1
|
Liang Q, Zhou L, Li Y, Liu J, Liu Y. Nano drug delivery system reconstruct tumour vasculature for the tumour vascular normalisation. J Drug Target 2021; 30:119-130. [PMID: 33960252 DOI: 10.1080/1061186x.2021.1927056] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The abnormal structure and function of blood vessels in the TME are obvious characteristics of the tumour. Abnormal blood vessels with high leakage support the occurrence of malignant tumours and increase the possibility of tumour cell invasion and metastasis. The formation of abnormal vascular also enhances immunosuppression and prevents the delivery of chemotherapy drugs to deeper tumours. Therefore, the normalisation of tumour blood vessels is a very promising approach to improve anti-tumour efficacy, aiming to restore the structural integrity of vessels and improve drug delivery efficiency and anti-tumour immunity. In this review, we have summarised strategies to improve cancer treatment that via nano drug delivery technology regulates the normalisation of tumour blood vessels. The treatment strategies related to the structure and function of tumour blood vessels such as angiogenesis factors, tumour-associated macrophages, tumour vascular endothelial cells, tumour-associated fibroblasts and immune checkpoints in the TME were mainly discussed. The normalisation of tumour blood vessels presents new opportunities and challenges for the more efficient delivery of nanoparticles to tumour tissues and cells and an innovative combination of treatments for cancer.
Collapse
Affiliation(s)
- Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Liyue Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yifan Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jinxia Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
2
|
Tarhriz V, Bandehpour M, Dastmalchi S, Ouladsahebmadarek E, Zarredar H, Eyvazi S. Overview of CD24 as a new molecular marker in ovarian cancer. J Cell Physiol 2018; 234:2134-2142. [PMID: 30317611 DOI: 10.1002/jcp.27581] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023]
Abstract
Ovarian cancer (OC) is the fifth leading cause of cancer-related death among women. The high mortality rate is due to lack of early symptoms, late diagnosis, limited treatment options, and also emerging of drug resistance. Todays, molecular markers have become promising in tumor-targeted therapy. Several molecular markers have been known in OC immunotherapy. Identification of the specific molecular markers with prognostic significance is interested. CD24 is a small sialoglycoprotein which is localized in lipid rafts through its glycosylphosphatidylinositol (GPI) anchor. It has been reported that CD24 is overexpressed in many cancers including OC. Also, CD24 is identified as a cancer stem cell marker in OC. The CD24 expression is associated with the development, invasion, and metastasis of cancer cells. The exact role of CD24 in cancer cells is not clearly understood. Recently, CD24 has been identified as an independent prognostic marker of survival in patients with OC. In this study, we reviewed the molecular targets in OC immune-targeted therapy and also presented an overview of the new molecular marker CD24 and its association with the OC by reviewing the recent literature.
Collapse
Affiliation(s)
- Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojgan Bandehpour
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Pharmacy, Near East University, Nicosia, North Cyprus, Turkey
| | - Elaheh Ouladsahebmadarek
- Women's Reproductive Health Research Center, Clinical Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Clinical Institute, Tabriz University of Medical Science, Tabriz, Iran
| | - Shirin Eyvazi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Biotechnology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Xing Y, Wang Y, Wang S, Wang X, Fan D, Zhou D, An J. Human cytomegalovirus infection contributes to glioma disease progression via upregulating endocan expression. Transl Res 2016; 177:113-126. [PMID: 27474433 DOI: 10.1016/j.trsl.2016.06.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/06/2016] [Accepted: 06/22/2016] [Indexed: 12/19/2022]
Abstract
The etiology of malignant glioma remains unclear. To examine the association between glioma and human cytomegalovirus (HCMV) infection and the possible mechanism through which HCMV contributes to malignant glioma, we investigated the expression of HCMV components and an angiogenesis marker, endocan, in 79 glioma specimens and 8 control brain samples. HCMV pp65 protein and DNA were detected in 65.8% (52 of 79) and 54.4% (43 of 79) of glioma specimens, respectively. The positive rate and expression levels of pp65 were significantly correlated with the glioma grades. The endocan expression was detected in 78.5% (62 of 79) of glioma specimens, and elevated endocan immunoreactivity was also significantly associated with high-grade glioma. The pp65 was predominantly detected and colocalized with endocan in the cytoplasm of tumor cells. Importantly, there was a significant positive correlation in detection rates between those 2 proteins. In control samples, neither HCMV pp65 nor endocan expression was detected. Moreover, the serum endocan levels in glioma patients were markedly higher than that in healthy subjects. In in vitro study, HCMV infection induced the expression of interleukin 6 and tumor necrosis factor-α in human glioblastoma U87 MG (U87) cells and human umbilical vein endothelial cells (HUVECs). Furthermore, elevated endocan levels were also observed in HCMV-infected U87 cells and HUVECs and antiviral treatment with ganciclovir reduced the endocan expression. These results suggest HCMV infection leads to glioma progression through an upregulation of endocan and the secretion of inflammatory cytokines. Thus, anti-HCMV treatment may represent a potentially novel therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Yan Xing
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yisong Wang
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shijie Wang
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xin Wang
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dongying Fan
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Jing An
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China; Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
4
|
Taurone S, Galli F, Signore A, Agostinelli E, Dierckx RAJO, Minni A, Pucci M, Artico M. VEGF in nuclear medicine: Clinical application in cancer and future perspectives (Review). Int J Oncol 2016; 49:437-47. [PMID: 27277340 DOI: 10.3892/ijo.2016.3553] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/28/2016] [Indexed: 11/06/2022] Open
Abstract
Clinical trials using antiangiogenic drugs revealed their potential against cancer. Unfortunately, a large percentage of patients does not yet benefit from this therapeutic approach highlighting the need of diagnostic tools to non-invasively evaluate and monitor response to therapy. It would also allow to predict which kind of patient will likely benefit of antiangiogenic therapy. Reasons for treatment failure might be due to a low expression of the drug targets or prevalence of other pathways. Molecular imaging has been therefore explored as a diagnostic technique of choice. Since the vascular endothelial growth factor (VEGF/VEGFR) pathway is the main responsible of tumor angiogenesis, several new drugs targeting either the soluble ligand or its receptor to inhibit signaling leading to tumor regression could be involved. Up today, it is difficult to determine VEGF or VEGFR local levels and their non-invasive measurement in tumors might give insight into the available target for VEGF/VEGFR-dependent antiangiogenic therapies, allowing therapy decision making and monitoring of response.
Collapse
Affiliation(s)
| | - Filippo Galli
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, 'Sapienza' University, Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, 'Sapienza' University, Rome, Italy
| | - Enzo Agostinelli
- Department of Biochemical Sciences 'A. Rossi Fanelli', 'Sapienza' University, Rome, Italy
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Antonio Minni
- Department of Sensory Organs, 'Sapienza' University, Rome, Italy
| | - Marcella Pucci
- Department of Sensory Organs, 'Sapienza' University, Rome, Italy
| | - Marco Artico
- Department of Sensory Organs, 'Sapienza' University, Rome, Italy
| |
Collapse
|
5
|
VEGF/NRP-1axis promotes progression of breast cancer via enhancement of epithelial-mesenchymal transition and activation of NF-κB and β-catenin. Cancer Lett 2016; 373:1-11. [PMID: 26805761 DOI: 10.1016/j.canlet.2016.01.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022]
Abstract
Autocrine vascular endothelial growth factor (VEGF) can regulate the survival and progression of cancers through its various receptors. But the mechanisms and mediators for these functions are largely uncovered, especially in breast cancer. We examined the potential roles and mechanisms of VEGF/neuropilin-1 (NRP-1) axis in regulating the tumorigenesis and metastasis of breast cancer and found the expression of VEGF and NRP-1 correlated with aggressiveness of breast cancer. Knockdown of VEGF or NRP-1 inhibited the proliferation, migration and invasion, but enhanced the apoptosis of MDA-MB-231 cells. In contrast, induction of NRP-1 over-expression promoted the proliferation, migration and invasion of MCF-7 cells. VEGF or NRP-1 silencing attenuated the epithelial-mesenchymal transition (EMT) process and the activation of NF-κBp65, but enhanced GSK-3β expression in MDA-MB-231 cells while NRP-1 over-expression reversed the effects in MCF-7 cells. Treatment with hVEGF165 did not change the inhibition in NRP-1 silencing MDA-MB-231 cells, but enhanced the aggressiveness of NRP-1 over-expressing MCF-7 cells. In addition, VEGF-silencing inhibited the growth and metastasis of implanted MDA-MB-231 tumors in vivo. Our novel data suggest that the positive regulation of the VEGF/NRP-1 axis on the tumorigenesis and metastasis of breast cancer may be associated with enhancing the EMT process and the NF-κB and β-catenin signaling. Hence, the VEGF/NRP-1 axis may be a valuable target for design of therapies for intervention of breast cancer.
Collapse
|
6
|
Singh S, Sharma B, Kanwar SS, Kumar A. Lead Phytochemicals for Anticancer Drug Development. FRONTIERS IN PLANT SCIENCE 2016; 7:1667. [PMID: 27877185 PMCID: PMC5099879 DOI: 10.3389/fpls.2016.01667] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 10/24/2016] [Indexed: 05/07/2023]
Abstract
Cancer is a serious concern at present. A large number of patients die each year due to cancer illnesses in spite of several interventions available. Development of an effective and side effects lacking anticancer therapy is the trending research direction in healthcare pharmacy. Chemical entities present in plants proved to be very potential in this regard. Bioactive phytochemicals are preferential as they pretend differentially on cancer cells only, without altering normal cells. Carcinogenesis is a complex process and includes multiple signaling events. Phytochemicals are pleiotropic in their function and target these events in multiple manners; hence they are most suitable candidate for anticancer drug development. Efforts are in progress to develop lead candidates from phytochemicals those can block or retard the growth of cancer without any side effect. Several phytochemicals manifest anticancer function in vitro and in vivo. This article deals with these lead phytomolecules with their action mechanisms on nuclear and cellular factors involved in carcinogenesis. Additionally, druggability parameters and clinical development of anticancer phytomolecules have also been discussed.
Collapse
|
7
|
Wyględowska-Promieńska D, Piotrowska-Gwóźdź A, Piotrowska-Seweryn A, Mazur-Piotrowska G. Combination of Aflibercept and Bromfenac Therapy in Age-Related Macular Degeneration: A Pilot Study Aflibercept and Bromfenac in AMD. Med Sci Monit 2015; 21:3906-12. [PMID: 26667262 PMCID: PMC4687982 DOI: 10.12659/msm.895977] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Among many protocols for treatment of exudative AMD, combined therapy of anti-VEGF agents and non-steroidal anti-inflammatory drugs (NSAIDs) seems to be an ideal alternative to monotherapy based on ranibizumab or bevacizumab. The aim of this study was to evaluate the effectiveness of aflibercept and bromfenac in the treatment of exudative AMD. MATERIAL AND METHODS The study was conducted on a group of 27 patients with exudative AMD who were administered intravitreal aflibercept and topical bromfenac (study group) once a month. Additional injections were administered up to 3 months after the third administration, depending on response to treatment. The control group consisted of subjects treated with aflibercept only. Visual acuity and anatomical outcomes in optical coherence tomography (OCT) were assessed at baseline visit, 4 months after the first dose, and 6 months after the start of the treatment. RESULTS Visual acuity improved over time in the study group and the differences between the groups were statistically significant. No statistically significant differences were found in OCT parameters. CONCLUSIONS Combined therapy of aflibercept and bromfenac in the treatment of wet AMD is more effective than single aflibercept therapy.
Collapse
Affiliation(s)
- Dorota Wyględowska-Promieńska
- Clinical Department of Ophthalmology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Anna Piotrowska-Gwóźdź
- Clinical Department of Ophthalmology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Piotrowska-Seweryn
- Clinical Department of Laryngology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Grażyna Mazur-Piotrowska
- Clinical Department of Ophthalmology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
8
|
Kadomatsu K, Bencsik P, Görbe A, Csonka C, Sakamoto K, Kishida S, Ferdinandy P. Therapeutic potential of midkine in cardiovascular disease. Br J Pharmacol 2014; 171:936-44. [PMID: 24286213 DOI: 10.1111/bph.12537] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/12/2013] [Accepted: 11/20/2013] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Ischaemic heart disease, stroke and their pathological consequences are life-threatening conditions that account for about half of deaths in developed countries. Pathology of these diseases includes cell death due to ischaemia/reperfusion injury, vascular stenosis and cardiac remodelling. The growth factor midkine plays a pivotal role in these events. Midkine shows an acute cytoprotective effect in ischaemia/reperfusion injury at least in part via its anti-apoptotic effect. Moreover, while midkine promotes endothelial cell proliferation, it also recruits inflammatory cells to lesions. These activities eventually enhance angiogenesis, thereby preventing cardiac tissue remodelling. However, midkine's activity in recruiting inflammatory cells into the vascular wall also triggers neointima formation, and consequently, vascular stenosis. Moreover, midkine is induced in cancer tissues where it enhances angiogenesis. Therefore, midkine may promote tumour formation through its angiogenic and anti-apoptotic activity. This review focuses on the roles of midkine in ischaemic cardiovascular disease and their pathological consequences, that is angiogenesis, vascular stenosis, and cardiac remodelling, and discusses the possible therapeutic potential of modulation of midkine in these diseases. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
9
|
Han K, Jin J, Maia M, Lowe J, Sersch MA, Allison DE. Lower exposure and faster clearance of bevacizumab in gastric cancer and the impact of patient variables: analysis of individual data from AVAGAST phase III trial. AAPS J 2014; 16:1056-63. [PMID: 24942210 PMCID: PMC4147052 DOI: 10.1208/s12248-014-9631-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/02/2014] [Indexed: 01/23/2023] Open
Abstract
Altered pharmacokinetics of antibody drugs has been reported in advanced gastric cancer (AGC). We aim to evaluate bevacizumab pharmacokinetics in AGC from the Phase III trial (AVAGAST), and explore the influence of patient variables. Bevacizumab concentrations (Cp) were measured in plasma samples taken following disease progression from 162 patients (7.5 mg/kg every 3 weeks). Predicted Cp [median and 90% prediction interval] was simulated using the population pharmacokinetic model established for other cancers (PPK model) and compared to observed Cp. Bevacizumab clearance was estimated using NONMEM and compared between subgroups. Patient characteristics of AGC are similar to other cancers except for lower body weight despite higher percentage of males. Eighty-five percent of observed Cp was below the median predicted Cp and 38% below the lower boundary of the 90% prediction interval. Median bevacizumab clearance in AGC was 4.5 versus 3 mL/day/kg in other cancers. Bevacizumab clearance was significantly faster (p < 0.05) in patients without gastrectomy (n = 42) or lower albumin. Clearance appeared to be faster in patients with lower total protein, higher ECOG scores, more metastatic sites, and poorer response. No significant difference in bevacizumab concentrations and clearance was observed between Asian and Non-Asian patients. AGC patients exhibited significantly lower bevacizumab exposure due to an approximate 50% increase in clearance versus other cancers. Bevacizumab is cleared faster in patients without prior gastrectomy. No significant difference in bevacizumab pharmacokinetics was observed between Asian and Non-Asian patients. The underlying mechanism for faster bevacizumab clearance in AGC is unknown and warrants further research.
Collapse
Affiliation(s)
- Kelong Han
- Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA,
| | | | | | | | | | | |
Collapse
|
10
|
Wyględowska-Promieńska D, Piotrowska-Gwóźdź A, Piotrowska-Seweryn A, Mazur-Piotrowska G, Rokicki W. Combination of bevacizumab and bromfenac therapy in age-related macular degeneration: a pilot study. Med Sci Monit 2014; 20:1168-75. [PMID: 25006692 PMCID: PMC4102605 DOI: 10.12659/msm.890671] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background According to recent studies, the newest strategy for the treatment of exudative age-related macular degeneration is to combine anti-VEGF agents with non-steroid anti-inflammatory drugs (NSAIDs) such as nepafenac and bromfenac to decrease the frequency of intravitreal injections. Since most research has focused on ranibizumab, the aim of this study is to evaluate whether an alternative drug such as bevacizumab could lead to similar outcomes. Material/Methods The study was conducted on a group of 26 patients who were administered intravitreal bevacizumab and topical bromfenac (study group) and 26 patients with single bevacizumab therapy (control group). Cases that were not qualified for ranibizumab therapy were included in the study group. Results The study revealed that the visual acuity and parameters observed in OCT improved more in the study group than in the control group. However, the correlations between the above factors and the frequency of intravitreal injections were statistically significant only in visual acuity. Conclusions We recommend the combined therapy of bevacizumab and bromfenac as an alternative and beneficial method of treatment in patients with exudative AMD who do not qualify for ranibizumab therapy. This combined therapy might efficiently reduce the number of intravitreal injections of bevacizumab.
Collapse
Affiliation(s)
| | | | | | | | - Wojciech Rokicki
- Clinical Department of Ophthalmology, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
11
|
Liu Y, Tian H, Blobe GC, Theuer CP, Hurwitz HI, Nixon AB. Effects of the combination of TRC105 and bevacizumab on endothelial cell biology. Invest New Drugs 2014; 32:851-9. [PMID: 24994097 PMCID: PMC4169868 DOI: 10.1007/s10637-014-0129-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 06/12/2014] [Indexed: 11/26/2022]
Abstract
Endoglin, or CD105, is a cell membrane glycoprotein that is overexpressed on proliferating endothelial cells (EC), including those found in malignancies and choroidal neovascularization. Endoglin mediates the transition from quiescent endothelium, characterized by the relatively dominant state of Smad 2/3 phosphorylation, to active angiogenesis by preferentially phosphorylating Smad 1/5/8. The monoclonal antibody TRC105 binds endoglin with high avidity and is currently being tested in phase 1b and phase 2 clinical trials. In this report, we evaluated the effects of TRC105 on primary human umbilical vascular endothelial cells (HUVEC) as a single agent and in combination with bevacizumab. As single agents, both TRC105 and bevacizumab efficiently blocked HUVEC tube formation, and the combination of both agents achieved even greater levels of inhibition. We further assessed the effects of each drug on various aspects of HUVEC function. While bevacizumab was observed to inhibit HUVEC viability in nutrient-limited medium, TRC105 had little effect on HUVEC viability, either alone or in combination with bevacizumab. Additionally, both drugs inhibited HUVEC migration and induced apoptosis. At the molecular level, TRC105 treatment of HUVEC lead to decreased Smad 1/5/8 phosphorylation in response to BMP-9, a primary ligand for endoglin. Together, these results indicate that TRC105 acts as an effective anti-angiogenic agent alone and in combination with bevacizumab.
Collapse
Affiliation(s)
- Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Box 2631, Durham, NC 27710 USA
| | - Hongyu Tian
- Department of Medicine, Duke University Medical Center, Box 2631, Durham, NC 27710 USA
| | - Gerard C. Blobe
- Department of Medicine, Duke University Medical Center, Box 2631, Durham, NC 27710 USA
| | | | - Herbert I. Hurwitz
- Department of Medicine, Duke University Medical Center, Box 2631, Durham, NC 27710 USA
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Box 2631, Durham, NC 27710 USA
| |
Collapse
|
12
|
Wållberg H, Ståhl S. Design and evaluation of radiolabeled tracers for tumor imaging. Biotechnol Appl Biochem 2014; 60:365-83. [PMID: 24033592 DOI: 10.1002/bab.1111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/20/2013] [Indexed: 12/22/2022]
Abstract
The growing understanding of tumor biology and the identification of tumor-specific genetic and molecular alterations, such as the overexpression of membrane receptors and other proteins, allows for personalization of patient management using targeted therapies. However, this puts stringent demands on the diagnostic tools used to identify patients who are likely to respond to a particular treatment. Radionuclide molecular imaging is a promising noninvasive method to visualize and characterize the expression of such targets. A number of different proteins, from full-length antibodies and their derivatives to small scaffold proteins and peptide receptor-ligands, have been applied to molecular imaging, each demonstrating strengths and weaknesses. Here, we discuss the concept of molecular targeting and, in particular, molecular imaging of cancer-associated targets. Additionally, we describe important biotechnological considerations and desired features when designing and developing tracers for radionuclide molecular imaging.
Collapse
Affiliation(s)
- Helena Wållberg
- Division of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|
13
|
Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014; 351:13-22. [PMID: 24836189 DOI: 10.1016/j.canlet.2014.05.002] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/31/2014] [Accepted: 05/01/2014] [Indexed: 01/01/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer therapies mainly focus on mass cell killing without high specificity and often cause severe side effects and toxicities. Peptides are a novel class of anticancer agents that could specifically target cancer cells with lower toxicity to normal tissues, which will offer new opportunities for cancer prevention and treatment. Anticancer peptides face several therapeutic challenges. In this review, we present the sources and mechanisms of anticancer peptides and further discuss modification strategies to improve the anticancer effects of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanfeng Gao
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanming Qi
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanfang Ma
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanzhang Li
- College of Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
14
|
Molecularly targeted therapies in multiple myeloma. LEUKEMIA RESEARCH AND TREATMENT 2014; 2014:976567. [PMID: 24829804 PMCID: PMC4009206 DOI: 10.1155/2014/976567] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 01/22/2023]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients will eventually relapse or become refractory to the treatments. Although the treatments have improved, the major problem in MM is the resistance to therapy. Novel agents are currently in development for the treatment of relapsed/refractory MM, including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, cell signaling targeted therapies, and strategies targeting the tumor microenvironment. We have previously reviewed in detail the contemporary immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies therapies for MM. Therefore, in this review, we focused on the role of molecular targeted therapies in the treatment of relapsed/refractory multiple myeloma, including cell signaling targeted therapies (HDAC, PI3K/AKT/mTOR, p38 MAPK, Hsp90, Wnt, Notch, Hedgehog, and cell cycle) and strategies targeting the tumor microenvironment (hypoxia, angiogenesis, integrins, CD44, CXCR4, and selectins). Although these novel agents have improved the therapeutic outcomes for MM patients, further development of new therapeutic agents is warranted.
Collapse
|
15
|
Parametric response maps of perfusion MRI may identify recurrent glioblastomas responsive to bevacizumab and irinotecan. PLoS One 2014; 9:e90535. [PMID: 24675671 PMCID: PMC3968002 DOI: 10.1371/journal.pone.0090535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/02/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Perfusion weighted imaging (PWI) can be used to measure key aspects of tumor vascularity in vivo and recent studies suggest that perfusion imaging may be useful in the early assessment of response to angiogenesis inhibitors. Aim of this work is to compare Parametric Response Maps (PRMs) with the Region Of Interest (ROI) approach in the analysis of tumor changes induced by bevacizumab and irinotecan in recurrent glioblastomas (rGBM), and to evaluate if changes in tumor blood volume measured by perfusion MRI may predict clinical outcome. METHODS 42 rGBM patients with KPS ≥ 50 were treated until progression, as defined by MRI with RANO criteria. Relative cerebral blood volume (rCBV) variation after 8 weeks of treatment was calculated through semi-automatic ROI placement in the same anatomic region as in baseline. Alternatively, rCBV variations with respect to baseline were calculated into the evolving tumor region using a voxel-by-voxel difference. PRMs were created showing where rCBV significantly increased, decreased or remained unchanged. RESULTS An increased blood volume in PRM (PRMCBV+) higher than 18% (first quartile) after 8 weeks of treatment was associated with increased progression free survival (PFS; 24 versus 13 weeks, p = 0.045) and overall survival (OS; 38 versus 25 weeks, p = 0.016). After 8 weeks of treatment ROI analysis showed that mean rCBV remained elevated in non responsive patients (4.8 ± 0.9 versus 5.1 ± 1.2, p = 0.38), whereas decreased in responsive patients (4.2 ± 1.3 versus 3.8 ± 1.6 p = 0.04), and re-increased progressively when patients approached tumor progression. CONCLUSIONS Our data suggest that PRMs can provide an early marker of response to antiangiogenic treatment and warrant further confirmation in a larger cohort of GBM patients.
Collapse
|
16
|
Tabouret E, Boudouresque F, Barrie M, Matta M, Boucard C, Loundou A, Carpentier A, Sanson M, Metellus P, Figarella-Branger D, Ouafik L, Chinot O. Association of matrix metalloproteinase 2 plasma level with response and survival in patients treated with bevacizumab for recurrent high-grade glioma. Neuro Oncol 2013; 16:392-9. [PMID: 24327581 DOI: 10.1093/neuonc/not226] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A predictive marker of bevacizumab activity is an unmet medical need. We evaluated the predictive value of selected circulating prebiomarkers involved in neoangiogenesis and invasion on patient outcome in recurrent high-grade glioma treated with bevacizumab. METHODS Analyzed in plasma were a set of 11 prebiomakers of interest (vascular endothelial growth factor receptor [VEGF]; VEGF receptor 2; basic fibroblast growth factor; stromal cell derived factor 1; placenta growth factor; urokinase-type plasminogen activator; plasminogen activator inhibitor 1; matrix metalloproteinases 2, 7, and 9; and adrenomedulline), using ELISA, at baseline and 2 weeks after bevacizumab initiation in a prospective cohort of 26 patients (Cohort 1). Correlations were validated in a separate retrospective cohort (Cohort 2; n = 50) and tested in cohort patients treated with cytotoxic agents without bevacizumab (Cohort 3; n = 34). Dosages were correlated to objective response, progression-free survival (PFS), and overall survival (OS). RESULTS In Cohort 1, high MMP2 baseline level was associated with a probability of objective response of 83.3% versus 15.4% for low MMP2 level (P = .001). In multivariate analysis, baseline level of MMP2 correlated with PFS (hazard ratio, 3.92; 95% confidence interval [CI]:1.46-10.52; P = .007) and OS (hazard ratio, 4.62; 95% CI: 1.58-13.53; P = .005), as decrease of VEGF (P = .038 for PFS and P = .013 for OS) and MMP9 (P = .016 for PFS and P = .025 for OS). In Cohort 2, MMP2, but not MMP9, confirmed its predictive significance. In Cohort 3, no association was found between MMP2, MMP9, and outcome. CONCLUSION In patients with recurrent high-grade glioma treated with bevacizumab, but not with cytotoxic agent, high MMP2 plasma levels are associated with prolonged tumor control and survival. MMP2 should be tested in randomized clinical trials that evaluate bevacizumab efficacy, and its biological role reassessed.
Collapse
Affiliation(s)
- Emeline Tabouret
- Aix-Marseille Université, INSERM CRO2, Marseille, France (E.T., F.B., D.F.-B., L.O., O.C.); Assistance Publique-Hôpitaux de Marseille (AP-HM), CHU Timone, Service de Neuro-Oncologie, Marseille, France (E.T., M.B., M.M., C.B.); Aix-Marseille Université, Public Health Research Unit-EA3279 AP-HM, Marseille, France (A.L.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France (A.C.); Université Paris Descartes, Laboratoire de Recherches Biochirurgicales, Hôpital Européen Georges Pompidou, Paris, France (A.C.); INSERM U 975, CNRS UMR 7225, Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moëlle Épinière UMR-S975, Paris, France (M.S.); AP-HP, Service Neurologie 2, Groupe Hospitalier Pitié-Salpêtrière, Paris, France (M.S.); AP-HM, CHU Timone, Service de Neurochirurgie, Marseille, France (P.M.); AP-HM, CHU Timone, Service d'Anatomopathologie, Marseille, France (D.F.-B.); CHU Nord, Laboratoire de Transfert, Marseille (L.O.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zorena K, Raczyńska D, Raczyńska K. Biomarkers in diabetic retinopathy and the therapeutic implications. Mediators Inflamm 2013; 2013:193604. [PMID: 24311895 PMCID: PMC3839118 DOI: 10.1155/2013/193604] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/09/2013] [Indexed: 02/06/2023] Open
Abstract
The main problem both in type 1 (T1DM) and type 2 (T2DM) diabetes is the development of chronic vascular complications encompassing micro- as well as macrocirculation. Chronic complications lower the quality of life, lead to disability, and are the cause of premature death in DM patients. One of the chronic vascular complications is a diabetic retinopathy (DR) which leads to a complete loss of sight in DM patients. Recent trials show that the primary cause of diabetic retinopathy is retinal neovascularization caused by disequilibrium between pro- and antiangiogenic factors. Gaining knowledge of the mechanisms of action of factors influencing retinal neovascularization as well as the search for new, effective treatment methods, especially in advanced stages of DR, puts special importance on research concentrating on the implementation of biological drugs in DR therapy. At present, it is antivascular endothelial growth factor and antitumor necrosis factor that gain particular significance.
Collapse
Affiliation(s)
- Katarzyna Zorena
- Department of Clinical and Experimental Endocrinology, Institute of Maritime and Tropical Medicine, Medical University of Gdańsk, Powstania Styczniowego 9b, 81-519 Gdynia, Poland
| | - Dorota Raczyńska
- Department of Anesthesiology and Intensive Care Medicine, Medical University of Gdańsk, Poland
- Department and Clinic of Ophthalmology, Medical University of Gdańsk, Poland
| | - Krystyna Raczyńska
- Department and Clinic of Ophthalmology, Medical University of Gdańsk, Poland
| |
Collapse
|
18
|
Bendell JC, Gordon MS, Hurwitz HI, Jones SF, Mendelson DS, Blobe GC, Agarwal N, Condon CH, Wilson D, Pearsall AE, Yang Y, McClure T, Attie KM, Sherman ML, Sharma S. Safety, pharmacokinetics, pharmacodynamics, and antitumor activity of dalantercept, an activin receptor-like kinase-1 ligand trap, in patients with advanced cancer. Clin Cancer Res 2013; 20:480-9. [PMID: 24173543 DOI: 10.1158/1078-0432.ccr-13-1840] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The angiogenesis inhibitor dalantercept (formerly ACE-041) is a soluble form of activin receptor-like kinase-1 (ALK1) that prevents activation of endogenous ALK1 by bone morphogenetic protein-9 (BMP9) and BMP10 and exhibits antitumor activity in preclinical models. This first-in-human study of dalantercept evaluated its safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity in adults with advanced solid tumors. EXPERIMENTAL DESIGN Patients in dose-escalating cohorts received dalantercept subcutaneously at one of seven dose levels (0.1-4.8 mg/kg) every 3 weeks until disease progression. Patients in an expansion cohort received dalantercept at 0.8 or 1.6 mg/kg every 3 weeks until disease progression. RESULTS In 37 patients receiving dalantercept, the most common treatment-related adverse events were peripheral edema, fatigue, and anemia. Edema and fluid retention were dose-limiting toxicities and responded to diuretic therapy. No clinically significant, treatment-related hypertension, proteinuria, gross hemorrhage, or gastrointestinal perforations were observed. One patient with refractory squamous cell cancer of the head and neck had a partial response, and 13 patients had stable disease according to RECISTv1.1, eight of whom had prolonged periods (≥12 weeks) of stable disease. Correlative pharmacodynamic markers included tumor metabolic activity and tumor blood flow, which decreased from baseline in 63% and 82% of evaluable patients, respectively, and telangiectasia in eight patients. CONCLUSION Dalantercept was well-tolerated at doses up to 1.6 mg/kg, with a safety profile distinct from inhibitors of the VEGF pathway. Dalantercept displayed promising antitumor activity in patients with advanced refractory cancer, and multiple phase II studies are underway.
Collapse
Affiliation(s)
- Johanna C Bendell
- Authors' Affiliations: Sarah Cannon Research Institute, Nashville, Tennessee; Pinnacle Oncology Hematology, Scottsdale, Arizona; Duke University Medical Center, Durham, North Carolina; Huntsman Cancer Institute, Salt Lake City, Utah; and Acceleron Pharma, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hsu HW, Wall NR, Hsueh CT, Kim S, Ferris RL, Chen CS, Mirshahidi S. Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol 2013; 50:19-26. [PMID: 24269532 DOI: 10.1016/j.oraloncology.2013.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/24/2013] [Accepted: 10/02/2013] [Indexed: 02/02/2023]
Abstract
Tumor angiogenesis is a hallmark of advanced cancers and promotes invasion and metastasis. Over 90% of head and neck squamous cell carcinomas (HNSCC) express angiogenic factors such as vascular endothelial growth factor (VEGF). Several preclinical studies support the prognostic implications of angiogenic markers for HNSCC and currently this is an attractive treatment target in solid tumors. Since radiotherapy is one of the most commonly used treatments for HNSCC, it is imperative to identify the interactions between antiangiogenic therapy and radiotherapy, and to develop combination therapy to improve clinical outcome. The mechanisms between antiangiogenic agents and ionizing radiation are complicated and involve many interactions between the vasculature, tumor stroma and tumor cells. The proliferation and metastasis of tumor cells rely on angiogenesis/blood vessel formation. Rapid growing tumors will cause hypoxia, which up-regulates tumor cell survival factors, such as hypoxia-inducing factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), giving rise to more tumor proliferation, angiogenesis and increased radioresistance. Thus, agents that target tumor vasculature and new tumor vessel formation can modulate the tumor microenvironment to improve tumor blood flow and oxygenation, leading to enhanced radiosensitivity. In this review, we discuss the mechanisms of how antiangiogenic therapies improve tumor response to radiation and data that support this combination strategy as a promising method for the treatment of HNSCC in the future.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Department of Pharmacology, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA
| | - Nathan R Wall
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA, USA
| | - Chung-Tsen Hsueh
- Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Seungwon Kim
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien-Shing Chen
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA; Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Saied Mirshahidi
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
20
|
Jeltsch M, Leppänen VM, Saharinen P, Alitalo K. Receptor tyrosine kinase-mediated angiogenesis. Cold Spring Harb Perspect Biol 2013; 5:5/9/a009183. [PMID: 24003209 DOI: 10.1101/cshperspect.a009183] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The endothelial cell is the essential cell type forming the inner layer of the vasculature. Two families of receptor tyrosine kinases (RTKs) are almost completely endothelial cell specific: the vascular endothelial growth factor (VEGF) receptors (VEGFR1-3) and the Tie receptors (Tie1 and Tie2). Both are key players governing the generation of blood and lymphatic vessels during embryonic development. Because the growth of new blood and lymphatic vessels (or the lack thereof) is a central element in many diseases, the VEGF and the Tie receptors provide attractive therapeutic targets in various diseases. Indeed, several drugs directed to these RTK signaling pathways are already on the market, whereas many are in clinical trials. Here we review the VEGFR and Tie families, their involvement in developmental and pathological angiogenesis, and the different possibilities for targeting them to either block or enhance angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Michael Jeltsch
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | | | |
Collapse
|
21
|
FUJIWARA YUTAKA, CHAYAHARA NAOKO, MUKOHARA TORU, KIYOTA NAOMI, TOMIOKA HIDEO, FUNAKOSHI YOHEI, MINAMI HIRONOBU. Hypothyroidism in patients with colorectal carcinoma treated with fluoropyrimidines. Oncol Rep 2013; 30:1802-6. [DOI: 10.3892/or.2013.2644] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/10/2013] [Indexed: 11/06/2022] Open
|
22
|
Chinot OL. Bevacizumab-based therapy in relapsed glioblastoma: rationale and clinical experience to date. Expert Rev Anticancer Ther 2013; 12:1413-27. [PMID: 23249106 DOI: 10.1586/era.12.128] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Relapsed glioblastoma (GBM) has an extremely poor prognosis and remains an invariably fatal disease, with a median overall survival of 6-7 months. Despite numerous clinical trials over the past 20-30 years, treatment options for relapsed GBM remain limited. In recent years, significant research efforts have focused on the use of antiangiogenic therapies for the treatment of GBM. Bevacizumab is a humanized monoclonal antibody that specifically inhibits the proangiogenic VEGF, with well-established clinical efficacy in a number of solid malignancies, which is now under investigation for the treatment of GBM. In this review, we discuss the available data regarding bevacizumab-based therapy in relapsed GBM, highlighting its potential and ongoing challenges in this difficult-to-treat disease.
Collapse
Affiliation(s)
- Olivier L Chinot
- Aix-Marseille University, Assistance Publique-Hopitaux de Marseille, Centre Hospitalo-Universitaire Timone, Service de Neuro-Oncologie, 13008 Marseille, France.
| |
Collapse
|
23
|
Noble P, Vyas M, Al-Attar A, Durrant S, Scholefield J, Durrant L. High levels of cleaved caspase-3 in colorectal tumour stroma predict good survival. Br J Cancer 2013; 108:2097-105. [PMID: 23591201 PMCID: PMC3670501 DOI: 10.1038/bjc.2013.166] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: The primary aim was to determine the prognostic significance of apoptosis in colorectal tumour cells and tumour-associated stroma. A secondary aim was to determine whether apoptosis was related to immune surveillance. Methods: Immunohistochemistry was performed using monoclonal antibodies recognising cleaved caspase-3 (CC3), cleaved poly (ADP-ribose) polymerase (PARP), p53, Bcl2, MHC-II, B cells (CD16), macrophages (CD68) and T cells (CD3), on a tissue microarray of 462 colorectal tumours. Results: Kaplan–Meier analysis demonstrated that patients with high expression of CC3 in the tumour or CC3 or cleaved PARP in tumour-associated stroma have a good prognosis. This suggests that tumour stroma is promoting tumourigenesis and that high levels of death within the stroma breaks this link. CC3 levels in the tumour correlated with cleaved PARP and MHC-II expression but not with CD16, CD68, CD3, p53 or Bcl2 expression. CC3 levels on tumour-associated stroma also correlated with cleaved PARP and MHC-II expression but not with CD16, CD68, CD3, p53 or Bcl2 expression. Tumour cells express MHC-II in response to IFN-γ, suggesting that this may be one of the initiators of apoptosis within the good prognosis tumours. Although 73% of the MHC-II-positive tumour had high levels of apoptosis, many tumours had high levels of apoptosis in the absence of MHC-II, implying that this is only one of many causes of apoptosis within tumours. On multivariate analysis, using Cox's proportional hazards model, tumour stage, vascular invasion and expression of CC3 in tumour-associated stroma were shown to be independent markers of prognosis. Conclusion: This study shows that a high level of apoptosis within colorectal tumour-associated stroma is an independent marker of good prognosis.
Collapse
Affiliation(s)
- P Noble
- Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Hucknall Road, Nottingham NG5 1PB, UK
| | | | | | | | | | | |
Collapse
|
24
|
Chiu CL, Morgan CT, Lupton SJ, Lind JM. Parent of origin influences the cardiac expression of vascular endothelial growth factor (Vegfa). BMC MEDICAL GENETICS 2013; 14:43. [PMID: 23560444 PMCID: PMC3626619 DOI: 10.1186/1471-2350-14-43] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 03/25/2013] [Indexed: 11/10/2022]
Abstract
Background Vascular endothelial growth factor A (VEGFA) is a major regulator of both physiological and pathological angiogenesis. Associations between polymorphisms in VEGFA and complex disease have been inconsistent. The parent from whom the allele was inherited may account for these inconsistencies. This study examined the parent of origin effect on the expression of murine Vegfa. Methods Two homozygous, inbred mouse strains A/J (AJ) and 129x1/SvJ (129) were crossed to produce reciprocal AJ129 and 129AJ offspring, respectively. RNA was extracted from cardiac tissue of 6 week old male (n = 8) and female (n = 8) parental, and male and female F1 offspring mice (AJ129 n = 8 and 129AJ n = 8). Vegfa and Hif1a expression levels were measured by qPCR and compared between the F1 offspring from the reciprocal crosses. Results We found significant differences in the expression of Vegfa in F1 offspring (AJ129 and 129AJ mice) of the reciprocal crosses between AJ and 129 mice. Offspring of male AJ mice had significantly higher expression of Vegfa than offspring of male 129 mice (p = 0.006). This difference in expression was not the result of preferential allele expression (allelic imbalance). Expression of Hif1a, a transcriptional regulator of Vegfa expression, was also higher in F1 offspring of an AJ father (p = 0.004). Conclusion Differences in Vegfa and Hif1a gene expression are likely the result of an upstream angiogenic regulator gene that is influenced by the parent of origin. These results highlight the importance of including inheritance information, such as parent of origin, when undertaking allelic association studies.
Collapse
Affiliation(s)
- Christine L Chiu
- University of Western Sydney, School of Medicine, Penrith, NSW 2751, Locked Bag 1797, Australia.
| | | | | | | |
Collapse
|
25
|
Okamoto K, Okamoto I, Miyazaki M, Tanaka K, Kaneda H, Nakagawa K. Bronchoscopic findings for bevacizumab-related pulmonary hemorrhage in advanced non-small cell lung cancer. Invest New Drugs 2013; 31:1364-6. [PMID: 23525757 DOI: 10.1007/s10637-013-9951-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/11/2013] [Indexed: 10/27/2022]
Abstract
We report contemporaneous bronchoscopic findings for a case of bevacizumab-related pulmonary hemorrhage in a patient with advanced non-small cell lung cancer (NSCLC). Flexible bronchoscopy at diagnosis revealed abnormal capillary dilation that was suggestive of endobronchial involvement at the primary tumor location. The patient developed massive hemoptysis despite of marked tumor shrinkage achieved by bevacizumab-containing chemotherapy. Emergency flexible bronchoscopy for hemoptysis suggested that the location of the primary tumor was the source of bleeding. Subsequent follow-up flexible bronchoscopy revealed an ulcerative mucosal-like lesion associated with a white necrotic substance as well as attenuation of the dilation of submucosal vessels compared with that apparent at diagnosis. Our case report highlights the potential mechanistic insights into bevacizumab-related bleeding and importance of performing bronchoscopy at diagnosis in NSCLC patients, given that abnormal bronchoscopic findings may be a risk factor for bleeding.
Collapse
Affiliation(s)
- Kunio Okamoto
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Li J, Gupta M, Jin D, Xin Y, Visich J, Allison DE. Characterization of the long-term pharmacokinetics of bevacizumab following last dose in patients with resected stage II and III carcinoma of the colon. Cancer Chemother Pharmacol 2012; 71:575-80. [PMID: 23228985 DOI: 10.1007/s00280-012-2031-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022]
Abstract
PURPOSE The study characterizes the long-term pharmacokinetics (PK) following last dose of bevacizumab as adjuvant therapy in patients with resected stage II and III colon carcinoma in a Phase III clinical study (AVF3077s). METHODS Patients in AVF3077s received bevacizumab (5 mg/kg every 2 weeks) as adjuvant therapy for 1 year. Following the last dose bevacizumab concentration, data at 3 and 6 months were used to characterize long-term bevacizumab PK based on the population-modeling approach. RESULTS The long-term bevacizumab PK were consistent with previously reported results based on short-term bevacizumab PK. The clearance (CL), central volume of distribution (V(1)), intercompartmental clearance (Q), and the peripheral volume of distribution (V(2)) were 214 mL/day, 2,830 mL, 636 mL/day, and 2,490 mL, which correspond to a disposition and elimination half-life of 1.33 and 19.1 days, respectively. The empirical Bayes estimates of median post-treatment bevacizumab drug levels at 3 and 6 months were 6.14 and 0.23 μg/mL, respectively. For test covariates, the change in CL and V(1) of bevacizumab was less than 20% of the typical value. Body weight is the important covariate explaining the inter-individual variability on CL and V(1). CONCLUSIONS Long-term bevacizumab PK in this study was predictable based on short-term PK data from metastatic settings in other tumor types. An exploratory analysis demonstrated no apparent association of the tested covariates with bevacizumab PK. Further, the extended serum persistence of bevacizumab following last dose should be considered in clinical study designs and post-treatment evaluations that may be affected by bevacizumab.
Collapse
Affiliation(s)
- Jing Li
- Genentech, Inc., South San Francisco, CA, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Sideras K, Dueck AC, Hobday TJ, Rowland KM, Allred JB, Northfelt DW, Lingle WL, Behrens RJ, Fitch TR, Nikcevich DA, Perez EA. North central cancer treatment group (NCCTG) N0537: phase II trial of VEGF-trap in patients with metastatic breast cancer previously treated with an anthracycline and/or a taxane. Clin Breast Cancer 2012; 12:387-91. [PMID: 23083501 PMCID: PMC3586936 DOI: 10.1016/j.clbc.2012.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 08/14/2012] [Accepted: 09/13/2012] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Angiogenesis is an established target for the treatment of MBC. Aflibercept (VEGF-Trap) is a humanized fusion protein, which binds VEGF-A, VEGF-B, and PIGF-1 and -2. PATIENTS AND METHODS A 2-stage phase II study with primary end points of confirmed tumor response and 6-month progression-free survival (PFS). If either end point was promising after the initial 21 patients, an additional 20 patients would be enrolled. Measurable disease, <2 previous chemotherapy treatments, previous anthracycline or taxane therapy, and Eastern Cooperative Oncology Group performance status of 0 or 1 were required. Aflibercept was given at a dose of 4 mg/kg intravenous every 14 days. RESULTS Twenty-one patients were enrolled; 71% had visceral disease, 57% were estrogen receptor negative, 19% had HER2(+) disease with previous trastuzumab treatment, and 33% had 2 previous chemotherapy regimens. Partial response rate was 4.8% (95% confidence interval [CI], 0.1%-23.8%) and 6-month PFS was 9.5% (95% CI, 1.2%-30.4%). Neither primary end point met efficacy goals and the study was terminated. A median of 3 cycles was given. Median PFS was 2.4 months. Common grade 3 or 4 adverse events were hypertension (33%), fatigue (19%), dyspnea (14%), and headache (14%). Two cases of severe left ventricular dysfunction were noted. CONCLUSIONS Aflibercept did not meet efficacy goals in patients previously treated with MBC. Toxicity was as expected for anti-VEGF therapy.
Collapse
|
28
|
Kieran MW, Kalluri R, Cho YJ. The VEGF pathway in cancer and disease: responses, resistance, and the path forward. Cold Spring Harb Perspect Med 2012; 2:a006593. [PMID: 23209176 DOI: 10.1101/cshperspect.a006593] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antiangiogenesis was proposed as a novel target for the treatment of cancer 40 years ago. Since the original hypothesis put forward by Judah Folkman in 1971, factors that mediate angiogenesis, their cellular targets, many of the pathways they signal, and inhibitors of the cytokines and receptors have been identified. Vascular endothelial growth factor (VEGF) is the most prominent among the angiogenic cytokines and is believed to play a central role in the process of neovascularization, both in cancer as well as other inflammatory diseases. This article reviews the biology of VEGF and its receptors, the use of anti-VEGF approaches in clinical disease, the toxicity of these therapies, and the resistance mechanisms that have limited the activity of these agents when used as monotherapy.
Collapse
Affiliation(s)
- Mark W Kieran
- Department of Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | | | | |
Collapse
|
29
|
Dolloff NG, Talamo G. Targeted Therapy of Multiple Myeloma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 779:197-221. [DOI: 10.1007/978-1-4614-6176-0_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Jiang T, Zhuang J, Duan H, Luo Y, Zeng Q, Fan K, Yan H, Lu D, Ye Z, Hao J, Feng J, Yang D, Yan X. CD146 is a coreceptor for VEGFR-2 in tumor angiogenesis. Blood 2012; 120:2330-2339. [PMID: 22718841 DOI: 10.1182/blood-2012-01-406108] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD146 is a novel endothelial biomarker and plays an essential role in angiogenesis; however, its role in the molecular mechanism underlying angiogenesis remains poorly understood. In the present study, we show that CD146 interacts directly with VEGFR-2 on endothelial cells and at the molecular level and identify the structural basis of CD146 binding to VEGFR-2. In addition, we show that CD146 is required in VEGF-induced VEGFR-2 phosphorylation, AKT/p38 MAPKs/NF-κB activation, and thus promotion of endothelial cell migration and microvascular formation. Furthermore, we show that anti-CD146 AA98 or CD146 siRNA abrogates all VEGFR-2 activation induced by VEGF. An in vivo angiogenesis assay showed that VEGF-promoted microvascular formation was impaired in the endothelial conditional knockout of CD146 (CD146(EC-KO)). Our animal experiments demonstrated that anti-CD146 (AA98) and anti-VEGF (bevacizumab) have an additive inhibitory effect on xenografted human pancreatic and melanoma tumors. The results of the present study suggest that CD146 is a new coreceptor for VEGFR-2 and is therefore a promising target for blocking tumor-related angiogenesis.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- CD146 Antigen/chemistry
- CD146 Antigen/genetics
- CD146 Antigen/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Female
- Humans
- Mice
- Mice, Knockout
- Mice, Nude
- Molecular Targeted Therapy
- Mutant Proteins/antagonists & inhibitors
- Mutant Proteins/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- RNA Interference
- RNA, Small Interfering
- Recombinant Proteins/antagonists & inhibitors
- Recombinant Proteins/metabolism
- Specific Pathogen-Free Organisms
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Tianxia Jiang
- Key Laboratory of Protein and Peptide Pharmaceutical, National Laboratory of Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
New vessel formation (angiogenesis) is an essential physiological process for embryologic development, normal growth, and tissue repair. Angiogenesis is tightly regulated at the molecular level; however, this process is dysregulated in several pathological conditions such as cancer. The imbalance between pro- and antiangiogenic signaling molecules within tumors creates an abnormal vascular network that is characterized by dilated, tortuous, and leaky vessels. The pathophysiological consequences of these vascular abnormalities include temporal and spatial heterogeneity in tumor blood flow, oxygenation, and increased tumor interstitial fluid pressure. The resultant microenvironment deeply impacts on tumor progression, and also leads to a reduction in therapy efficacy. The discovery of vascular endothelial growth factor (VEGF) as a major driver of tumor angiogenesis has led to efforts to develop novel therapeutics aimed at inhibiting its activity. Anti-VEGF therapy has become an important option for the management of several human malignancies; however, a significant number of patients do not respond to anti-VEGF therapy when used either as single agent or in combination with chemotherapy. In addition, the benefit of antiangiogenic therapy is relatively short lived and the majority of patients relapse and progress. An increasing amount of reports suggest several potential mechanisms of resistance to antiangiogenic therapy including, but not limited to, tumor hypoxia. This chapter discusses the role of the VEGF axis in tumor biology and highlights the clinical application of anti-VEGF therapies elaborating on pitfalls and strategies to improve clinical outcome.
Collapse
Affiliation(s)
- Annamaria Rapisarda
- SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | |
Collapse
|
32
|
Qin L, Bromberg-White JL, Qian CN. Opportunities and challenges in tumor angiogenesis research: back and forth between bench and bed. Adv Cancer Res 2012; 113:191-239. [PMID: 22429856 DOI: 10.1016/b978-0-12-394280-7.00006-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis is essential for tumor growth and metastasis. Many signaling pathways are involved in regulating tumor angiogenesis, with the vascular endothelial growth factor pathway being of particular interest. The recognition of the heterogeneity in tumor vasculature has led to better predictions of prognosis through differential analyses of the vasculature. However, the clinical benefits from antiangiogenic therapy are limited, because many antiangiogenic agents cannot provide long-term survival benefits, suggesting the development of drug resistance. Activation of the hypoxia and c-Met pathways, as well as other proangiogenic factors, has been shown to be responsible for such resistance. Vessel co-option could be another important mechanism. For future development, research to improve the efficacy of antiangiogenic therapy includes (a) using tumor-derived endothelial cells for drug screening; (b) developing the drugs focusing on specific tumor types; (c) developing a better preclinical model for drug study; (d) developing more accurate biomarkers for patient selection; (e) targeting the c-Met pathway or other pathways; and (f) optimizing the dose and schedule of antiangiogenic therapy. In summary, the future of antiangiogenic therapy for cancer patients depends on our efforts to develop the right drugs, select the right patients, and optimize the treatment conditions.
Collapse
Affiliation(s)
- Li Qin
- State Key Laboratory on Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | | | | |
Collapse
|
33
|
Cao Y, E G, Wang E, Pal K, Dutta SK, Bar-Sagi D, Mukhopadhyay D. VEGF exerts an angiogenesis-independent function in cancer cells to promote their malignant progression. Cancer Res 2012; 72:3912-8. [PMID: 22693250 DOI: 10.1158/0008-5472.can-11-4058] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
VEGF/vascular permeability factor (VEGF/VPF or VEGF-A) is a pivotal driver of cancer angiogenesis that is a central therapeutic target in the treatment of malignancy. However, little work has been devoted to investigating functions of VEGF that are independent of its proangiogenic activity. Here, we report that VEGF produced by tumor cells acts in an autocrine manner to promote cell growth through interaction with the VEGF receptor neuropilin-1 (NRP-1). Reducing VEGF expression by tumor cells induced a differentiated phenotype in vitro and inhibited tumor forming capacity in vivo, independent of effects on angiogenesis. Autocrine activation of tumor cell growth was dependent on signaling through NRP-1, and Ras was determined to be a critical effector signaling molecule downstream of NRP-1. Our findings define a novel function for VEGF in dedifferentiation of tumor cells expanding its role in cancer beyond its known proangiogenic function.
Collapse
Affiliation(s)
- Ying Cao
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Insufficient radiofrequency ablation promotes angiogenesis of residual hepatocellular carcinoma via HIF-1α/VEGFA. PLoS One 2012; 7:e37266. [PMID: 22615958 PMCID: PMC3352883 DOI: 10.1371/journal.pone.0037266] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 04/14/2012] [Indexed: 12/21/2022] Open
Abstract
Background The mechanism of rapid growth of the residual tumor after radiofrequency (RF) ablation is poorly understood. In this study, we investigated the effect of hyperthermia on HepG2 cells and generated a subline with enhanced viability and dys-regulated angiogenesis in vivo, which was used as a model to further determine the molecular mechanism of the rapid growth of residual HCC after RF ablation. Methodology/Principal Findings Heat treatment was used to establish sublines of HepG2 cells. A subline (HepG2 k) with a relatively higher viability and significant heat tolerance was selected. The cellular protein levels of VEGFA, HIF-1α and p-Akt, VEGFA mRNA and secreted VEGFA were measured, and all of these were up-regulated in this subline compared to parental HepG2 cells. HIF-1α inhibitor YC-1 and VEGFA siRNA inhibited the high viability of the subline. The conditioned media from the subline exerted stronger pro-angiogenic effects. Bevacizumab, VEGFA siRNA and YC-1 inhibited proangiogenic effects of the conditioned media of HepG2 k cells and abolished the difference between parental HepG2 cells and HepG2 k cells. For in vivo studies, a nude mouse model was used, and the efficacy of bavacizumab was determined. HepG2 k tumor had stronger pro-angiogenic effects than parental HepG2 tumor. Bevacizumab could inhibit the tumor growth and angiogenesis, and also eliminate the difference in tumor growth and angiogenesis between parental HepG2 tumor and HepG2 k tumor in vivo. Conclusions/Significance The angiogenesis induced by HIF1α/VEGFA produced by altered cells after hyperthermia treatment may play an important role in the rapid growth of residual HCC after RF ablation. Bevacizumab may be a good candidate drug for preventing and treating the process.
Collapse
|
35
|
Su Y, Yang WB, Li S, Ye ZJ, Shi HZ, Zhou Q. Effect of angiogenesis inhibitor bevacizumab on survival in patients with cancer: a meta-analysis of the published literature. PLoS One 2012; 7:e35629. [PMID: 22539986 PMCID: PMC3335091 DOI: 10.1371/journal.pone.0035629] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/19/2012] [Indexed: 02/07/2023] Open
Abstract
Bevacizumab is a recombinant humanized monoclonal antibody against vascular endothelial growth factor which has been used in conjunction with other anti-cancer agents in the treatment of patients with many cancers. It remains controversial whether bevacizumab can prolong survival in cancer patients. This meta-analysis was therefore performed to evaluate effect of bevacizumab on survival in cancer patients. PubMed, EMBASE, and Web of Science databases were searched for English-language studies of randomized controlled trials comparing bevacizumab with control therapy published through February 8, 2012. Progression-free survival, overall survival, and one-year survival rate were analyzed using random- or fixed-effects model. Thirty one assessable randomized controlled trials were identified. A significant improvement in progression-free survival in cancer patients was attributable to bevacizumab compared with control therapy (hazard ratio, 0.72; 95% confidence interval, 0.68 to 0.76; p<0.001). Overall survival was also significantly longer in patients were treated with bevacizumab (hazard ratio, 0.87; 95% confidence interval, 0.83 to 0.91; p<0.001). The significant benefit in one-year survival rate was further seen in cancer patients receiving bevacizumab (odds ratio, 1.30; 95% confidence interval, 1.20 to 1.41; p<0.001). Current evidences showed that bevacizumab prolong progression-free survival and overall survival, and increase one-year survival rate in cancer patients as compared with control therapy.
Collapse
Affiliation(s)
| | | | | | | | - Huan-Zhong Shi
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhou
- Key Laboratory of Pulmonary Diseases of Health Ministry, Department of Respiratory Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Nielsen DL, Sengeløv L. Inhibition of placenta growth factor with TB-403: a novel antiangiogenic cancer therapy. Expert Opin Biol Ther 2012; 12:795-804. [PMID: 22506966 DOI: 10.1517/14712598.2012.679655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION There is clinical evidence that therapies targeting the vascular endothelial growth factor pathway are effective in delaying cancer progression. However, tumors may be either intrinsically resistant or evolve resistance to such therapies. Hence, there is a need for new therapies targeting angiogenesis. AREAS COVERED The data are obtained by searching in the PubMed database. The search terms used included antiangiogenic therapy, TB-403 (RO5323441), placenta growth factor (PlGF) and VEGFR-1 (Flt-1). We review preclinical data concerning the function and inhibition of PlGF and summarize data on expression of PlGF in cancer patients. Data from early-phase clinical trials of TB-403 (RO5323441), a monoclonal antibody inhibiting PlGF, are discussed. Future development strategies, therapeutic potentials and limitations of TB-403 are further evaluated. EXPERT OPINION There are some conflicting data on the function of PlGF and the importance of its role in primary tumor growth. Data from some preclinical models of PlGF inhibition and early-phase clinical trials with TB-403 are, however, promising, although the true potential of the drug is yet to be determined. Further clinical development should be preceded by molecular studies in the context of well-designed preclinical models and/or small translational studies. Future challenges involve identifying predictive biomarkers.
Collapse
Affiliation(s)
- Dorte Lisbet Nielsen
- Department of Oncology, University of Copenhagen, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | | |
Collapse
|
37
|
Wan J, Csaszar E, Chen WQ, Li K, Lubec G. Proteins from Avastin® (bevacizumab) show tyrosine nitrations for which the consequences are completely unclear. PLoS One 2012; 7:e34511. [PMID: 22523550 PMCID: PMC3327692 DOI: 10.1371/journal.pone.0034511] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/05/2012] [Indexed: 12/21/2022] Open
Abstract
Avastin® (bevacizumab) is a protein drug widely used for cancer treatment although its further use is questionable due to serious side effects reported. As no systematic proteomic study on posttranslational modifications (PTMs) was reported so far, it was the aim of the current study to use a gel-based proteomics method for determination of Avastin®-protein(s). Avastin® was run on two-dimensional gel electrophoresis (2-DE), spots were picked, followed by multi-enzyme in-gel digestion. Subsequently, the resulting peptides and posttranslational modifications were identified by mass spectrometry (nano-LC-ESI-MS/MS; HCT and LTQ Orbitrap MS). Heavy and light chains were observed and the 9 spots that were picked from 2DE-gels were identified as bevacizumab with high sequence coverage. MS/MS results showed multiple tyrosine nitrations on the Avastin® light and heavy chains that were either represented as nitrotyrosine or as aminotyrosine, which was shown to be generated from nitrotyrosine under reducing conditions. Protein nitration is known to significantly change protein functions and interactions and it may well be that some of the adverse effects of the protein drug Avastin® may be due to this PTM, which may have been generated during production- thus, nitration of Avastin® is a challenge for the pharmaceutical industry.
Collapse
Affiliation(s)
- Jia Wan
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | | | | | | |
Collapse
|
38
|
Vision, retinal thickness, and foveal avascular zone size after intravitreal bevacizumab for diabetic macular edema. Adv Ther 2012; 29:359-69. [PMID: 22402896 DOI: 10.1007/s12325-012-0009-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Indexed: 10/28/2022]
Abstract
INTRODUCTION To investigate three monthly intravitreal bevacizumab (IVB) injections effects in chronic diabetic macular edema (DME). METHODS A prospective, noncomparative study in which inclusion criteria were; DME with central macular thickness (CMT) of at least 250 μm, and no treatment for diabetic retinopathy (DR) within 4 months before the first injection. All eyes received three monthly 1.25 mg IVB injections. CMT by optical coherence tomography, visual acuity (VA), foveal avascular zone (FAZ) greatest linear dimension (GLD), and area of FAZ by fundus fluorescein angiography were documented initially and 1 month after last injection. Outcomes (P<0.05 were significant) and correlations (r values) were analyzed. RESULTS A total of 29 eyes of 29 patients (group 1, 19 female, 10 male), aged 60.7±6.6 years were analyzed. The patients were split into two groups; group 2 included 15 mild-to-moderate nonproliferative DR, and group 3 included 14 more-severe DR. VA gain was significant in all groups (P<0.05). Mean CMT decrease was approximately 46, 36, and 55 μm in groups 1, 2, and 3, respectively (P<0.05 only in group 1). A 0.045-mm2 increase in FAZ area was obtained in group 1 (P<0.05). In group 2, an increase in GLD and area of FAZ was 0.048 mm and 0.058 mm2, respectively (P<0.05), whereas in group 3, FAZ enlargement was nonsignificant. VA and CMT were significantly correlated (r values=0.5-0.6), except for the final VA-final CMT in group 2. FAZ dimensions and other parameters (VA and CMT) were noncorrelated. CONCLUSION According to the authors' short-term results, three monthly IVB injections can be used for chronic DME regardless of VA, CMT, or FAZ dimensions, despite the FAZ enlargement encountered, especially in cases with milder DR.
Collapse
|
39
|
Abstract
PURPOSE To review the current literature concerning the use of bevacizumab in treating neovascular disorders affecting the anterior segment ocular structures. METHODS The authors reviewed the literature on anti-vascular endothelial growth factor (VEGF) therapy with bevacizumab for various anterior segment neovascular disorders that was indexed in MEDLINE (up to January 2011). RESULTS Response to bevacizumab anti-VEGF therapy is variable, based on the amount of scarring, the chronicity and extent of corneal neovascularization, the disease process, and the medication formulation and its route of administration. Anti-VEGF agents are especially effective when administered early, before anatomical changes, such as corneal neovascularization and/or angle closure, are established. Neovascularization can recur if the ischemic or inflammatory process is not reversed, so eyes with long-standing diseases, such as autoimmune disorders that involve ongoing inflammation and VEGF production, seem to be less responsive to bevacizumab anti-VEGF therapy. For established neovascularization, combining anti-VEGF agents with the removal of established vessels may be more effective than anti-VEGF therapy alone. Subconjunctival bevacizumab may be more appropriate for focal, deep, and peripheral neovascularization, whereas diffuse superficial neovascularization with central corneal involvement may be best treated via topical application. CONCLUSIONS Besides the widely accepted use of bevacizumab in cancer therapy and chorioretinal neovascularization, the initial, striking, short-term response and patients' high tolerance of local bevacizumab therapy offer encouraging results for the potential role of anti-VEGF agents in treating anterior segment neovascular disorders. Controlled prospective trials are needed to establish the long-term safety, efficacy, and dosing guidelines for the use of anti-VEGF agents in anterior segment neovascularization.
Collapse
|
40
|
Ferreira MBA, Lima JPSN, Cohen EEW. Novel targeted therapies in head and neck cancer. Expert Opin Investig Drugs 2012; 21:281-95. [PMID: 22239178 DOI: 10.1517/13543784.2012.651455] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Molecularly targeted therapy, with the potential for increased selectivity and fewer adverse effects, hold promise in the treatment of HNSCC. AREAS COVERED Targeted agents for HNSCC expected to improve the effectiveness of current therapy including HER family, Src-family kinase, cell cycle, MET, AKT, HDAC, PARP, COX inhibitors and antiangiogenesis. EXPERT OPINION Epidermal growth factor receptor inhibitors are established in HNSCC and the need now is to find biomarkers for sensitivity to better select patients. Moreover, other pathway inhibitors hold significant promise and are being tested in clinical trials. Angiogenesis inhibition is likely to yield only modest efficacy alone but may augment existing standards. Lastly, one clinical arena where targeted therapies may find secure purchase is in the adjuvant or prevention setting where minimal or preneoplastic disease can be affected by inhibition of a single or few targets.
Collapse
|
41
|
Bonifazi M, Rossi M, Moja L, Scigliano VD, Franchi M, La Vecchia C, Zocchetti C, Negri E. Bevacizumab in clinical practice: prescribing appropriateness relative to national indications and safety. Oncologist 2011; 17:117-24. [PMID: 22210090 DOI: 10.1634/theoncologist.2011-0184] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The aim of this study was to describe the clinical use of bevacizumab in Lombardy (9.5 million inhabitants), Italy, during 2006-2007 in patients with metastatic colorectal cancer (mCRC) to evaluate compliance with the Italian Medicine Agency (AIFA) indications, the incidence of adverse events, and the survival rate. We performed computerized record linkage among three different Lombardy health care databases: File F registry, Regional discharge database, and Registry Office records. Patients were classified into approved and off-label uses according to the AIFA indications. Treatment with bevacizumab was administered to 780 patients, of whom 81.7% (n = 637) had mCRC. Among these, 37.8% (n = 241) of patients received the drug in observance of AIFA indications. Overall, ∼10% of patients had serious treatment-related toxicities (fistula, 3.5%; venous thromboembolism, 2.8%; hemorrhage, 1.9%; intestinal perforation and arterial thromboembolism, <1%). The 1-year survival rate was 74.3% and the 2-year survival rate was 39.2%. The median survival time was 20.5 months, and there were no meaningful differences between gender and age groups. There was a gap between the bevacizumab approved indication and clinical practice pattern: overall, less than one half of the patients received bevacizumab in observance with the regulatory indication. The main reason for nonadherence to the indication was use as a second-line or advanced line of therapy. The incidence of serious adverse events and the survival rates of mCRC patients were similar to those reported in clinical trials.
Collapse
Affiliation(s)
- Martina Bonifazi
- Department of Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri, via Giuseppe La Masa 19, 20156 Milan, Italia
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ghadimi MP, Liu P, Peng T, Bolshakov S, Young ED, Torres KE, Colombo C, Hoffman A, Broccoli D, Hornick JL, Lazar AJ, Pisters P, Pollock RE, Lev D. Pleomorphic liposarcoma: clinical observations and molecular variables. Cancer 2011; 117:5359-69. [PMID: 21598240 PMCID: PMC3161152 DOI: 10.1002/cncr.26195] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/12/2011] [Accepted: 03/28/2011] [Indexed: 11/12/2022]
Abstract
BACKGROUND Pleomorphic liposarcoma (PLS) is a rare high-grade sarcoma that has lipoblastic differentiation. In this study, the authors evaluated PLS natural history, patient outcomes, and commonly deregulated protein biomarkers. METHODS Medical records from patients (n = 155) who had PLS from 1993 to 2010 were reviewed. Univariate and multivariate analyses were conducted to identify independent prognosticators. A PLS tissue microarray (TMA) (n = 56 patient specimens) was constructed for immunohistochemical analysis of molecular markers, and p53 gene sequencing (exons 5-9) was conducted. RESULTS The average patient age was 57 years, and the patients presented with primary disease (n = 102), recurrent disease (n = 16), and metastatic disease (n = 37). Lower extremity was the most common disease site (40%), and the average tumor size was 11 cm. Complete follow-up data were available for 83 patients, and their median follow-up was 22.6 months. The 5-year disease-specific survival rate was 53%; and recurrent disease, unresectability, and microscopic positive margins were identified as predictors of a poor prognosis. Systemic relapse (the strongest poor prognostic determinant) developed in 35% of patients with localized PLS. Immunohistochemical analysis revealed increased expression of peroxisome proliferator-activated receptor gamma (an adipogenic marker), B-cell leukemia 2 and survivin (survival factors), vascular endothelial growth factor (an angiogenic factor), matrix metalloproteinase 2, and other biomarkers. Frequent loss of retinoblastoma protein expression and high p53 mutation rates (approximately 60%) were observed. CONCLUSIONS PLS is an aggressive, metastasizing sarcoma. Identifying ubiquitous molecular events underlying PLS progression is crucial for progress in patient management and outcomes.
Collapse
Affiliation(s)
- Markus P. Ghadimi
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Ping Liu
- The Division of Quantitative Sciences, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Tingsheng Peng
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Svetlana Bolshakov
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Eric D. Young
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Keila E. Torres
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Chiara Colombo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Aviad Hoffman
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Dominique Broccoli
- The Department of Laboratory Oncology Research, Memorial University Medical Center, Savannah, USA
| | - Jason L. Hornick
- The Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Alexander J. Lazar
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, USA
- The Graduate School of Biomedical Sciences, Houston, USA
| | - Peter Pisters
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Raphael E. Pollock
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Dina Lev
- Sarcoma Research Center, University of Texas MD Anderson Cancer Center, Houston, USA
- The Graduate School of Biomedical Sciences, Houston, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
43
|
Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 2011; 30:83-95. [PMID: 21249423 DOI: 10.1007/s10555-011-9281-4] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system regulates angiogenesis in cancer with both pro- and antiangiogenic activities. The induction of angiogenesis is mediated by tumor-associated macrophages and myeloid-derived suppressor cells (MDSC) which produce proinflammatory cytokines, endothelial growth factors (VEGF, bFGF…), and protease (MMP9) implicated in neoangiogenesis. Some cytokines (IL-6, IL-17…) activated Stat3 which also led to the production of VEGF and bFGF. In contrast, other cytokines (IFN, IL-12, IL-21, and IL-27) display an antiangiogenic activity. Recently, it has been shown that some antiangiogenic molecules alleviates immunosuppression associated with cancer by decreasing immunosuppressive cells (MDSC, regulatory T cells), immunosuppressive cytokines (IL-10, TGFβ), and inhibitory molecules on T cells (PD-1). Some of these broad effects may result from the ability of some antiangiogenic molecules, especially cytokines to inhibit the Stat3 transcription factor. The association often observed between angiogenesis and immunosuppression may be related to hypoxia which induces both neoangiogenesis via activation of HIF-1 and VEGF and favors the intratumor recruitment and differentiation of regulatory T cells and MDSC. Preliminary studies suggest that modulation of immune markers (intratumoral MDSC and IL-8, peripheral regulatory T cells…) may predict clinical response to antiangiogenic therapy. In preclinical models, a synergy has been observed between antiangiogenic molecules and immunotherapy which may be explained by an improvement of immune status in tumor-bearing mice after antiangiogenic therapy. In preclinical models, antiangiogenic molecules promoted intratumor trafficking of effector cells, enhance endogenous anti-tumor response, and synergyzed with immunotherapy protocols to cure established murine tumors. All these results warrant the development of clinical trials combining antiangiogenic drugs and immunotherapy.
Collapse
|
44
|
Ługowska I, Woźniak W, Klepacka T, Michalak E, Szamotulska K. A prognostic evaluation of vascular endothelial growth factor in children and young adults with osteosarcoma. Pediatr Blood Cancer 2011; 57:63-8. [PMID: 21416580 DOI: 10.1002/pbc.23021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Accepted: 12/10/2010] [Indexed: 11/09/2022]
Abstract
BACKGROUND The potential role of VEGF in osteosarcoma has been evaluated in several studies. The majority of them included heterogeneous and limited series of patients, giving conflicting results. The aim of presented study is to evaluate the prognostic role of VEGF-A in biopsy samples of clinically homogeneous group of osteosarcoma patients with at least 5 years of follow up. MATERIALS AND METHODS VEGF-A was assessed immunohistochemically in the pre-treatment biopsy samples of 91 patients (mean age 14 years; range, 4-23 years) with primary, high-grade, non-metastatic osteosarcoma localized in extremities. The survival of each patient was assessed after at least 5-year follow-up period. RESULTS VEGF-A over 50% of positive tumor cells was observed in 39% of cases and was linked to patients age below 14-year old (P = 0.025) and tumor size more than 8 cm (P = 0.054). VEGF-A was associated with a significantly decreased both overall survival (P = 0.006) and progression-free survival (P = 0.011). In the Cox proportional hazard model it was confirmed that VEGF-A expression in the biopsy samples was an independent prognostic factor of unfavorable survival in osteosarcoma (HR 2.51; 95% CI: 1.12-5.66). CONCLUSION The expression of VEGF-A in the biopsy sample is the potential marker for predicting the course and outcome of osteosarcoma.
Collapse
Affiliation(s)
- Iwona Ługowska
- Institute of Mother and Child, Department of Pediatrics and Adolescent Oncological Surgery, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
45
|
Weidlich S, Walsh K, Crowther D, Burczynski ME, Feuerstein G, Carey FA, Steele RJC, Wolf CR, Miele G, Smith G. Pyrosequencing-based methods reveal marked inter-individual differences in oncogene mutation burden in human colorectal tumours. Br J Cancer 2011; 105:246-54. [PMID: 21712828 PMCID: PMC3142798 DOI: 10.1038/bjc.2011.197] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 05/06/2011] [Accepted: 05/11/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The epidermal growth factor receptor-targeted monoclonal antibody cetuximab (Erbitux) was recently introduced for the treatment of metastatic colorectal cancer. Treatment response is dependent on Kirsten-Ras (K-Ras) mutation status, in which the majority of patients with tumour-specific K-Ras mutations fail to respond to treatment. Mutations in the oncogenes B-Raf and PIK3CA (phosphoinositide-3-kinase) may also influence cetuximab response, highlighting the need for a sensitive, accurate and quantitative assessment of tumour mutation burden. METHODS Mutations in K-Ras, B-Raf and PIK3CA were identified by both dideoxy and quantitative pyrosequencing-based methods in a cohort of unselected colorectal tumours (n=102), and pyrosequencing-based mutation calls correlated with various clinico-pathological parameters. RESULTS The use of quantitative pyrosequencing-based methods allowed us to report a 13.7% increase in mutation burden, and to identify low-frequency (<30% mutation burden) mutations not routinely detected by dideoxy sequencing. K-Ras and B-Raf mutations were mutually exclusive and independently associated with a more advanced tumour phenotype. CONCLUSION Pyrosequencing-based methods facilitate the identification of low-frequency tumour mutations and allow more accurate assessment of tumour mutation burden. Quantitative assessment of mutation burden may permit a more detailed evaluation of the role of specific tumour mutations in the pathogenesis and progression of colorectal cancer and may improve future patient selection for targeted drug therapies.
Collapse
Affiliation(s)
- S Weidlich
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - K Walsh
- Roche Pharmaceuticals, Nutley, NJ, USA
| | | | | | | | - F A Carey
- Department of Pathology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - R J C Steele
- Centre for Academic Clinical Practice, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - C R Wolf
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
- Cancer Research UK Molecular Pharmacology Laboratory, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - G Miele
- Roche Pharmaceuticals, Nutley, NJ, USA
| | - G Smith
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| |
Collapse
|
46
|
Schell JC, Koenig SB, Bastin K, Wirostko WJ. Intracameral bevacizumab administered for non-small cell lung cancer metastasis to iris. Clin Pract 2011; 1:e39. [PMID: 24765300 PMCID: PMC3981238 DOI: 10.4081/cp.2011.e39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 05/11/2011] [Indexed: 11/23/2022] Open
Abstract
Ocular iris metastasis from lung cancer is uncommon. We report a patient with metastatic non-small cell lung cancer who was found to have a metastatic lesion to the iris. Local therapy for pain control and vision loss was administered with intracameral bevacizumab. Complete resolution of pain, improvement in vision, and near complete resolution of iris tumor were seen within two months. No ocular toxicity to anterior segment structures was detectable on corneal pachymetry and corneal specular microscopy. This is the first case report demonstrating safety and efficacy of intracameral bevacizumab for iris metastasis from non-small cell lung cancer.
Collapse
Affiliation(s)
- Jonathan C Schell
- The Eye Institute, Department of Ophthalmology, Medical College of Wisconsin
| | - Steven B Koenig
- The Eye Institute, Department of Ophthalmology, Medical College of Wisconsin
| | - Kenneth Bastin
- Radiation Oncology Associates Ltd., St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - William J Wirostko
- The Eye Institute, Department of Ophthalmology, Medical College of Wisconsin
| |
Collapse
|
47
|
Sultan MB, Zhou D, Loftus J, Dombi T, Ice KS. A phase 2/3, multicenter, randomized, double-masked, 2-year trial of pegaptanib sodium for the treatment of diabetic macular edema. Ophthalmology 2011; 118:1107-18. [PMID: 21529957 DOI: 10.1016/j.ophtha.2011.02.045] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Revised: 01/24/2011] [Accepted: 02/25/2011] [Indexed: 02/07/2023] Open
Abstract
PURPOSE To confirm the safety and compare the efficacy of intravitreal pegaptanib sodium 0.3 mg versus sham injections in subjects with diabetic macular edema (DME) involving the center of the macula associated with vision loss not due to ischemia. DESIGN Randomized (1:1), sham-controlled, multicenter, parallel-group trial. PARTICIPANTS Subjects with DME. INTERVENTION Subjects received pegaptanib 0.3 mg or sham injections every 6 weeks in year 1 (total = 9 injections) and could receive focal/grid photocoagulation beginning at week 18. During year 2, subjects received injections as often as every 6 weeks per prespecified criteria. MAIN OUTCOME MEASURES The primary efficacy endpoint was the proportion gaining ≥ 10 letters of visual acuity (VA) from baseline to year 1. Safety was monitored throughout. RESULTS In all, 260 (pegaptanib, n = 133; sham, n = 127) and 207 (pegaptanib, n = 107; sham, n = 100) subjects were included in years 1 and 2 intent-to-treat analyses, respectively. A total of 49 of the 133 (36.8%) subjects from the pegaptanib group and 25 of the 127 (19.7%) from the sham group experienced a VA improvement of ≥ 10 letters at week 54 compared with baseline (odds ratio [OR], 2.38; 95% confidence interval, 1.32-4.30; P = 0.0047). For pegaptanib-treated subjects, change in mean VA from baseline by visit was superior (P<0.05) to sham at weeks 6, 24, 30, 36, 42, 54, 78, 84, 90, 96, and 102. At week 102, pegaptanib-treated subjects gained, on average, 6.1 letters versus 1.3 letters for sham (P<0.01). Fewer pegaptanib- than sham-treated subjects received focal/grid laser treatment (week 54, 31/133 [23.3%] vs 53/127 [41.7%], respectively, P = 0.002; week 102, 27/107 [25.2%] vs 45/100 [45.0%], respectively, P = 0.003). The pegaptanib treatment group showed significantly better results on the National Eye Institute-Visual Functioning Questionnaire than sham for subscales important in this population. Pegaptanib was well tolerated; the frequencies of discontinuations, adverse events, treatment-related adverse events, and serious adverse events were comparable in the pegaptanib and sham groups. CONCLUSIONS Patients with DME derive clinical benefit from treatment with the selective vascular endothelial growth factor antagonist pegaptanib 0.3 mg. These findings indicate that intravitreal pegaptanib is effective in the treatment of DME and, taken together with prior study data, support a positive safety profile in this population. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found after the references.
Collapse
|
48
|
Sato Y. Persistent vascular normalization as an alternative goal of anti-angiogenic cancer therapy. Cancer Sci 2011; 102:1253-6. [PMID: 21401807 DOI: 10.1111/j.1349-7006.2011.01929.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Angiogenesis is recognized as one of the principal hallmarks of cancers. Cancers contain newly formed immature vessels devoid of firm coverage by pericytes. Several drugs targeting vascular endothelial growth factor signals are now in clinical use for anti-angiogenic cancer treatment. Those drugs transiently normalize tumor vessels and ultimately provoke vascular regression. This regression causes tumor hypoxia, which could trigger certain cancer cells to become more invasive and metastatic. Normalized vessels do not induce tumor hypoxia, and may protect from cancer cell intravasation and enhance anticancer treatment with chemotherapeutic agents, radiation, or immune therapy. Thus, persistent vascular normalization could be an alternative goal of anti-angiogenic cancer treatment.
Collapse
Affiliation(s)
- Yasufumi Sato
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| |
Collapse
|
49
|
Tugues S, Koch S, Gualandi L, Li X, Claesson-Welsh L. Vascular endothelial growth factors and receptors: anti-angiogenic therapy in the treatment of cancer. Mol Aspects Med 2011; 32:88-111. [PMID: 21565214 DOI: 10.1016/j.mam.2011.04.004] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Accepted: 04/27/2011] [Indexed: 12/21/2022]
Abstract
Vascular endothelial growth factors (VEGFs) are critical regulators of vascular and lymphatic function during development, in health and in disease. There are five mammalian VEGF ligands and three VEGF receptor tyrosine kinases. In addition, several VEGF co-receptors that lack intrinsic catalytic activity, but that indirectly modulate the responsiveness to VEGF contribute to the final biological effect. This review describes the molecular features of VEGFs, VEGFRs and co-receptors with focus on their role in the treatment of cancer.
Collapse
Affiliation(s)
- Sònia Tugues
- Uppsala University, Dept. of Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsv. 20, 751 85 Uppsala, Sweden
| | | | | | | | | |
Collapse
|
50
|
Ensinger C. Models of partnership between the pharmaceutical and diagnostics industries around companion diagnostics for cancer and beyond. ACTA ACUST UNITED AC 2011; 5:91-4. [PMID: 23480583 DOI: 10.1517/17530059.2011.552497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
An increase of targeted anticancer therapies has led to the beginning of a new era of cancer treatment, partly by replacing classical chemotherapies, partly supplementing these. Whereas for some substances only clinical experiences are relevant for treatment decisions, for some major cancer groups predictive markers are known that indicate probable tumor responses. To identify the latter, a need for companion diagnostics is given, often already existing as successful cooperation between pharmaceutical and diagnostic industries. This editorial focuses on the impact of companion diagnostic tests in personalized anticancer medicine, reporting recent advances in identifying and characterizing tumor subgroups responding to selected drugs. The most successful targeted therapies are directed against the EGFR/Her-2/neu receptors with regard to their downstream molecules in major cancer groups, including breast, gastric, lung and colorectal carcinomas. The development of biomarkers provides great opportunities to identify subpopulations with differential drug responses. On the one hand patients themselves are gaining major advantages of personalized and better tolerable cancer treatment, on the other hand, owing to very focused targeted therapies, these developments make possible cost-intensive targeted drug investigations and trials, especially in a situation of limited healthcare budgets.
Collapse
Affiliation(s)
- Christian Ensinger
- Medical University of Innsbruck, Institute of Pathology, Muellerstrasse 44, A-6020 Innsbruck, Austria +43 512 9003 71315 ; +43 512 9003 73301 ;
| |
Collapse
|