with epithelial ovarian cancer. Methods We enrolled 132 patients with ovarian neoplasms and 32 unaffected women in this study. Serum samples were collected preoperatively at the time of diagnosis and the levels of angiogenesis biomarkers were measured with an ELISA. Results Levels of Ang-1, Ang-2, VEGF, VEGF-D, VEGF/sVEGFR-2 and Ang-2/ sVEGFR-2 ratios were elevated whereas sVEGFR-2 was lower in patients with ovarian carcinoma than in women with normal ovaries, benign and/or borderline ovarian neoplasms. In ROC analysis, the area under the curve for serum Ang-2/sVEGFR-2 ratio (0.76) was greater than Ang-2 (0.75) and VEGF (0.65) but lower than for CA 125 (0.90) to differentiate ovarian cancer from benign or borderline ovarian tumors. In ovarian cancer high Ang-2/sVEGFR-2 ratio was associated with the presence of ascites, high stage and grade of ovarian cancer, with the size of primary residual tumor >1 cm and with recurrence of disease. Elevated Ang-2, VEGF, VEGF/sVEGFR-2, Ang-2/VEGF and Ang-2/sVEGFR-2 ratios and low level of sVEGFR-2 were significant predictors of poor overall survival (OS) and recurrence free survival (RFS) in univariate survival analyses. Conclusions Ovarian cancer patients had elevated levels of angiogenesis related growth factors in circulation reflecting increased angiogenesis and poor prognosis. The serum level of Ang-2 predicted most accurately poor OS and Ang-2/sVEGFR-2 ratio malignancy of ovarian neoplasms and short RFS. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-696) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maarit Anttila
- Department of Gynecology, Kuopio University Hospital, P,O, Box 900, Kuopio, FIN 70029 KYS, Finland.
| |
Collapse
|
43
|
Benzekry S, Lamont C, Beheshti A, Tracz A, Ebos JML, Hlatky L, Hahnfeldt P. Classical mathematical models for description and prediction of experimental tumor growth. PLoS Comput Biol 2014; 10:e1003800. [PMID: 25167199 PMCID: PMC4148196 DOI: 10.1371/journal.pcbi.1003800] [Citation(s) in RCA: 280] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 07/08/2014] [Indexed: 01/03/2023] Open
Abstract
Despite internal complexity, tumor growth kinetics follow relatively simple laws that can be expressed as mathematical models. To explore this further, quantitative analysis of the most classical of these were performed. The models were assessed against data from two in vivo experimental systems: an ectopic syngeneic tumor (Lewis lung carcinoma) and an orthotopically xenografted human breast carcinoma. The goals were threefold: 1) to determine a statistical model for description of the measurement error, 2) to establish the descriptive power of each model, using several goodness-of-fit metrics and a study of parametric identifiability, and 3) to assess the models' ability to forecast future tumor growth. The models included in the study comprised the exponential, exponential-linear, power law, Gompertz, logistic, generalized logistic, von Bertalanffy and a model with dynamic carrying capacity. For the breast data, the dynamics were best captured by the Gompertz and exponential-linear models. The latter also exhibited the highest predictive power, with excellent prediction scores (≥80%) extending out as far as 12 days in the future. For the lung data, the Gompertz and power law models provided the most parsimonious and parametrically identifiable description. However, not one of the models was able to achieve a substantial prediction rate (≥70%) beyond the next day data point. In this context, adjunction of a priori information on the parameter distribution led to considerable improvement. For instance, forecast success rates went from 14.9% to 62.7% when using the power law model to predict the full future tumor growth curves, using just three data points. These results not only have important implications for biological theories of tumor growth and the use of mathematical modeling in preclinical anti-cancer drug investigations, but also may assist in defining how mathematical models could serve as potential prognostic tools in the clinic.
Collapse
Affiliation(s)
- Sébastien Benzekry
- Inria Bordeaux Sud-Ouest, Institut de Mathématiques de Bordeaux, Bordeaux, France
- Center of Cancer Systems Biology, GRI, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Clare Lamont
- Center of Cancer Systems Biology, GRI, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Afshin Beheshti
- Center of Cancer Systems Biology, GRI, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Amanda Tracz
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - John M. L. Ebos
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Lynn Hlatky
- Center of Cancer Systems Biology, GRI, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Philip Hahnfeldt
- Center of Cancer Systems Biology, GRI, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
44
|
Cremolini C, Loupakis F, Bocci G, Falcone A. Biomarkers and Response to Bevacizumab—Letter. Clin Cancer Res 2014; 20:1056-7. [DOI: 10.1158/1078-0432.ccr-13-2763] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Wong HH, Eisen T. Tivozanib for the treatment of metastatic renal cancer. Expert Rev Anticancer Ther 2014; 13:649-60. [PMID: 23773100 DOI: 10.1586/era.13.40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tyrosine kinase inhibitors have revolutionized the treatment of metastatic renal cell carcinoma (RCC). Drugs such as sorafenib, sunitinib and pazopanib act on the VEGF receptor pathway, but they can also inhibit other kinases, resulting in off-target toxicities. Tivozanib was developed due to its potency and selectivity against VEGF receptors 1-3. It has a favorable pharmacokinetic profile after oral administration and a long plasma half-life. In the Phase III TIVO-1 trial, it demonstrated a higher response rate and longer progression-free survival than sorafenib with a better side-effect profile. It is currently awaiting approval to be used in the first-line treatment of metastatic RCC. An early-phase trial has also shown its tolerability at full dose when given with the mTOR inhibitor temsirolimus, suggesting its potential in combination treatment. This article examines tivozanib from its laboratory to clinical development, as well as its relevance and future role in the treatment of RCC in the era of the tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Han Hsi Wong
- Cambridge University Health Partners, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
46
|
Sakakura H, Murakumo Y, Mii S, Hagiwara S, Kato T, Asai M, Hoshino A, Yamamoto N, Sobue S, Ichihara M, Ueda M, Takahashi M. Detection of a soluble form of CD109 in serum of CD109 transgenic and tumor xenografted mice. PLoS One 2014; 9:e83385. [PMID: 24400073 PMCID: PMC3882221 DOI: 10.1371/journal.pone.0083385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/26/2013] [Indexed: 12/25/2022] Open
Abstract
CD109, a glycosylphosphatidylinositol-anchored glycoprotein, is expressed at high levels in some human tumors including squamous cell carcinomas. As CD109 is reportedly cleaved by furin and its soluble form is secreted into culture medium in vitro, we hypothesized that CD109 could serve as a tumor marker in vivo. In this study, we investigated CD109 as a novel serum tumor marker using transgenic mice that overexpress mouse CD109 (mCD109-TG mice) and tumor xenografted mice inoculated with human CD109 (hCD109)-overexpressing HEK293 cells. In sera and urine of mCD109-TG mice, mCD109 was detected using western blotting. In xenografted mice, hCD109 secreted from inoculated tumors was detected in sera, using western blotting and CD109 ELISA. Concentrations of tumor-secreted CD109 increased proportionally as tumors enlarged. Concentrations of secreted CD109 decreased notably by 17 h after tumor resection, and became undetectable 48 h after resection. The half-life of tumor-secreted CD109 was about 5.86±0.17 h. These results indicate that CD109 is present in serum as a soluble form, and suggest its potential as a novel tumor marker in patients with cancers that express CD109.
Collapse
Affiliation(s)
- Hiroki Sakakura
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshiki Murakumo
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sumitaka Hagiwara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takuya Kato
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masato Asai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akiyoshi Hoshino
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Noriyuki Yamamoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sayaka Sobue
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan
| | - Masatoshi Ichihara
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahide Takahashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|
47
|
Morgenstern DA, Marzouki M, Bartels U, Irwin MS, Sholler GLS, Gammon J, Yankanah R, Wu B, Samson Y, Baruchel S. Phase I study of vinblastine and sirolimus in pediatric patients with recurrent or refractory solid tumors. Pediatr Blood Cancer 2014; 61:128-33. [PMID: 23956145 DOI: 10.1002/pbc.24656] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/23/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND The combination of vinblastine and mammalian target of rapamycin (mTOR) inhibitor sirolimus inhibits the growth of neuroblastoma xenografts through pro-apoptotic and anti-angiogenic mechanisms. This phase I study aimed to explore the safety and toxicity of this combination in pediatric patients with advanced solid tumors. PROCEDURE Patients ≤21 years of age with recurrent/refractory solid tumors (including CNS) were eligible. Sirolimus was administered daily by mouth or nasogastric (NG) tube, with doses adjusted to achieve a target trough concentration of 10-15 ng/ml, with weekly intravenous vinblastine (dose escalated 4-6 mg/m(2)/dose according to 3 + 3 phase I design). RESULTS Fourteen patients were enrolled (median age 8.7 years; range 2.3-19) of whom 12 were evaluable for toxicity and 11 for response. One patient experienced a dose-limiting toxicity (grade 3 mucositis) at the highest vinblastine dose level. Myelosuppression was the most common toxicity. Dose-adjusted sirolimus trough concentrations were significantly lower in patients receiving drug via NG tube (1.50 ± 0.75 ng/ml/mg vs. 2.25 ± 1.07 ng/ml/mg for oral administration). Correlative biomarker analysis demonstrated a significant reduction in serum concentration of soluble vascular endothelial growth factor receptor (sVEGFR2) at 28 days compared to baseline consistent with inhibition of angiogenesis. One patient had a partial response and three had stable disease for more than 3 months. CONCLUSIONS The combination of mTOR inhibitor and vinblastine given over an extended continuous schedule is safe, associated with a reduction in circulating angiogenic factor (CAF) VEGFR2 and resulted in clinical responses. Future studies using the intravenously administered mTOR inhibitor temsirolimus are planned.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Department of Paediatrics, University of Toronto and New Agent and Innovative Therapy Programme, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Vilgrain I, Sidibé A, Polena H, Cand F, Mannic T, Arboleas M, Boccard S, Baudet A, Gulino-Debrac D, Bouillet L, Quesada JL, Mendoza C, Lebas JF, Pelletier L, Berger F. Evidence for post-translational processing of vascular endothelial (VE)-cadherin in brain tumors: towards a candidate biomarker. PLoS One 2013; 8:e80056. [PMID: 24358106 PMCID: PMC3864785 DOI: 10.1371/journal.pone.0080056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 09/30/2013] [Indexed: 12/28/2022] Open
Abstract
Vessel abnormalities are among the most important features in malignant glioma. Vascular endothelial (VE)-cadherin is of major importance for vascular integrity. Upon cytokine challenge, VE-cadherin structural modifications have been described including tyrosine phosphorylation and cleavage. The goal of this study was to examine whether these events occurred in human glioma vessels. We demonstrated that VE-cadherin is highly expressed in human glioma tissue and tyrosine phosphorylated at site Y(685), a site previously found phosphorylated upon VEGF challenge, via Src activation. In vitro experiments showed that VEGF-induced VE-cadherin phosphorylation, preceded the cleavage of its extracellular adhesive domain (sVE, 90 kDa). Interestingly, metalloproteases (MMPs) secreted by glioma cell lines were responsible for sVE release. Because VEGF and MMPs are important components of tumor microenvironment, we hypothesized that VE-cadherin proteolysis might occur in human brain tumors. Analysis of glioma patient sera prior treatment confirmed the presence of sVE in bloodstream. Furthermore, sVE levels studied in a cohort of 53 glioma patients were significantly predictive of the overall survival at three years (HR 0.13 [0.04; 0.40] p ≤ 0.001), irrespective to histopathological grade of tumors. Altogether, these results suggest that VE-cadherin structural modifications should be examined as candidate biomarkers of tumor vessel abnormalities, with promising applications in oncology.
Collapse
Affiliation(s)
- Isabelle Vilgrain
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Adama Sidibé
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Helena Polena
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Francine Cand
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Tiphaine Mannic
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Mélanie Arboleas
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Sandra Boccard
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Antoine Baudet
- Grenoble University Hospital, Division of Internal Medicine, Grenoble, France
| | - Danielle Gulino-Debrac
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
| | - Laurence Bouillet
- INSERM, Unit 1036, Biology of Cancer and Infection, Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, Grenoble, France
- Grenoble University Hospital, Division of Internal Medicine, Grenoble, France
| | - Jean-Louis Quesada
- INSERM 003, Clinical Investigation Center, Grenoble University Hospital, Grenoble, France
| | - Christophe Mendoza
- INSERM 003, Clinical Investigation Center, Grenoble University Hospital, Grenoble, France
| | | | - Laurent Pelletier
- INSERM, Unit 836 Brain Nanomedicine, Grenoble Neurosciences Institut Grenoble, Grenoble, France
- Joseph Fourier University, Medicine School, Saint-Martin-d'Hères, France
- Grenoble University Hospital, Biology and Pathology Institute, Grenoble, France
| | - François Berger
- INSERM, Unit 836 Brain Nanomedicine, Grenoble Neurosciences Institut Grenoble, Grenoble, France
- Joseph Fourier University, Medicine School, Saint-Martin-d'Hères, France
- Grenoble University Hospital, Division of Oncology, Grenoble, France
| |
Collapse
|
49
|
Tan HT, Ling LH, Dolor-Torres MC, Yip JWL, Richards AM, Chung MC. Proteomics discovery of biomarkers for mitral regurgitation caused by mitral valve prolapse. J Proteomics 2013; 94:337-45. [DOI: 10.1016/j.jprot.2013.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 01/26/2023]
|
50
|
Hansson EK, Amantea MA, Westwood P, Milligan PA, Houk BE, French J, Karlsson MO, Friberg LE. PKPD Modeling of VEGF, sVEGFR-2, sVEGFR-3, and sKIT as Predictors of Tumor Dynamics and Overall Survival Following Sunitinib Treatment in GIST. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2013; 2:e84. [PMID: 24257372 PMCID: PMC3852160 DOI: 10.1038/psp.2013.61] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/06/2013] [Indexed: 01/26/2023]
Abstract
The predictive value of longitudinal biomarker data (vascular endothelial growth factor (VEGF), soluble VEGF receptor (sVEGFR)-2, sVEGFR-3, and soluble stem cell factor receptor (sKIT)) for tumor response and survival was assessed based on data from 303 patients with imatinib-resistant gastrointestinal stromal tumors (GIST) receiving sunitinib and/or placebo treatment. The longitudinal tumor size data were well characterized by a tumor growth inhibition model, which included, as significant descriptors of tumor size change, the model-predicted relative changes from baseline over time for sKIT (most significant) and sVEGFR-3, in addition to sunitinib exposure. Survival time was best described by a parametric time-to-event model with baseline tumor size and relative change in sVEGFR-3 over time as predictive factors. Based on the proposed modeling framework to link longitudinal biomarker data with overall survival using pharmacokinetic-pharmacodynamic models, sVEGFR-3 demonstrated the greatest predictive potential for overall survival following sunitinib treatment in GIST.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e84; doi:10.1038/psp.2013.61; advance online publication 20 November 2013.
Collapse
Affiliation(s)
- E K Hansson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|