1
|
Ryu J, Boylan KLM, Twigg CAI, Evans R, Skubitz APN, Thomas SN. Quantification of putative ovarian cancer serum protein biomarkers using a multiplexed targeted mass spectrometry assay. Clin Proteomics 2024; 21:1. [PMID: 38172678 PMCID: PMC10762856 DOI: 10.1186/s12014-023-09447-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecologic malignancy in women, and high-grade serous ovarian cancer (HGSOC) is the most common subtype. Currently, no clinical test has been approved by the FDA to screen the general population for ovarian cancer. This underscores the critical need for the development of a robust methodology combined with novel technology to detect diagnostic biomarkers for HGSOC in the sera of women. Targeted mass spectrometry (MS) can be used to identify and quantify specific peptides/proteins in complex biological samples with high accuracy, sensitivity, and reproducibility. In this study, we sought to develop and conduct analytical validation of a multiplexed Tier 2 targeted MS parallel reaction monitoring (PRM) assay for the relative quantification of 23 putative ovarian cancer protein biomarkers in sera. METHODS To develop a PRM method for our target peptides in sera, we followed nationally recognized consensus guidelines for validating fit-for-purpose Tier 2 targeted MS assays. The endogenous target peptide concentrations were calculated using the calibration curves in serum for each target peptide. Receiver operating characteristic (ROC) curves were analyzed to evaluate the diagnostic performance of the biomarker candidates. RESULTS We describe an effort to develop and analytically validate a multiplexed Tier 2 targeted PRM MS assay to quantify candidate ovarian cancer protein biomarkers in sera. Among the 64 peptides corresponding to 23 proteins in our PRM assay, 24 peptides corresponding to 16 proteins passed the assay validation acceptability criteria. A total of 6 of these peptides from insulin-like growth factor-binding protein 2 (IBP2), sex hormone-binding globulin (SHBG), and TIMP metalloproteinase inhibitor 1 (TIMP1) were quantified in sera from a cohort of 69 patients with early-stage HGSOC, late-stage HGSOC, benign ovarian conditions, and healthy (non-cancer) controls. Confirming the results from previously published studies using orthogonal analytical approaches, IBP2 was identified as a diagnostic biomarker candidate based on its significantly increased abundance in the late-stage HGSOC patient sera compared to the healthy controls and patients with benign ovarian conditions. CONCLUSIONS A multiplexed targeted PRM MS assay was applied to detect candidate diagnostic biomarkers in HGSOC sera. To evaluate the clinical utility of the IBP2 PRM assay for HGSOC detection, further studies need to be performed using a larger patient cohort.
Collapse
Affiliation(s)
- Joohyun Ryu
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Kristin L M Boylan
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Carly A I Twigg
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Richard Evans
- Clinical and Translational Research Institute, University of Minnesota, Minneapolis, MN, USA
| | - Amy P N Skubitz
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Stefani N Thomas
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Mei C, Gong W, Wang X, Lv Y, Zhang Y, Wu S, Zhu C. Anti-angiogenic therapy in ovarian cancer: Current understandings and prospects of precision medicine. Front Pharmacol 2023; 14:1147717. [PMID: 36959862 PMCID: PMC10027942 DOI: 10.3389/fphar.2023.1147717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Ovarian cancer (OC) remains the most fatal disease of gynecologic malignant tumors. Angiogenesis refers to the development of new vessels from pre-existing ones, which is responsible for supplying nutrients and removing metabolic waste. Although not yet completely understood, tumor vascularization is orchestrated by multiple secreted factors and signaling pathways. The most central proangiogenic signal, vascular endothelial growth factor (VEGF)/VEGFR signaling, is also the primary target of initial clinical anti-angiogenic effort. However, the efficiency of therapy has so far been modest due to the low response rate and rapidly emerging acquiring resistance. This review focused on the current understanding of the in-depth mechanisms of tumor angiogenesis, together with the newest reports of clinical trial outcomes and resistance mechanism of anti-angiogenic agents in OC. We also emphatically summarized and analyzed previously reported biomarkers and predictive models to describe the prospect of precision therapy of anti-angiogenic drugs in OC.
Collapse
Affiliation(s)
- Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijing Gong
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Xu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongning Lv
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Chunqi Zhu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Clinical Significance of Tie-2-Expressing Monocytes/Macrophages and Angiopoietins in the Progression of Ovarian Cancer-State-of-the-Art. Cells 2022; 11:cells11233851. [PMID: 36497114 PMCID: PMC9737633 DOI: 10.3390/cells11233851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Tumour growth and metastasis are specific to advanced stages of epithelial ovarian cancer (EOC). Tumour angiogenesis is an essential part of these processes. It is responsible for providing tumours with nutrients, metabolites, and cytokines and facilitates tumour and immune cell relocation. Destabilised vasculature, a distinctive feature of tumours, is also responsible for compromising drug delivery into the bulk. Angiogenesis is a complex process that largely depends on how the tumour microenvironment (TME) is composed and how a specific organ is formed. There are contrary reports on whether Tie-2-expressing monocytes/macrophages (TEMs) reported as the proangiogenic population of monocytes have any impact on tumour development. The aim of this paper is to summarise knowledge about ovarian-cancer-specific angiogenesis and the unique role of Tie-2-expressing monocytes/macrophages in this process. The significance of this cell subpopulation for the pathophysiology of EOC remains to be investigated.
Collapse
|
4
|
Drocaş I, Crăiţoiu Ş, Stepan AE, Simionescu CE, Marinescu D. VEGF Immunoexpression in Endometrioid Endometrial Carcinomas. CURRENT HEALTH SCIENCES JOURNAL 2022; 48:155-161. [PMID: 36320870 PMCID: PMC9590369 DOI: 10.12865/chsj.48.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/20/2022] [Indexed: 11/05/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the most important stimulator of endometrial tumor angiogenesis, a mechanism that may be a therapeutic target in the context of an incidence and persistent mortality of endometrial endometrial carcinomas (EEC). In this study, VEGF immunoexpression was analyzed for 50 cases of EEC in relation to the histopathological parameters of tumor aggressiveness. High VEGF scores have been associated with the high grade and advanced stage of EEC, but unrelated to the depth of myometrial invasion, the pattern of tumor invasion, or vascular invasion. VEGF may be useful for assessing EEC aggression, but also for tumor angiogenic potential, which recommends it as a possible mark for specific antitumor therapy.
Collapse
Affiliation(s)
- Ileana Drocaş
- PhD Student, Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ştefania Crăiţoiu
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Alex Emilian Stepan
- Department of Pathology, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Daniela Marinescu
- Department of General Surgery, University of Medicine and Pharmacy of Craiova
| |
Collapse
|
5
|
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
|
6
|
Angiopoietin-2 as a Prognostic Factor in Patients with Incurable Stage IV Colorectal Cancer. J Gastrointest Cancer 2021; 52:237-242. [PMID: 32166589 DOI: 10.1007/s12029-020-00392-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE Angiopoietin (Ang), a ligand of the endothelium-specific receptor Tie-2 system, is associated with tumor growth and progression that depend on angiogenesis. The present study aimed to investigate the predictive potential of angiopoietin factors in incurable stage IV colorectal cancer (CRC) patients who have undergone primary tumor resection. METHODS The study included 40 consecutive patients with incurable stage IV CRC who underwent primary tumor resection at our hospital between 2011 and 2015. Patients were divided into subgroups of low and high Ang-1, Ang-2, and Tie-2. Patient age and sex, tumor location, TNM stages, vascular invasion, chemotherapy, and overall survival were assessed. RESULTS The cut-off values of Ang-1, Ang-2, and Tie-2 were 0.4, 1.8, and 15.0 ng/mL, respectively. Overall survival was significantly longer in the low Ang-2 group than in the high Ang-2 group. High Ang-2 levels were associated with age, N stage, and chemotherapy. Immunofluorescent staining of Ang-2 revealed that endothelial cells and cancer cells expressed Ang-2 in each case. CONCLUSIONS Our findings suggest that the serum Ang-2 level is associated with disease progression and is an important predictor of mortality in incurable stage IV CRC patients. Thus, it may be a useful prognostic biomarker in these patients.
Collapse
|
7
|
Grimes MM, Kenney SR, Dominguez DR, Brayer KJ, Guo Y, Wandinger-Ness A, Hudson LG. The R-enantiomer of ketorolac reduces ovarian cancer tumor burden in vivo. BMC Cancer 2021; 21:40. [PMID: 33413202 PMCID: PMC7791840 DOI: 10.1186/s12885-020-07716-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rho-family GTPases, including Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42), are important modulators of cancer-relevant cell functions and are viewed as promising therapeutic targets. Based on high-throughput screening and cheminformatics we identified the R-enantiomer of an FDA-approved drug (ketorolac) as an inhibitor of Rac1 and Cdc42. The corresponding S-enantiomer is a non-steroidal anti-inflammatory drug (NSAID) with selective activity against cyclooxygenases. We reported previously that R-ketorolac, but not the S-enantiomer, inhibited Rac1 and Cdc42-dependent downstream signaling, growth factor stimulated actin cytoskeleton rearrangements, cell adhesion, migration and invasion in ovarian cancer cell lines and patient-derived tumor cells. METHODS In this study we treated mice with R-ketorolac and measured engraftment of tumor cells to the omentum, tumor burden, and target GTPase activity. In order to gain insights into the actions of R-ketorolac, we also performed global RNA-sequencing (RNA-seq) analysis on tumor samples. RESULTS Treatment of mice with R-ketorolac decreased omental engraftment of ovarian tumor cells at 18 h post tumor cell injection and tumor burden after 2 weeks of tumor growth. R-ketorolac treatment inhibited tumor Rac1 and Cdc42 activity with little impact on mRNA or protein expression of these GTPase targets. RNA-seq analysis revealed that R-ketorolac decreased expression of genes in the HIF-1 signaling pathway. R-ketorolac treatment also reduced expression of additional genes associated with poor prognosis in ovarian cancer. CONCLUSION These findings suggest that R-ketorolac may represent a novel therapeutic approach for ovarian cancer based on its pharmacologic activity as a Rac1 and Cdc42 inhibitor. R-ketorolac modulates relevant pathways and genes associated with disease progression and worse outcome.
Collapse
Affiliation(s)
- Martha M. Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - S. Ray Kenney
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
- Division of Molecular Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Dayna R. Dominguez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - Kathryn J. Brayer
- Analytical and Translational Genomics Shared Resource, Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico USA
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Yuna Guo
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Angela Wandinger-Ness
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Laurie G. Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| |
Collapse
|
8
|
High expression of Tie-2 predicts poor prognosis in primary high grade serous ovarian cancer. PLoS One 2020; 15:e0241484. [PMID: 33151982 PMCID: PMC7644024 DOI: 10.1371/journal.pone.0241484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
Background Antiangiogenic therapy, although part of standard treatment in ovarian cancer, has variable efficacy. Furthermore, little is known about the prognostic biomarkers and factors influencing angiogenesis in cancer tissue. We evaluated the expression of angiopoietin-2 and two endothelial tyrosine kinase receptors, Tie-1 and Tie-2, and assessed their value in the prediction of survival in patients with malignant epithelial ovarian cancer. We also compared the expression of these factors between primary high grade serous tumors and their distant metastasis. Materials and methods We evaluated 86 women with primary epithelial ovarian cancer. Matched distal omental metastasis were investigated in 18.6% cases (N = 16). The expression levels of angiogenic factors were evaluated by immunohistochemistry in 306 specimens and by qRT-PCR in 111 samples. Results A high epithelial expression level of Tie-2 is a significant prognostic factor in primary high grade serous ovarian cancer. It predicted significantly shorter overall survival both in univariate (p<0.001) and multivariate survival analyses (p = 0.022). Low angiopoietin-2 expression levels in primary ovarian tumors were significantly associated with shorter overall survival (p = 0.015) in the univariate survival analysis. A low expression of angiopoietin-2 was also significantly related to high grade tumors, size of residual tumor after primary surgery and the recurrence of cancer (p = 0.008; p = 0.012; p = 0.018) in the whole study population. The expression of angiopoietin-2 and Tie-2 was stronger in distal omental metastasis than in primary high grade serous tumors in matched-pair analysis (p = 0.001; p = 0.002). Conclusions The angiogenic factor, angiopoietin-2, and its receptor Tie-2 seem to be significant prognostic factors in primary epithelial ovarian cancer. Their expression levels are also increased in metastatic lesions in comparison with primary tumors.
Collapse
|
9
|
Liu M, Cai L, Li Q, Chen X, Gao L, Jiang L. The Expression of VEGF and CD31 in Endometrial Lesions and Its Associations with Blood Flow Parameters of Transvaginal 3D Power Doppler Ultrasonography: A Preliminary Study. Cancer Manag Res 2020; 12:11211-11218. [PMID: 33177872 PMCID: PMC7649237 DOI: 10.2147/cmar.s277274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/09/2020] [Indexed: 11/23/2022] Open
Abstract
Aim To investigate the association of the blood flow parameters measured by transvaginal three-dimensional power Doppler ultrasound and the angiogenesis of endometrial cancer. Material and Methods The expressions of vascular endothelial growth factor (VEGF) and CD31 in benign and malignant endometrial lesions, and in malignant lesions with different clinical and pathological features were analyzed. The correlations of the blood flow parameters (vascularization index [VI], blood flow index [FI], and vascularization-blood flow index [VFI]) of transvaginal 3D power Doppler ultrasound, and VEGF expressions, microvessel density (MVD) were also evaluated. Results The VEGF-positive rates and the MVD values in benign and malignant endometrial lesions were significantly different (both P<0.001). The differences of VEGF-positive rates (P < 0.001) and MVD values (P = 0.021) between type I and type II lesions of endometrial cancer were statistically significant. There was no significant difference in the VEGF-positive rate and MVD value between stage IA and IB (P=0.443, P=0.311). The difference of VEGF expression and MVD in stage IA, stage IB and stage II and above was statistically significant (P=0.003, P=0.017). The VEGF-positive rate and MVD value were not significantly different in IAG1 and IAG2 lesions of endometrioid adenocarcinoma (P=0.709, P=0.792). There was no significant correlation between VI, FI, VFI and VEGF expression and MVD in endometrial cancer. Conclusion The VEGF-positive rates and MVD values were relatively high in malignant endometrial lesions, type II and stage II and above lesions of type I endometrial cancer, indicating that the angiogenesis of endometrial cancer tissues might play a crucial role in the tumor classification, and pelvic metastasis.
Collapse
Affiliation(s)
- Meijuan Liu
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Li Cai
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Qifan Li
- Department of Ultrasound, RongCheng Maternal and Child Care Service Centre, Weihai, People's Republic of China
| | - Xiaoran Chen
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lingyun Gao
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lei Jiang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| |
Collapse
|
10
|
Abdelgawad ME, Darwish H, Nabawy MM, El-mezayen H. Development of novel score based on Angiogenic panel for accurate diagnosis of hepatocellular carcinoma among hepatitis C virus high-risk patients. INFECTION GENETICS AND EVOLUTION 2020; 85:104572. [DOI: 10.1016/j.meegid.2020.104572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
|
11
|
Pan X, Ma X. A Novel Six-Gene Signature for Prognosis Prediction in Ovarian Cancer. Front Genet 2020; 11:1006. [PMID: 33193589 PMCID: PMC7593580 DOI: 10.3389/fgene.2020.01006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer (OC) is the most malignant tumor in the female reproductive tract. Although abundant molecular biomarkers have been identified, a robust and accurate gene expression signature is still essential to assist oncologists in evaluating the prognosis of OC patients. In this study, samples from 367 patients in The Cancer Genome Atlas (TCGA) database were subjected to mRNA expression profiling. Then, we used a gene set enrichment analysis (GSEA) to screen genes correlated with epithelial–mesenchymal transition (EMT) and assess their prognostic power with a Cox proportional regression model. Six genes (TGFBI, SFRP1, COL16A1, THY1, PPIB, BGN) associated with overall survival (OS) were used to construct a risk assessment model, after which the patients were divided into high-risk and low-risk groups. The six-gene signature was an independent prognostic biomarker of OS for OC patients based on the multivariate Cox regression analysis. In addition, the six-gene model was validated with samples from the Gene Expression Omnibus (GEO) database. In summary, we established a six-gene signature relevant to the prognosis of OC, which might become a therapeutic tool with clinical applications in the future.
Collapse
Affiliation(s)
- Xin Pan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoxin Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Yang Y, Yang Y, Yang J, Zhao X, Wei X. Tumor Microenvironment in Ovarian Cancer: Function and Therapeutic Strategy. Front Cell Dev Biol 2020; 8:758. [PMID: 32850861 PMCID: PMC7431690 DOI: 10.3389/fcell.2020.00758] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is one of the leading causes of death in patients with gynecological malignancy. Despite optimal cytoreductive surgery and platinum-based chemotherapy, ovarian cancer disseminates and relapses frequently, with poor prognosis. Hence, it is urgent to find new targeted therapies for ovarian cancer. Recently, the tumor microenvironment has been reported to play a vital role in the tumorigenesis of ovarian cancer, especially with discoveries from genome-, transcriptome- and proteome-wide studies; thus tumor microenvironment may present potential therapeutic target for ovarian cancer. Here, we review the interactions between the tumor microenvironment and ovarian cancer and various therapies targeting the tumor environment.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Lee JM, Annunziata CM, Hays JL, Cao L, Choyke P, Yu M, An D, Turkbey IB, Minasian LM, Steinberg SM, Chen H, Wright J, Kohn EC. Phase II trial of bevacizumab and sorafenib in recurrent ovarian cancer patients with or without prior-bevacizumab treatment. Gynecol Oncol 2020; 159:88-94. [PMID: 32747013 DOI: 10.1016/j.ygyno.2020.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To examine whether blocking multiple points of the angiogenesis pathway by addition of sorafenib, a multi-kinase inhibitor against VEGFR2/3, Raf, c-Kit, and PDGFR, to bevacizumab would yield clinical activity in ovarian cancer (OvCa). METHODS This phase II study tested bevacizumab plus sorafenib in two cohorts; bevacizumab-naïve and bevacizumab-exposed patients. Bevacizumab (5 mg/kg IV every 2 weeks) was given with sorafenib 200 mg bid 5 days-on/2 days-off. The primary objective was response rate using a Simon two-stage optimal design. Progression-free survival (PFS) and toxicity were the secondary endpoints. Exploratory correlative studies included plasma cytokine concentrations, tissue proteomics and dynamic contrast-enhanced-magnetic resonance imaging (DCE-MRI). RESULTS Between March 2007 and August 2012, 54 women were enrolled, 41 bevacizumab-naive and 13 bevacizumab-prior, with median 5 (2-9) and 6 (5-9) prior systemic therapies, respectively. Nine of 35 (26%) evaluable bevacizumab-naive patients attained partial responses (PR), and 18 had stable disease (SD) ≥ 4 months. No responses were seen in the bevacizumab-prior group and 7 (54%) patients had SD ≥ 4 months, including one exceptional responder with SD of 27 months. The overall median PFS was 5.5 months (95%CI: 4.0-6.8 months). Treatment-related grade 3/4 adverse events (≥5%) included hypertension (17/54 [31%]; grade 3 in 16 patients and grade 4 in one patient) and venous thrombosis or pulmonary embolism (5/54 [9%]; grade 3 in 4 patients and grade 4 in one patient). Pretreatment low IL8 concentration was associated with PFS ≥ 4 months (p = .031). CONCLUSIONS The bevacizumab and sorafenib combination did not meet the pre-specified primary endpoint although some clinical activity was seen in heavily-pretreated bevacizumab-naive OvCa patients with platinum-resistant disease. Anticipated class toxicities required close monitoring and dose modifications.
Collapse
Affiliation(s)
- Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America.
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America
| | - John L Hays
- Division of Medical Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, United States of America
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, United States of America
| | - Peter Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, United States of America
| | - Minshu Yu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America
| | - Daniel An
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America
| | - Ismail Baris Turkbey
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, United States of America
| | - Lori M Minasian
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, United States of America
| | - Helen Chen
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, MD, United States of America
| | - John Wright
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, MD, United States of America
| | - Elise C Kohn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, United States of America
| |
Collapse
|
14
|
Sun L, Yang M, Zhang X, Li H, Wu L, Zhang Y, Cai S. Anlotinib combined with etoposide for platinum-resistant recurrent ovarian cancer: A case report. Medicine (Baltimore) 2020; 99:e20053. [PMID: 32443311 PMCID: PMC7253621 DOI: 10.1097/md.0000000000020053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Platinum-resistant ovarian cancer is characterized by its poor prognosis and limited treatment options. Angiogenesis plays a fundamental role in the development of drug-resistance in ovarian cancer. Anlotinib, a novel oral multi-targeted tyrosine kinase inhibitor which targets a board spectrum of angiogenesis-associated growth factor receptors, has shown promising anti-tumor efficacy in clinical trials. Herein, we report a case of ovarian cancer treated with anlotinib plus etoposide after secondary cytoreductive surgery. PATIENT CONCERNS A 45-year-old female with primary platinum-resistant ovarian cancer who progressed rapidly after the first cytoreductive surgery, the second cytoreductive surgery, and several lines of treatment. The patient refused to receive intravenous chemotherapy any more. DIAGNOSIS Primary platinum-resistant ovarian cancer. INTERVENTIONS The oral combination treatment of anlotinib (12 mg, qd) and etoposide (100 mg, qd) were delivered. OUTCOMES Finally, the patient was responsive to the orally treatment of anlotinib combined with etoposide. The patient has been alive with no evidence of disease progression for 18 weeks. CONCLUSION Our case suggests that oral treatment of anlotinib combined with etoposide which is acceptable and convenient, may be an additional option for the management of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Li Sun
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Meng Yang
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Xuan Zhang
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Hua Li
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Lingying Wu
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yuzi Zhang
- The Medical Department, 3D Medicines Inc, Shanghai, People's Republic of China
| | - Shangli Cai
- The Medical Department, 3D Medicines Inc, Shanghai, People's Republic of China
| |
Collapse
|
15
|
Catani MV, Savini I, Tullio V, Gasperi V. The "Janus Face" of Platelets in Cancer. Int J Mol Sci 2020; 21:ijms21030788. [PMID: 31991775 PMCID: PMC7037171 DOI: 10.3390/ijms21030788] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022] Open
Abstract
Besides their vital role in hemostasis and thrombosis, platelets are also recognized to be involved in cancer, where they play an unexpected central role: They actively influence cancer cell behavior, but, on the other hand, platelet physiology and phenotype are impacted by tumor cells. The existence of this platelet-cancer loop is supported by a large number of experimental and human studies reporting an association between alterations in platelet number and functions and cancer, often in a way dependent on patient, cancer type and treatment. Herein, we shall report on an update on platelet-cancer relationships, with a particular emphasis on how platelets might exert either a protective or a deleterious action in all steps of cancer progression. To this end, we will describe the impact of (i) platelet count, (ii) bioactive molecules secreted upon platelet activation, and (iii) microvesicle-derived miRNAs on cancer behavior. Potential explanations of conflicting results are also reported: Both intrinsic (heterogeneity in platelet-derived bioactive molecules with either inhibitory or stimulatory properties; features of cancer cell types, such as aggressiveness and/or tumour stage) and extrinsic (heterogeneous characteristics of cancer patients, study design and sample preparation) factors, together with other confounding elements, contribute to “the Janus face” of platelets in cancer. Given the difficulty to establish the univocal role of platelets in a tumor, a better understanding of their exact contribution is warranted, in order to identify an efficient therapeutic strategy for cancer management, as well as for better prevention, screening and risk assessment protocols.
Collapse
Affiliation(s)
- Maria Valeria Catani
- Correspondence: (M.V.C.); (V.G.); Tel.: +39-06-72596465 (M.V.C.); +39-06-72596465 (V.G.)
| | | | | | - Valeria Gasperi
- Correspondence: (M.V.C.); (V.G.); Tel.: +39-06-72596465 (M.V.C.); +39-06-72596465 (V.G.)
| |
Collapse
|
16
|
Ayala-Domínguez L, Olmedo-Nieva L, Muñoz-Bello JO, Contreras-Paredes A, Manzo-Merino J, Martínez-Ramírez I, Lizano M. Mechanisms of Vasculogenic Mimicry in Ovarian Cancer. Front Oncol 2019; 9:998. [PMID: 31612116 PMCID: PMC6776917 DOI: 10.3389/fonc.2019.00998] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022] Open
Abstract
Solid tumors carry out the formation of new vessels providing blood supply for growth, tumor maintenance, and metastasis. Several processes take place during tumor vascularization. In angiogenesis, new vessels are derived from endothelial cells of pre-existing vessels; while in vasculogenesis, new vessels are formed de novo from endothelial progenitor cells, creating an abnormal, immature, and disorganized vascular network. Moreover, highly aggressive tumor cells form structures similar to vessels, providing a pathway for perfusion; this process is named vasculogenic mimicry (VM), where vessel-like channels mimic the function of vessels and transport plasma and blood cells. VM is developed by numerous types of aggressive tumors, including ovarian carcinoma which is the second most common cause of death among gynecological cancers. VM has been associated with poor patient outcome and survival in ovarian cancer, although the involved mechanisms are still under investigation. Several signaling molecules have an important role in VM in ovarian cancer, by regulating the expression of genes related to vascular, embryogenic, and hypoxic signaling pathways. In this review, we provide an overview of the current knowledge of the signaling molecules involved in the promotion and regulation of VM in ovarian cancer. The clinical implications and the potential benefit of identification and targeting of VM related molecules for ovarian cancer treatment are also discussed.
Collapse
Affiliation(s)
- Lízbeth Ayala-Domínguez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Leslie Olmedo-Nieva
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - J Omar Muñoz-Bello
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adriana Contreras-Paredes
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Imelda Martínez-Ramírez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
17
|
Efficacy of a Bispecific Antibody Co-Targeting VEGFA and Ang-2 in Combination with Chemotherapy in a Chemoresistant Colorectal Carcinoma Xenograft Model. Molecules 2019; 24:molecules24162865. [PMID: 31394786 PMCID: PMC6719918 DOI: 10.3390/molecules24162865] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 01/13/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) inhibition by the addition of bevacizumab to the chemotherapy regimen of metastatic colorectal cancer leads to an improved outcome. However, anti-angiogenic tumor therapy targeting a single factor may be limited by complementary mechanisms. Angiopoietin-2 (Ang-2, ANGPT2) is another important factor that cooperates with VEGF to drive tumor angiogenesis. It was shown that high Ang-2 levels are associated with a poor clinical outcome of colorectal cancer patients treated with bevacizumab-containing therapy. Therefore, combined inhibition of VEGF and Ang-2 was supposed to improve anti-angiogenic therapy. Here, we evaluated the efficacy of a bispecific antibody (CrossMab) co-targeting VEGF and Ang-2 in combination with chemotherapy in a chemoresistant colorectal carcinoma model. Antitumor activity was evaluated in athymic nude mice bearing subcutaneous DLD1 xenograft tumors and treated with anti-VEGF (B20), anti-Ang-2 (LC06) and anti-VEGF/Ang-2 (CrossMab) antibodies. Chemotherapy consisted of 5-FU and irinotecan. Resected tumors were analyzed immunohistochemically. First, an impact of targeting each single factor but also a clear advantage of co-targeting both factors could be demonstrated. Accordingly, tumor tissue showed strong staining for VEGF and Ang-2. Chemotherapy alone was less effective. Efficient tumor growth inhibition could be achieved by treatment with anti-VEGF/chemotherapy, single CrossMab and CrossMab/chemotherapy, which resulted in 3 out of 10, 6 out of 10 and 10 out of 10 complete responses, respectively, during seven weeks. Complete retarded tumors were characterized by massive intratumoral necrosis surrounded by layers of vital tumor cells and connective tissue with CD31-positive vessels at the periphery. In some cases, a distinct feature known as vessel co-option could be observed. In conclusion, the data from this model clearly support the strategy of co-targeting VEGF and Ang-2 and further demonstrate the beneficial impact of co-treatment with chemotherapy. The clear superiority of the CrossMab-containing regimen compared to clinical standard anti-VEGF/chemotherapy warrants further analyses in other models.
Collapse
|
18
|
Zhao L, Ma X, Liang M, Li D, Ma P, Wang S, Wu Z, Zhao X. Prediction for early recurrence of intrahepatic mass-forming cholangiocarcinoma: quantitative magnetic resonance imaging combined with prognostic immunohistochemical markers. Cancer Imaging 2019; 19:49. [PMID: 31307551 PMCID: PMC6631577 DOI: 10.1186/s40644-019-0234-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/25/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Partial hepatectomy is the first option for intrahepatic mass-forming cholangiocarcinoma (IMCC) treatment, which would prolong survival. The main reason for the poor outcome after curative resection is the high incidence of early recurrence (ER). The aim of this study was to investigate the combined predictive performance of qualitative and quantitative magnetic resonance imaging (MRI) features and prognostic immunohistochemical markers for the ER of IMCC. METHODS Forty-seven patients with pathologically proven IMCC were enrolled in this retrospective study. Preoperative contrast-enhanced MRI and post-operative immunohistochemical staining of epidermal growth factor receptors (EGFR), vascular endothelial growth factor receptor (VEGFR), P53 and Ki67 were performed. Univariate analysis identified clinic-radiologic and pathological risk factors of ER. Radiomics analysis was performed based on four MRI sequences including fat suppression T2-weighted imaging (T2WI/FS), arterial phase (AP), portal venous phase (PVP), and delayed phase (DP) contrast enhanced imaging. A clinicoradiologic-pathological (CRP) model, radiomics model, and combined model were developed. And ROC curves were used to explore their predictive performance for ER stratification. RESULTS Enhancement patterns and VEGFR showed significant differences between the ER group and non-ER group (P = 0.001 and 0.034, respectively). The radiomics model based on AP, PVP and DP images presented superior AUC (0.889, 95% confidence interval (CI): 0.783-0.996) among seven radiomics models with a sensitivity of 0.938 and specificity of 0.839. The combined model, containing enhancement patterns, VEGFR and radiomics features, showed a preferable ER predictive performance compared to the radiomics model or CRP model alone, with AUC, sensitivity and specificity of 0.949, 0.875 and 0.774, respectively. CONCLUSIONS The combined model was the superior predictive model of ER. Combining qualitative and quantitative MRI features and VEGFR enables ER prediction, thus facilitating personalized treatment for patients with IMCC.
Collapse
Affiliation(s)
- Li Zhao
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Meng Liang
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Peiqing Ma
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Sicong Wang
- Department of Pharmaceutical Diagnosis, GE Healthcare, Life Sciences, No.1 Tongji South Road, Beijing, 100176, China
| | - Zhiyuan Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College , No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and PekingUnion Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
19
|
Sawada M, Oishi T, Komatsu H, Sato S, Chikumi J, Nonaka M, Kudoh A, Osaku D, Harada T. Serum vascular endothelial growth factor A and vascular endothelial growth factor receptor 2 as prognostic biomarkers for uterine cervical cancer. Int J Clin Oncol 2019; 24:1612-1619. [PMID: 31236742 DOI: 10.1007/s10147-019-01495-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/17/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND There are few studies on serum vascular endothelial growth factors and receptors (VEGF/VEGFRs) in patients with uterine cervical cancer (CC). The aim of this study was to determine whether VEGF/VEGFRs could be used as prognostic biomarkers in patients with CC. METHODS A total of 107 patients with stage IB to IIB CC, who underwent radical hysterectomy at Tottori University Hospital between 2006 and 2015, were included in this study. Serum samples were collected prior to radical hysterectomy, and levels of VEGF-A, VEGF-C, VEGFR-1, and VEGFR-2 were analyzed by enzyme-linked immunosorbent assays. We evaluated the association between the levels of these angiogenic factors and clinicopathologic variables. Survival analysis of 93 patients treated between 2006 and 2013 was performed. RESULTS The levels of VEGF-A in patients with bulky tumor, pelvic lymph-node involvement (PLNI), and parametrial infiltration (PI) were significantly higher than those in patients without these factors (P = 0.022, P = 0.020, and P = 0.0013, respectively). The overall survival (OS) of patients with high VEGF-A and VEGFR-2 defined by median levels was significantly lower than the OS of patients with low levels of VEGF-A and VEGFR-2 (P = 0.014, P = 0.012, respectively). Multivariate analysis revealed that PLNI, serum VEGF-A levels, and serum VEGFR-2 levels were independent prognostic factors for OS (hazard ratio for VEGF-A 3.42, 95% CI 1.07-13.2; hazard ratio for VEGFR-2 6.37, 95% CI 1.59-43.5). CONCLUSION Our results suggest that serum VEGF-A and VEGFR-2 may be promising prognostic biomarkers for CC.
Collapse
Affiliation(s)
- Mayumi Sawada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan.
| | - Hiroaki Komatsu
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Shinya Sato
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Jun Chikumi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Michiko Nonaka
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Akiko Kudoh
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Daiken Osaku
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| | - Tasuku Harada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago, 683-8504, Japan
| |
Collapse
|
20
|
Sopo M, Anttila M, Hämäläinen K, Kivelä A, Ylä-Herttuala S, Kosma VM, Keski-Nisula L, Sallinen H. Expression profiles of VEGF-A, VEGF-D and VEGFR1 are higher in distant metastases than in matched primary high grade epithelial ovarian cancer. BMC Cancer 2019; 19:584. [PMID: 31200683 PMCID: PMC6570919 DOI: 10.1186/s12885-019-5757-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/27/2019] [Indexed: 01/11/2023] Open
Abstract
Background In many malignancies including ovarian cancer, different angiogenic factors have been related to poor prognosis. However, data on their relations to each other or importance as a prognostic factor in ovarian cancer is missing. Therefore, we investigated the expressions of VEGF-A, VEGF-C, and VEGF-D, and the receptors VEGFR1, VEGFR2, and VEGFR3 in patients with malignant epithelial ovarian neoplasms. We further compared expression levels between primary tumors and related distant omental metastases. Methods This study included 86 patients with malignant ovarian epithelial tumors and 16 related distant metastases. Angiogenic factor expression was evaluated using immunohistochemistry (n = 102) and qRT-PCR (n = 29). Results Compared to primary high grade serous ovarian tumors, the related omental metastases showed higher expressions of VEGF-A (p = 0.022), VEGF-D (p = 0.010), and VEGFR1 (p = 0.046). In univariate survival analysis, low epithelial expression of VEGF-A in primary tumors was associated with poor prognosis (p = 0.024), and short progression-free survival was associated with high VEGF-C (p = 0.034) and low VEGFR3 (p = 0.002). The relative expressions of VEGF-D, VEGFR1, VEGFR2, and VEGFR3 mRNA determined by qRT-PCR analyses were significantly correlated with the immunohistochemically detected levels of these proteins in primary high grade serous ovarian cancer and metastases (p = 0.004, p = 0.009, p = 0.015, and p = 0.018, respectively). Conclusions The expressions of VEGF receptors and their ligands significantly differed between malignant ovarian tumors and paired distant metastases. VEGF-A, VEGF-D, and VEGFR1 protein expressions seem to be higher in distant metastases than in the primary high grade serous ovarian cancer lesions.
Collapse
Affiliation(s)
- Minna Sopo
- Department of Gynecology, Kuopio University Hospital, Kuopio, Finland
| | - Maarit Anttila
- Department of Gynecology, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine, School of Medicine, Gynaecology, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Hämäläinen
- Department of Pathology and Forensic Medicine, Kuopio University Hospital, Kuopio, Finland.,Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
| | - Annukka Kivelä
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Veli-Matti Kosma
- Department of Pathology and Forensic Medicine, Kuopio University Hospital, Kuopio, Finland.,Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland.,Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
| | - Leea Keski-Nisula
- Department of Gynecology, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine, School of Medicine, Gynaecology, University of Eastern Finland, Kuopio, Finland
| | - Hanna Sallinen
- Department of Gynecology, Kuopio University Hospital, Kuopio, Finland. .,Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland. .,Institute of Clinical Medicine, School of Medicine, Gynaecology, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
21
|
Guan J, Darb-Esfahani S, Richter R, Taube ET, Ruscito I, Mahner S, Woelber L, Prieske K, Concin N, Vergote I, Van Nieuwenhuysen E, Achimas-Cadariu P, Glajzer J, Woopen H, Stanske M, Kulbe H, Denkert C, Sehouli J, Braicu EI. Vascular endothelial growth factor receptor 2 (VEGFR2) correlates with long-term survival in patients with advanced high-grade serous ovarian cancer (HGSOC): a study from the Tumor Bank Ovarian Cancer (TOC) Consortium. J Cancer Res Clin Oncol 2019; 145:1063-1073. [PMID: 30810838 DOI: 10.1007/s00432-019-02877-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The impact of angiogenesis on long-term survival of high-grade serous ovarian cancer (HGSOC) patients remains unclear. This study investigated whether angiogenic markers correlated with 5-year progression-free survival (PFS) in a large cohort of matched advanced HGSOC tissue samples. METHODS Tumor samples from 124 primary HGSOC patients were retrospectively collected within the Tumor Bank Ovarian Cancer ( http://www.toc-network.de ). All patients were in advanced stages (FIGO stage III-IV). No patient had received anti-angiogenesis therapy. The cohort contains 62 long-term survivors and 62 controls matched by age and post-surgical tumor residuals. Long-term survivors were defined as patients with no relapse within 5 years after the end of first-line chemotherapy. Controls were patients who suffered from first relapse within 6-36 months after primary treatment. Samples were assessed for immunohistochemical expression of vascular endothelial growth factor (VEGF) A and VEGF receptor 2 (VEGFR2). Expression profiles of VEGFA and VEGFR2 were compared between the two groups. RESULTS Significant correlation between VEGFA and VEGFR2 expression was observed (p < 0.0001, Spearman coefficient 0.347). A high expression of VEGFR2 (VEGFR2high) was found more frequently in long-term survivors (77.4%, 48/62) than in controls (51.6%, 30/62, p = 0.001), independent of FIGO stage and VEGFA expression in multivariate analysis (p = 0.005). Also, VEGFR2high was found the most frequently in women with PFS ≥ 10 years (p = 0.001) among all 124 patients. However, no significant association was detected between VEGFA expression and 5-year PFS (p = 0.075). CONCLUSIONS VEGFR2 overexpression significantly correlated with long-term PFS in HGSOC patients, independent of age, FIGO stage, tumor residual and VEGFA expression.
Collapse
Affiliation(s)
- Jun Guan
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Silvia Darb-Esfahani
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Berlin Institute of Health, Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rolf Richter
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Eliane T Taube
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Berlin Institute of Health, Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ilary Ruscito
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany.,Laboratory of Cell Therapy and Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Sven Mahner
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology, University-Medical-Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Linn Woelber
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology, University-Medical-Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Prieske
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology, University-Medical-Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Concin
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ignace Vergote
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology and Obstetrics, University Hospital Leuven, Leuven, Belgium
| | - Els Van Nieuwenhuysen
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Gynecology and Obstetrics, University Hospital Leuven, Leuven, Belgium
| | - Patriciu Achimas-Cadariu
- Tumorbank Ovarian Cancer Network, Berlin, Germany.,Department of Surgical and Gynecological Oncology, The Oncology Institute Cluj-Napoca, University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Joanna Glajzer
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Hannah Woopen
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Mandy Stanske
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hagen Kulbe
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Carsten Denkert
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Tumorbank Ovarian Cancer Network, Berlin, Germany
| | - Elena Ioana Braicu
- Department of Gynecology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany. .,Tumorbank Ovarian Cancer Network, Berlin, Germany.
| |
Collapse
|
22
|
Himbert C, Ose J, Nattenmüller J, Warby CA, Holowatyj AN, Böhm J, Lin T, Haffa M, Gigic B, Hardikar S, Scherer D, Zielske L, Schrotz-King P, Kölsch T, Siegel EM, Shibata D, Ulrich A, Schneider M, Hursting SD, Kauczor HU, Ulrich CM. Body Fatness, Adipose Tissue Compartments, and Biomarkers of Inflammation and Angiogenesis in Colorectal Cancer: The ColoCare Study. Cancer Epidemiol Biomarkers Prev 2018; 28:76-82. [PMID: 30333223 DOI: 10.1158/1055-9965.epi-18-0654] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Adiposity has been linked to both risk and prognosis of colorectal cancer; however, the impact of different fat areas [visceral (VFA) vs. subcutaneous fat area (SFA)] is unclear. We investigated associations between adiposity and biomarkers of inflammation and angiogenesis among patients with colorectal cancer. METHODS Preoperative serum samples and computed tomography scans were obtained from 188 patients diagnosed with primary invasive stage I-IV colorectal cancer enrolled in the ColoCare Study. Adiposity was assessed by area-based quantification of VFA, SFA, and VFA:SFA ratio on spinal levels L3/L4 and L4/L5. Circulating levels of inflammation (CRP, SAA, sICAM-1, and sVCAM-1) and angiogenesis (VEGF-A and VEGF-D) were assessed from patient sera on the Meso Scale Discovery platform. Partial correlations and regression analyses, adjusted for age, sex, and tumor stage, were performed. RESULTS VFA was moderately correlated with CRP and SAA (CRP: L3/L4 and L4/L5:r = 0.21, P = 0.01; SAA: L3/L4:r = 0.17, P = 0.04). The correlation between SFA and the measured biomarkers were weak (r ≤ 0.13, not significant). The ratio of VFA:SFA at L3/L4 was moderately correlated with VEGF-A (r = 0.28, P = 0.0008) and SAA (r = 0.24, P = 0.006), and less so with CRP (r = 0.18, P = 0.04) and sICAM-1 (r = 0.18, P = 0.04). Similar correlations were found for the VFA:SFA ratio at L4/L5. CONCLUSIONS We observed an association between visceral adiposity and biomarkers of inflammation and angiogenesis in colorectal cancer. In particular, the VFA:SFA ratio was correlated with circulating levels of the proangiogenic biomarker VEGF-A. IMPACT Our findings support a direct association of visceral adipose tissue with inflammatory and angiogenic processes, which play fundamental roles in the development and progression of colorectal cancer.
Collapse
Affiliation(s)
- Caroline Himbert
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Johanna Nattenmüller
- Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
| | - Christy A Warby
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Andreana N Holowatyj
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Jürgen Böhm
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Tengda Lin
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Mariam Haffa
- National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Sheetal Hardikar
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Dominique Scherer
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Lin Zielske
- National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petra Schrotz-King
- National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Torsten Kölsch
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Erin M Siegel
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - David Shibata
- University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | | | - Hans-Ulrich Kauczor
- Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Salt Lake City, Utah.
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
23
|
Zhang C, Wang N, Tan HY, Guo W, Li S, Feng Y. Targeting VEGF/VEGFRs Pathway in the Antiangiogenic Treatment of Human Cancers by Traditional Chinese Medicine. Integr Cancer Ther 2018; 17:582-601. [PMID: 29807443 PMCID: PMC6142106 DOI: 10.1177/1534735418775828] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bearing in mind the doctrine of tumor angiogenesis hypothesized by Folkman
several decades ago, the fundamental strategy for alleviating numerous cancer
indications may be the strengthening application of notable antiangiogenic
therapies to inhibit metastasis-related tumor growth. Under physiological
conditions, vascular sprouting is a relatively infrequent event unless when
specifically stimulated by pathogenic factors that contribute to the
accumulation of angiogenic activators such as the vascular endothelial growth
factor (VEGF) family and basic fibroblast growth factor (bFGF). Since VEGFs have
been identified as the principal cytokine to initiate angiogenesis in tumor
growth, synthetic VEGF-targeting medicines containing bevacizumab and sorafenib
have been extensively used, but prominent side effects have concomitantly
emerged. Traditional Chinese medicines (TCM)–derived agents with distinctive
safety profiles have shown their multitarget curative potential by impairing
angiogenic stimulatory signaling pathways directly or eliciting synergistically
therapeutic effects with anti-angiogenic drugs mainly targeting VEGF-dependent
pathways. This review aims to summarize (a) the up-to-date
understanding of the role of VEGF/VEGFR in correlation with proangiogenic
mechanisms in various tissues and cells; (b) the elaboration of
antitumor angiogenesis mechanisms of 4 representative TCMs, including
Salvia miltiorrhiza, Curcuma longa, ginsenosides, and
Scutellaria baicalensis; and (c)
circumstantial clarification of TCM-driven therapeutic actions of suppressing
tumor angiogenesis by targeting VEGF/VEGFRs pathway in recent years, based on
network pharmacology.
Collapse
Affiliation(s)
- Cheng Zhang
- 1 The University of Hong Kong, Hong Kong SAR
| | - Ning Wang
- 1 The University of Hong Kong, Hong Kong SAR
| | - Hor-Yue Tan
- 1 The University of Hong Kong, Hong Kong SAR
| | - Wei Guo
- 1 The University of Hong Kong, Hong Kong SAR
| | - Sha Li
- 1 The University of Hong Kong, Hong Kong SAR
| | - Yibin Feng
- 1 The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
24
|
Combined Gene Therapy Using AdsVEGFR2 and AdsTie2 With Chemotherapy Reduces the Growth of Human Ovarian Cancer and Formation of Ascites in Mice. Int J Gynecol Cancer 2018; 27:879-886. [PMID: 28498260 DOI: 10.1097/igc.0000000000000973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Ovarian cancer is highly dependent on tumor microvessels and angiogenesis regulated by vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) and angiopoietins (Ang) and their Tie receptors. We studied the efficacy of adenoviral (Ad) gene therapy with soluble VEGFR2 and Tie2 combined with paclitaxel and carboplatin for the treatment of ovarian cancer. METHODS An intraperitoneal human ovarian cancer xenograft model in nude mice (n = 44) was used in this study. Gene therapy was given intravenously when the presence of sizable tumors was confirmed in magnetic resonance imaging. The study groups were as follows: AdCMV as a control (group I), AdCMV with chemotherapy (group II), AdsVEGFR2 and AdsTie2 (group III), and AdsVEGFR2 and AdsTie2 with chemotherapy (group IV). Antitumor effectiveness was assessed by overall tumor growth, ascites, immunohistochemistry, microvessel density, and sequential magnetic resonance imaging analyses. RESULTS AdsVEGFR2 and AdsTie2 gene therapy (group III) significantly reduced tumor weights as compared with group II (P = 0.007). Accumulation of ascites was significantly reduced when the mice were treated with AdsVEGFR2 and AdsTie2 gene therapy or with combined gene therapy and chemotherapy as compared with controls (P = 0.029 and P = 0.010, respectively). Vascular endothelial growth factor and Ang2 levels in ascites fluid were elevated after the gene therapy. CONCLUSIONS Combined inhibition of VEGF/VEGFR2 and Ang/Tie2 pathways provided efficient therapy for ovarian cancer in mice. In addition, antiangiogenic gene therapy has potential as a treatment for the accumulation of ascites.
Collapse
|
25
|
Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, Taus A, Albanell J, Sablin MP, Alt M, Bahleda R, Varga A, Boetsch C, Franjkovic I, Heil F, Lahr A, Lechner K, Morel A, Nayak T, Rossomanno S, Smart K, Stubenrauch K, Krieter O. First-in-Human Phase I Study of Single-agent Vanucizumab, A First-in-Class Bispecific Anti-Angiopoietin-2/Anti-VEGF-A Antibody, in Adult Patients with Advanced Solid Tumors. Clin Cancer Res 2017; 24:1536-1545. [PMID: 29217526 DOI: 10.1158/1078-0432.ccr-17-1588] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/23/2017] [Accepted: 11/30/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Vanucizumab is an investigational antiangiogenic, first-in-class, bispecific mAb targeting VEGF-A and angiopoietin-2 (Ang-2). This first-in-human study evaluated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of vanucizumab in adults with advanced solid tumors refractory to standard therapies.Experimental Design: Patients received escalating biweekly (3-30 mg/kg) or weekly (10-30 mg/kg) intravenous doses guided by a Bayesian logistic regression model with overdose control.Results: Forty-two patients were treated. One dose-limiting toxicity, a fatal pulmonary hemorrhage from a large centrally located mediastinal mass judged possibly related to vanucizumab, occurred with the 19 mg/kg biweekly dose. Arterial hypertension (59.5%), asthenia (42.9%), and headache (31%) were the most common toxicities. Seventeen (41%) patients experienced treatment-related grade ≥3 toxicities. Toxicity was generally higher with weekly than biweekly dosing. A MTD of vanucizumab was not reached in either schedule. Pharmacokinetics were dose-linear with an elimination half-life of 6-9 days. All patients had reduced plasma levels of free VEGF-A and Ang-2; most had reductions in KTRANS (measured by dynamic contrast-enhanced MRI). Two patients (renal cell and colon cancer) treated with 30 mg/kg achieved confirmed partial responses. Ten patients were without disease progression for ≥6 months. A flat-fixed 2,000 mg biweekly dose (phamacokinetically equivalent to 30 mg/kg biweekly) was recommended for further investigation.Conclusions: Biweekly vanucizumab had an acceptable safety and tolerability profile consistent with single-agent use of selective inhibitors of the VEGF-A and Ang/Tie2 pathway. Vanucizumab modulated its angiogenic targets, impacted tumor vascularity, and demonstrated encouraging antitumor activity in this heterogeneous population. Clin Cancer Res; 24(7); 1536-45. ©2017 AACR.
Collapse
Affiliation(s)
- Manuel Hidalgo
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain. .,START Madrid-CIOCC, HM Sanchinarro, Madrid, Spain
| | | | | | | | | | | | - Alvaro Taus
- Medical Oncology Department, Hospital del Mar., Barcelona, Spain
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar., Barcelona, Spain
| | - Marie-Paule Sablin
- Department of Medical Oncology, Institut Curie, Saint-Cloud and Paris, France
| | - Marie Alt
- Department of Medical Oncology, Institut Curie, Saint-Cloud and Paris, France
| | - Ratislav Bahleda
- Department of Drug Development, Gustave Roussy, Villejuif, France
| | - Andrea Varga
- Department of Drug Development, Gustave Roussy, Villejuif, France
| | | | | | - Florian Heil
- Roche Innovation Center Munich, Penzberg, Germany
| | | | | | | | - Tapan Nayak
- Roche Innovation Center Basel, Basel, Switzerland
| | | | - Kevin Smart
- Roche Innovation Center Welwyn, Welwyn Garden City, United Kingdom
| | | | | |
Collapse
|
26
|
Boylan KLM, Geschwind K, Koopmeiners JS, Geller MA, Starr TK, Skubitz APN. A multiplex platform for the identification of ovarian cancer biomarkers. Clin Proteomics 2017; 14:34. [PMID: 29051715 PMCID: PMC5634875 DOI: 10.1186/s12014-017-9169-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023] Open
Abstract
Background Currently, there are no FDA approved screening tools for detecting early stage ovarian cancer in the general population. Development of a biomarker-based assay for early detection would significantly improve the survival of ovarian cancer patients.
Methods We used a multiplex approach to identify protein biomarkers for detecting early stage ovarian cancer. This new technology (Proseek® Multiplex Oncology Plates) can simultaneously measure the expression of 92 proteins in serum based on a proximity extension assay. We analyzed serum samples from 81 women representing healthy, benign pathology, early, and advanced stage serous ovarian cancer patients.
Results Principle component analysis and unsupervised hierarchical clustering separated patients into cancer versus non-cancer subgroups. Data from the Proseek® plate for CA125 levels exhibited a strong correlation with current clinical assays for CA125 (correlation coefficient of 0.89, 95% CI 0.83, 0.93). CA125 and HE4 were present at very low levels in healthy controls and benign cases, while higher levels were found in early stage cases, with highest levels found in the advanced stage cases. Overall, significant trends were observed for 38 of the 92 proteins (p < 0.001), many of which are novel candidate serum biomarkers for ovarian cancer. The area under the ROC curve (AUC) for CA125 was 0.98 and the AUC for HE4 was 0.85 when comparing early stage ovarian cancer versus healthy controls. In total, 23 proteins had an estimated AUC of 0.7 or greater. Using a naïve Bayes classifier that combined 12 proteins, we improved the sensitivity corresponding to 95% specificity from 93 to 95% when compared to CA125 alone. Although small, a 2% increase would have a significant effect on the number of women correctly identified when screening a large population. Conclusions These data demonstrate that the Proseek® technology can replicate the results established by conventional clinical assays for known biomarkers, identify new candidate biomarkers, and improve the sensitivity and specificity of CA125 alone. Additional studies using a larger cohort of patients will allow for validation of these biomarkers and lead to the development of a screening tool for detecting early stage ovarian cancer in the general population. Electronic supplementary material The online version of this article (doi:10.1186/s12014-017-9169-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kristin L M Boylan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, MMC 395, 420 Delaware Street, S.E, Minneapolis, MN 55455 USA.,Ovarian Cancer Early Detection Program, University of Minnesota, Minneapolis, MN USA
| | - Kate Geschwind
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, MMC 395, 420 Delaware Street, S.E, Minneapolis, MN 55455 USA.,Ovarian Cancer Early Detection Program, University of Minnesota, Minneapolis, MN USA
| | - Joseph S Koopmeiners
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Melissa A Geller
- Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, MN USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Timothy K Starr
- Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, MN USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA.,Department of Genetics, Cell Biology and Genetics, University of Minnesota, Minneapolis, MN USA
| | - Amy P N Skubitz
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, MMC 395, 420 Delaware Street, S.E, Minneapolis, MN 55455 USA.,Ovarian Cancer Early Detection Program, University of Minnesota, Minneapolis, MN USA.,Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, MN USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
27
|
Serum Vascular Endothelial Growth Factor-A as a Prognostic Biomarker for Epithelial Ovarian Cancer. Int J Gynecol Cancer 2017; 27:1325-1332. [DOI: 10.1097/igc.0000000000001027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BackgroundBevacizumab, 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.MethodsA 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.ResultsThe 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.ConclusionsSerum 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
|
28
|
Demethylation of the MIR145 promoter suppresses migration and invasion in breast cancer. Oncotarget 2017; 8:61731-61741. [PMID: 28977900 PMCID: PMC5617460 DOI: 10.18632/oncotarget.18686] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 05/12/2017] [Indexed: 12/16/2022] Open
Abstract
miR-145 has been implicated in the progression of breast cancer. Here, we report that its expression is decreased in breast cancer specimens and cell lines and that this low level of expression is associated with DNA methylation of its gene, MIR145. Methylation of MIR145 has previously been correlated with cell migration and invasion, both in vivo and in vitro. We found that demethylation of MIR145 reactivates miR-145 and contributes to the anti-cancer properties of 5-aza-2'-deoxyazacytidine (5-AzaC). Therefore, miR-145 is a potentially valuable biomarker for breast cancer.
Collapse
|
29
|
Yang P, Chen N, Yang D, Crane J, Yang S, Wang H, Dong R, Yi X, Xie L, Jing G, Cai J, Wang Z. The ratio of serum Angiopoietin-1 to Angiopoietin-2 in patients with cervical cancer is a valuable diagnostic and prognostic biomarker. PeerJ 2017; 5:e3387. [PMID: 28584715 PMCID: PMC5452943 DOI: 10.7717/peerj.3387] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/07/2017] [Indexed: 01/15/2023] Open
Abstract
Objectives Angiopoietins have been found to play essential roles in tumor angiogenesis. The present study was aimed at investigating the diagnostic and prognostic values of serum angiopoietin 1 and 2 (sAng-1 and sAng-2) in cervical cancer. Methods The sAng-1 and sAng-2 concentrations were analyzed in 77 women with cervical cancer, 44 women with cervical intraepithelial neoplasia (CIN) and 43 women without cervical lesions by enzyme-linked immunosorbent assay. The diagnostic values of sAng-1, sAng-2 and sAng-1/sAng-2 were evaluated by receiver operating characteristic (ROC) curves. The Ang-1 and Ang-2 expression in cervical cancer tissues as well as microvessel density (MVD), were assessed by immunohistochemistry. Results The concentration of sAng-2 gradually increased and the sAng-1/Ang-2 ratio was gradually decreased from normal control to CIN, then to squamous cell cancer, and the sAng-1/sAng-2 ratio was also significantly decreased in adenocarcinoma. The area under ROC curves of sAng-2 and sAng-1/sAng-2 ratio for discriminating cervical cancer from normal were 0.744 and 0.705, respectively. Decreased sAng-1/sAng-2 was significantly associated with advanced tumor stage, poor differentiation, lymph-vascular space invasion and high MVD. sAng-2 was positively correlated with the Ang-2 expression in cervix epithelia. A high sAng-1/sAng-2 ratio was associated with a longer progression-free survival and a longer overall survival in cervical cancer patients. Conclusions These findings suggest that sAng-2 and the sAng-1/sAng-2 ratio may be valuable diagnostic and prognostic biomarkers for cervical cancer.
Collapse
Affiliation(s)
- Ping Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Na Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyun Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Janet Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America.,Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States of America
| | - Shouhua Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hangyu Wang
- School of Pharmacy, Shihezi University, Shihezi, Xinjiang, China
| | - Ruiqing Dong
- Department of Obstetrics and Gynecology, Tianyou Hospital attended to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoqing Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisha Xie
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guo Jing
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Gadomska G, Stankowska K, Boinska J, Ślusarz R, Tylicka M, Michalska M, Jachalska A, Rość D. VEGF-A, sVEGFR-1, and sVEGFR-2 in BCR-ABL negative myeloproliferative neoplasms. MEDICINA-LITHUANIA 2017; 53:34-39. [PMID: 28237691 DOI: 10.1016/j.medici.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 12/13/2016] [Accepted: 01/09/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND OBJECTIVE Data from the literature indicate the relationship between the bone marrow microvessel density and the blood parameters of angiogenesis. The aim of this study was to evaluate selected parameters of angiogenesis (VEGF-A, sVEGFR-1, and sVEGFR-2) and their correlations with white blood cells, platelets, and red blood cells. MATERIALS AND METHODS The study included 72 patients (mean age, 61.84 years) with myeloproliferative neoplasms (MPNs): essential thrombocythemia (ET) (n=46), polycythemia vera (PV) (n=19), and primary myelofibrosis (PMF) (n=7). Serum VEGF-A, sVEGFR-1, and sVEGFR-2 were determined using the ELISA assay. RESULTS We observed a significantly higher level of VEGF-A and reduced concentrations of sVEGFR-1 and sVEGFR-2 in the whole group of patients with MPNs as compared to controls. Detailed analysis confirmed significantly higher level of VEGF-A and lower concentration of sVEGFR-2 in each subgroups of MPNs patients. However, sVEGFR-1 concentrations were significantly lower only in PV and ET patients. CONCLUSIONS The study showed an increased level of VEGF-A, which may indicate the intensity of neoangiogenesis in the bone marrow. Decreased sVEGFR-1 and sVEGFR-2 in the blood of patients with MPNs may reflect consumption of these soluble receptors.
Collapse
Affiliation(s)
- Grażyna Gadomska
- Department of Hematology and Malignant Diseases of Hematopoietic System, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Katarzyna Stankowska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland.
| | - Joanna Boinska
- Department of Neurological and Neurosurgical Nursing, Faculty of Health Sciences, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Robert Ślusarz
- Department of Neurological and Neurosurgical Nursing, Faculty of Health Sciences, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Marzena Tylicka
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Małgorzata Michalska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Anna Jachalska
- Department of Hematology and Malignant Diseases of Hematopoietic System, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| |
Collapse
|
31
|
Scholz A, Harter PN, Cremer S, Yalcin BH, Gurnik S, Yamaji M, Di Tacchio M, Sommer K, Baumgarten P, Bähr O, Steinbach JP, Trojan J, Glas M, Herrlinger U, Krex D, Meinhardt M, Weyerbrock A, Timmer M, Goldbrunner R, Deckert M, Braun C, Schittenhelm J, Frueh JT, Ullrich E, Mittelbronn M, Plate KH, Reiss Y. Endothelial cell-derived angiopoietin-2 is a therapeutic target in treatment-naive and bevacizumab-resistant glioblastoma. EMBO Mol Med 2016; 8:39-57. [PMID: 26666269 PMCID: PMC4718155 DOI: 10.15252/emmm.201505505] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is treated by surgical resection followed by radiochemotherapy. Bevacizumab is commonly deployed for anti‐angiogenic therapy of recurrent GBM; however, innate immune cells have been identified as instigators of resistance to bevacizumab treatment. We identified angiopoietin‐2 (Ang‐2) as a potential target in both naive and bevacizumab‐treated glioblastoma. Ang‐2 expression was absent in normal human brain endothelium, while the highest Ang‐2 levels were observed in bevacizumab‐treated GBM. In a murine GBM model, VEGF blockade resulted in endothelial upregulation of Ang‐2, whereas the combined inhibition of VEGF and Ang‐2 leads to extended survival, decreased vascular permeability, depletion of tumor‐associated macrophages, improved pericyte coverage, and increased numbers of intratumoral T lymphocytes. CD206+ (M2‐like) macrophages were identified as potential novel targets following anti‐angiogenic therapy. Our findings imply a novel role for endothelial cells in therapy resistance and identify endothelial cell/myeloid cell crosstalk mediated by Ang‐2 as a potential resistance mechanism. Therefore, combining VEGF blockade with inhibition of Ang‐2 may potentially overcome resistance to bevacizumab therapy.
Collapse
Affiliation(s)
- Alexander Scholz
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Patrick N Harter
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
| | - Sebastian Cremer
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Burak H Yalcin
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Stefanie Gurnik
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Maiko Yamaji
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Mariangela Di Tacchio
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Kathleen Sommer
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany
| | - Peter Baumgarten
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany Department of Neurosurgery, Goethe University Medical School, Frankfurt, Germany
| | - Oliver Bähr
- Senckenberg Institute of Neurooncology, Goethe University Medical School, Frankfurt, Germany
| | - Joachim P Steinbach
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany Senckenberg Institute of Neurooncology, Goethe University Medical School, Frankfurt, Germany
| | - Jörg Trojan
- Medical Clinic I, Goethe University Medical School, Frankfurt, Germany
| | - Martin Glas
- Klinische Kooperationseinheit Neuroonkologie, Robert Janker Klinik, Bonn, Germany
| | | | - Dietmar Krex
- Klinik und Poliklinik für Neurochirurgie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Matthias Meinhardt
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Astrid Weyerbrock
- Klinik für Neurochirurgie, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Marco Timmer
- Zentrum für Neurochirurgie, Uniklinik Köln, Köln, Germany
| | | | | | - Christian Braun
- Zentrum für Neuroonkologie, Universitätsklinik Tübingen, Tübingen, Germany
| | - Jens Schittenhelm
- Abteilung Neuropathologie, Universitätsklinik Tübingen, Tübingen, Germany
| | - Jochen T Frueh
- LOEWE Center for Cell and Gene Therapy, Goethe University Medical School, Frankfurt, Germany Pediatric Hematology & Oncology, Children's Hospital, Goethe University Medical School, Frankfurt, Germany
| | - Evelyn Ullrich
- LOEWE Center for Cell and Gene Therapy, Goethe University Medical School, Frankfurt, Germany Pediatric Hematology & Oncology, Children's Hospital, Goethe University Medical School, Frankfurt, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
| | - Karl H Plate
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
| | - Yvonne Reiss
- Institute of Neurology (Edinger Institute), Goethe University Medical School, Frankfurt, Germany German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
| |
Collapse
|
32
|
Monk BJ, Poveda A, Vergote I, Raspagliesi F, Fujiwara K, Bae DS, Oaknin A, Ray-Coquard I, Provencher DM, Karlan BY, Lhommé C, Richardson G, Rincón DG, Coleman RL, Marth C, Brize A, Fabbro M, Redondo A, Bamias A, Ma H, Vogl FD, Bach BA, Oza AM. Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2. Gynecol Oncol 2016; 143:27-34. [PMID: 27546885 DOI: 10.1016/j.ygyno.2016.07.112] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/12/2016] [Accepted: 07/23/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Trebananib, a peptibody that blocks binding of angiopoietin-1 and -2 to Tie2, significantly prolonged progression-free survival (PFS) in patients with recurrent epithelial ovarian cancer in the phase 3 TRINOVA-1 study. We report overall survival (OS) in the intent-to-treat population and clinically relevant subgroups and time to second disease progression (PFS-2). PATIENTS AND METHODS Women with recurrent disease (platinum-free interval<12months) were randomized to receive intravenous paclitaxel 80mg/m(2) (3weeks on/1week off) plus intravenous trebananib 15mg/kg or placebo, weekly. OS in the intent-to-treat population was a key secondary endpoint. Exploratory analysis of PFS-2 was conducted according to guidance by the European Medicines Agency. RESULTS Median OS was not significantly improved with trebananib compared with placebo (19.3 versus 18.3months; HR, 0.95; 95% CI, 0.81-1.11; P=0.52) in the intent-to-treat population (n=919). In subgroup analysis, trebananib improved median OS compared with placebo (14.5 versus 12.3months; HR, 0.72; 95% CI, 0.55-0.93; P=0.011) in patients with ascites at baseline (n=295). In the intent-to-treat population, trebananib significantly improved median PFS-2 compared with placebo (12.5 versus 10.9months; HR, 0.85; 95% CI, 0.74-0.98; P=0.024). The incidence and type of adverse events in this updated analysis was consistent with that described in the primary analysis; no new safety signals were detected. CONCLUSIONS OS was not significantly longer in the intent-to-treat population, although there was an improvement in OS in patients with ascites receiving trebananib. PFS-2 confirmed that the PFS benefit associated with trebananib was maintained through the second disease progression independent of the choice of subsequent therapy.
Collapse
Affiliation(s)
- Bradley J Monk
- Department of Obstetrics and Gynecology, University of Arizona Cancer Center at Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - Andrés Poveda
- Area Clinica de Oncologia Ginecológica, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Ignace Vergote
- Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven Cancer Institute, KU Leuven, European Union, Belgium
| | - Francesco Raspagliesi
- Gynecologic Oncology Unit, Fondazione IRCCS, Istituto Nazionale per la Cura e lo Studio dei Tumori, Milano, Italy
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka-Shi, Japan
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, South Korea
| | - Ana Oaknin
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Diane M Provencher
- Division of Gynecologic Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Catherine Lhommé
- Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Gary Richardson
- Academic Haematology and Oncology, Cabrini Hospital, Malvern, VIC, Australia
| | | | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christian Marth
- Universitätsklinik für Gynäkologie und Geburtshilfe, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Arija Brize
- Latvian Oncology Center, Riga Eastern Clinical University Hospital, Riga, Latvia
| | - Michel Fabbro
- Regional Cancer Institute Montpellier, Montpellier, France
| | | | - Aristotelis Bamias
- Alexandra Hospital, Department of Clinical Therapeutics, National & Kapodistrian University of Athens, Athens, Greece
| | - Haijun Ma
- Global Biostatistical Science, Amgen Inc., Thousand Oaks, CA, USA
| | - Florian D Vogl
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Bruce A Bach
- Global Development Oncology, Amgen Inc., Thousand Oaks, CA, USA
| | - Amit M Oza
- Department of Medicine, Princess Margaret Hospital, University of Toronto, ON, Canada
| |
Collapse
|
33
|
Trachana SP, Pilalis E, Gavalas NG, Tzannis K, Papadodima O, Liontos M, Rodolakis A, Vlachos G, Thomakos N, Haidopoulos D, Lykka M, Koutsoukos K, Kostouros E, Terpos E, Chatziioannou A, Dimopoulos MA, Bamias A. The Development of an Angiogenic Protein "Signature" in Ovarian Cancer Ascites as a Tool for Biologic and Prognostic Profiling. PLoS One 2016; 11:e0156403. [PMID: 27258020 PMCID: PMC4892506 DOI: 10.1371/journal.pone.0156403] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 05/13/2016] [Indexed: 01/07/2023] Open
Abstract
Advanced ovarian cancer (AOC) is one of the leading lethal gynecological cancers in developed countries. Based on the important role of angiogenesis in ovarian cancer oncogenesis and expansion, we hypothesized that the development of an "angiogenic signature" might be helpful in prediction of prognosis and efficacy of anti-angiogenic therapies in this disease. Sixty-nine samples of ascitic fluid- 35 from platinum sensitive and 34 from platinum resistant patients managed with cytoreductive surgery and 1st-line carboplatin-based chemotherapy- were analyzed using the Proteome ProfilerTM Human Angiogenesis Array Kit, screening for the presence of 55 soluble angiogenesis-related factors. A protein profile based on the expression of a subset of 25 factors could accurately separate resistant from sensitive patients with a success rate of approximately 90%. The protein profile corresponding to the "sensitive" subset was associated with significantly longer PFS (8 [95% Confidence Interval {CI}: 8-9] vs. 20 months [95% CI: 15-28]; Hazard ratio {HR}: 8.3, p<0.001) and OS (20.5 months [95% CI: 13.5-30] vs. 74 months [95% CI: 36-not reached]; HR: 5.6 [95% CI: 2.8-11.2]; p<0.001). This prognostic performance was superior to that of stage, histology and residual disease after cytoreductive surgery and the levels of vascular endothelial growth factor (VEGF) in ascites. In conclusion, we developed an "angiogenic signature" for patients with AOC, which can be used, after appropriate validation, as a prognostic marker and a tool for selection for anti-angiogenic therapies.
Collapse
Affiliation(s)
- Sofia-Paraskevi Trachana
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
- * E-mail:
| | - Eleftherios Pilalis
- Metabolic Engineering and Bioinformatics Program Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Nikos G. Gavalas
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Kimon Tzannis
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Olga Papadodima
- Metabolic Engineering and Bioinformatics Program Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Alexandros Rodolakis
- First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Georgios Vlachos
- First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Nikolaos Thomakos
- First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Dimitrios Haidopoulos
- First Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Maria Lykka
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Konstantinos Koutsoukos
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Efthimios Kostouros
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Evagelos Terpos
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Aristotelis Chatziioannou
- Metabolic Engineering and Bioinformatics Program Institute of Biology, Medicinal Chemistry & Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece
| |
Collapse
|
34
|
Photopsias during Systemic Bevacizumab Therapy. Case Rep Ophthalmol Med 2016; 2016:1926178. [PMID: 27069702 PMCID: PMC4812264 DOI: 10.1155/2016/1926178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/28/2016] [Indexed: 12/11/2022] Open
Abstract
Background. The authors describe a case of recurrent photopsias in a 56-year-old woman following repeat treatments with systemic intravenous bevacizumab for stage IV ovarian cancer. To our knowledge, this is the first report of photopsias following systemic bevacizumab treatments in a patient with a normal eye exam. Case Presentation. A 56-year-old Caucasian female complained of onset of floaters and flashes in the temporal peripheral field of the right eye 1 day after receiving intravenous of 30 g of 25 mg/mL of systemic bevacizumab for treatment of stage IV ovarian cancer. Ophthalmic examination, including dilated fundus exam, spectral domain optical coherence tomography (SD-OCT) of the optic nerve head, and enhanced depth imaging SD-OCT of the macula, revealed no significant abnormalities. Possible mechanisms are reviewed. Conclusion. We propose that patients who undergo intravenous bevacizumab treatments are questioned for any ocular symptoms and that more systematic evaluations of retinal nerve fiber layer and choroidal effects are obtained in those patients who are on long-term treatment at high doses.
Collapse
|
35
|
Al Wadi K, Ghatage P. Efficacy of trebananib (AMG 386) in treating epithelial ovarian cancer. Expert Opin Pharmacother 2016; 17:853-60. [PMID: 26933765 DOI: 10.1517/14656566.2016.1161027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the leading cause of death among gynecologic cancers. The majority of women are diagnosed with advanced stage disease. It is considered a chemosensitive cancer with a high initial response rate to first-line platinum and taxane-based chemotherapy. However, most patients with advanced EOC will relapse with subsequent resistance to conventional chemotherapy and ultimately succumb to their disease. Therefore, new therapeutic agents and strategies are desperately needed to improve the outcomes in patients with advanced EOC. AREAS COVERED This review focuses on the use of Trebananib (a non-VEGF-dependent angiogenesis pathway inhibitor) in EOC. Angiogenesis has been recognized as an important process promoting EOC growth and metastasis. Targeting angiogenesis in EOC have been developed and studied with demonstrated clinical efficacy. Bevacizumab, a humanized monoclonal antibody, that targets vascular endothelial growth factor A (VEGF-A), has been the most well evaluated molecular targeted therapy in the treatment of advanced and recurrent EOC with proven clinical efficacy. However, VEGF-dependent angiogenesis pathway inhibitors are often associated with serious toxicities and drug resistance ultimately develops. Hence, new therapeutic approach targeting the angiopoietin-Tie-2 complex pathway (a non-VEGF-dependent angiogenesis pathway) has gained interest over the past few years as an alternative strategy to overcome VEGF-dependent anti-angiogenesis-related toxicity and resistance. EXPERT OPINION Targeting angiopoietin-Tie-2 pathway represents a promising alternative approach to tumor anti-angiogenesis with a distinct toxicity profile from the VEGF-dependent pathway inhibitors. However, there are still many questions to be answered regarding the optimal treatment schedules, maintenance regimens, duration of maintenance therapy, and the best combination strategy. Currently there is no reliable surrogate molecular, cellular, or genetic marker that would definitively predict response to anti-angiogenic therapy. Identification of certain relevant and predictive biomarkers in the future may optimize treatment's efficacy by distinguishing the subset group of patients with EOC that would derive the most benefit from existing antiangiogenic treatment regimens.
Collapse
Affiliation(s)
- Khalid Al Wadi
- a Division of Gynecologic Oncology , Tom Baker Cancer Centre , Calgary , AB , Canada.,b Women's Specialized Hospital, King Fahad Medical City , Riyadh , Saudi Arabia
| | - Prafull Ghatage
- a Division of Gynecologic Oncology , Tom Baker Cancer Centre , Calgary , AB , Canada
| |
Collapse
|
36
|
Grenga I, Kwilas AR, Donahue RN, Farsaci B, Hodge JW. Inhibition of the angiopoietin/Tie2 axis induces immunogenic modulation, which sensitizes human tumor cells to immune attack. J Immunother Cancer 2015; 3:52. [PMID: 26579226 PMCID: PMC4647578 DOI: 10.1186/s40425-015-0096-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/21/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The angiopoietin/Tie2 pathway is an attractive target for cancer therapy due to its well-known role in regulating angiogenesis. Trebananib, a recombinant peptide-Fc fusion protein, or peptibody, that binds to angiopoietin-1 (Ang1) and Ang2 to block their interaction with the Tie2 receptor, is under active clinical investigation. We investigated whether suppressing the angiopoietin/Tie2 pathway, using the preclinical version of Trebananib (mL4-3 and L1-7(N)), could increase the sensitivity of human tumor cells to immune-mediated lysis through immunogenic modulation, which would make Trebananib a promising candidate for combination with immunotherapy. METHODS We assessed human carcinoma cells for expression and activation of Ang1 and Ang2 and their receptor tyrosine kinase Tie2. In vitro, we exposed tumor cell lines expressing Tie2 to the peptibodies mL4-3 and L1-7(N), which inhibit the binding of Ang1 and Ang2 to Tie2, and assessed the cells for changes in viability, proliferation, surface phenotype, and sensitivity to attack by antigen-specific cytotoxic T lymphocytes (CTLs). RESULTS Suppression of the angiopoietin/Tie2 pathway using mL4-3 and L1-7(N) had no effect on the proliferation or viability of tumor cells. However, these inhibitors markedly altered tumor cell phenotype, rendering tumor cells significantly more sensitive to antigen-specific CTL killing. ICAM-1 was shown to be mechanistically involved in these inhibitors' ability to sensitize tumor cells to immune-mediated attack by functional blocking studies. CONCLUSION Our findings provide a rationale for the combination of agents targeting the angiopoietin/Tie2 pathway with cancer immunotherapies.
Collapse
Affiliation(s)
- Italia Grenga
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Anna R Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| |
Collapse
|
37
|
Papadopoulos KP, Kelley RK, Tolcher AW, Razak ARA, Van Loon K, Patnaik A, Bedard PL, Alfaro AA, Beeram M, Adriaens L, Brownstein CM, Lowy I, Kostic A, Trail PA, Gao B, DiCioccio AT, Siu LL. A Phase I First-in-Human Study of Nesvacumab (REGN910), a Fully Human Anti-Angiopoietin-2 (Ang2) Monoclonal Antibody, in Patients with Advanced Solid Tumors. Clin Cancer Res 2015; 22:1348-55. [PMID: 26490310 DOI: 10.1158/1078-0432.ccr-15-1221] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/25/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Nesvacumab (REGN910) is a fully human immunoglobulin G1 (IgG1) monoclonal antibody that specifically binds and inactivates the Tie2 receptor ligand Ang2 with high affinity, but shows no binding to Ang1. The main objectives of this trial were to determine the safety, tolerability, dose-limiting toxicities (DLT), and recommended phase II dose (RP2D) of nesvacumab. EXPERIMENTAL DESIGN Nesvacumab was administered intravenously every two weeks with dose escalations from 1 to 20 mg/kg in patients with advanced solid tumors. RESULTS A total of 47 patients were treated with nesvacumab. No patients in the dose escalation phase experienced DLTs, therefore a maximum tolerated dose (MTD) was not reached. The most common nesvacumab-related adverse events were fatigue (23.4%), peripheral edema (21.3%), decreased appetite, and diarrhea (each 10.6%; all grade ≤ 2). Nesvacumab was characterized by linear kinetics and had a terminal half-life of 6.35 to 9.66 days in a dose-independent manner. Best response by RECIST 1.1 in 43 evaluable patients included 1 partial response (adrenocortical carcinoma) of 24 weeks duration. Two patients with hepatocellular carcinoma had stable disease (SD) > 16 weeks, with tumor regression and >50% decrease in α-fetoprotein. Analyses of putative angiogenesis biomarkers in serum and tumor biopsies were uninformative for treatment duration. CONCLUSIONS Nesvacumab safety profile was acceptable at all dose levels tested. Preliminary antitumor activity was observed in patients with treatment-refractory advanced solid tumors. On the basis of cumulative safety, antitumor activity, pharmacokinetic and pharmacodynamic data, the 20 mg/kg dose was determined to be the RP2D.
Collapse
Affiliation(s)
| | - Robin Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Anthony W Tolcher
- South Texas Accelerated Research Therapeutics (START), San Antonio, Texas
| | | | - Katherine Van Loon
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Amita Patnaik
- South Texas Accelerated Research Therapeutics (START), San Antonio, Texas
| | | | - Ariceli A Alfaro
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Muralidhar Beeram
- South Texas Accelerated Research Therapeutics (START), San Antonio, Texas
| | | | | | - Israel Lowy
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Ana Kostic
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Bo Gao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | |
Collapse
|
38
|
Pöss J, Fuernau G, Denks D, Desch S, Eitel I, de Waha S, Link A, Schuler G, Adams V, Böhm M, Thiele H. Angiopoietin-2 in acute myocardial infarction complicated by cardiogenic shock-a biomarker substudy of the IABP-SHOCK II-Trial. Eur J Heart Fail 2015; 17:1152-60. [DOI: 10.1002/ejhf.342] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/29/2015] [Accepted: 07/06/2015] [Indexed: 11/06/2022] Open
Affiliation(s)
- Janine Pöss
- Medical Clinic II/Cardiology/Angiology/Intensive Care Medicine; University Heart Centre Lübeck, University Hospital Schleswig-Holstein (UKSH); Ratzeburger Allee 160 23538 Lübeck Germany
| | - Georg Fuernau
- Medical Clinic II/Cardiology/Angiology/Intensive Care Medicine; University Heart Centre Lübeck, University Hospital Schleswig-Holstein (UKSH); Ratzeburger Allee 160 23538 Lübeck Germany
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Daniel Denks
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Steffen Desch
- Medical Clinic II/Cardiology/Angiology/Intensive Care Medicine; University Heart Centre Lübeck, University Hospital Schleswig-Holstein (UKSH); Ratzeburger Allee 160 23538 Lübeck Germany
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Ingo Eitel
- Medical Clinic II/Cardiology/Angiology/Intensive Care Medicine; University Heart Centre Lübeck, University Hospital Schleswig-Holstein (UKSH); Ratzeburger Allee 160 23538 Lübeck Germany
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Suzanne de Waha
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
- Herzzentrum, Segeberger Kliniken GmbH; Bad Segeberg Germany
| | - Andreas Link
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin; Universitätsklinikum des Saarlandes; Homburg/Saar Germany
| | - Gerhard Schuler
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Volker Adams
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin; Universitätsklinikum des Saarlandes; Homburg/Saar Germany
| | - Holger Thiele
- Medical Clinic II/Cardiology/Angiology/Intensive Care Medicine; University Heart Centre Lübeck, University Hospital Schleswig-Holstein (UKSH); Ratzeburger Allee 160 23538 Lübeck Germany
- Clinic for Internal Medicine/Cardiology; University of Leipzig - Heart Centre; Leipzig Germany
| |
Collapse
|
39
|
Lee JM, Trepel JB, Choyke P, Cao L, Sissung T, Houston N, Yu M, Figg WD, Turkbey IB, Steinberg SM, Lee MJ, Ivy SP, Liu JF, Matulonis UA, Kohn EC. CECs and IL-8 Have Prognostic and Predictive Utility in Patients with Recurrent Platinum-Sensitive Ovarian Cancer: Biomarker Correlates from the Randomized Phase-2 Trial of Olaparib and Cediranib Compared with Olaparib in Recurrent Platinum-Sensitive Ovarian Cancer. Front Oncol 2015; 5:123. [PMID: 26082895 PMCID: PMC4450585 DOI: 10.3389/fonc.2015.00123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/17/2015] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Olaparib (O), a polyADPribose polymerase (PARP) inhibitor, and cediranib (C), a VEGF receptor (VEGFR)1-3 inhibitor together had greater activity than O alone in women with recurrent platinum-sensitive ovarian cancer (OvCa). The objective of this study is to identify potential lead biomarker candidates for response to O + C in the setting of a multi-institutional phase II study of O with and without C in recurrent platinum-sensitive OvCa. METHODS A self-selected group of patients participated in a prospectively planned exploratory biomarker substudy of the randomized phase II study of O versus O + C. Whole blood for peripheral blood mononuclear cell (PBMC) and plasma isolation was collected prior to and on day 3 of treatment. Quantitation of circulating endothelial cells (CEC), IL-6, IL-8, VEGF, and soluble VEGFR-2 plasma concentrations, and polyADPribose (PAR) incorporation were performed. Single nucleotide polymorphism analysis of XRCC1 280H, R194W, and Q399R was done. Dynamic contrast-enhanced-magnetic resonance imaging (DCE-MRI) was performed at baseline and day 3 of treatment. Parameter changes were compared between the two arms using an exact Wilcoxon rank sum test. Kaplan-Meier and log-rank tests were used to examine survival outcome. RESULTS Thirteen patients elected to participate in the translational substudy, seven patients on O and six patients on O + C. Patients on O + C had a greater decrease in IL-8 concentration and larger CEC fold increase compared with those on O alone (p = 0.026, p = 0.032). The fold increase in CEC on day 3 was associated with duration of progression-free survival (PFS) (R (2) = 0.77, 95% CI 0.55-0.97, p < 0.001). IL-8 post-pretreatment changes correlate with PFS (p = 0.028). XRCC1 DNA polymorphisms were not related to PFS. All patients had reduction in PAR incorporation, and all except one had reduction in vascular flow on DCE-MRI. CONCLUSION Our exploratory correlative studies indicate that CEC and IL-8 changes may be predictive for response to O + C and prognostic in recurrent platinum-sensitive OvCa, requiring prospective validation.
Collapse
Affiliation(s)
| | | | | | - Liang Cao
- Center for Cancer Research , Bethesda, MD , USA
| | | | | | - Minshu Yu
- Center for Cancer Research , Bethesda, MD , USA
| | | | | | | | | | - S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute , Bethesda, MD , USA
| | - Joyce F Liu
- Dana-Farber Cancer Institute , Boston, MA , USA
| | | | | |
Collapse
|