51
|
Cooper SL, Carter JJ, March J, Woolard J. Long-term cardiovascular effects of vandetanib and pazopanib in normotensive rats. Pharmacol Res Perspect 2019; 7:e00477. [PMID: 31164986 PMCID: PMC6543457 DOI: 10.1002/prp2.477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/08/2019] [Indexed: 01/24/2023] Open
Abstract
Vandetanib and pazopanib are clinically available, multi-targeted inhibitors of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinases. Short-term VEGF receptor inhibition is associated with hypertension in 15%-60% of patients, which may limit the use of these anticancer therapies over the longer term. To evaluate the longer-term cardiovascular implications of treatment, we investigated the "on"-treatment (21 days) and "off"-treatment (10 days) effects following daily administration of vandetanib, pazopanib, or vehicle, in conscious rats. Cardiovascular variables were monitored in unrestrained Sprague-Dawley rats instrumented with radiotelemetric devices. In Study 1, rats were randomly assigned to receive either daily intraperitoneal injections of vehicle (volume 0.5 mL; n = 5) or vandetanib 25 mg/kg/day (volume 0.5 mL; n = 6). In Study 2, rats received either vehicle (volume 0.5 mL; n = 4) or pazopanib 30 mg/kg/day (volume 0.5 mL; n = 7), dosed once every 24 hours for 21 days. All solutions were in 2% Tween, 5% propylene glycol in 0.9% saline solution. Vandetanib caused sustained increases in mean arterial pressure (MAP), systolic blood pressure (SBP), and diastolic blood pressure (DBP) compared to baseline and vehicle. Vandetanib also significantly altered the circadian cycling of MAP, SBP, and DBP. Elevations in SBP were detectable 162 hours after the last dose of vandetanib. Pazopanib also caused increases in MAP, SBP, and DBP. However, compared to vandetanib, these increases were of slower onset and a smaller magnitude. These data suggest that the cardiovascular consequences of vandetanib and pazopanib treatment are sustained, even after prolonged cessation of drug treatment.
Collapse
Affiliation(s)
- Samantha L. Cooper
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| | - Joanne J. Carter
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Julie March
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| |
Collapse
|
52
|
Lane N, Nyman J, Uppuganti S, Chaudhari A, Aguirre J, Shidara K, Liu X, Yao W, Kimmel D. Inhibition of vascular endothelial growth factor in young adult mice causes low bone blood flow and bone strength with no effect on bone mass in trabecular regions. Bone Rep 2019; 10:100210. [PMID: 31193542 PMCID: PMC6535464 DOI: 10.1016/j.bonr.2019.100210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine the effect of an antibody to vascular endothelial growth factor (VEGF) on bone blood flow, bone strength, and bone mass in the young adult mouse. METHODS Ten-week-old male BALB/cJ mice were body weight-randomized into either a rodent anti-VEGF monoclonal antibody (anti-VEGF, B20-4.1.1; 5 mg/kg 2×/wk.; n = 12) group or a vehicle (VEH; n = 12) group. After 42 days, mice were evaluated for bone blood flow at the distal femur by 18F-NaF-PET/CT and then necropsied. Samples from trabecular and cortical bone regions were evaluated for bone strength by mechanical testing, bone mass by peripheral quantitative computed tomography (pQCT), and micoarchitecture (MicroCT). Hydration of the whole femur was studied by proton nuclear magnetic resonance relaxometry (1H NMR). RESULTS Distal femur blood flow was 43% lower in anti-VEGF mice than in VEH mice (p = 0.009). Ultimate load in the lumbar vertebral body was 25% lower in anti-VEGF than in VEH mice (p = 0.013). Bone mineral density (BMD) in the trabecular region of the proximal humeral metaphysis by pQCT, and bone volume fraction and volumetric BMD by MicroCT were the same in the two groups. Volume fraction of bound water (BW) of the whole femur was 14% lower in anti-VEGF than in VEH mice (p = 0.003). Finally, BW, but not cortical tissue mineral density, helped section modulus explain the variance in the ultimate moment experienced by the femur in three-point bending. CONCLUSION Anti-VEGF caused low bone blood flow and bone strength in trabecular bone regions without influencing BMD and microarchitecture. Low bone strength was also associated with low bone hydration. These data suggest that bone blood flow is a novel bone property that affects bone quality.
Collapse
Affiliation(s)
- N.E. Lane
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - J.S. Nyman
- Department of Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - S. Uppuganti
- Department of Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - A.J. Chaudhari
- Center for Molecular and Genomic Imaging, Department of Radiology, University of California at Davis, Davis, CA 95616, USA
| | - J.I. Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - K. Shidara
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - X.P. Liu
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - W. Yao
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - D.B. Kimmel
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
53
|
Qian Z, Sharma D, Jia W, Radke D, Kamp T, Zhao F. Engineering stem cell cardiac patch with microvascular features representative of native myocardium. Theranostics 2019; 9:2143-2157. [PMID: 31149034 PMCID: PMC6531308 DOI: 10.7150/thno.29552] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
The natural myocardium is a highly aligned tissue with an oriented vasculature. Its characteristic cellular as well as nanoscale extracellular matrix (ECM) organization along with an oriented vascular network ensures appropriate blood supply and functional performance. Although significant efforts have been made to develop anisotropic cardiac structure, currently neither an ideal biomaterial nor an effective vascularization strategy to engineer oriented and high-density capillary-like microvessels has been achieved for clinical cardiovascular therapies. A naturally derived oriented ECM nanofibrous scaffold mimics the physiological structure and components of tissue ECM and guides neovascular network formation. The objective of this study was to create an oriented and dense microvessel network with physiological myocardial microvascular features. METHODS Highly aligned decellularized human dermal fibroblast sheets were used as ECM scaffold to regulate physiological alignment of microvascular networks by co-culturing human mesenchymal stem cells (hMSCs) and endothelial cells (ECs). The influence of topographical features on hMSC and EC interaction was investigated to understand underlying mechanisms of neovasculature formation. RESULTS Results demonstrate that the ECM topography can be translated to ECs via CD166 tracks and significantly improved hMSC-EC crosstalk and vascular network formation. The aligned ECM nanofibers enhanced structure, length, and density of microvascular networks compared to randomly organized nanofibrous ECM. Moreover, hMSC-EC co-culture promoted secretion of pro-angiogenic growth factors and matrix remodeling via metalloprotease-2 (MMP-2) activation, which resulted in highly dense vascular network formation with intercapillary distance (20 μm) similar to the native myocardium. CONCLUSION HMSC-EC co-culture on the highly aligned ECM generates physiologically oriented and dense microvascular network, which holds great potential for cardiac tissue engineering.
Collapse
Affiliation(s)
- Zichen Qian
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Wenkai Jia
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Daniel Radke
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Timothy Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
54
|
Groppa E, Brkic S, Uccelli A, Wirth G, Korpisalo-Pirinen P, Filippova M, Dasen B, Sacchi V, Muraro MG, Trani M, Reginato S, Gianni-Barrera R, Ylä-Herttuala S, Banfi A. EphrinB2/EphB4 signaling regulates non-sprouting angiogenesis by VEGF. EMBO Rep 2018; 19:embr.201745054. [PMID: 29643120 PMCID: PMC5934775 DOI: 10.15252/embr.201745054] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 03/03/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis, whose best-understood mechanism is sprouting. However, therapeutic VEGF delivery to ischemic muscle induces angiogenesis by the alternative process of intussusception, or vascular splitting, whose molecular regulation is essentially unknown. Here, we identify ephrinB2/EphB4 signaling as a key regulator of intussusceptive angiogenesis and its outcome under therapeutically relevant conditions. EphB4 signaling fine-tunes the degree of endothelial proliferation induced by specific VEGF doses during the initial stage of circumferential enlargement of vessels, thereby limiting their size and subsequently enabling successful splitting into normal capillary networks. Mechanistically, EphB4 neither inhibits VEGF-R2 activation by VEGF nor its internalization, but it modulates VEGF-R2 downstream signaling through phospho-ERK1/2. In vivo inhibitor experiments show that ERK1/2 activity is required for EphB4 regulation of VEGF-induced intussusceptive angiogenesis. Lastly, after clinically relevant VEGF gene delivery with adenoviral vectors, pharmacological stimulation of EphB4 normalizes dysfunctional vascular growth in both normoxic and ischemic muscle. These results identify EphB4 as a druggable target to modulate the outcome of VEGF gene delivery and support further investigation of its therapeutic potential.
Collapse
Affiliation(s)
- Elena Groppa
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Sime Brkic
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Andrea Uccelli
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Galina Wirth
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | - Maria Filippova
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Veronica Sacchi
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Manuele Giuseppe Muraro
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Marianna Trani
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Silvia Reginato
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Roberto Gianni-Barrera
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.,Department of Surgery, University Hospital, Basel, Switzerland
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland.,Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Andrea Banfi
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland .,Department of Surgery, University Hospital, Basel, Switzerland
| |
Collapse
|
55
|
Ciuffreda MC, Malpasso G, Chokoza C, Bezuidenhout D, Goetsch KP, Mura M, Pisano F, Davies NH, Gnecchi M. Synthetic extracellular matrix mimic hydrogel improves efficacy of mesenchymal stromal cell therapy for ischemic cardiomyopathy. Acta Biomater 2018; 70:71-83. [PMID: 29341932 DOI: 10.1016/j.actbio.2018.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) repair infarcted hearts mainly through paracrine mechanisms. Low cell engraftment limits the release of soluble paracrine factors (SF) over time and, consequently, MSC efficacy. We tested whether a synthetic extracellular matrix mimic, a hydrogel containing heparin (H-HG), could ameliorate MSC engraftment and binding/release of SF, thus improving MSC therapy efficacy. METHODS AND RESULTS In vitro, rat bone-marrow MSC (rBM-MSC) were seeded and grown into H-HG. Under normoxia, the hydrogel did not affect cell survival (rBM-MSC survival >90% at each time point tested); vice versa, under hypoxia the biomaterial resulted to be protective for the cells (p < .001 vs rBM-MSC alone). H-HG or control PEG hydrogels (HG) were incubated with VEGF or bFGF for binding/release quantification. Data showed significantly higher amount of VEGF and bFGF bound by H-HG compared with HG (p < .05) and a constant release over time. In vivo, myocardial infarction (MI) was induced in female Sprague Dawley rats by permanent coronary ligation. One week later, saline, rBM-MSC, H-HG or rBM-MSC/H-HG were injected in the infarct zone. The co-injection of rBM-MSC/H-HG into infarcted hearts significantly increased cardiac function. Importantly, we observed a significant gain in MSC engraftment, reduction of ventricular remodeling and stimulation of neo-vasculogenesis. We also documented higher amounts of several pro-angiogenic factors in hearts treated with rBM-MSC/H-HG. CONCLUSIONS Our data show that H-HG increases MSC engraftment, efficiently fine tunes the paracrine MSC actions and improves cardiac function in infarcted rat hearts. STATEMENT OF SIGNIFICANCE Transplantation of MSC is a promising treatment for ischemic heart disease, but low cell engraftment has so far limited its efficacy. The enzymatically degradable H-HG that we developed is able to increase MSC retention/engraftment and, at the same time, to fine-tune the paracrine effects mediated by the cells. Most importantly, the co-transplantation of MSC and H-HG in a rat model of ischemic cardiomyopathy improved heart function through a significant reduction in ventricular remodeling/scarring and amelioration in neo-vasculogenesis/endogenous cardiac regeneration. These beneficial effects are comparable to those obtained by others using a much greater number of cells, strengthening the efficacy of the biomaterial used in increasing the therapeutic effects of MSC. Given its efficacy and safety, documented by the absence of immunoreaction, our strategy appears readily translatable to clinical scenarios.
Collapse
Affiliation(s)
- Maria Chiara Ciuffreda
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Giuseppe Malpasso
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Cindy Chokoza
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Deon Bezuidenhout
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Kyle P Goetsch
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Manuela Mura
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Federica Pisano
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy
| | - Neil H Davies
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Department of Health Sciences, Cape Town, South Africa
| | - Massimiliano Gnecchi
- Department of Medical Sciences and Infectious Diseases - Coronary Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Italy; Department of Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
56
|
Kress S, Baur J, Otto C, Burkard N, Braspenning J, Walles H, Nickel J, Metzger M. Evaluation of a Miniaturized Biologically Vascularized Scaffold in vitro and in vivo. Sci Rep 2018; 8:4719. [PMID: 29549334 PMCID: PMC5856827 DOI: 10.1038/s41598-018-22688-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/28/2018] [Indexed: 02/07/2023] Open
Abstract
In tissue engineering, the generation and functional maintenance of dense voluminous tissues is mainly restricted due to insufficient nutrient supply. Larger three-dimensional constructs, which exceed the nutrient diffusion limit become necrotic and/or apoptotic in long-term culture if not provided with an appropriate vascularization. Here, we established protocols for the generation of a pre-vascularized biological scaffold with intact arterio-venous capillary loops from rat intestine, which is decellularized under preservation of the feeding and draining vascular tree. Vessel integrity was proven by marker expression, media/blood reflow and endothelial LDL uptake. In vitro maintenance persisted up to 7 weeks in a bioreactor system allowing a stepwise reconstruction of fully vascularized human tissues and successful in vivo implantation for up to 4 weeks, although with time-dependent decrease of cell viability. The vascularization of the construct lead to a 1.5× increase in cellular drug release compared to a conventional static culture in vitro. For the first time, we performed proof-of-concept studies demonstrating that 3D tissues can be maintained within a miniaturized vascularized scaffold in vitro and successfully implanted after re-anastomosis to the intrinsic blood circulation in vivo. We hypothesize that this technology could serve as a powerful platform technology in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sebastian Kress
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany
| | - Johannes Baur
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Christoph Otto
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Natalie Burkard
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Joris Braspenning
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany
| | - Heike Walles
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany.,Fraunhofer Institute of Silicate Research ISC, Translational Center for Regenerative Therapies, 97070, Würzburg, Germany
| | - Joachim Nickel
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany.
| | - Marco Metzger
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany. .,Fraunhofer Institute of Silicate Research ISC, Translational Center for Regenerative Therapies, 97070, Würzburg, Germany.
| |
Collapse
|
57
|
Abstract
The therapeutic options available to treat a wide range of malignancies are rapidly increasing. At the same time, the population being treated is aging with more cardiovascular risk factors, comorbid conditions, and associated poor cardiac reserve. Both traditional chemotherapeutic agents (for example, anthracyclines) and newer therapies (for example, targeted tyrosine kinase inhibitors and immune checkpoint inhibitors) have demonstrated profound cardiovascular toxicities. It is important to understand the mechanisms of these toxicities to establish strategies for the prevention and management of complications—arrhythmias, heart failure, and even death. In the first of this two-part review series, we focus on what is known and hypothesized about the mechanisms of cardiovascular toxicity from anthracyclines, HER2/ErbB2 inhibitors, immune checkpoint inhibitors, and vascular endothelial growth factor inhibitors.
Collapse
Affiliation(s)
- Timothy M Markman
- Department of Medicine, Cardiovascular Division, Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maurie Markman
- Cancer Treatment Centers of America at Eastern Regional Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
58
|
Pombero A, Garcia-Lopez R, Estirado A, Martinez S. Vascular pattern of the dentate gyrus is regulated by neural progenitors. Brain Struct Funct 2018; 223:1971-1987. [PMID: 29306978 DOI: 10.1007/s00429-017-1603-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/28/2017] [Indexed: 01/19/2023]
Abstract
Neurogenesis is a vital process that begins during early embryonic development and continues until adulthood, though in the latter case, it is restricted to the subventricular zone and the subgranular zone of the dentate gyrus (DG). In particular, the DG's neurogenic properties are structurally and functionally unique, which may be related to its singular vascular pattern. Neurogenesis and angiogenesis share molecular signals and act synergistically, supporting the concept of a neurogenic niche as a functional unit between neural precursors cells and their environment, in which the blood vessels play an important role. Whereas it is well known that vascular development controls neural proliferation in the embryonary and in the adult brain, by releasing neurotrophic factors; the potential influence of neural cells on vascular components during angiogenesis is largely unknown. We have demonstrated that the reduction of neural progenitors leads to a significant impairment of vascular development. Since VEGF is a potential regulator in the neurogenesis-angiogenesis crosstalk, we were interested in assessing the possible role of this molecule in the hippocampal neurovascular development. Our results showed that VEGF is the molecule involved in the regulation of vascular development by neural progenitor cells in the DG.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Blood Vessels/physiology
- CD13 Antigens/metabolism
- Cell Differentiation
- Cell Proliferation
- Dentate Gyrus/anatomy & histology
- Dentate Gyrus/embryology
- Dentate Gyrus/growth & development
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental/physiology
- Ki-67 Antigen/metabolism
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neovascularization, Physiologic/physiology
- Nerve Tissue Proteins/metabolism
- Nestin/genetics
- Nestin/metabolism
- Neural Stem Cells/physiology
- Neurogenesis/physiology
- RNA, Messenger
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
Collapse
Affiliation(s)
- Ana Pombero
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Raquel Garcia-Lopez
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Alicia Estirado
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Salvador Martinez
- Instituto de Neurociencias, UMH-CSIC, Campus de San Juan, 03550, Alicante, Spain.
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
59
|
Morita A, Mori A, Arima S, Sakamoto K, Nagamitsu T, Ishii K, Nakahara T. Transient phenotypic changes in endothelial cells and pericytes in neonatal mouse retina following short-term blockade of vascular endothelial growth factor receptors. Dev Dyn 2018; 247:699-711. [DOI: 10.1002/dvdy.24614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/23/2017] [Accepted: 12/04/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Akane Morita
- Department of Molecular Pharmacology; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Asami Mori
- Department of Molecular Pharmacology; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Shiho Arima
- Department of Organic Synthesis; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Tohru Nagamitsu
- Department of Organic Synthesis; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology; Kitasato University School of Pharmaceutical Sciences; Tokyo Japan
| |
Collapse
|
60
|
Watson EC, Grant ZL, Coultas L. Endothelial cell apoptosis in angiogenesis and vessel regression. Cell Mol Life Sci 2017; 74:4387-4403. [PMID: 28646366 PMCID: PMC11107683 DOI: 10.1007/s00018-017-2577-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Blood vessel regression is an essential process for ensuring blood vessel networks function at optimal efficiency and for matching blood supply to the metabolic needs of tissues as they change over time. Angiogenesis is the major mechanism by which new blood vessels are produced, but the vessel growth associated with angiogenesis must be complemented by remodeling and maturation events including the removal of redundant vessel segments and cells to fashion the newly forming vasculature into an efficient, hierarchical network. This review will summarize recent findings on the role that endothelial cell apoptosis plays in vascular remodeling during angiogenesis and in vessel regression more generally.
Collapse
Affiliation(s)
- Emma C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Zoe L Grant
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Leigh Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
61
|
Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis 2017; 20:519-531. [PMID: 28667352 PMCID: PMC5660148 DOI: 10.1007/s10456-017-9563-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/21/2017] [Indexed: 12/02/2022]
Abstract
Fibronectin containing alternatively spliced EIIIA and EIIIB domains is largely absent from mature quiescent vessels in adults, but is highly expressed around blood vessels during developmental and pathological angiogenesis. The precise functions of fibronectin and its splice variants during developmental angiogenesis however remain unclear due to the presence of cardiac, somitic, mesodermal and neural defects in existing global fibronectin KO mouse models. Using a rare family of surviving EIIIA EIIIB double KO mice, as well as inducible endothelial-specific fibronectin-deficient mutant mice, we show that vascular development in the neonatal retina is regulated in an autocrine manner by endothelium-derived fibronectin, and requires both EIIIA and EIIIB domains and the RGD-binding α5 and αv integrins for its function. Exogenous sources of fibronectin do not fully substitute for the autocrine function of endothelial fibronectin, demonstrating that fibronectins from different sources contribute differentially to specific aspects of angiogenesis.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- University of Suffolk, James Hehir Building, University Avenue, Ipswich, Suffolk, IP3 0FS, UK
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- Duke University Medical Center, 307 Research Drive, Durham, NC, 27710, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA.
| |
Collapse
|
62
|
Molinaro E, Romei C, Biagini A, Sabini E, Agate L, Mazzeo S, Materazzi G, Sellari-Franceschini S, Ribechini A, Torregrossa L, Basolo F, Vitti P, Elisei R. Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol 2017; 13:644-660. [PMID: 28707679 DOI: 10.1038/nrendo.2017.76] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare malignancy, accounting for 1-2% of all thyroid cancers. Although rare, ATC accounts for the majority of deaths from thyroid carcinoma. ATC often originates in a pre-existing thyroid cancer lesion, as suggested by the simultaneous presence of areas of differentiated or poorly differentiated thyroid carcinoma. ATC is characterized by the accumulation of several oncogenic alterations, and studies have shown that an increased number of oncogenic alterations equates to an increased level of dedifferentiation and aggressiveness. The clinical management of ATC requires a multidisciplinary approach; according to recent American Thyroid Association guidelines, surgery, radiotherapy and/or chemotherapy should be considered. In addition to conventional therapies, novel molecular targeted therapies are the most promising emerging treatment modalities. These drugs are often multiple receptor tyrosine kinase inhibitors, several of which have been tested in clinical trials with encouraging results so far. Accordingly, clinical trials are ongoing to evaluate the safety, efficacy and effectiveness of these new agents. This Review describes the updated clinical and pathological features of ATC and provides insight into the molecular biology of this disease. The most recent literature regarding conventional, newly available and future therapies for ATC is also discussed.
Collapse
Affiliation(s)
- Eleonora Molinaro
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Agnese Biagini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Elena Sabini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Laura Agate
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Salvatore Mazzeo
- Diagnostic and Interventional Radiology Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa
| | - Gabriele Materazzi
- Division of Endocrine Surgery, Department of Surgical Pathology, University Hospital of Pisa
| | | | | | - Liborio Torregrossa
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Fulvio Basolo
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Paolo Vitti
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| |
Collapse
|
63
|
Vilanova G, Colominas I, Gomez H. A mathematical model of tumour angiogenesis: growth, regression and regrowth. J R Soc Interface 2017; 14:rsif.2016.0918. [PMID: 28100829 DOI: 10.1098/rsif.2016.0918] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/19/2016] [Indexed: 12/14/2022] Open
Abstract
Cancerous tumours have the ability to recruit new blood vessels through a process called angiogenesis. By stimulating vascular growth, tumours get connected to the circulatory system, receive nutrients and open a way to colonize distant organs. Tumour-induced vascular networks become unstable in the absence of tumour angiogenic factors (TAFs). They may undergo alternating stages of growth, regression and regrowth. Following a phase-field methodology, we propose a model of tumour angiogenesis that reproduces the aforementioned features and highlights the importance of vascular regression and regrowth. In contrast with previous theories which focus on vessel remodelling due to the absence of flow, we model an alternative regression mechanism based on the dependency of tumour-induced vascular networks on TAFs. The model captures capillaries at full scale, the plastic dynamics of tumour-induced vessel networks at long time scales, and shows the key role played by filopodia during angiogenesis. The predictions of our model are in agreement with in vivo experiments and may prove useful for the design of antiangiogenic therapies.
Collapse
Affiliation(s)
- Guillermo Vilanova
- Departamento de Métodos Matemáticos e de Representación, Grupo de Métodos Numéricos en Ingeniería-Universidade da Coruña, Campus de Elviña, 15071 A Coruña, Spain
| | - Ignasi Colominas
- Departamento de Métodos Matemáticos e de Representación, Grupo de Métodos Numéricos en Ingeniería-Universidade da Coruña, Campus de Elviña, 15071 A Coruña, Spain
| | - Hector Gomez
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| |
Collapse
|
64
|
Li M, Kroetz DL. Bevacizumab-induced hypertension: Clinical presentation and molecular understanding. Pharmacol Ther 2017; 182:152-160. [PMID: 28882537 DOI: 10.1016/j.pharmthera.2017.08.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bevacizumab is a vascular endothelial growth factor-A-specific angiogenesis inhibitor indicated as an adjunct to chemotherapy for the treatment of several types of cancer. Hypertension is commonly observed during bevacizumab treatment, and high-grade toxicity can limit therapy and lead to other cardiovascular complications. The factors that contribute to interindividual variability in blood pressure response to bevacizumab treatment are not well understood. In this review, we outline research efforts to understand the mechanisms and pathophysiology of hypertension resulting from bevacizumab treatment. Moreover, we highlight current knowledge of the pharmacogenetics of bevacizumab-induced hypertension, which may be used to develop strategies to prevent or minimize this toxicity.
Collapse
Affiliation(s)
- Megan Li
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
65
|
High-circulating Tie2 Is Associated With Pathologic Complete Response to Chemotherapy and Antiangiogenic Therapy in Breast Cancer. Am J Clin Oncol 2017; 39:248-54. [PMID: 24577164 DOI: 10.1097/coc.0000000000000046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Vascular endothelial growth factor (VEGF) is a central mediator of angiogenesis in breast cancer. Research in antiangiogenic cancer treatment has been marked by the development of the monoclonal antibody bevacizumab, which targets VEGF in many solid tumors. As patients do not equally benefit from bevacizumab, it has become necessary to define the profile of patients who will benefit from the drug. MATERIALS AND METHODS We have conducted a prospective phase II study in 39 patients using bevacizumab in breast cancer in the neoadjuvant setting, and found improved pathologic complete response (pCR) when bevacizumab was added to chemotherapy in patients with hormone receptor negative and invasive ductal carcinoma. Blood samples were collected at baseline and serially while patients were on treatment. Circulating angiogenesis-related proteins angiopoietin (ANG)1, ANG2, basic fibroblast growth factor, IL-1a, matrix metalloproteinase 9, platelet derived growth factor - BB, platelet endothelial cell adhesion molecule -1, Tie2, VEGF, and vascular endothelial growth factor receptor 2 were measured at baseline and during treatment. This correlative study was conducted to identify specific serum angiogenic factor profiles that might be associated with pCR in the neoadjuvant setting in breast cancer patients receiving bevacizumab and chemotherapy. RESULTS Elevated baseline serum Tie2 and basic fibroblast growth factor were associated with pCR in response to this combination. Changes in serum levels of these proteins were seen during treatment but were not significantly different between the pCR and non-pCR groups. CONCLUSIONS Baseline-circulating Tie2 levels may help distinguish patients who will have pCR from those who will not and may form the basis for future development of antiangiogenic therapy in breast cancer. Larger studies are needed to validate these findings. ClinicalTrials.gov Identifier: NCT00203502.
Collapse
|
66
|
Molecular mechanisms for vascular complications of targeted cancer therapies. Clin Sci (Lond) 2017; 130:1763-79. [PMID: 27612952 DOI: 10.1042/cs20160246] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/22/2016] [Indexed: 12/15/2022]
Abstract
Molecularly targeted anti-cancer therapies have revolutionized cancer treatment by improving both quality of life and survival in cancer patients. However, many of these drugs are associated with cardiovascular toxicities that are sometimes dose-limiting. Moreover, the long-term cardiovascular consequences of these drugs, some of which are used chronically, are not yet known. Although the scope and mechanisms of the cardiac toxicities are better defined, the mechanisms for vascular toxicities are only beginning to be elucidated. This review summarizes what is known about the vascular adverse events associated with three classes of novel anti-cancer therapies: vascular endothelial growth factor (VEGF) inhibitors, breakpoint cluster-Abelson (BCR-ABL) kinase inhibitors used to treat chronic myelogenous leukaemia (CML) and immunomodulatory agents (IMiDs) used in myeloma therapeutics. Three of the best described vascular toxicities are reviewed including hypertension, increased risk of acute cardiovascular ischaemic events and arteriovenous thrombosis. The available data regarding the mechanism by which each therapy causes vascular complication are summarized. When data are limited, potential mechanisms are inferred from the known effects of inhibiting each target on vascular cell function and disease. Enhanced understanding of the molecular mechanisms of vascular side effects of targeted cancer therapy is necessary to effectively manage cancer patients and to design safer targeted cancer therapies for the future.
Collapse
|
67
|
Ayar O, Alpay A, Koban Y, Akdemir MO, Yazgan S, Canturk Ugurbas S, Ugurbas SH. The Effect of Dexamethasone Intravitreal Implant on Retinal Nerve Fiber Layer in Patients Diagnosed with Branch Retinal Vein Occlusion. Curr Eye Res 2017. [PMID: 28632411 DOI: 10.1080/02713683.2017.1313430] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate the effect of a single dose of intravitreal dexamethasone (DEX) implant on retinal nerve fiber layer (RNFL) thickness in patients with branch retinal vein occlusion (BRVO) in a 6-month period. MATERIALS AND METHODS This retrospective observational study included the patients with BRVO who received intravitreal DEX implant and whose assessment included the baseline RNFL thickness measurements. The data of 26 eyes of 24 patients were retrospectively analyzed. Spectral domain optic coherence tomography was used to measure peripapillary RNFL thickness in six regional subfields. Intraocular pressure (IOP) values at each visit were recorded. The data of single dose DEX implant during 6 months were assessed. RESULTS The mean preoperative and postoperative 6th month nasal RNFL values were 85.4 ± 23.0 μm and 82.1 ± 17.6 μm, respectively, and the difference between the measurements was not statistically significant (p = 0.372). There was a slight decrease in the mean RNFL values postoperatively compared to the baseline values in all quadrants except supero-temporal quadrant; however, none of them reached statistically significant level (p > 0.05). The mean IOP values before and 6 months after implantation were 15.7 ± 2.9 mmHg and 16.5 ± 4.2 mmHg, respectively. The difference between the 6th month IOP values and baseline IOP values was not statistically significant (p = 0.236). CONCLUSION Intravitreal DEX implant seems to have no adverse effect on RNFL thickness in BRVO patients in a 6-month period.
Collapse
Affiliation(s)
- Orhan Ayar
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Atilla Alpay
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Yaran Koban
- b Faculty of Medicine, Department of Ophthalmology , Kafkas University , Kars , Turkey
| | - Mehmet Orcun Akdemir
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Serpil Yazgan
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Sılay Canturk Ugurbas
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Suat Hayri Ugurbas
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| |
Collapse
|
68
|
Matrone A, Valerio L, Pieruzzi L, Giani C, Cappagli V, Lorusso L, Agate L, Puleo L, Viola D, Bottici V, Del Re M, Molinaro E, Danesi R, Elisei R. Protein kinase inhibitors for the treatment of advanced and progressive radiorefractory thyroid tumors: From the clinical trials to the real life. Best Pract Res Clin Endocrinol Metab 2017; 31:319-334. [PMID: 28911728 DOI: 10.1016/j.beem.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The last ten years have been characterized by the introduction in the clinical practice of new drugs named tyrosine kinase inhibitors for the treatment of several human tumors. After the positive conclusion of two international multicentric, randomized phase III clinical trials, two of these drugs, sorafenib and lenvatinib, have been recently approved and they are now available for the treatment of advanced and progressive radioiodine refractory thyroid tumors. We have been involved in most clinical trials performed with different tyrosine kinase inhibitors in different histotypes of thyroid cancer thus acquiring a lot of experience in the management of both drugs and their adverse events. Aim of this review is to give an overview of both the rationale for the use of these inhibitors in thyroid cancer and the major results of the clinical trials. Some suggestions for the management of treated patients in the real life are also provided.
Collapse
Affiliation(s)
- Antonio Matrone
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Laura Valerio
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Letizia Pieruzzi
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Carlotta Giani
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Virginia Cappagli
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Loredana Lorusso
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Laura Agate
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Luciana Puleo
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - David Viola
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Valeria Bottici
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Marzia Del Re
- Unit of Pharmacology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Eleonora Molinaro
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Romano Danesi
- Unit of Pharmacology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Rossella Elisei
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy.
| |
Collapse
|
69
|
Heinolainen K, Karaman S, D'Amico G, Tammela T, Sormunen R, Eklund L, Alitalo K, Zarkada G. VEGFR3 Modulates Vascular Permeability by Controlling VEGF/VEGFR2 Signaling. Circ Res 2017; 120:1414-1425. [PMID: 28298294 DOI: 10.1161/circresaha.116.310477] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 01/02/2023]
Abstract
RATIONALE Vascular endothelial growth factor (VEGF) is the main driver of angiogenesis and vascular permeability via VEGF receptor 2 (VEGFR2), whereas lymphangiogenesis signals are transduced by VEGFC/D via VEGFR3. VEGFR3 also regulates sprouting angiogenesis and blood vessel growth, but to what extent VEGFR3 signaling controls blood vessel permeability remains unknown. OBJECTIVE To investigate the role of VEGFR3 in the regulation of VEGF-induced vascular permeability. METHODS AND RESULTS Long-term global Vegfr3 gene deletion in adult mice resulted in increased fibrinogen deposition in lungs and kidneys, indicating enhanced vascular leakage at the steady state. Short-term deletion of Vegfr3 in blood vascular endothelial cells increased baseline leakage in various tissues, as well as in tumors, and exacerbated vascular permeability in response to VEGF, administered via intradermal adenoviral delivery or through systemic injection of recombinant protein. VEGFR3 gene silencing upregulated VEGFR2 protein levels and phosphorylation in cultured endothelial cells. Consistent with elevated VEGFR2 activity, vascular endothelial cadherin showed reduced localization at endothelial cell-cell junctions in postnatal retinas after Vegfr3 deletion, or after VEGFR3 silencing in cultured endothelial cells. Furthermore, concurrent deletion of Vegfr2 prevented VEGF-induced excessive vascular leakage in mice lacking Vegfr3. CONCLUSIONS VEGFR3 limits VEGFR2 expression and VEGF/VEGFR2 pathway activity in quiescent and angiogenic blood vascular endothelial cells, thereby preventing excessive vascular permeability.
Collapse
Affiliation(s)
- Krista Heinolainen
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Sinem Karaman
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Gabriela D'Amico
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Tuomas Tammela
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Raija Sormunen
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Lauri Eklund
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Kari Alitalo
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Georgia Zarkada
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.).
| |
Collapse
|
70
|
Hargreaves A, Bigley A, Price S, Kendrew J, Barry ST. Automated image analysis of intra-tumoral and peripheral endocrine organ vascular bed regression using 'Fibrelength' as a novel structural biomarker. J Appl Toxicol 2017; 37:902-912. [PMID: 28186326 DOI: 10.1002/jat.3438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The study of vascular modulation has received a great deal of attention in recent years as knowledge has increased around the role of angiogenesis within disease contexts such as cancer. Despite rapidly expanding insights into the molecular processes involved and the concomitant generation of a number of anticancer vascular modulating chemotherapeutics, techniques used in the measurement of structural vascular change have advanced more modestly, particularly with regard to the preclinical quantification of off-target vascular regression within systemic, notably endocrine, blood vessels. Such changes translate into a number of major clinical side effects and there remains a need for improved preclinical screening and analysis. Here we present the generation of a novel structural biomarker, which can be incorporated into a number of contemporary image analysis platforms and used to compare tumour versus systemic host tissue vascularity. By contrasting the measurements obtained, the preclinical efficacy of vascular modulating chemotherapies can be evaluated in light of the predicted therapeutic window. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Adam Hargreaves
- PathCelerate Ltd, The BioHub, Alderley Park, Mereside, Alderley Edge, Cheshire, UK
| | - Alison Bigley
- Oraclebio Ltd, BioCity Scotland, North Lanarkshire, Scotland, UK
| | - Shirley Price
- University of Surrey, 3660 Office of the Vice-Provost, Guildford, Surrey, UK
| | - Jane Kendrew
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| | - Simon T Barry
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| |
Collapse
|
71
|
Abstract
PURPOSE To compare the structural characteristics of the choroid in the areas with greater retinal degeneration to the areas with less retinal degeneration in eyes with retinitis pigmentosa (RP). METHODS Patients with RP who had a hyperautofluorescent ring were studied. The choroidal images obtained by enhanced depth imaging optical coherence tomography located 7,500 μm from the optic disk in the horizontal plane were analyzed. The cross-sectional areas of the total, luminal, and stromal choroid were measured. The area within the hyperautofluorescent ring was defined as the "central choroid" with less retinal degeneration. RESULTS Thirty-seven eyes of 24 patients with RP were studied. The cross-sectional area of the total choroid was significantly smaller in the RP eyes than that in the control eyes (P < 0.01). The stromal areas of the choroid were not significantly different from the stromal areas of the controls. However, the luminal areas of the nasal and temporal choroid in the RP eyes were significantly smaller than that of the corresponding areas of the controls. The ratio of the luminal area to the total choroidal area in the central choroid was 68.0 ± 3.3% which was significantly larger than that of the nasal or the temporal choroid (P < 0.01). CONCLUSION The choroidal structure is differentially altered in eyes with RP. The changes in the choroid were dependent on whether they were located within the hyperautofluorescent or outside the hyperautofluorescent ring.
Collapse
|
72
|
Payne SL, Peacock HM, Vickaryous MK. Blood vessel formation during tail regeneration in the leopard gecko (Eublepharis macularius): The blastema is not avascular. J Morphol 2017; 278:380-389. [PMID: 28078708 DOI: 10.1002/jmor.20648] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
Unique among amniotes, many lizards are able to self-detach (autotomize) their tail and then regenerate a replacement. Tail regeneration involves the formation of a blastema, an accumulation of proliferating cells at the site of autotomy. Over time, cells of the blastema give rise to most of the tissues in the replacement tail. In non-amniotes capable of regenerating (such as urodeles and some teleost fish), the blastema is reported to be essentially avascular until tissue differentiation takes place. For tail regenerating lizards less is known. Here, we investigate neovascularization during tail regeneration in the leopard gecko (Eublepharis macularius). We demonstrate that the gecko tail blastema is not an avascular structure. Beginning with the onset of regenerative outgrowth, structurally mature (mural cell supported) blood vessels are found within the blastema. Although the pattern of blood vessel distribution in the regenerate tail differs from that of the original, a hierarchical network is established, with vessels of varying luminal diameters and wall thicknesses. Using immunostaining, we determine that blastema outgrowth and tissue differentiation is characterized by a dynamic interplay between the pro-angiogenic protein vascular endothelial growth factor (VEGF) and the anti-angiogenic protein thrombospondin-1 (TSP-1). VEGF-expression is initially widespread, but diminishes as tissues differentiate. In contrast, TSP-1 expression is initially restricted but becomes more abundant as VEGF-expression wanes. We predict that variation in the neovascular response observed between different regeneration-competent species likely relates to the volume of the blastema. J. Morphol. 278:380-389, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Hanna M Peacock
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Matthew K Vickaryous
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
73
|
Semeniuk-Wojtaś A, Lubas A, Stec R, Szczylik C, Niemczyk S. Influence of Tyrosine Kinase Inhibitors on Hypertension and Nephrotoxicity in Metastatic Renal Cell Cancer Patients. Int J Mol Sci 2016; 17:ijms17122073. [PMID: 27941701 PMCID: PMC5187873 DOI: 10.3390/ijms17122073] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/06/2016] [Accepted: 12/02/2016] [Indexed: 01/09/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common kidney malignancies. An upgraded comprehension of the molecular biology implicated in the development of cancer has stimulated an increase in research and development of innovative antitumor therapies. The aim of the study was to analyze the medical literature for hypertension and renal toxicities as the adverse events of the vascular endothelial growth factor (VEGF) signaling pathway inhibitor (anti-VEGF) therapy. Relevant studies were identified in PubMed and ClinicalTrials.gov databases. Eligible studies were phase III and IV prospective clinical trials, meta-analyses and retrospective studies that had described events of hypertension or nephrotoxicity for patients who received anti-VEGF therapy. A total of 48 studies were included in the systematic review. The incidence of any grade hypertension ranged from 17% to 49.6%. Proteinuria and increased creatinine levels were ascertained in 8% to 73% and 5% to 65.6% of patients, respectively. These adverse events are most often mild in severity but may sometimes lead to treatment discontinuation. Nephrotoxicity and hypertension are related to multiple mechanisms; however, one of the main disturbances in those patients is VEGF inhibition. There is a significant risk of developing hypertension and renal dysfunction among patients receiving anti-VEGF treatment; however, there is also some evidence that these side effects may be used as biomarkers of response to antiangiogenic agents.
Collapse
Affiliation(s)
| | - Arkadiusz Lubas
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Rafał Stec
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Cezary Szczylik
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Stanisław Niemczyk
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| |
Collapse
|
74
|
Burkhardt D, Bartosova M, Schaefer B, Grabe N, Lahrmann B, Nasser H, Freise C, Schneider A, Lingnau A, Degenhardt P, Ranchin B, Sallay P, Cerkauskiene R, Malina M, Ariceta G, Schmitt CP, Querfeld U. Reduced Microvascular Density in Omental Biopsies of Children with Chronic Kidney Disease. PLoS One 2016; 11:e0166050. [PMID: 27846250 PMCID: PMC5113061 DOI: 10.1371/journal.pone.0166050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/21/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Endothelial dysfunction is an early manifestation of cardiovascular disease (CVD) and consistently observed in patients with chronic kidney disease (CKD). We hypothesized that CKD is associated with systemic damage to the microcirculation, preceding macrovascular pathology. To assess the degree of "uremic microangiopathy", we have measured microvascular density in biopsies of the omentum of children with CKD. PATIENTS AND METHODS Omental tissue was collected from 32 healthy children (0-18 years) undergoing elective abdominal surgery and from 23 age-matched cases with stage 5 CKD at the time of catheter insertion for initiation of peritoneal dialysis. Biopsies were analyzed by independent observers using either a manual or an automated imaging system for the assessment of microvascular density. Quantitative immunohistochemistry was performed for markers of autophagy and apoptosis, and for the abundance of the angiogenesis-regulating proteins VEGF-A, VEGF-R2, Angpt1 and Angpt2. RESULTS Microvascular density was significantly reduced in uremic children compared to healthy controls, both by manual imaging with a digital microscope (median surface area 0.61% vs. 0.95%, p<0.0021 and by automated quantification (total microvascular surface area 0.89% vs. 1.17% p = 0.01). Density measured by manual imaging was significantly associated with age, height, weight and body surface area in CKD patients and healthy controls. In multivariate analysis, age and serum creatinine level were the only independent, significant predictors of microvascular density (r2 = 0.73). There was no immunohistochemical evidence for apoptosis or autophagy. Quantitative staining showed similar expression levels of the angiogenesis regulators VEGF-A, VEGF-receptor 2 and Angpt1 (p = 0.11), but Angpt2 was significantly lower in CKD children (p = 0.01). CONCLUSIONS Microvascular density is profoundly reduced in omental biopsies of children with stage 5 CKD and associated with diminished Angpt2 signaling. Microvascular rarefaction could be an early systemic manifestation of CKD-induced cardiovascular disease.
Collapse
Affiliation(s)
- Dorothea Burkhardt
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Grabe
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Bernd Lahrmann
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Hamoud Nasser
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Freise
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Schneider
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Lingnau
- Department of Pediatric Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Degenhardt
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Surgery, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Bruno Ranchin
- Hospices Civils de Lyon, Service de Nephrologie Pediatrique and Epicime-Centre d’Investigation Clinique 1407, Hopital Femme Mere Enfant, Lyon, France
| | - Peter Sallay
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Rimante Cerkauskiene
- Coordinating Centre for Children’s Rare Diseases, Children´s Hospital, Affiliate of Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Michal Malina
- Department of Pediatrics, Second Faculty of Medicine, Charles University-Prague, Prague 5, Czech Republic
| | - Gema Ariceta
- Servicio de Nefrología Pediátrica, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Uwe Querfeld
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
75
|
Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain. Biomaterials 2016; 105:145-155. [PMID: 27521617 DOI: 10.1016/j.biomaterials.2016.07.028] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/15/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
Stem cell therapies have shown promise in promoting recovery in stroke but have been limited by poor cell survival and differentiation. We have developed a hyaluronic acid (HA)-based self-polymerizing hydrogel that serves as a platform for adhesion of structural motifs and a depot release for growth factors to promote transplant stem cell survival and differentiation. We took an iterative approach in optimizing the complex combination of mechanical, biochemical and biological properties of an HA cell scaffold. First, we optimized stiffness for a minimal reaction of adjacent brain to the transplant. Next hydrogel crosslinkers sensitive to matrix metalloproteinases (MMP) were incorporated as they promoted vascularization. Finally, candidate adhesion motifs and growth factors were systemically changed in vitro using a design of experiment approach to optimize stem cell survival or proliferation. The optimized HA hydrogel, tested in vivo, promoted survival of encapsulated human neural progenitor cells (iPS-NPCs) after transplantation into the stroke core and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. This HA hydrogel can be tracked in vivo with MRI. A hydrogel can serve as a therapeutic adjunct in a stem cell therapy through selective control of stem cell survival and differentiation in vivo.
Collapse
|
76
|
Karovic S, Shiuan EF, Zhang SQ, Cao H, Maitland ML. Patient-Level Adverse Event Patterns in a Single-Institution Study of the Multi-Kinase Inhibitor Sorafenib. Clin Transl Sci 2016; 9:260-266. [PMID: 27443985 PMCID: PMC5350995 DOI: 10.1111/cts.12408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/27/2016] [Indexed: 01/05/2023] Open
Abstract
Novel characterization of patterns of adverse events (AEs) of kinase inhibitors (KIs) could reveal new insights on human molecular physiology and methods to improve the therapeutic index of KIs. Incidence and severity of AEs for each of 157 patients enrolled in sorafenib clinical trials were determined for three clinically relevant treatment intervals: weeks 0–3, weeks 3–7, and after 7 weeks. The most common within patient co‐occurrences were mucositis with dermatologic events: hand‐foot syndrome (HFS; odds ratio [OR] = 4.36; p = 0.0017) and rash (OR = 5.32; p < 0.001). Prevalence of severe: alopecia (p = 0.02), diarrhea (p < 0.001), and fatigue (p = 0.005) increased over the course of therapy. Incidence of HFS (60%) and diarrhea (25%) increased up to a minimum steady‐state concentration (approximately 5 mcg mL‐1) and plateaued thereafter. Common AEs of sorafenib occur in distinct temporal and tissue distribution patterns and this analysis identified unrecognized relationships among mechanism‐dependent and independent effects of a KI.
Collapse
Affiliation(s)
- S Karovic
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - E F Shiuan
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - S Q Zhang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - H Cao
- Department of Health Studies, University of Chicago, Chicago, Illinois, USA
| | - M L Maitland
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, USA.,Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
77
|
Hollanders K, Hove IV, Sergeys J, Bergen TV, Lefevere E, Kindt N, Castermans K, Vandewalle E, van Pelt J, Moons L, Stalmans I. AMA0428, A Potent Rock Inhibitor, Attenuates Early and Late Experimental Diabetic Retinopathy. Curr Eye Res 2016; 42:260-272. [PMID: 27399806 DOI: 10.1080/02713683.2016.1183030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE Diabetic retinopathy (DR) is characterized by an early stage of inflammation and vessel leakage, and an advanced vasoproliferative stage. Also, neurodegeneration might play an important role in disease pathogenesis. The aim of this study was to investigate the effect of the Rho kinase (ROCK) inhibitor, AMA0428, on these processes. METHODS The response to ROCK inhibition by AMA0428 (1 µg) was studied in vivo using the murine model for streptozotocin (STZ)-induced diabetes, focusing on early non-proliferative DR features and the oxygen-induced retinopathy (OIR) model to investigate proliferative DR. Intravitreal (IVT) administration of AMA0428 was compared with murine anti-VEGF-R2 antibody (DC101, 6.2 µg) and placebo (H2O/PEG; 1C8). Outcome was assessed by analyzing leukostasis using fluorescein isothiocyanate coupled concanavalin A (FITC-ConA) and vessel leakage (bovine serum albumin conjugated with fluorescein isothiocyanate; FITC-BSA)/neovascularization and neurodegeneration by immunohistological approaches (hematoxylin and eosin (H&E), terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL), Brn3a). ELISA and Western blotting were employed to unravel the consequences of ROCK inhibition (1 µM AMA0428) on myosin phosphatase target protein (MYPT)-1 phosphorylation, endothelial nitric oxide synthase (eNOS) phosphorylation, and vascular endothelial growth factor (VEGF) levels in retinas of diabetic mice, on NF-κβ activity and ICAM-1 expression in endothelial cells (ECs). RESULTS In vivo, AMA0428 significantly reduced vessel leakage and neovascularization, respectively, in the STZ and OIR model, comparable to DC101 therapy. Additionally, the ROCK inhibitor decreased neurodegeneration in both models and inhibited leukostasis by 30% (p < 0.05) in the STZ model (p < 0.05), while DC101 had no positive effect on the outcome of these latter processes. ROCK activity was upregulated in the diabetic retina and AMA0428 administration resulted in decreased phospho-MYPT-1, enhanced phospho-eNOS, and reduced VEGF levels. In vitro, AMA0428 interfered with NF-κβ activity, thereby inhibiting ICAM-1 expression in ECs. CONCLUSIONS Targeting ROCK with AMA0428 effectively attenuated outcome in an early DR model (STZ) and a late vasoproliferative retinopathy model (OIR). These findings make AMA0428 a promising candidate with an additional anti-inflammatory and neuroprotective benefit for DR patients, as compared with anti-VEGF treatment.
Collapse
Affiliation(s)
- Karolien Hollanders
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,b Department of Ophthalmology , University Hospitals Ghent , Ghent , Belgium
| | - Inge Van Hove
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Jurgen Sergeys
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Tine Van Bergen
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium
| | - Evy Lefevere
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | | | | | - Evelien Vandewalle
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,e Department of Ophthalmology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| | - Jos van Pelt
- f Department of Hepatology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| | - Lieve Moons
- c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Ingeborg Stalmans
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,e Department of Ophthalmology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| |
Collapse
|
78
|
Abdel-Rahman O, ElHalawani H. Risk of cardiovascular adverse events in patients with solid tumors treated with ramucirumab: A meta analysis and summary of other VEGF targeted agents. Crit Rev Oncol Hematol 2016; 102:89-100. [PMID: 27129437 DOI: 10.1016/j.critrevonc.2016.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We performed a systematic review and meta-analysis of the risk of cardiovascular adverse events associated with ramucirumab. PATIENTS AND METHODS Eligible studies included randomized phase II and III trials of patients with solid tumors on ramucirumab; describing events of hypertension, bleeding, arterial/venous thrombosis and congestive heart failure. RESULTS Our search strategy yielded 160 potentially relevant citations from Pubmed/Medline, CENTRAL Cochrane registry and ASCO meeting library. After exclusion of ineligible studies, a total of 11 clinical trials were considered eligible for the meta-analysis. The RR of all-grade hypertension, bleeding, ATE, VTE and congestive heart failure were 2.83 (95% CI 2.43-3.29; p<0.0001), 1.98 (95% CI 1.77-2.21; p<0.0001); 0.97 (95% CI 0.62-1.52; p=0.91), 0.83 (95% CI 0.52-1.35; p=0.46), 1.36 (95% CI 0.77-2.4; p=0.28), respectively. CONCLUSIONS Our meta-analysis has demonstrated that ramucirumab is associated with an increased risk of hypertension and bleeding. Clinicians should be aware of this risk and perform regular clinical monitoring.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
79
|
Abstract
Aflibercept is a recombinant fusion protein that acts as a soluble decoy receptor for vascular endothelial growth factor (VEGF), a key regulator of angiogenesis. It binds to all isoforms of VEGF-A as well as VEGF-B and placental growth factor, and, thus, prevents them from binding to and activating their cognate receptors. In the USA and EU, intravenously administered aflibercept in combination with an infusion of leucovorin, fluorouracil and irinotecan (FOLFIRI) is approved for the treatment of patients with metastatic colorectal cancer that is resistant to or has progressed after treatment with an oxaliplatin-containing regimen. The efficacy of aflibercept in this indication was assessed in a multinational, pivotal phase 3 trial (VELOUR), in which the approved regimen of aflibercept 4 mg/kg every 2 weeks plus FOLFIRI significantly prolonged median overall survival by 1.44 months compared with FOLFIRI alone (primary endpoint). The addition of aflibercept also significantly prolonged progression-free survival and significantly increased the objective response rate compared with FOLFIRI alone. Addition of aflibercept to FOLFIRI was associated with anti-VEGF-related adverse events and an increased incidence of FOLFIRI-related adverse events, but the tolerability of the combination was generally acceptable in this pre-treated population. The most common grade 3 or 4 adverse events with aflibercept plus FOLFIRI included neutropenia, diarrhoea and hypertension. In conclusion, aflibercept plus FOLFIRI is a useful treatment option for patients with metastatic colorectal cancer previously treated with an oxaliplatin-containing regimen, with or without bevacizumab.
Collapse
Affiliation(s)
- Yahiya Y Syed
- Springer, Private Bag 65901, Mairangi Bay 0754, Auckland, New Zealand,
| | | |
Collapse
|
80
|
Tjostheim SS, Stepien RL, Markovic LE, Stein TJ. Effects of Toceranib Phosphate on Systolic Blood Pressure and Proteinuria in Dogs. J Vet Intern Med 2016; 30:951-7. [PMID: 27149912 PMCID: PMC5084777 DOI: 10.1111/jvim.13951] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/06/2016] [Accepted: 04/06/2016] [Indexed: 12/30/2022] Open
Abstract
Background Systemic hypertension and proteinuria are established adverse effects of tyrosine kinase inhibitor treatment in people. Objective The objective of this study was to investigate changes in systolic blood pressure and the incidence of proteinuria secondary to treatment with toceranib phosphate in dogs with cancer. Animals Twenty‐six control dogs and 30 dogs with cancer were evaluated for the first part of the study (baseline characteristics). For the second part (effect of toceranib phosphate treatment), 48 client‐owned dogs were evaluated, including 20 control dogs and 28 dogs with various types of neoplasia. Methods Prospective cohort study. Client‐owned healthy control dogs and dogs with cancer were enrolled. Blood pressure and urine protein:creatinine ratios were measured before treatment and 2 weeks after initiation of toceranib phosphate treatment. Results Systolic blood pressure was significantly (P = 0.0013) higher in previously normotensive treatment dogs after initiation of treatment with toceranib phosphate (152 mmHg ± 19) compared to baseline (136 mmHg ± 14). 37% of treated dogs developed SBP ≥ 160 mmHg. The prevalence of systemic hypertension (37%) and proteinuria (21%) at baseline in treatment dogs did not differ from that of age‐matched healthy controls (15% [P = 0.13] and 0% [P = 0.069], respectively). Conclusions and Clinical Importance Toceranib phosphate treatment might result in increased systolic blood pressures in dogs. Systemic hypertension should be considered a potential adverse effect of this drug in dogs. Systemic hypertension and proteinuria were detected at clinically relevant frequencies in the dogs with cancer before antineoplastic therapies suggesting that monitoring of these variables might be warranted in this population.
Collapse
Affiliation(s)
- S S Tjostheim
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
| | - R L Stepien
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
| | - L E Markovic
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
| | - T J Stein
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
| |
Collapse
|
81
|
Watson EC, Whitehead L, Adams RH, Dewson G, Coultas L. Endothelial cell survival during angiogenesis requires the pro-survival protein MCL1. Cell Death Differ 2016; 23:1371-9. [PMID: 26943318 DOI: 10.1038/cdd.2016.20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 01/07/2016] [Accepted: 01/29/2016] [Indexed: 01/31/2023] Open
Abstract
Angiogenesis is essential to match the size of blood vessel networks to the metabolic demands of growing tissues. While many genes and pathways necessary for regulating angiogenesis have been identified, those responsible for endothelial cell (EC) survival during angiogenesis remain largely unknown. We have investigated the in vivo role of myeloid cell leukemia 1 (MCL1), a pro-survival member of the BCL2 family, in EC survival during angiogenesis. EC-specific deletion of Mcl1 resulted in a dose-dependent increase in EC apoptosis in the angiogenic vasculature and a corresponding decline in vessel density. Our results suggest this apoptosis was independent of the BH3-only protein BIM. Despite the known link between apoptosis and blood vessel regression, this was not the cause of reduced vessel density observed in the absence of endothelial MCL1. Rather, the reduction in vessel density was linked to ectopic apoptosis in regions of the angiogenic vasculature where EC proliferation and new vessel growth occurs. We have therefore identified MCL1 as an essential survival factor for ECs that is required for blood vessel production during angiogenesis.
Collapse
Affiliation(s)
- E C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia
| | - L Whitehead
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia.,Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia
| | - R H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, Münster D, Germany.,Faculty of Medicine, University of Münster, Münster D, Germany
| | - G Dewson
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia.,Cell Signalling and Cell Death Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia
| | - L Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, 1G Royal Parade, VIC, Australia
| |
Collapse
|
82
|
Brinda BJ, Viganego F, Vo T, Dolan D, Fradley MG. Anti-VEGF-Induced Hypertension: a Review of Pathophysiology and Treatment Options. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2016; 18:33. [DOI: 10.1007/s11936-016-0452-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
83
|
Favia A, Pafumi I, Desideri M, Padula F, Montesano C, Passeri D, Nicoletti C, Orlandi A, Del Bufalo D, Sergi M, Ziparo E, Palombi F, Filippini A. NAADP-Dependent Ca(2+) Signaling Controls Melanoma Progression, Metastatic Dissemination and Neoangiogenesis. Sci Rep 2016; 6:18925. [PMID: 26733361 PMCID: PMC4702115 DOI: 10.1038/srep18925] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
A novel transduction pathway for the powerful angiogenic factor VEGF has been recently shown in endothelial cells to operate through NAADP-controlled intracellular release of Ca2+. In the present report the possible involvement of NAADP-controlled Ca2+ signaling in tumor vascularization, growth and metastatic dissemination was investigated in a murine model of VEGF-secreting melanoma. Mice implanted with B16 melanoma cells were treated with NAADP inhibitor Ned-19 every second day for 4 weeks and tumor growth, vascularization and metastatization were evaluated. Control specimens developed well vascularized tumors and lung metastases, whereas in Ned-19-treated mice tumor growth and vascularization as well as lung metastases were strongly inhibited. In vitro experiments showed that Ned-19 treatment controls the growth of B16 cells in vitro, their migratory ability, adhesive properties and VEGFR2 expression, indicating NAADP involvement in intercellular autocrine signaling. To this regard, Ca2+ imaging experiments showed that the response of B16 cells to VEGF stimulation is NAADP-dependent. The whole of these observations indicate that NAADP-controlled Ca2+ signaling can be relevant not only for neoangiogenesis but also for direct control of tumor cells.
Collapse
Affiliation(s)
- Annarita Favia
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Irene Pafumi
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Marianna Desideri
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 53 Via E. Chianesi, 00144, Rome, Italy
| | - Fabrizio Padula
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Camilla Montesano
- Department of Chemistry, SAPIENZA University of Rome, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Daniela Passeri
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Via di Tor Vegata, 00173 Rome, Italy
| | - Carmine Nicoletti
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Via di Tor Vegata, 00173 Rome, Italy
| | - Donatella Del Bufalo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 53 Via E. Chianesi, 00144, Rome, Italy
| | - Manuel Sergi
- Faculty of Biosciences and Technologies for Food, Agriculture and Environment, University of Teramo, 1 Via R. Balzarini, 64023 Teramo, Italy
| | - Elio Ziparo
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Fioretta Palombi
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology,SAPIENZA University of Rome, 16 Via A. Scarpa, 00161 Rome, Italy
| |
Collapse
|
84
|
Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovascularization: What's new? Pharmacol Res 2016; 103:253-69. [DOI: 10.1016/j.phrs.2015.11.027] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/19/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
|
85
|
Yoon MH, Kim YJ, Lee SY, Lee KW, Chin HS. Effects of Intravitreal Injection of Bevacizumab or Ranibizumab on Systemic Circulation. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2016. [DOI: 10.3341/jkos.2016.57.3.429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Myung Hun Yoon
- Department of Ophthalmology, Inha University School of Medicine, Incheon, Korea
| | - Young Jun Kim
- Department of Ophthalmology, Inha University School of Medicine, Incheon, Korea
| | - So Young Lee
- Department of Ophthalmology, Inha University School of Medicine, Incheon, Korea
| | - Kang Won Lee
- Department of Ophthalmology, Inha University School of Medicine, Incheon, Korea
| | - Hee Seung Chin
- Department of Ophthalmology, Inha University School of Medicine, Incheon, Korea
| |
Collapse
|
86
|
Takyar S, Zhang Y, Haslip M, Jin L, Shan P, Zhang X, Lee PJ. An endothelial TLR4-VEGFR2 pathway mediates lung protection against oxidant-induced injury. FASEB J 2015; 30:1317-27. [PMID: 26655705 DOI: 10.1096/fj.15-275024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/23/2015] [Indexed: 02/05/2023]
Abstract
TLR4 deficiency causes hypersusceptibility to oxidant-induced injury. We investigated the role of TLR4 in lung protection, using used bone marrow chimeras; cell-specific transgenic modeling; and lentiviral delivery in vivo to knock down or express TLR4 in various lung compartments; and lung-specific VEGF transgenic mice to investigate the effect of TLR4 on VEGF-mediated protection. C57/BL6 mice were exposed to 100% oxygen in an enclosed chamber and assessed for survival and lung injury. Primary endothelial cells were stimulated with recombinant VEGF and exposed to hyperoxia or hydrogen peroxide. Endothelium-specific expression of human TLR4 (as opposed to its expression in epithelium or immune cells) increased the survival of TLR4-deficent mice in hyperoxia by 24 h and decreased LDH release and lung cell apoptosis after 72 h of exposure by 30%. TLR4 expression was necessary and sufficient for the protective effect of VEGF in the lungs and in primary endothelial cells in culture. TLR4 knockdown inhibited VEGF signaling through VEGF receptor 2 (VEGFR2), Akt, and ERK pathways in lungs and primary endothelial cells and decreased the availability of VEGFR2 at the cell surface. These findings demonstrate a novel mechanism through which TLR4, an innate pattern receptor, interacts with an endothelial survival pathway.
Collapse
Affiliation(s)
- Seyedtaghi Takyar
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Yi Zhang
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Maria Haslip
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Lei Jin
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Peiying Shan
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Xuchen Zhang
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Patty J Lee
- *Section of Pulmonary, Critical Care, and Sleep Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA; and Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut, USA
| |
Collapse
|
87
|
O'Donnell PH, Karovic S, Karrison TG, Janisch L, Levine MR, Harris PJ, Polite BN, Cohen EEW, Fleming GF, Ratain MJ, Maitland ML. Serum C-Telopeptide Collagen Crosslinks and Plasma Soluble VEGFR2 as Pharmacodynamic Biomarkers in a Trial of Sequentially Administered Sunitinib and Cilengitide. Clin Cancer Res 2015. [PMID: 26199386 DOI: 10.1158/1078-0432.ccr-15-0427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Fit-for-purpose pharmacodynamic biomarkers could expedite development of combination antiangiogenic regimens. Plasma sVEGFR2 concentrations ([sVEGFR2]) mark sunitinib effects on the systemic vasculature. We hypothesized that cilengitide would impair microvasculature recovery during sunitinib withdrawal and could be detected through changes in [sVEGFR2]. EXPERIMENTAL DESIGN Advanced solid tumor patients received 50 mg sunitinib daily for 14 days. For the next 14 days, patients were randomized to arm A (cilengitide 2,000 mg administered intravenously twice weekly) or arm B (no treatment). The primary endpoint was change in [sVEGFR2] between days 14 and 28. A candidate pharmacodynamic biomarker of cilengitide inhibition of integrin αvβ3, serum c-telopeptide collagen crosslinks (CTx), was also measured. RESULTS Of 21 patients, 14 (7 per arm) received all treatments without interruption and had all blood samples available for analysis. The mean change and SD of [sVEGFR2] for all sunitinib-treated patients was consistent with previous data. There was no significant difference in the mean change in [sVEGFR2] from days 14 to 28 between the arms [arm A: 2.8 ng/mL; 95% confidence interval (CI), 2.1-3.6 vs. arm B: 2.0 ng/mL; 95% CI, 0.72-3.4; P = 0.22, 2-sample t test]. Additional analyses suggested (i) prior bevacizumab therapy to be associated with unusually low baseline [sVEGFR2] and (ii) sunitinib causes measurable changes in CTx. CONCLUSIONS Cilengitide had no measurable effects on any circulating biomarkers. Sunitinib caused measurable declines in serum CTx. The properties of [sVEGFR2] and CTx observed in this study inform the design of future combination antiangiogenic therapy trials.
Collapse
Affiliation(s)
- Peter H O'Donnell
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Sanja Karovic
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Theodore G Karrison
- Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois. Department of Health Studies, The University of Chicago, Chicago, Illinois
| | - Linda Janisch
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Matthew R Levine
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Pamela J Harris
- Cancer Therapy Evaluation Program of the National Cancer Institute, Bethesda, Maryland
| | - Blase N Polite
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Ezra E W Cohen
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Gini F Fleming
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Michael L Maitland
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
88
|
Verreault M, Wehbe M, Strutt D, Masin D, Anantha M, Walker D, Chu F, Backstrom I, Kalra J, Waterhouse D, Yapp DT, Bally MB. Determination of an optimal dosing schedule for combining Irinophore C™ and temozolomide in an orthotopic model of glioblastoma. J Control Release 2015; 220:348-357. [PMID: 26528901 DOI: 10.1016/j.jconrel.2015.10.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/22/2015] [Accepted: 10/28/2015] [Indexed: 12/30/2022]
Abstract
Our laboratory reported that Irinophore C™ (IrC™; a lipid-based nanoparticulate formulation of irinotecan) is effective against an orthotopic model of glioblastoma (GBM) and that treatment with IrC™ was associated with vascular normalization within the tumor. Here, the therapeutic effects of IrC™ when used in combination with temozolomide (TMZ) in concurrent and sequential treatment schedules were tested. It was anticipated that IrC™ engendered vascular normalization would increase the delivery of TMZ to the tumor and that this would be reflected by improved treatment outcomes. The approach compared equally efficacious doses of irinotecan (IRN; 50 mg/kg) and IrC™ (25 mg/kg) in order to determine if there was a unique advantage achieved when combining TMZ with IrC™. The TMZ sensitive U251MG(O) cell line (null expression of O-6-methylguanine-DNA methyltransferase (MGMT)) modified to express the fluorescent protein mKate2 was inoculated orthotopically into NOD.CB17-SCID mice and treatment was initiated 14 days later. Our results demonstrated that IrC™ and TMZ administered concurrently resulted in optimal treatment outcomes, with 50% long term survivors (>180 days) in comparison to 17% long term survivors in animals treated with IRN and TMZ or TMZ alone. Indeed, the different treatments resulted in a 353%, 222% and 280% increase in median survival time (MST) compared to untreated animals for, respectively, IrC™ combined with TMZ, IRN combined with TMZ, and TMZ alone. When TMZ was administered after completion of IRN or IrC™ dosing, an increase in median survival time of 167-174% was observed compared to untreated animals and of 67% and 74%, respectively, when IRN (50 mg/kg) and IrC™ (25mg/kg) were given as single agents. We confirmed in these studies that after completion of the Q7D×3 dosing of IrC™, but not IRN, the tumor-associated vascular was normalized as compared to untreated tumors. Specifically, reductions in the fraction of collagen IV-free CD31 staining (p<0.05) and reductions in tumor vessel diameter were observed in tumors from IrC™-treated animals when compared to tumors from untreated or IRN treated animals. Analysis by transmission electron microscopy of the ultra-structure of tumors from IrC™-treated and untreated animals revealed that tumor-associated vessels from treated animals were smaller, more organized and exhibited a morphology comparable to normal blood vessels. In conclusion, optimal treatment outcomes were achieved when IrC™ and TMZ were administered concurrently, whereas IrC™ followed by TMZ treatment given sequentially did not confer any therapeutic advantage.
Collapse
Affiliation(s)
- M Verreault
- Brain and Spine Institute Research Center, 47 Bd de l'Hopital, Paris, 75013, France.
| | - M Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver V6T 1Z3, BC, Canada
| | - D Strutt
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - D Masin
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - M Anantha
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - D Walker
- Ultrastructural Imaging, UBC James Hogg Research Laboratories (iCAPTURE), Providence Heart and Lung Institute, St. Paul's Hospital, Rm 166, 1081 Burrard St, Vancouver, BC, Canada
| | - F Chu
- Ultrastructural Imaging, UBC James Hogg Research Laboratories (iCAPTURE), Providence Heart and Lung Institute, St. Paul's Hospital, Rm 166, 1081 Burrard St, Vancouver, BC, Canada
| | - I Backstrom
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - J Kalra
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - D Waterhouse
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - D T Yapp
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver V6T 1Z3, BC, Canada
| | - M B Bally
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver V6T 1Z3, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver V6T 2B5, BC, Canada; Center for Drug Research and Development, Vancouver V6T 1Z4, BC, Canada.
| |
Collapse
|
89
|
Carter CA, Caroen SZ, Oronsky AL, Oronsky BT. Dysphonia after Bevacizumab Rechallenge: A Case Report. Case Rep Oncol 2015; 8:423-5. [PMID: 26600773 PMCID: PMC4649738 DOI: 10.1159/000441122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Inhibition of vascular endothelial growth factor (VEGF) signaling, an initiator of tumor angiogenesis, inhibits tumor growth and invasion. Bevacizumab, a monoclonal antibody to VEGF, in common use as an adjunct to standard chemotherapy like irinotecan in advanced colorectal cancer, also affects the normal (nontumor) vasculature. Dysphonia or voice changes have been anecdotally reported in patients that have been exposed to antiangiogenics. In this case report, we present an occurrence of severe dysphonia in a 60-year-old male with metastatic colorectal cancer after reintroduction of irinotecan and bevacizumab. To our knowledge, this is the first case of dysphonia associated with bevacizumab rechallenge.
Collapse
Affiliation(s)
- Corey A Carter
- Walter Reed National Military Medical Center, Bethesda, Md., USA
| | | | | | | |
Collapse
|
90
|
Mechanisms of Vessel Pruning and Regression. Dev Cell 2015; 34:5-17. [PMID: 26151903 DOI: 10.1016/j.devcel.2015.06.004] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/26/2015] [Accepted: 06/03/2015] [Indexed: 01/27/2023]
Abstract
The field of angiogenesis research has primarily focused on the mechanisms of sprouting angiogenesis. Yet vascular networks formed by vessel sprouting subsequently undergo extensive vascular remodeling to form a functional and mature vasculature. This "trimming" includes distinct processes of vascular pruning, the regression of selected vascular branches. In some situations complete vascular networks may undergo physiological regression. Vessel regression is an understudied yet emerging field of research. This review summarizes the state-of-the-art of vessel pruning and regression with a focus on the cellular processes and the molecular regulators of vessel maintenance and regression.
Collapse
|
91
|
Retinal region-dependent susceptibility of capillaries to high-concentration oxygen exposure and vascular endothelial growth factor receptor inhibition in neonatal mice. J Pharmacol Sci 2015; 129:107-18. [PMID: 26403253 DOI: 10.1016/j.jphs.2015.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 01/08/2023] Open
Abstract
Retinal blood flow insufficiency due to capillary loss induces hypoxia in the retina, leading to an abnormal angiogenesis, relating to ischemic retinopathy. To better understand the mechanism and process of retinal capillary regression, we examined the process of hyperoxia- and vascular endothelial growth factor receptor (VEGFR) inhibitor-induced retinal capillary regression in neonatal mice. We also investigated the effects of Ca(2+) channel blockers, amlodipine and nicardipine, on hyperoxia-induced capillary regression. The regression of capillaries adjacent to arteries began immediately after the mice were exposed to 80% oxygen on postnatal day 7. An apparent avascular zone was established within 24 h after the initiation of oxygen exposure, whereas capillaries in the retinal vascular front were not affected. Axitinib, an inhibitor of VEGFR tyrosine kinase, induced capillary regression throughout the retinal vasculature. High-concentration oxygen exposure affected the capillaries on the arterial side of the retinal circulation more preferentially than axitinib. The Ca(2+) channel blockers significantly delayed hyperoxia-induced capillary regression and changes in the capillaries on the arterial side. These results suggest that the decreased blood flow due to arterial constriction contributes to hyperoxia-induced capillary regression. Compounds that improve the retinal blood flow may prevent ischemia by preventing capillary loss.
Collapse
|
92
|
Abstract
Angiogenesis is the process through which new blood vessels are formed from pre-existing vessels and is essential for the growth of all solid tumors. Vascular endothelial growth factor (VEGF) is a regulator of angiogenesis, which is crucial for tumor growth and metastasis. Its inhibition with antiangiogenic drugs is thought to improve delivery of chemotherapy through vascular normalization and disruption of tumor vasculature. Aflibercept is a recombinant fusion protein of the VEGF receptor (VEGFR)1 and VEGFR2 extracellular domains that binds to VEGF-A, VEGF-B, placental growth factor (PlGF) 1 and 2. Aflibercept has demonstrated preclinical efficacy in different tumor types and exerts its antiangiogenic effects through regression of tumor vasculature, remolding of vasculature, and inhibition of new tumor vessel growth. This review examines the effects of aflibercept on tumor vasculature and on different types of solid tumors, and explores the preclinical and clinical benefits of inclusion aflibercept into anticancer treatment strategies.
Collapse
Affiliation(s)
- Vincenzo Ricci
- Oncology Department, San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| | - Monica Ronzoni
- Oncology Department, San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| | - Teresa Fabozzi
- San Raffaele Institute, 60, Olgettina St., 20132 Milan, Italy
| |
Collapse
|
93
|
Lopez-Pastrana J, Ferrer LM, Li YF, Xiong X, Xi H, Cueto R, Nelson J, Sha X, Li X, Cannella AL, Imoukhuede PI, Qin X, Choi ET, Wang H, Yang XF. Inhibition of Caspase-1 Activation in Endothelial Cells Improves Angiogenesis: A NOVEL THERAPEUTIC POTENTIAL FOR ISCHEMIA. J Biol Chem 2015; 290:17485-94. [PMID: 26037927 DOI: 10.1074/jbc.m115.641191] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 12/12/2022] Open
Abstract
Deficient angiogenesis may contribute to worsen the prognosis of myocardial ischemia, peripheral arterial disease, ischemic stroke, etc. Dyslipidemic and inflammatory environments attenuate endothelial cell (EC) proliferation and angiogenesis, worsening the prognosis of ischemia. Under these dyslipidemic and inflammatory environments, EC-caspase-1 becomes activated and induces inflammatory cell death that is defined as pyroptosis. However, the underlying mechanism that correlates caspase-1 activation with angiogenic impairment and the prognosis of ischemia remains poorly defined. By using flow cytometric analysis, enzyme and receptor inhibitors, and hind limb ischemia model in caspase-1 knock-out (KO) mice, we examined our novel hypothesis, i.e. inhibition of caspase-1 in ECs under dyslipidemic and inflammatory environments attenuates EC pyroptosis, improves EC survival mediated by vascular endothelial growth factor receptor 2 (VEGFR-2), angiogenesis, and the prognosis of ischemia. We have made the following findings. Proatherogenic lipids induce higher caspase-1 activation in larger sizes of human aortic endothelial cells (HAECs) than in smaller sizes of HAECs. Proatherogenic lipids increase pyroptosis significantly more in smaller sizes of HAECs than in larger sizes of the cells. VEGFR-2 inhibition increases caspase-1 activation in HAECs induced by lysophosphatidylcholine treatment. Caspase-1 activation inhibits VEGFR-2 expression. Caspase-1 inhibition improves the tube formation of lysophosphatidylcholine-treated HAECs. Finally, caspase-1 depletion improves angiogenesis and blood flow in mouse hind limb ischemic tissues. Our results have demonstrated for the first time that inhibition of proatherogenic caspase-1 activation in ECs improves angiogenesis and the prognosis of ischemia.
Collapse
Affiliation(s)
- Jahaira Lopez-Pastrana
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Lucas M Ferrer
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, the Department of Bioengineering, University of Illinois-Urbana Champaign, Urbana, Illinois 61801
| | - Ya-Feng Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xinyu Xiong
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Hang Xi
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Ramon Cueto
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Jun Nelson
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xiaojin Sha
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xinyuan Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Ann L Cannella
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Princess I Imoukhuede
- the Department of Bioengineering, University of Illinois-Urbana Champaign, Urbana, Illinois 61801
| | | | - Eric T Choi
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Surgery, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Hong Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Xiao-Feng Yang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology,
| |
Collapse
|
94
|
Yukiura H, Kano K, Kise R, Inoue A, Aoki J. Autotaxin overexpression causes embryonic lethality and vascular defects. PLoS One 2015; 10:e0126734. [PMID: 25992708 PMCID: PMC4438000 DOI: 10.1371/journal.pone.0126734] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/07/2015] [Indexed: 11/19/2022] Open
Abstract
Autotaxin (ATX) is a secretory protein, which converts lysophospholipids to lysophosphatidic acid (LPA), and is essential for embryonic vascular formation. ATX is abundantly detected in various biological fluids and its level is elevated in some pathophysiological conditions. However, the roles of elevated ATX levels remain to be elucidated. In this study, we generated conditional transgenic (Tg) mice overexpressing ATX and examined the effects of excess LPA signalling. We found that ATX overexpression in the embryonic period caused severe vascular defects and was lethal around E9.5. ATX was conditionally overexpressed in the neonatal period using the Cre/loxP system, which resulted in a marked increase in the plasma LPA level. This resulted in retinal vascular defects including abnormal vascular plexus and increased vascular regression. Our findings indicate that the ATX level must be carefully regulated to ensure coordinated vascular formation
Collapse
Affiliation(s)
- Hiroshi Yukiura
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3, Aoba, Aramaki, Aoba-ku, Sendai, 980–8578, Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3, Aoba, Aramaki, Aoba-ku, Sendai, 980–8578, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3, Aoba, Aramaki, Aoba-ku, Sendai, 980–8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3, Aoba, Aramaki, Aoba-ku, Sendai, 980–8578, Japan
- PREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3, Aoba, Aramaki, Aoba-ku, Sendai, 980–8578, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
- * E-mail:
| |
Collapse
|
95
|
Alameddine RS, Yakan AS, Skouri H, Mukherji D, Temraz S, Shamseddine A. Cardiac and vascular toxicities of angiogenesis inhibitors: The other side of the coin. Crit Rev Oncol Hematol 2015; 96:195-205. [PMID: 26037841 DOI: 10.1016/j.critrevonc.2015.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/02/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the best-described tumor hallmarks. Targeting angiogenesis is becoming a successful strategy to suppress cancer growth. Vascular endothelial growth factor (VEGF), the fulcrum of angiogenesis, contributes to vascular and cardiac homeostasis. Angiogenesis inhibitors classically associated with vascular side effects are increasingly recognized for cardiac adverse effects as reflected by several meta-analyses. A global approach to these findings is a pressing need, and future strategies involving collaboration among different medical specialties are highly encouraged.
Collapse
Affiliation(s)
- Raafat S Alameddine
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | | | - Hadi Skouri
- Division of Cardiology, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
96
|
Effect of intravitreal bevacizumab on retrobulbar blood flow of patients with diabetic macular edema. Eur J Ophthalmol 2015; 25:539-45. [PMID: 25907286 DOI: 10.5301/ejo.5000617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2015] [Indexed: 11/20/2022]
Abstract
PURPOSE To determine the effect of intravitreal bevacizumab on retrobulbar blood flow of patients with diabetic macular edema. METHODS Peak systolic velocity, end-diastolic velocity, resistance, and pulsatility indices of central retinal artery (CRA), nasal posterior ciliary artery (NPCA), temporal posterior ciliary artery (TPCA), and ophthalmic artery (OA) were assessed by color Doppler ultrasonography in injected and uninjected eyes of 37 patients at baseline and at day 1 and day 7 after the injection. RESULTS At day 1 after the injection, peak systolic and end diastolic velocities of the CRA and NPCA in the injected eyes and peak systolic velocity of NPCA in the uninjected eyes decreased significantly (p<0.05, p = 0.025, respectively). Peak systolic and end diastolic velocities were not significantly different in the injected and uninjected eyes at day 7 after the injection (p>0.05). In the injected eyes, there was no significant change in resistance and pulsatility indices (p>0.05), but a decrease in resistance index of NPCA and resistance and pulsatility indices of TPCA occurred in uninjected eyes at day 7 after the injection (p = 0.016, p = 0.023, and p = 0.025, respectively). CONCLUSIONS Intravitreal bevacizumab affects retrobulbar blood flow in injected and uninjected eyes of patients with diabetic macular edema.
Collapse
|
97
|
Wang W, Sivakumar W, Torres S, Jhaveri N, Vaikari VP, Gong A, Howard A, Golden EB, Louie SG, Schönthal AH, Hofman FM, Chen TC. Effects of convection-enhanced delivery of bevacizumab on survival of glioma-bearing animals. Neurosurg Focus 2015; 38:E8. [DOI: 10.3171/2015.1.focus14743] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECT
Bevacizumab (Avastin), an antibody to vascular endothelial growth factor (VEGF), alone or in combination with irinotecan (Camptosar [CPT-11]), is a promising treatment for recurrent glioblastoma. However, the intravenous (IV) administration of bevacizumab produces a number of systemic side effects, and the increase in survival it provides for patients with recurrent glioblastoma is still only a few months. Because bevacizumab is an antibody against VEGF, which is secreted into the extracellular milieu by glioma cells, the authors hypothesized that direct chronic intratumoral delivery techniques (i.e., convection-enhanced delivery [CED]) can be more effective than IV administration. To test this hypothesis, the authors compared outcomes for these routes of bevacizumab application with respect to animal survival, microvessel density (MVD), and inflammatory cell distribution.
METHODS
Two human glioma cell lines, U87 and U251, were used as sources of intracranial tumor cells. The glioma cell lines were implanted into the brains of mice in an orthotopic xenograft mouse tumor model. After 7 days, the mice were treated with one of the following: 1) vehicle, 2) CED bevacizumab, 3) IV bevacizumab, 4) intraperitoneal (IP) irinotecan, 5) CED bevacizumab plus IP irinotecan, or 6) IV bevacizumab plus IP irinotecan. Alzet micro-osmotic pumps were used to introduce bevacizumab directly into the tumor. Survival was monitored. Excised tumor tissue samples were immunostained to measure MVD and inflammatory cell and growth factor levels.
RESULTS
The results demonstrate that mice treated with CED of bevacizumab alone or in combination with irinotecan survived longer than those treated systemically; CED-treated animals survived 30% longer than IV-treated animals. In combination studies, CED bevacizumab plus CPT-11 increased survival by more than 90%, whereas IV bevacizumab plus CPT-11 increased survival by 40%. Furthermore, CED bevacizumab-treated tissues exhibited decreased MVD compared with that of IV-treated tissues. In additional studies, the infiltration of macrophages and dendritic cells into CED-treated animals were increased compared with those in IV-treated animals, suggesting a highly active inflammatory response taking place in CED-treated mice.
CONCLUSIONS
The administration of bevacizumab via CED increases survival over that of treatment with IV bevacizumab. Thus, CED of bevacizumab alone or in combination with chemotherapy can be an effective protocol for treating gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Stan G. Louie
- 4Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Axel H. Schönthal
- 3Molecular Microbiology and Immunology, Keck School of Medicine; and
| | | | | |
Collapse
|
98
|
Nakahara T, Hoshino M, Hoshino SI, Mori A, Sakamoto K, Ishii K. Structural and functional changes in retinal vasculature induced by retinal ischemia-reperfusion in rats. Exp Eye Res 2015; 135:134-45. [PMID: 25728136 DOI: 10.1016/j.exer.2015.02.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/25/2015] [Accepted: 02/25/2015] [Indexed: 12/17/2022]
Abstract
Recent studies have shown retinal blood vessel damage in experimental models of retinal degeneration. The present study aimed to provide a detailed description of the structural and functional changes in retinal vasculature induced by retinal ischemia-reperfusion (I/R) in rats. Retinal ischemia was induced for 60 min by raising the intraocular pressure to 130 mmHg. Morphological changes in vascular components (endothelial cells, pericytes, and basement membranes), the patency and perfusion of blood vessels, and expression of vascular endothelial growth factor (VEGF) were assessed in the retinas at 2, 7, and 14 days after I/R. Significant reductions in vascular densities were observed at 7 and 14 days after I/R. Pericyte loss occurred after the appearance of endothelial cell degeneration, whereas the vascular basement membranes remained unchanged. Some vessels showed no perfusion in damaged retina. A decrease in the immunoreactivity of VEGF in the region extending from the ganglion cell layer to the outer plexiform layer was evident 2 days after I/R. In retinal I/R model, retinal ganglion cells are rapidly (<2 day) damaged following reperfusion, therefore, the current results suggest that neuronal cell damage precedes capillary degeneration, and neuronal cells may play an important role in maintaining vascular structure and function through the production and release of endothelial cell survival factors, including VEGF. Neuronal cell damage could be an additional cause of progression of ischemic retinal damage by reducing blood supply to the retinal neurons due to the destruction of the blood vessel network.
Collapse
Affiliation(s)
- Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Maya Hoshino
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shin-Ichiro Hoshino
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
99
|
Ricca AM, Morshedi RG, Wirostko BM. High Intraocular Pressure Following Anti-Vascular Endothelial Growth Factor Therapy: Proposed Pathophysiology due to Altered Nitric Oxide Metabolism. J Ocul Pharmacol Ther 2015; 31:2-10. [DOI: 10.1089/jop.2014.0062] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Aaron M. Ricca
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - R. Grant Morshedi
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | |
Collapse
|
100
|
Schäker K, Bartsch S, Patry C, Stoll SJ, Hillebrands JL, Wieland T, Kroll J. The bipartite rac1 Guanine nucleotide exchange factor engulfment and cell motility 1/dedicator of cytokinesis 180 (elmo1/dock180) protects endothelial cells from apoptosis in blood vessel development. J Biol Chem 2015; 290:6408-18. [PMID: 25586182 DOI: 10.1074/jbc.m114.633701] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Engulfment and cell motility 1/dedicator of cytokinesis 180 (Elmo1/Dock180) is a bipartite guanine nucleotide exchange factor for the monomeric GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1). Elmo1/Dock180 regulates Rac1 activity in a specific spatiotemporal manner in endothelial cells (ECs) during zebrafish development and acts downstream of the Netrin-1/Unc5-homolog B (Unc5B) signaling cascade. However, mechanistic details on the pathways by which Elmo1/Dock180 regulates endothelial function and vascular development remained elusive. In this study, we aimed to analyze the vascular function of Elmo1 and Dock180 in human ECs and during vascular development in zebrafish embryos. In vitro overexpression of Elmo1 and Dock180 in ECs reduced caspase-3/7 activity and annexin V-positive cell number upon induction of apoptosis. This protective effect of Elmo1 and Dock180 is mediated by activation of Rac1, p21-activated kinase (PAK) and AKT/protein kinase B (AKT) signaling. In zebrafish, Elmo1 and Dock180 overexpression reduced the total apoptotic cell and apoptotic EC number and promoted the formation of blood vessels during embryogenesis. In conclusion, Elmo1 and Dock180 protect ECs from apoptosis by the activation of the Rac1/PAK/AKT signaling cascade in vitro and in vivo. Thus, Elmo1 and Dock180 facilitate blood vessel formation by stabilization of the endothelium during angiogenesis.
Collapse
Affiliation(s)
- Kathrin Schäker
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany, and
| | - Susanne Bartsch
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Christian Patry
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Sandra J Stoll
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany
| | - Jens Kroll
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany, and
| |
Collapse
|