1
|
Lozano LP, Jensen R, Jennisch M, Pandala NG, Jamshidi F, Boldt HC, Tucker BA, Binkley EM. Genetics and Current Research Models of Mendelian Tumor Predisposition Syndromes with Ocular Involvement. Prog Retin Eye Res 2025:101359. [PMID: 40274012 DOI: 10.1016/j.preteyeres.2025.101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
In this review, we aim to provide a survey of hereditable tumor predisposition syndromes with a mendelian inheritance pattern and ocular involvement. We focus our discussion on von Hippel-Lindau disease, neurofibromatosis type 1, NF2-related schwannomatosis, tuberous sclerosis complex, retinoblastoma, and the BAP1 Tumor Predisposition Syndrome. For each of the six diseases, we discuss the clinical presentation and the molecular pathophysiology. We emphasize the genetics, current research models, and therapeutic developments. After reading each disease section, readers should possess an understanding of the clinical presentation, genetic causes and inheritance patterns, and current state of research in disease modeling and treatment.
Collapse
Affiliation(s)
- Lola P Lozano
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Renato Jensen
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Madeleine Jennisch
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Narendra G Pandala
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Farzad Jamshidi
- Department of Ophthalmology, University of Pittsburgh/UPMC, Pittsburgh, PA, 15213, USA.
| | - H Culver Boldt
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Budd A Tucker
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Elaine M Binkley
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Zhu X, Qi B, Ren Z, Cong L, Pan X, Zhou Q, Zhang BN, Xie L. Targeted Neuroprotection of Retinal Ganglion Cells Via AAV2-hSyn-NGF Gene Therapy in Glaucoma Models. Invest Ophthalmol Vis Sci 2025; 66:48. [PMID: 40244606 PMCID: PMC12013675 DOI: 10.1167/iovs.66.4.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose The purpose of this study was to evaluate the neuroprotective effects of delivering nerve growth factor (NGF) to retinal ganglion cells (RGCs) through adeno-associated virus serotype 2 (AAV2) carrying a neuronal-specific human synapsin (hSyn) promoter. Methods AAV2-hSyn-NGF was injected intravitreally in three glaucoma models: optic nerve crush (ONC), microbead-induced ocular hypertension (MB), and genetic glaucoma model (DBA). Quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA) determined the optimal injection concentration of AAV vector. Flow cytometry monitored immune responses. Transduction efficiency was quantified using green fluorescent protein (GFP) co-localization with RGC-specific marker RNA-binding protein with multiple splicing (RBPMS). The RGCs' density, retinal nerve fiber density, ganglion cell complex thickness, and positive scotopic threshold response (pSTR) were measured to assess structural and functional outcomes of the RGCs. Non-parametric Mann-Whitney U tests or Kruskal-Wallis tests were utilized to ascertain the statistical significance (P < 0.05). Results The optimal concentration of AAV vector for intravitreal injection was determined to be 1 × 1010 vector particles (VPs) per eye. The use of the hSyn promoter significantly enhanced targeting specificity to RGCs, resulting in a transduction efficiency of 46.64% ± 2.18%. Administration of AAV2-hSyn-NGF effectively preserved the RGCs' density, nerve fiber layer integrity, and the thickness of ganglion cell complex, while maintaining the RGCs' function across three glaucoma models. Furthermore, this gene delivery system did not elicit detectable immune responses or structural damage to the retina. Conclusions The AAV2-hSyn-NGF gene therapy offers a safe and effective neuroprotective strategy for RGCs across multiple glaucoma models, making it a promising candidate for future clinical trials in patients with glaucoma.
Collapse
Affiliation(s)
- Xinlei Zhu
- Qingdao Medical College, Qingdao University, Qingdao, China
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Benxiang Qi
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Zhongmei Ren
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Lin Cong
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Xiaojing Pan
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Bi Ning Zhang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
- School of Ophthalmology, Shandong First Medical University, Qingdao, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, China
| |
Collapse
|
3
|
Latini L, De Araujo DSM, Amato R, Canovai A, Buccarello L, De Logu F, Novelli E, Vlasiuk A, Malerba F, Arisi I, Florio R, Asari H, Capsoni S, Strettoi E, Villetti G, Imbimbo BP, Dal Monte M, Nassini R, Geppetti P, Marinelli S, Cattaneo A. A p75 neurotrophin receptor-sparing nerve growth factor protects retinal ganglion cells from neurodegeneration by targeting microglia. Br J Pharmacol 2024; 181:4890-4919. [PMID: 39252503 DOI: 10.1111/bph.17316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Retinal ganglion cells (RGCs) are the output stage of retinal information processing, via their axons forming the optic nerve (ON). ON damage leads to axonal degeneration and death of RGCs, and results in vision impairment. Nerve growth factor (NGF) signalling is crucial for RGC operations and visual functions. Here, we investigate a new neuroprotective mechanism of a novel therapeutic candidate, a p75-less, TrkA-biased NGF agonist (hNGFp) in rat RGC degeneration, in comparison with wild type human NGF (hNGFwt). EXPERIMENTAL APPROACH Both neonate and adult rats, whether subjected or not to ON lesion, were treated with intravitreal injections or eye drops containing either hNGFp or hNGFwt. Different doses of the drugs were administered at days 1, 4 or 7 after injury for a maximum of 10 days, when immunofluorescence, electrophysiology, cellular morphology, cytokine array and behaviour studies were carried out. Pharmacokinetic evaluation was performed on rabbits treated with hNGFp ocular drops. RESULTS hNGFp exerted a potent RGC neuroprotection by acting on microglia cells, and outperformed hNGFwt in rescuing RGC degeneration and reducing inflammatory molecules. Delayed use of hNGFp after ON lesion resulted in better outcomes compared with treatment with hNGFwt. Moreover, hNGFp-based ocular drops were less algogenic than hNGFwt. Pharmacokinetic measurements revealed that biologically relevant quantities of hNGFp were found in the rabbit retina. CONCLUSIONS AND IMPLICATIONS Our data point to microglia as a new cell target through which NGF-induced TrkA signalling exerts neuroprotection of the RGC, emphasizing hNGFp as a powerful treatment to tackle retinal degeneration.
Collapse
Affiliation(s)
- Laura Latini
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Anastasiia Vlasiuk
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Hiroki Asari
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Simona Capsoni
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Gino Villetti
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
4
|
Marangoni D, Placidi G, D'Agostino E, De Siena E, Attinà G, Mastrangelo S, Ruggiero A, Colosimo C, Falsini B. Longitudinal changes in retinal ganglion cell function in optic pathway glioma evaluated by photopic negative response. Exp Eye Res 2024; 246:110012. [PMID: 39059735 DOI: 10.1016/j.exer.2024.110012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Photopic negative response (PhNR), an index of retinal ganglion cell (RGC) function, is impaired in patients with optic pathway gliomas (OPGs). The aim of this longitudinal study was to evaluate whether PhNR deteriorates over time in OPG patients. Fourteen pediatric patients affected by OPG (4 males and 10 females, mean age 12.4 ± 5.7 years, 8 with neurofibromatosis type 1 [NF1]) with ≥12 months of follow-up and ≥2 evaluations, were included in this retrospective study. All patients had received chemotherapy, with or without OPG surgical resection, at least 5 years prior to the study. At baseline, all patients underwent a complete ophthalmological examination. Follow-up included clinical examination and PhNR measurement as well as brain MRI (according to pediatric oncologist indications) every 6 or 12 months. Mean follow-up duration was 16.7 ± 7.5 months (range 12-36 months). Photopic electroretinograms were elicited by 2.0 cd-s/m2 Ganzfeld white flashes presented on a steady 20 cd/m2 white background. The PhNR amplitude was measured as the difference between baseline and the maximal negative amplitude (minimum) of the negative wave, following the photopic b-wave. Compared to baseline, mean PhNR amplitude was significantly decreased at the end of follow-up (p = 0.008). NF1-related OPGs exhibited a decline in PhNR amplitude (p = 0.005) and an increase in PhNR peak-time during the follow-up (p = 0.013), whereas sporadic OPGs showed no significant changes. Tumor size remained stable in all patients on MRI. PhNR amplitude decreased over the observation period, suggesting progressive RGC dysfunction in NF1-related pediatric OPGs, despite stable size on MRI imaging. PhNR could serve as a non-invasive objective tool for assessing longitudinal changes in RGC function in the clinical management of childhood OPG.
Collapse
Affiliation(s)
- Dario Marangoni
- University Eye Clinic, Department of Medicine, Surgery and Health Science, University of Trieste, Strada di Fiume, 447, 34129, Trieste, Italy.
| | - Giorgio Placidi
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Elena D'Agostino
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Elisa De Siena
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Cesare Colosimo
- Radiology and Neuroradiology Department, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Benedetto Falsini
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| |
Collapse
|
5
|
Capossela L, Gatto A, Ferretti S, Di Sarno L, Graglia B, Massese M, Soligo M, Chiaretti A. Multifaceted Roles of Nerve Growth Factor: A Comprehensive Review with a Special Insight into Pediatric Perspectives. BIOLOGY 2024; 13:546. [PMID: 39056738 PMCID: PMC11273967 DOI: 10.3390/biology13070546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Nerve growth factor (NGF) is a neurotrophic peptide largely revealed for its ability to regulate the growth and survival of peripheral sensory, sympathetic, and central cholinergic neurons. The pro-survival and regenerative properties of neurotrophic factors propose a therapeutic potential in a wide range of brain diseases, and NGF, in particular, has appeared as an encouraging potential treatment. In this review, a summary of clinical studies regarding NGF and its therapeutic effects published to date, with a specific interest in the pediatric context, will be attempted. NGF has been studied in neurological disorders such as hypoxic-ischemic encephalopathy, traumatic brain injury, neurobehavioral and neurodevelopmental diseases, congenital malformations, cerebral infections, and in oncological and ocular diseases. The potential of NGF to support neuronal survival, repair, and plasticity in these contexts is highlighted. Emerging therapeutic strategies for NGF delivery, including intranasal administration as well as advanced nanotechnology-based methods, are discussed. These techniques aim to enhance NGF bioavailability and target specificity, optimizing therapeutic outcomes while minimizing systemic side effects. By synthesizing current research, this review underscores the promise and challenges of NGF-based therapies in pediatric neurology, advocating for continued innovation in delivery methods to fully harness NGF's therapeutic potential.
Collapse
Affiliation(s)
- Lavinia Capossela
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Antonio Gatto
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Serena Ferretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Lorenzo Di Sarno
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Benedetta Graglia
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Miriam Massese
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Marzia Soligo
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), 00133 Rome, Italy;
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| |
Collapse
|
6
|
Chen Y, Yu J, Ge S, Jia R, Song X, Wang Y, Fan X. An Overview of Optic Pathway Glioma With Neurofibromatosis Type 1: Pathogenesis, Risk Factors, and Therapeutic Strategies. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 38837168 PMCID: PMC11160950 DOI: 10.1167/iovs.65.6.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
Optic pathway gliomas (OPGs) are most predominant pilocytic astrocytomas, which are typically diagnosed within the first decade of life. The majority of affected children with OPGs also present with neurofibromatosis type 1 (NF1), the most common tumor predisposition syndrome. OPGs in individuals with NF1 primarily affect the optic pathway and lead to visual disturbance. However, it is challenging to assess risk in asymptomatic patients without valid biomarkers. On the other hand, for symptomatic patients, there is still no effective treatment to prevent or recover vision loss. Therefore, this review summarizes current knowledge regarding the pathogenesis of NF1-associated OPGs (NF1-OPGs) from preclinical studies to seek potential prognostic markers and therapeutic targets. First, the loss of the NF1 gene activates 3 distinct Ras effector pathways, including the PI3K/AKT/mTOR pathway, the MEK/ERK pathway, and the cAMP pathway, which mediate glioma tumorigenesis. Meanwhile, non-neoplastic cells from the tumor microenvironment (microglia, T cells, neurons, etc.) also contribute to gliomagenesis via various soluble factors. Subsequently, we investigated potential genetic risk factors, molecularly targeted therapies, and neuroprotective strategies for tumor prevention and vision recovery. Last, potential directions and promising preclinical models of NF1-OPGs are presented for further research. On the whole, NF1-OPGs develop as a result of the interaction between glioma cells and the tumor microenvironment. Developing effective treatments require a better understanding of tumor molecular characteristics, as well as multistage interventions targeting both neoplastic cells and non-neoplastic cells.
Collapse
Affiliation(s)
- Ying Chen
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Jie Yu
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xin Song
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Yefei Wang
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
7
|
Gavioli E, Mantelli F, Cesta MC, Sacchetti M, Allegretti M. The History of Nerve Growth Factor: From Molecule to Drug. Biomolecules 2024; 14:635. [PMID: 38927039 PMCID: PMC11201509 DOI: 10.3390/biom14060635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/12/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Nerve growth factor (NGF), the first neurotrophin to be discovered, has a long and eventful research journey with a series of turning points, setbacks, and achievements. Since the groundbreaking investigations led by Nobel Prize winner Rita Levi-Montalcini, advancements in the comprehension of NGF's functions have revolutionized the field of neuroscience, offering new insights and opportunities for therapeutic innovation. However, the clinical application of NGF has historically been hindered by challenges in determining appropriate dosing, administration strategies, and complications related to the production process. Recent advances in the production and scientific knowledge of recombinant NGF have enabled its clinical development, and in 2018, the United States Food and Drug Administration approved cenegermin-bkbj, a recombinant human NGF, for the treatment of all stages of neurotrophic keratitis. This review traces the evolutionary path that transformed NGF from a biological molecule into a novel therapy with potential research applications beyond the eye. Special emphasis is put on the studies that advanced NGF from discovery to the first medicinal product approved to treat a human disease.
Collapse
Affiliation(s)
| | - Flavio Mantelli
- Dompé farmaceutici S.p.A., Via Santa Lucia, 6, 20122 Milano, Italy; (F.M.); (M.C.C.); (M.S.)
| | - Maria Candida Cesta
- Dompé farmaceutici S.p.A., Via Santa Lucia, 6, 20122 Milano, Italy; (F.M.); (M.C.C.); (M.S.)
| | - Marta Sacchetti
- Dompé farmaceutici S.p.A., Via Santa Lucia, 6, 20122 Milano, Italy; (F.M.); (M.C.C.); (M.S.)
| | - Marcello Allegretti
- Dompé farmaceutici S.p.A., Via Santa Lucia, 6, 20122 Milano, Italy; (F.M.); (M.C.C.); (M.S.)
| |
Collapse
|
8
|
Miyagishima KJ, Qiao F, Stasheff SF, Nadal-Nicolás FM. Visual Deficits and Diagnostic and Therapeutic Strategies for Neurofibromatosis Type 1: Bridging Science and Patient-Centered Care. Vision (Basel) 2024; 8:31. [PMID: 38804352 PMCID: PMC11130890 DOI: 10.3390/vision8020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is an inherited autosomal dominant disorder primarily affecting children and adolescents characterized by multisystemic clinical manifestations. Mutations in neurofibromin, the protein encoded by the Nf1 tumor suppressor gene, result in dysregulation of the RAS/MAPK pathway leading to uncontrolled cell growth and migration. Neurofibromin is highly expressed in several cell lineages including melanocytes, glial cells, neurons, and Schwann cells. Individuals with NF1 possess a genetic predisposition to central nervous system neoplasms, particularly gliomas affecting the visual pathway, known as optic pathway gliomas (OPGs). While OPGs are typically asymptomatic and benign, they can induce visual impairment in some patients. This review provides insight into the spectrum and visual outcomes of NF1, current diagnostic techniques and therapeutic interventions, and explores the influence of NF1-OPGS on visual abnormalities. We focus on recent advancements in preclinical animal models to elucidate the underlying mechanisms of NF1 pathology and therapies targeting NF1-OPGs. Overall, our review highlights the involvement of retinal ganglion cell dysfunction and degeneration in NF1 disease, and the need for further research to transform scientific laboratory discoveries to improved patient outcomes.
Collapse
Affiliation(s)
- Kiyoharu J. Miyagishima
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Fengyu Qiao
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Steven F. Stasheff
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
- Center for Neuroscience and Behavioral Medicine, Gilbert Neurofibromatosis Institute, Children’s National Health System, Washington, DC 20010, USA
- Neurology Department, George Washington University School of Medicine, Washington, DC 20037, USA
| | - Francisco M. Nadal-Nicolás
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| |
Collapse
|
9
|
Asteriti S, Marino V, Avesani A, Biasi A, Dal Cortivo G, Cangiano L, Dell'Orco D. Recombinant protein delivery enables modulation of the phototransduction cascade in mouse retina. Cell Mol Life Sci 2023; 80:371. [PMID: 38001384 PMCID: PMC10673981 DOI: 10.1007/s00018-023-05022-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
Inherited retinal dystrophies are often associated with mutations in the genes involved in the phototransduction cascade in photoreceptors, a paradigmatic signaling pathway mediated by G protein-coupled receptors. Photoreceptor viability is strictly dependent on the levels of the second messengers cGMP and Ca2+. Here we explored the possibility of modulating the phototransduction cascade in mouse rods using direct or liposome-mediated administration of a recombinant protein crucial for regulating the interplay of the second messengers in photoreceptor outer segments. The effects of administration of the free and liposome-encapsulated human guanylate cyclase-activating protein 1 (GCAP1) were compared in biological systems of increasing complexity (in cyto, ex vivo, and in vivo). The analysis of protein biodistribution and the direct measurement of functional alteration in rod photoresponses show that the exogenous GCAP1 protein is fully incorporated into the mouse retina and photoreceptor outer segments. Furthermore, only in the presence of a point mutation associated with cone-rod dystrophy in humans p.(E111V), protein delivery induces a disease-like electrophysiological phenotype, consistent with constitutive activation of the retinal guanylate cyclase. Our study demonstrates that both direct and liposome-mediated protein delivery are powerful complementary tools for targeting signaling cascades in neuronal cells, which could be particularly important for the treatment of autosomal dominant genetic diseases.
Collapse
Affiliation(s)
- Sabrina Asteriti
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
- Department of Translational Research, University of Pisa, 56123, Pisa, Italy
| | - Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | - Anna Avesani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | - Amedeo Biasi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | - Lorenzo Cangiano
- Department of Translational Research, University of Pisa, 56123, Pisa, Italy.
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy.
| |
Collapse
|
10
|
Tringali G, Pizzoferrato M, Lisi L, Marinelli S, Buccarello L, Falsini B, Cattaneo A, Navarra P. A Vicious NGF-p75 NTR Positive Feedback Loop Exacerbates the Toxic Effects of Oxidative Damage in the Human Retinal Epithelial Cell Line ARPE-19. Int J Mol Sci 2023; 24:16237. [PMID: 38003427 PMCID: PMC10671591 DOI: 10.3390/ijms242216237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
In spite of its variety of biological activities, the clinical exploitation of human NGF (hNGF) is currently limited to ocular pathologies. It is therefore interesting to test the effects of hNGF in preclinical models that may predict their efficacy and safety in the clinical setting of ocular disorders and compare the effects of hNGF with those of its analogs. We used a human retinal pigment cell line, ARPE-19 cells, to investigate the effects of hNGF and its analogs, mouse NGF (mNGF) and painless NGF (pNGF), on cell viability under basal conditions and after exposure to oxidative stimuli, i.e., hydrogen peroxide (H2O2) and ultraviolet (UV)-A rays. The effects of hNGF and pNGF were also tested on the gene expression and protein synthesis of the two NGF receptor subtypes, p75 neurotrophic receptors (p75NTR) and tyrosine kinase A (TrkA) receptors. We drew the following conclusions: (i) the exposure of ARPE-19 cells to H2O2 or UV-A causes a dose-dependent decrease in the number of viable cells; (ii) under baseline conditions, hNGF, but not pNGF, causes a concentration-dependent decrease in cell viability in the range of doses 1-100 ng/mL; (iii) hNGF, but not pNGF, significantly potentiates the toxic effects of H2O2 or of UV-A on ARPE-19 cells in the range of doses 1-100 ng/mL, while mNGF at the same doses presents an intermediate behavior; (iv) 100 ng/mL of hNGF triggers an increase in p75NTR expression in H2O2-treated ARPE-19 cells, while pNGF at the same dose does not; (v) pNGF, but not hNGF (both given at 100 ng/mL), increases the total cell fluorescence intensity for TrkA receptors in H2O2-treated ARPE-19 cells. The present findings suggest a vicious positive feedback loop through which NGF-mediated upregulation of p75NTR contributes to worsening the toxic effects of oxidative damage in the human retinal epithelial cell line ARPE-19. Looking at the possible clinical relevance of these findings, one can postulate that pNGF might show a better benefit/risk ratio than hNGF in the treatment of ocular disorders.
Collapse
Affiliation(s)
- Giuseppe Tringali
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Michela Pizzoferrato
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Benedetto Falsini
- UOC Ophtalmology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Ophthalmology, Bambino Gesù IRCCS Children’s Hospital, 00133 Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
- Bio@SNS Laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| |
Collapse
|
11
|
Chernov A, Kudryavtsev I, Komlev A, Alaverdian D, Tsapieva A, Galimova E, Shamova O. Nerve Growth Factor, Antimicrobial Peptides and Chemotherapy: Glioblastoma Combination Therapy to Improve Their Efficacy. Biomedicines 2023; 11:3009. [PMID: 38002009 PMCID: PMC10669874 DOI: 10.3390/biomedicines11113009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/07/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive and lethal malignancy of the central nervous system with a median survival rate of 15 months. We investigated the combined anticancer effects of nerve growth factor (NGF), cathelicidin (LL-37), and protegrin-1 (PG-1) with chemotherapy (temozolomide, doxorubicin, carboplatin, cisplatin, and etoposide) in the glioblastoma U251 cell line to overcome the limitations of conventional chemotherapy and to guarantee specific treatments to succeed. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to study cell viability and to determine the cytotoxic effects of NGF, LL-37, and PG-1 and their combination with chemotherapy in U251 cells. Synergism or antagonism was determined using the combination index (CI) method. Caspase-3 activity was evaluated spectrophotometrically using a caspase-3 activity assay kit. Apoptosis was analyzed with flow cytometry using propidium iodide (PI) and YO-PRO-1. NGF and the peptides showed a strong cytotoxic effect on U251 glioma cells in the MTT test (IC50 0.0214, 3.1, and 26.1 μM, respectively) compared to chemotherapy. The combination of PG-1 + etoposide had a synergistic effect on apoptosis of U251 glioma cells. It should be noted that the cells were in the early and late stages of apoptosis, respectively, compared with the control cells. The caspase-3 activation analysis revealed that the caspase-3 level was not significantly (p > 0.05) increased in U251 cells following PG-1 with etoposide treatment compared with that in the untreated cells, suggesting that the combination of PG-1 and etoposide may induce caspase-independent apoptosis in U251 cells. NGF, LL-37, and PG-1 represent promising drug candidates as the treatment regimen for GBM. Furthermore, the synergistic efficacy of the combined protocol using PG-1 and etoposide may overcome some of the typical limitations of the conventional therapeutic protocols, thus representing a promising approach for GBM therapy.
Collapse
Affiliation(s)
- Alexandr Chernov
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Aleksei Komlev
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Diana Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Anna Tsapieva
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Elvira Galimova
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg 194223, Russia
| | - Olga Shamova
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
- Department of Biochemistry, Saint Petersburg State University, Saint-Petersburg 199034, Russia
| |
Collapse
|
12
|
Tang Y, Gutmann DH. Neurofibromatosis Type 1-Associated Optic Pathway Gliomas: Current Challenges and Future Prospects. Cancer Manag Res 2023; 15:667-681. [PMID: 37465080 PMCID: PMC10351533 DOI: 10.2147/cmar.s362678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Optic pathway glioma (OPG) occurs in as many as one-fifth of individuals with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Generally considered low-grade and slow growing, many children with NF1-OPGs remain asymptomatic. However, due to their location within the optic pathway, ~20-30% of those harboring NF1-OPGs will experience symptoms, including progressive vision loss, proptosis, diplopia, and precocious puberty. While treatment with conventional chemotherapy is largely effective at attenuating tumor growth, it is not clear whether there is much long-term recovery of visual function. Additionally, because these tumors predominantly affect young children, there are unique challenges to NF1-OPG diagnosis, monitoring, and longitudinal management. Over the past two decades, the employment of authenticated genetically engineered Nf1-OPG mouse models have provided key insights into the function of the NF1 protein, neurofibromin, as well as the molecular and cellular pathways that contribute to optic gliomagenesis. Findings from these studies have resulted in the identification of new molecular targets whose inhibition blocks murine Nf1-OPG growth in preclinical studies. Some of these promising compounds have now entered into early clinical trials. Future research focused on defining the determinants that underlie optic glioma initiation, expansion, and tumor-induced optic nerve injury will pave the way to personalized risk assessment strategies, improved tumor monitoring, and optimized treatment plans for children with NF1-OPG.
Collapse
Affiliation(s)
- Yunshuo Tang
- Department of Ophthalmology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
13
|
Bianchi F, Cocilovo FM, Ruggiero A, Tamburrini G. Optic Pathway Gliomas: The Trends of Basic Research to Reduce the Impact of the Disease on Visual Function. Adv Tech Stand Neurosurg 2023; 48:123-137. [PMID: 37770684 DOI: 10.1007/978-3-031-36785-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Pediatric optic pathway gliomas (OPG) are low-grade brain tumors characterized by slow progression and invalidating visual loss. Common therapeutic strategies include surgery, radiotherapy, chemotherapy, and combinations of these modalities, but despite the different treatment strategies, no actual treatment exists to prevent or revert visual impairment. Nowadays, several reports of the literature show promising results regarding NGF eye drop instillation and improvement of visual outcome. Such results seem to be related with the NGF-linked prevention in caspase activation, which reduces retinal ganglion cell loss.Reducing retinal ganglion cell loss results clinically in visual field improvement as well as visual electric potential and optical coherence tomography gain. Nonetheless, visual acuity fails to show significant changes.Visual impairment represents nowadays one of the major issues in dealing with OPGs. Secondary to the interesting results offered by NGF eye drop administration, further studies are warranted to better comprehend potential treatment strategies.
Collapse
Affiliation(s)
| | | | - Antonio Ruggiero
- Fondazione Policlinico Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Milan, Italy
| | - Gianpiero Tamburrini
- Fondazione Policlinico Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Milan, Italy
| |
Collapse
|
14
|
Ruggiero A, Attinà G, Campanelli A, Maurizi P, Triarico S, Romano A, Massimi L, Tamburrini G, Verdolotti T, Mastrangelo S. Pediatric low-grade glioma and neurofibromatosis type 1: A single-institution experience. J Cancer Res Ther 2023; 19:228-234. [PMID: 37313902 DOI: 10.4103/jcrt.jcrt_1677_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Neurofibromatosis type 1 (NF1)-related gliomas appear to have a clinical behavior different from that of sporadic cases. The purpose of the study was to investigate the role of different factors in influencing the tumor response rate of children receiving chemotherapy for their symptomatic glioma. Methods Between 1995 and 2015, 60 patients with low-grade glioma (42 sporadic cases and 18 cases with NF1) were treated. Patients with brainstem gliomas were excluded. Thirty-nine patients underwent exclusive or postsurgical chemotherapy (vincristine/carboplatin-based regimen). Results Disease reduction was achieved in 12 of the 28 patients (42.8%) with sporadic low-grade glioma and in 9 of the 11 patients (81.8%) with NF1, with a significant difference between the 2 groups (P < 0.05). The response to chemotherapy in both the patient groups was not significantly influenced by sex, age, tumor site, and histopathology, although disease reduction occurred more frequently in children aged under 3 years. Conclusions Our study showed that pediatric patients with low-grade glioma and NF1 are more likely to respond to chemotherapy than those with non-NF1.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Anastasia Campanelli
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Massimi
- Pediatric Neurosurgery Unit, Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Gianpiero Tamburrini
- Pediatric Neurosurgery Unit, Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Tommaso Verdolotti
- Radiology and Neuroradiology Unit, Department of Radiological Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli Hospital Foundation IRCCS, Universita' Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
15
|
Walker DA, Aquilina K, Spoudeas H, Pilotto C, Gan HW, Meijer L. A new era for optic pathway glioma: A developmental brain tumor with life-long health consequences. Front Pediatr 2023; 11:1038937. [PMID: 37033188 PMCID: PMC10080591 DOI: 10.3389/fped.2023.1038937] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
Optic pathway and hypothalamic glioma (OPHG) are low-grade brain tumors that arise from any part of the visual pathways frequently involving the hypothalamus. The tumors grow slowly and present with features driven by their precise anatomical site, their age at presentation and the stage of growth and development of the host neural and orbital bony tissues. Up to 50% of optic pathway glioma arise in association with Neurofibromatosis type 1 (NF1), which affects 1 in 3,000 births and is a cancer predisposition syndrome. As low-grade tumors, they almost never transform to malignant glioma yet they can threaten life when they present under two years of age. The main risks are to threaten vision loss by progressive tumor damage to optic pathways; furthermore, invasion of the hypothalamus can lead to diencephalic syndrome in infancy and hypopituitarism later in life. Progressive cognitive and behavioural dysfunction can occur, as part of NF1 syndromic features and in sporadic cases where large bulky tumors compress adjacent structures and disrupt neuro-hypothalamic pathways. Persistently progressive tumors require repeated treatments to attempt to control vision loss, other focal brain injury or endocrine dysfunction. In contrast tumors presenting later in childhood can be seen to spontaneously arrest in growth and subsequently progress after periods of stability. These patterns are influenced by NF status as well as stages of growth and development of host tissues. The past two decades has seen an expansion in our understanding and knowledge of the clinical and scientific features of these tumors, their modes of presentation, the need for careful visual and endocrine assessment. This influences the decision-making surrounding clinical management with surgery, radiotherapy, chemotherapy and most recently, the potential benefit of molecularly targeted drug therapy. This article, based upon the authors' clinical and research experience and the published literature will highlight advances in approach to diagnosis, the established role of vision loss as justification of treatments and the emerging evidence of endocrine and neurological consequences that need to be incorporated into judgements for case selection for therapy or observation. Consideration is given to the current state of biological evidence justifying current trials of new therapies, the genetic studies of the NF1 gene and the potential for new approaches to OPHG detection and treatment. The outstanding health system priorities from the perspective of children, their parents and health system commissioners or insurers are discussed.
Collapse
Affiliation(s)
- David A. Walker
- Emeritus Professor Paediatric Oncology, University of Nottingham, Nottingham, United Kingdom
- Correspondence: David A. Walker
| | - Kristian Aquilina
- Department of NeuroEndocrinology, Great Ormond Street Hospital, London, United Kingdom
| | - Helen Spoudeas
- Department of NeuroEndocrinology, Great Ormond Street Hospital, London, United Kingdom
| | - Chiara Pilotto
- Pediatric Clinic, ASUFC Santa Maria Della Misericordia, Udine, Italy
| | - Hoong-Wei Gan
- Department of NeuroEndocrinology, Great Ormond Street Hospital, London, United Kingdom
| | - Lisethe Meijer
- Kinderoncologie, Prinses Máxima Centrum Voor Kinderoncologie BV, Utrecht, Netherlands
| |
Collapse
|
16
|
Intravitreal Fluocinolone Acetonide for Diabetic Macular Edema: Long-Term Effect and Structure/Function Correlation. Diagnostics (Basel) 2022; 12:diagnostics12102415. [PMID: 36292104 PMCID: PMC9600657 DOI: 10.3390/diagnostics12102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The long-term effect of intravitreal Fluocinolone acetonide (FAc) on retinal morphology and function in diabetic macular edema (DME) was investigated. Seventeen eyes of twelve consecutive DME patients, treated by intravitreal FAc, were retrospectively evaluated. Retinal morphology was assessed with central macular thickness (CMT). Retinal function was assessed by best-corrected visual acuity (BCVA) and cone b-wave and photopic negative response (PhNR). The main outcome was a mean change in CMT at month 24. The secondary outcomes were changes in cone b-wave and PhNR at month 24. The incidence of adverse events was also recorded. Mean CMT decreased from 406.52 µm (±138.74) at baseline to 310 µm (±130.39) at 24 months (p = 0.008). No significant changes in the other parameters were found. At baseline, BCVA and PhNR amplitude were negatively correlated (r = −0.55) with CMT. At the end of follow-up, the change in CMT was negatively correlated with baseline CMT (r = −0.53, p = 0.03) and positively correlated with baseline PhNR amplitude (r = 0.58, p < 0.01). A significant, long-term reduction in CMT was observed in DME patients after FAc implant. The anti-edema effect tended to be stronger in patients with the poorest baseline retinal morphology (CMT) and function (PhNR). Structure/function correlations might help to characterize the patients who may benefit from this treatment.
Collapse
|
17
|
Management of Optic Pathway Glioma: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14194781. [PMID: 36230704 PMCID: PMC9563939 DOI: 10.3390/cancers14194781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background: OPG accounts for 3−5% of childhood central nervous system (CNS) tumors and about 2% of pediatric glial lesions. Methods: Article selection was performed by searching PubMed, Web of Science, and Cochrane databases. Results: The pooled mortality rate was 0.12 (95%CI 0.09−0.14). Due to the unrepresentative data, improved and not changed outcomes were classified as favorable outcomes and worsened as unfavorable. Meta-analyses were performed to determine the rate of clinical and radiological favorable outcomes. In terms of visual assessment, the pooled rate of a favorable outcome in chemotherapy, radiotherapy, and surgery was 0.74, 0.81, and 0.65, respectively, and the overall pooled rate of the favorable outcome was 0.75 (95%CI 0.70−0.80). In terms of radiological assessment, the rate of a favorable outcome following chemotherapy, radiotherapy, and surgery was 0.71, 0.74, and 0.67, respectively, and the overall pooled rate of the favorable outcome is 0.71 (95%CI 0.65−0.77). The subgroup analysis revealed no significant difference in the rate of clinical and radiological favorable outcomes between the different treatment modalities (p > 0.05). Conclusion: Our analyses showed that each therapeutic modality represents viable treatment options to achieve remission for these patients.
Collapse
|
18
|
Lisi L, Ciotti GMP, Chiavari M, Martire M, Navarra P. The effects of CHF6467, a new mutated form of NGF, on cell models of human glioblastoma. A comparison with wild-type NGF. Growth Factors 2022; 40:37-45. [PMID: 35442129 DOI: 10.1080/08977194.2022.2060095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
CHF6467 is a mutated form of human recombinant nerve growth factor (NGF). The mutation selectively disrupts the binding of NGF to its p75NTR receptor while maintaining the affinity toward TrkA receptor. Because of such different profile of receptor interaction, CHF6467 maintains unaltered the neurotrophic and neuroprotective properties of wild-type NGF but shows reduced algogenic activity.In this study, we investigated the effects of CHF6467 on mortality, proliferation, cell-damage and migration in three human glioblastoma cell lines (U87MG, T98G, LN18), and in the rat astrocytoma C6 cells. Both CHF6467 and wild-type NGF, given in the range 1-50 ng/ml, did not modify cell proliferation, metabolism and migration, as well as the number of live/dead cells.The present in vitro data are predictive of a lack of tumorigenic activity by both wild-type NGF and CHF6467 on these cell types in vivo, and warrant for CHF6467 further clinical development.
Collapse
Affiliation(s)
- Lucia Lisi
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Gabriella Maria Pia Ciotti
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Marta Chiavari
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Maria Martire
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
19
|
Parmigiani E, Scalera M, Mori E, Tantillo E, Vannini E. Old Stars and New Players in the Brain Tumor Microenvironment. Front Cell Neurosci 2021; 15:709917. [PMID: 34690699 PMCID: PMC8527006 DOI: 10.3389/fncel.2021.709917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the direct interaction between cancer cells and tumor microenvironment (TME) has emerged as a crucial regulator of tumor growth and a promising therapeutic target. The TME, including the surrounding peritumoral regions, is dynamically modified during tumor progression and in response to therapies. However, the mechanisms regulating the crosstalk between malignant and non-malignant cells are still poorly understood, especially in the case of glioma, an aggressive form of brain tumor. The presence of unique brain-resident cell types, namely neurons and glial cells, and an exceptionally immunosuppressive microenvironment pose additional important challenges to the development of effective treatments targeting the TME. In this review, we provide an overview on the direct and indirect interplay between glioma and neuronal and glial cells, introducing new players and mechanisms that still deserve further investigation. We will focus on the effects of neural activity and glial response in controlling glioma cell behavior and discuss the potential of exploiting these cellular interactions to develop new therapeutic approaches with the aim to preserve proper brain functionality.
Collapse
Affiliation(s)
- Elena Parmigiani
- Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marta Scalera
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | | | - Elena Tantillo
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| |
Collapse
|
20
|
Amadoro G, Latina V, Balzamino BO, Squitti R, Varano M, Calissano P, Micera A. Nerve Growth Factor-Based Therapy in Alzheimer's Disease and Age-Related Macular Degeneration. Front Neurosci 2021; 15:735928. [PMID: 34566573 PMCID: PMC8459906 DOI: 10.3389/fnins.2021.735928] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disease which is the most common cause of dementia among the elderly. Imbalance in nerve growth factor (NGF) signaling, metabolism, and/or defect in NGF transport to the basal forebrain cholinergic neurons occurs in patients affected with AD. According to the cholinergic hypothesis, an early and progressive synaptic and neuronal loss in a vulnerable population of basal forebrain involved in memory and learning processes leads to degeneration of cortical and hippocampal projections followed by cognitive impairment with accumulation of misfolded/aggregated Aβ and tau protein. The neuroprotective and regenerative effects of NGF on cholinergic neurons have been largely demonstrated, both in animal models of AD and in living patients. However, the development of this neurotrophin as a disease-modifying therapy in humans is challenged by both delivery limitations (inability to cross the blood-brain barrier (BBB), poor pharmacokinetic profile) and unwanted side effects (pain and weight loss). Age-related macular degeneration (AMD) is a retinal disease which represents the major cause of blindness in developed countries and shares several clinical and pathological features with AD, including alterations in NGF transduction pathways. Interestingly, nerve fiber layer thinning, degeneration of retinal ganglion cells and changes of vascular parameters, aggregation of Aβ and tau protein, and apoptosis also occur in the retina of both AD and AMD. A protective effect of ocular administration of NGF on both photoreceptor and retinal ganglion cell degeneration has been recently described. Besides, the current knowledge about the detection of essential trace metals associated with AD and AMD and their changes depending on the severity of diseases, either systemic or locally detected, further pave the way for a promising diagnostic approach. This review is aimed at describing the employment of NGF as a common therapeutic approach to AMD and AD and the diagnostic power of detection of essential trace metals associated with both diseases. The multiple approaches employed to allow a sustained release/targeting of NGF to the brain and its neurosensorial ocular extensions will be also discussed, highlighting innovative technologies and future translational prospects.
Collapse
Affiliation(s)
- Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT)-CNR, Rome, Italy
- European Brain Research Institute, Rome, Italy
| | | | | | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Monica Varano
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Rome, Italy
| | | | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Rome, Italy
| |
Collapse
|
21
|
Minnella AM, Picardi SM, Maceroni M, Albanesi F, De Siena E, Placidi G, Caputo CG, De Vico U, Rizzo S, Falsini B. Retinal Morpho-Functional Changes Following 0.19 mg Fluocinolone Acetonide Intravitreal Implant for Chronic Diabetic Macular Edema. Adv Ther 2021; 38:3143-3153. [PMID: 33948926 PMCID: PMC8096132 DOI: 10.1007/s12325-021-01751-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/16/2021] [Indexed: 11/23/2022]
Abstract
Purpose To evaluate morpho-functional outcomes of the intravitreal fluocinolone acetonide (FAc) implant. Methods Retrospective, observational, single-center study. Primary endpoint was the mean change in central macular thickness (CMT) from baseline to month 1–3. Secondary endpoints included mean CMT change from baseline to month 4–8 and 9–14 and mean best corrected visual acuity (BCVA), photopic negative response (PhNR) and b-wave of flash full-field electroretinogram (ERG) changes from baseline to month 1–3, 4–8, and 9–14. Results Fourteen patients (18 eyes) were included. Mean (standard deviation) CMT decreased from 473 (196) µm at baseline to 371 (163) µm at month 1–3 (mean difference − 102.3 ± 98.35 µm, 95% CI ± 46.4 µm; p < 0.0001) and this decrease tended to endure up to month 9–14. BCVA did not change significantly. There was an improvement in mean PhNR amplitude from 2.76 (1.65) µV at baseline to 3.73 (2.32) µV at month 1–3 (mean difference 0.91 (1.14) µV, 95% CI ± 0.54 µV, p = 0.003); b-wave amplitude improved from 8.83 (4.52) µV at baseline versus 10.05 (5.04) µV at month 1–3 (mean difference 1.22 (2.23) µV, 95% CI ± 1.08 µV, p = 0.0384). These ERG positive changes tended to endure up to month 9–14, although they did not reach statistical significance after month 3. Conclusions Intravitreal FAc implant significantly improved anatomic as well as functional outcomes related to middle and inner retinal layers, known to be altered in diabetic retinopathy. Our findings support the hypothesis that intravitreal FAc implant may exert a protective effect in diabetic retinas with diabetic macular edema.
Collapse
|
22
|
Franzese O, Di Francesco AM, Meco D, Graziani G, Cusano G, Levati L, Riccardi R, Ruggiero A. hTERT Transduction Extends the Lifespan of Primary Pediatric Low-Grade Glioma Cells While Preserving the Biological Response to NGF. Pathol Oncol Res 2021; 27:612375. [PMID: 34257579 PMCID: PMC8262147 DOI: 10.3389/pore.2021.612375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/10/2021] [Indexed: 12/21/2022]
Abstract
The neurotrophin nerve growth factor (NGF) modulates the growth of human gliomas and is able to induce cell differentiation through the engagement of tropomyosin receptor kinase A (TrkA) receptor, although the role played in controlling glioma survival has proved controversial. Unfortunately, the slow growth rate of low-grade gliomas (LGG) has made it difficult to investigate NGF effects on these tumors in preclinical models. In fact, patient-derived low-grade human astrocytoma cells duplicate only a limited number of times in culture before undergoing senescence. Nevertheless, replicative senescence can be counteracted by overexpression of hTERT, the catalytic subunit of telomerase, which potentially increases the proliferative potential of human cells without inducing cancer-associated changes. We have extended, by hTERT transduction, the proliferative in vitro potential of a human LGG cell line derived from a pediatric pilocytic astrocytoma (PA) surgical sample. Remarkably, the hTERT-transduced LGG cells showed a behavior similar to that of the parental line in terms of biological responses to NGF treatment, including molecular events associated with induction of NGF-related differentiation. Therefore, transduction of LGG cells with hTERT can provide a valid approach to increase the in vitro life-span of patient-derived astrocytoma primary cultures, characterized by a finite proliferative potential.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela M Di Francesco
- Institute of Internal Medicine, Periodic Fever and Rare Diseases Center, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Daniela Meco
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gabriella Cusano
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | | | - Riccardo Riccardi
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | - Antonio Ruggiero
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| |
Collapse
|
23
|
Kanu LN, Ciolino JB. Nerve Growth Factor as an Ocular Therapy: Applications, Challenges, and Future Directions. Semin Ophthalmol 2021; 36:224-231. [PMID: 33641595 DOI: 10.1080/08820538.2021.1890793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nerve growth factor (NGF), the prototypical neurotrophin first discovered in the 1950s, has recently garnered increased interest as a therapeutic agent promoting neuronal health and regeneration. After gaining orphan drug status within the last decade, NGF-related research and drug development has accelerated. The purpose of this article is to review the preclinical and clinical evidence of NGF in various applications, including central and peripheral nervous system, skin, and ophthalmic disorders. We focus on the ophthalmic applications including not only the FDA-approved indication of neurotrophic keratitis but also retinal disease and glaucoma. NGF represents a promising therapy whose therapeutic profile is evolving. The challenges related to this therapy are reviewed, along with possible solutions and future directions.
Collapse
Affiliation(s)
- Levi N Kanu
- 1. Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Joseph B Ciolino
- 1. Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Eftimiadi G, Soligo M, Manni L, Di Giuda D, Calcagni ML, Chiaretti A. Topical delivery of nerve growth factor for treatment of ocular and brain disorders. Neural Regen Res 2021; 16:1740-1750. [PMID: 33510063 PMCID: PMC8328750 DOI: 10.4103/1673-5374.306062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are a family of proteins that support neuronal proliferation, survival, and differentiation in the central and peripheral nervous systems, and are regulators of neuronal plasticity. Nerve growth factor is one of the best-described neurotrophins and has advanced to clinical trials for treatment of ocular and brain diseases due to its trophic and regenerative properties. Prior trials over the past few decades have produced conflicting results, which have principally been ascribed to adverse effects of systemic nerve growth factor administration, together with poor penetrance of the blood-brain barrier that impairs drug delivery. Contrastingly, recent studies have revealed that topical ocular and intranasal nerve growth factor administration are safe and effective, suggesting that topical nerve growth factor delivery is a potential alternative to both systemic and invasive intracerebral delivery. The therapeutic effects of local nerve growth factor delivery have been extensively investigated for different ophthalmic diseases, including neurotrophic keratitis, glaucoma, retinitis pigmentosa, and dry eye disease. Further, promising pharmacologic effects were reported in an optic glioma model, which indicated that topically administered nerve growth factor diffused far beyond where it was topically applied. These findings support the therapeutic potential of delivering topical nerve growth factor preparations intranasally for acquired and degenerative brain disorders. Preliminary clinical findings in both traumatic and non-traumatic acquired brain injuries are encouraging, especially in pediatric patients, and clinical trials are ongoing. The present review will focus on the therapeutic effects of both ocular and intranasal nerve growth factor delivery for diseases of the brain and eye.
Collapse
Affiliation(s)
- Gemma Eftimiadi
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Daniela Di Giuda
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Lucia Calcagni
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
25
|
Attina G, Maurizi P, Triarico S, Capozza MA, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2020; 13:1601-1606. [DOI: 10.13005/bpj/2035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Optic pathway gliomas (OPG) are a common cancer in children with neurofibromatosis type 1. OPGs can cause clinical symptoms such as reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.The current guidelines provide for wait and see as the main approach if the tumor is not causing visual deterioration and adopting treatment only in the event of significant impairment of the visual function. Therefore, it is essential to early detect the visual deterioration changes as well as the identification of children eligible for treatment.
Collapse
Affiliation(s)
- Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
26
|
Minnella AM, Maceroni M, Picardi SM, Placidi G, De Siena E, Rizzo S, Falsini B. Combined Intravitreal Dexamethasone Implant and Cataract Surgery in Patients with Diabetic Retinopathy: Effect on Retinal Morphology and Function. Adv Ther 2020; 37:4675-4684. [PMID: 32970316 PMCID: PMC7547947 DOI: 10.1007/s12325-020-01506-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 09/14/2020] [Indexed: 01/30/2023]
Abstract
Introduction Cataract surgery can be associated with vision-threatening complications in patients with diabetes. This study aimed to assess the functional and anatomic outcomes of the intravitreal dexamethasone (DEX) implant, administered at the time as cataract surgery, in patients with diabetic retinopathy and diabetic macular edema (DME). Methods This was a retrospective, observational, and single-center study. The primary endpoint was the mean change in central macular thickness (CMT) from baseline to month 1. Secondary endpoints included mean change in best corrected visual acuity (BCVA) from baseline to month 1 and 3, mean change in CMT from baseline to month 3, the photopic negative response (PhNR) and the b wave of flash full-field electroretinogram from baseline to month 1, and the incidence of adverse events. Results Twenty-four eyes of 21 patients were included in the study. The mean (range) age of patients was 69 (63–87) years and 13 (61.9%) were men. Mean (standard deviation) CMT significantly decreased from 447 (134) µm at baseline to 341 (134) µm at month 1 (mean difference − 106 ± 134 µm, 95% CI − 183.9 to − 28.1 µm; p = 0.0087). BCVA significantly improved from 46 (20) ETDRS letters at baseline to 59 (22) ETDRS letters at month 1 (mean difference 13 ± 21 letters, 95% CI 0.8–25.2 letters; p = 0.0375). Regarding electrophysiology, there was a statistically significant reduction in mean PhNR from 5.24 (1.67) µV at baseline to 3.73 (1.19) µV at month 1 (mean difference − 1.51 ± 0.42 µV, 95% CI − 2.4 to − 0.7 µV, p = 0.0008); whereas b wave amplitude did not change (12.69 ± 6.89 µV at baseline versus 12.29 ± 6.30 µV at month 1; p = 0.8347). Four (16.7%) eyes developed ocular hypertension over the course of follow-up, which was successfully controlled with topical hypotensive medication. Conclusion Perioperative DEX implant significantly improved both anatomic and functional outcomes in patients with DME who underwent cataract surgery.
Collapse
|
27
|
Attina G, Ariano A, Maurizi P, Triarico S, Capozza MA, Coccia P, Rizzo D, Mastrangelo S, Ruggiero A. Treatment and Long-Term Sequelae in Childhood Brain Tumors. Open Neurol J 2020. [DOI: 10.2174/1874205x02014010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In children treated for brain tumors, important deficits in cognitive development have been described. The reduction of Intelligence Quotient (IQ) is correlated with multiple conditions such as tumor location, obstructive hydrocephalus, surgical intervention, and above all, the use of radiotherapy, especially in young children. Demyelinization represents the most striking microscopic alteration following radiation: cerebral white matter’s loss and failure to white matter development could partly account for changes in IQ score.Recently, combined chemo-radiotherapeutic approaches and the improvement of radiotherapy techniques have enabled the reduction of neurocognitive symptoms and improved the standard of life of childhood brain tumor survivors.
Collapse
|
28
|
Intranasal nerve growth factor administration improves neurological outcome after GBS meningitis. Childs Nerv Syst 2020; 36:2083-2088. [PMID: 32274529 DOI: 10.1007/s00381-020-04590-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/23/2020] [Indexed: 01/26/2023]
Abstract
Nerve growth factor (NGF) is a neurotrophin that promotes neural recovery and plasticity after experimental brain injury, supporting neuronal growth, differentiation, and survival of brain cells. Only a few studies reported NGF administration in pediatric patients with impaired brain functions after traumatic injuries, ischemic or infectious diseases, such as meningitis. We described the beneficial therapeutic effects of human-recombinant nerve growth factor (hr-NGF) treatment in an infant with persistent unresponsive wakefulness syndrome (UWS), due to late-onset group B Streptococcus meningitis. The infant received five monthly cycles of intranasal hr-NGF (0.1 mg/kg, 3 times daily for 7 consecutive days) through a mucosal atomizer device (MAD). NGF administration improved functional [positron emission tomography/computed tomography (PET/CT), single-photon emission/computed tomography (SPECT/CT), and magnetic resonance imaging (MRI)] assessments, electrophysiological [Electroencephalogram (EEG)] studies, as well as main cognitive processes and clinical and neurological functions. After hr-NGF treatment, significant improvements in facial mimicry, attention, motor reactions, oral motility, and feeding capacity were observed. She also recovered some hypothalamic functions and her cough reflex was restored. No side effects were reported during and after the treatment. For the first time ever, hr-NGF has been successfully utilized in an infant with UWS and severe neurologic outcome due to a bacterial meningitis. Although further studies are needed for better understanding the neuroprotective role of this neurotrophin, intranasal hr-NGF administration appears to be a promising and save rescuing strategy treatment in infants with severe neurological impairment after brain damage.
Collapse
|
29
|
Ciavarella C, Buzzi M, Bergantin E, Di Marco S, Giannaccare G, Campos E, Bisti S, Versura P. Effects of Cord Blood Serum (CBS) on viability of retinal Müller glial cells under in vitro injury. PLoS One 2020; 15:e0234145. [PMID: 32497126 PMCID: PMC7272066 DOI: 10.1371/journal.pone.0234145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/19/2020] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress and inflammation determine retinal ganglion cell degeneration, leading to retinal impairment and vision loss. Müller glial cells regulate retinal repair under injury, through gliosis. Meanwhile, reactive gliosis can turn in pathological effects, contributing to neurodegeneration. In the present study, we tested whether Cord Blood Serum (CBS), rich of growth factors, might improve the viability of Müller cells under in vitro damage. BDNF, NGF, TGF-α, GDNF and EGF levels were measured in CBS samples by Human Magnetic Luminex Assay. CBS effects were evaluated on rat (rMC-1) and human (MIO-M1) Müller cells, under H2O2 and IL-1β damage. Cells grown with FBS or CBS both at 5% were exposed to stress and analyzed in terms of cell viability, GFAP, IL-6 and TNF-α expression. CBS was also administrated after treatment with K252a, inhibitor of the neurotrophin receptor Trk. Cell viability of rMC-1 and MIO-M1 resulted significantly improved when pretreated with CBS and exposed to H2O2 and IL-1β, in comparison to the standard culture with FBS. Accordingly, the gliosis marker GFAP resulted down-regulated following CBS priming. In parallel, we observed a lower expression of the inflammatory mediators in rMC-1 (TNF-α) and MIO-M1 (IL-6, TNF- α), especially in presence of inflammatory damage. Trk inhibition through K252a administration impaired the effects of CBS under stress conditions on MIO-M1 and rMC-1 viability, not significantly different from FBS condition. CBS is enriched with neurotrophins and its administration to rMC-1 and MIO-M1 attenuates the cytotoxic effects of H2O2 and IL-1β. Moreover, the decrease of the main markers of gliosis and inflammation suggests a promising use of CBS for neuroprotection aims. This study is a preliminary basis that prompts future investigations to deeply explore and confirm the CBS potential.
Collapse
Affiliation(s)
- Carmen Ciavarella
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Marina Buzzi
- Emilia Romagna Cord Blood Bank-Transfusion Service, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Elisa Bergantin
- Emilia Romagna Cord Blood Bank-Transfusion Service, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | | | - Giuseppe Giannaccare
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Emilio Campos
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Silvia Bisti
- Vision Lab, DISCAB, University of L’Aquila, L’Aquila, Italy
- Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Piera Versura
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
- * E-mail:
| |
Collapse
|
30
|
Ruggiero A, Ariano A, Triarico S, Capozza MA, Romano A, Maurizi P, Mastrangelo S, Attinà G. Temozolomide and oral etoposide in children with recurrent malignant brain tumors. Drugs Context 2020; 9:dic-2020-3-1. [PMID: 32547627 PMCID: PMC7271709 DOI: 10.7573/dic.2020-3-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/01/2020] [Accepted: 05/08/2020] [Indexed: 11/26/2022] Open
Abstract
Despite advances in the treatment of brain tumors, the prognosis of children with recurrent malignant brain tumors remains poor. Etoposide (VP-16), an inhibitor of nuclear enzyme deoxyribonucleic acid (DNA)-topoisomerase II, has shown activity in brain tumors. Its efficacy appears schedule dependent but, to date, the most effective schedule of administration has not been well defined. Temozolomide (TMZ), like VP-16, penetrates the blood–brain barrier and has activity against malignant brain tumors. This novel alkylating agent is rapidly absorbed and is highly bioavailable after oral administration. The antitumor activity of TMZ has been shown to be schedule dependent. Based on the evidence of different mechanisms of cytotoxicity, TMZ and VP-16 have been utilized in combination in patients with malignant brain tumors. This review evaluates the results derived from the combination use of TMZ and oral VP-16. The reported data suggest potential activity of oral VP-16 and TMZ alone or in combination. Further clinical trials are needed to explore and confirm their promising activity in relapsed brain neoplasms.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Anna Ariano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
31
|
Di Marco S, Riccitelli S, Di Paolo M, Campos E, Buzzi M, Bisti S, Versura P. Cord Blood Serum (CBS)-Based Eye Drops Modulate Light-Induced Neurodegeneration in Albino Rat Retinas. Biomolecules 2020; 10:biom10050678. [PMID: 32354031 PMCID: PMC7277721 DOI: 10.3390/biom10050678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of visual loss in western countries, it has no cure, and its incidence will grow in the future, for the overall population aging. Albino rats with retinal degeneration induced by exposure to high-intensity light (light-damage, LD) have been extensively used as a model of AMD to test neuroprotective agents. Among them, trophic factors (NGF and BDNF) have been shown to play a significant role in photoreceptors' survival. Interestingly, cord blood serum (CBS) is an extract full of chemokines and trophic factors; we, therefore, hypothesized that CBS could be an excellent candidate for neuroprotection. Here, we investigate whether CBS-based eye drops might mitigate the effects of light-induced retinal degeneration in albino rats. CBS treatment significantly preserved flash-electroretinogram (f-ERG) response after LD and reduced the "hot-spot" extension. Besides, CBS-treated animals better preserved the morphology of the outer nuclear layer, together with a reduction in microglia migration and activation. Interestingly, the treatment did not modulate reactive gliosis and activation of the self-protective mechanism (FGF2). In conclusion, our results suggest that CBS-based eye drops might be successfully used to mitigate retinal neurodegenerative processes such as AMD.
Collapse
Affiliation(s)
- Stefano Di Marco
- Department of Applied Clinical Science and Biotechnology, University of L’Aquila, Via Vetoio, Coppito II, 67100 L’Aquila, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via Medaglie d’Oro 305, 00136 Roma, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 16132 Genova, Italy
- IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi, 16132 Genova, Italy
- Correspondence: (S.D.M.); (S.B.)
| | - Serena Riccitelli
- Department of Applied Clinical Science and Biotechnology, University of L’Aquila, Via Vetoio, Coppito II, 67100 L’Aquila, Italy
| | - Mattia Di Paolo
- Department of Applied Clinical Science and Biotechnology, University of L’Aquila, Via Vetoio, Coppito II, 67100 L’Aquila, Italy
| | - Emilio Campos
- Ophthalmology Unit, University of Bologna and S. Orsola-Malpighi Teaching Hospital, 40138 Bologna, Italy
| | - Marina Buzzi
- Emilia Romagna, Cord Blood Bank-Transfusion Service, S. Orsola-Malpighi Teaching Hospital, 40138 Bologna, Italy
| | - Silvia Bisti
- Department of Applied Clinical Science and Biotechnology, University of L’Aquila, Via Vetoio, Coppito II, 67100 L’Aquila, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via Medaglie d’Oro 305, 00136 Roma, Italy
- NetS3 Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genova, Italy
- Correspondence: (S.D.M.); (S.B.)
| | - Piera Versura
- Ophthalmology Unit, University of Bologna and S. Orsola-Malpighi Teaching Hospital, 40138 Bologna, Italy
| |
Collapse
|
32
|
Optic Pathway Glioma in Type 1 Neurofibromatosis: Review of Its Pathogenesis, Diagnostic Assessment, and Treatment Recommendations. Cancers (Basel) 2019; 11:cancers11111790. [PMID: 31739524 PMCID: PMC6896195 DOI: 10.3390/cancers11111790] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Type 1 neurofibromatosis (NF1) is a dominantly inherited condition predisposing to tumor development. Optic pathway glioma (OPG) is the most frequent central nervous system tumor in children with NF1, affecting approximately 15-20% of patients. The lack of well-established prognostic markers and the wide clinical variability with respect to tumor progression and visual outcome make the clinical management of these tumors challenging, with significant differences among distinct centers. We reviewed published articles on OPG diagnostic protocol, follow-up and treatment in NF1. Cohorts of NF1 children with OPG reported in the literature and patients prospectively collected in our center were analyzed with regard to clinical data, tumor anatomical site, diagnostic workflow, treatment and outcome. In addition, we discussed the recent findings on the pathophysiology of OPG development in NF1. This review provides a comprehensive overview about the clinical management of NF1-associated OPG, focusing on the most recent advances from preclinical studies with genetically engineered models and the ongoing clinical trials.
Collapse
|
33
|
The photopic negative response of the Light-adapted 3.0 ERG in clinical settings. Doc Ophthalmol 2019; 140:115-128. [DOI: 10.1007/s10633-019-09723-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 09/16/2019] [Indexed: 11/27/2022]
|
34
|
Xie C, Lu D, Xu M, Qu Z, Zhang W, Wang H. Knockdown of RAD18 inhibits glioblastoma development. J Cell Physiol 2019; 234:21100-21112. [PMID: 31081138 DOI: 10.1002/jcp.28713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
This study aimed at investigating the role of RAD18 in the regulation of glioblastoma development as well as the underlying mechanisms. The human glioblastoma U251 and U87MG cells were transfected with siRNAs specifically targeting RAD18, and the effects of knockdown of RAD18 on the viability, apoptosis, migration, and invasion of U251 and U87MG cells were investigated. Transcriptome sequencing of the siRNA-RAD18-tranfected and siRNA-NC-transfected U251 cells was performed, followed by bioinformatic analyses for sequencing data. The results showed that knockdown of RAD18 significantly inhibited cell viability, promoted apoptosis, and suppressed migration and invasion of U251 and U87MG cells. Bioinformatic analyses of sequencing data identified 1,051 differentially expressed genes (DEGs) (369 up- and 682 downregulated genes) in the siRNA-RAD18-transfected U251 cells compared with siRNA-NC-transfected U251 cells. Eleven DEGs, including nerve growth factor (NGF), colony-stimulating factor 2 (CSF2), matrix metallopeptidase 1 (MMP1), platelet-derived growth factor receptor α (PDGFRA), and heme oxygenase 1 (HMOX1), were identified as the hub nodes in protein-protein interaction (PPI) network. Moreover, the aforementioned 11 hub genes were significantly enriched in PI3K-Akt signaling pathway and GO functions associated with the extracellular region. Notably, quantitative real-time polymerase chain reaction further confirmed that the expression levels of NGF, CSF2, HMOX1, and MMP1 were significantly downregulated, while that of PDGFRA was markedly upregulated in the siRNA-RAD18-transfected U251 cells than in the siRNA-NC cells. In conclusion, the knockdown of RAD18 may inhibit glioblastoma development by regulating the expression of the aforementioned key DEGs.
Collapse
Affiliation(s)
- Chen Xie
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Dejuan Lu
- Department of Endoscope, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meng Xu
- Department of Neurosurgery, First People's Hospital of Heihe, Heihe, Heilongjiang, People's Republic of China
| | - Zhengyi Qu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Weiguang Zhang
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Hongwei Wang
- Department of Minimally Invasive Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
35
|
Rakotonjanahary J, Gravier N, Lambron J, De Carli E, Toulgoat F, Delion M, Pellier I, Rialland X. Long-term visual acuity in patients with optic pathway glioma treated during childhood with up-front BB-SFOP chemotherapy-Analysis of a French pediatric historical cohort. PLoS One 2019; 14:e0212107. [PMID: 30849081 PMCID: PMC6407847 DOI: 10.1371/journal.pone.0212107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/28/2019] [Indexed: 11/18/2022] Open
Abstract
Background Visual outcome is one of the main issues in the treatment of optic pathway glioma in childhood. Although the prognostic factors of low vision have been discussed extensively, no reliable indicators for visual loss exist. Therefore, we aimed to define initial and evolving factors associated with long-term vision loss. Methods We conducted a multicenter historical cohort study of children treated in France with up-front BB-SFOP chemotherapy between 1990 and 2004. Visual acuity performed at the long-term follow-up visit or within 6 months prior was analyzed. Logistic regression analysis was used to estimate the effects of clinical and radiological factors on long-term visual outcome. Findings Of the 180 patients in the cohort, long-term visual acuity data were available for 132 (73.3%) patients (median follow-up: 14.2 years; range: 6.1–25.6). At the last follow-up, 61/132 patients (46.2%) had impaired vision, and 35 of these patients (57.3%) were partially sighted or blind. Multivariate analysis showed that factors associated with a worse prognosis for long-term visual acuity were an age at diagnosis of < 1 year (OR 3.5 [95% CI: 1.1–11.2], p = 0.04), tumor extent (OR 4.7 [95% CI: 1.2–19.9], p = 0.03), intracranial hypertension requiring one or more surgical procedures (OR 5.6 [95% CI: 1.8–18.4], p = 0.003), and the need for additional treatment after initial BB-SFOP chemotherapy (OR 3.5 [95% CI: 1.1–11.9], p = 0.04). NF1 status did not appear as a prognostic factor, but in non-NF1 patients, a decrease in tumor volume with contrast enhancement after BB-SFOP chemotherapy was directly associated with a better visual prognosis (OR 0.8 [95% CI: 0.8–0.9], p = 0.04). Interpretation Our study confirms that a large proportion of children with optic pathway glioma have poor long-term outcomes of visual acuity. These data suggest new prognostic factors for visual acuity, but these results need to be confirmed further by large- and international-scale studies.
Collapse
Affiliation(s)
| | - Nicolas Gravier
- Kervision - Polyclinique de l’Atlantique, Nantes-Saint Herblain, France
| | - Julien Lambron
- Department of Pediatric Radiology, Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University Paris Diderot, Sorbonne Paris Cité, France
| | - Emilie De Carli
- Department of Pediatric Oncology, University Hospital, Angers, France
| | | | - Matthieu Delion
- Department of Neurosurgery, University Hospital, Angers, France
| | - Isabelle Pellier
- Department of Pediatric Oncology, University Hospital, Angers, France
- UMR Inserm 892 - CNRS 6299 - CRCNA, Angers, France
| | - Xavier Rialland
- Department of Pediatric Oncology, University Hospital, Angers, France
| |
Collapse
|
36
|
Granata G, Iodice F, Romanello R, Placidi G, Gasparini G, Falsini B, Rossini PM. Neurophysiological effect of transorbital electrical stimulation: Early results in advanced optic atrophy. Brain Stimul 2019; 12:800-802. [PMID: 30777642 DOI: 10.1016/j.brs.2019.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 10/27/2022] Open
Affiliation(s)
- Giuseppe Granata
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | | | | | - Giorgio Placidi
- Ophtalmology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Gasparini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Paolo Maria Rossini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Neurology Institute, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
37
|
Mesentier-Louro LA, Rosso P, Carito V, Mendez-Otero R, Santiago MF, Rama P, Lambiase A, Tirassa P. Nerve Growth Factor Role on Retinal Ganglion Cell Survival and Axon Regrowth: Effects of Ocular Administration in Experimental Model of Optic Nerve Injury. Mol Neurobiol 2019; 56:1056-1069. [PMID: 29869196 DOI: 10.1007/s12035-018-1154-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/24/2018] [Indexed: 01/04/2023]
Abstract
Retinal ganglion cell (RGC) degeneration occurs within 2 weeks following optic nerve crush (ONC) as a consequence of reduced retro-transport of growth factors including nerve growth factor (NGF). The hypothesis that intravitreal (ivt) and eye drop (ed) administration of recombinant human NGF (rhNGF) might counteract ONC in adult rats is explored in this study. We found that both ivt- and ed-rhNGF reduced RGC loss and stimulated axonal regrowth. Chiefly, survival and regenerative effects of rhNGF were associated with a reduction of cells co-expressing Nogo-A/p75NTR at crush site borders, which contribute to glia scar formation following nerve injury, and induce further degeneration. We also found that ocular application of rhNGF reduced p75NTR and proNGF and enhanced phosphorylation of TrkA and its intracellular signals at retina level. Nogo-R and Rock2 expression was also normalized by ed-rhNGF treatment in both ONC and contralateral retina. Our findings that ocular applied NGF reaches and exerts biological actions on posterior segment of the eye give a further insight into the neurotrophin diffusion/transport through eye structures and/or their trafficking in optic nerve. In addition, the use of a highly purified NGF form in injury condition in which proNGF/p75NTR binding is favored indicates that increased availability of mature NGF restores the balance between TrkA and p75NGF, thus resulting in RGC survival and axonal growth. In conclusion, ocular applied NGF is confirmed as a good experimental paradigm to study mechanisms of neurodegeneration and regeneration, disclose biomarkers, and time windows for efficacy treatment following cell or nerve injury.
Collapse
Affiliation(s)
- Louise A Mesentier-Louro
- Eye Repair Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pamela Rosso
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo F Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paolo Rama
- Eye Repair Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Lambiase
- Section of Ophthalmology, Department of Sense Organs, University Sapienza, Rome, Italy
| | - Paola Tirassa
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Via di Fosso di Fiorano, 64 (00143), Rome, Italy.
| |
Collapse
|
38
|
Zeng L, Yang C, Ming Y, Luo S, Chen L. Bioinformatics Analysis Reveals Potential Candidate Genes for Different Glioma Subtypes (Astrocytoma, Ependymoma, and Oligodendroglioma). Cancer Biother Radiopharm 2018. [DOI: 10.1089/cbr.2018.2475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Liangnan Zeng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changmei Yang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Ming
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shihong Luo
- Department of Neurosurgery, Hospital of Stomatology Southwest Medical University, Luzhou, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
39
|
Freret ME, Gutmann DH. Insights into optic pathway glioma vision loss from mouse models of neurofibromatosis type 1. J Neurosci Res 2018; 97:45-56. [PMID: 29704429 DOI: 10.1002/jnr.24250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 gene. The NF1-encoded protein (neurofibromin) is an inhibitor of the oncoprotein RAS and controls cell growth and survival. Individuals with NF1 are prone to developing low-grade tumors of the optic nerves, chiasm, tracts, and radiations, termed optic pathway gliomas (OPGs), which can cause vision loss. A paucity of surgical tumor specimens and of patient-derived xenografts for investigative studies has limited our understanding of human NF1-associated OPG (NF1-OPG). However, mice genetically engineered to harbor Nf1 gene mutations develop optic gliomas that share many features of their human counterparts. These genetically engineered mouse (GEM) strains have provided important insights into the cellular and molecular determinants that underlie mouse Nf1 optic glioma development, maintenance, and associated vision loss, with relevance by extension to human NF1-OPG disease. Herein, we review our current understanding of NF1-OPG pathobiology and describe the mechanisms responsible for tumor initiation, growth, and associated vision loss in Nf1 GEM models. We also discuss how Nf1 GEM and other preclinical models can be deployed to identify and evaluate molecularly targeted therapies for OPG, particularly as they pertain to future strategies aimed at preventing or improving tumor-associated vision loss in children with NF1.
Collapse
Affiliation(s)
- Morgan E Freret
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
40
|
Chai C, Song LJ, Han SY, Li XQ, Li M. MicroRNA-21 promotes glioma cell proliferation and inhibits senescence and apoptosis by targeting SPRY1 via the PTEN/PI3K/AKT signaling pathway. CNS Neurosci Ther 2018; 24:369-380. [PMID: 29316313 DOI: 10.1111/cns.12785] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022] Open
Abstract
AIMS Our study aims to investigate the effect of microRNA-21 (miR-21) on the proliferation, senescence, and apoptosis of glioma cells by targeting SPRY1 via the PTEN/PI3K/AKT signaling pathway. METHODS Glioma tissues and brain tissues were collected for this study after surgical decompression for traumatic brain injury. RT-qPCR was employed to measure mRNA levels of miR-21, SPRY1, PTEN, PI3K, and AKT, and Western blotting was conducted to determine protein levels of SPRY1, PTEN, PI3K, AKT, p-AKT, Caspase-3, Caspase-9, P53, GSK3, and p-GSK3. Human glioma U87 cells were assigned into the blank, negative control (NC), miR-21 mimics, miR-21 inhibitors, siRNA-SPRY1, and miR-21 inhibitors + siRNA-SPRY1 groups, with human HEB cells serving as the normal group. Cell proliferation, cell cycle, and apoptosis were determined by MTT and flow cytometry, respectively. RESULTS Compared with control group, an increased expression of miR-21, PI3K, AKT, p-AKT, P53, and p-GSK3, and a decreased expression of SPRY1, PTEN, Caspase-3, and Caspase-9 were observed in the glioma group, and no significant differences were found in the expression of GSK3. SPRY1 was verified to be the target gene of miR-21. Compared with the blank and NC groups, levels of PI3K, AKT, p-AKT, P53, and p-GSK3 increased while levels of SPRY1, PTEN, Caspase-3, and Caspase-9 decreased in the miR-21 mimics and siRNA-SPRY1 groups; the miR-21 inhibitors group reversed the tendency; furthermore, the miR-21 inhibitors group showed decreased cell proliferation but promoted apoptosis, which were opposite to the results of the miR-21 mimics and siRNA-SPRY1 groups. CONCLUSION MicroRNA-21 might promote cell proliferation and inhibit cell senescence and apoptosis of human glioma cells by targeting SPRY1 via the PTEN/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Chang Chai
- Henan Eye Institute, Henan Provincial People's Hospital, Zhengzhou, China
| | - Lai-Jun Song
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang-Yin Han
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xi-Qing Li
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Ming Li
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
41
|
Rocco ML, Soligo M, Manni L, Aloe L. Nerve Growth Factor: Early Studies and Recent Clinical Trials. Curr Neuropharmacol 2018; 16:1455-1465. [PMID: 29651949 PMCID: PMC6295934 DOI: 10.2174/1570159x16666180412092859] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/23/2018] [Accepted: 04/04/2018] [Indexed: 12/02/2022] Open
Abstract
Since its discovery, nerve growth factor (NGF) has long occupied a critical role in developmental and adult neurobiology for its many important regulatory functions on the survival, growth and differentiation of nerve cells in the peripheral and central nervous system. NGF is the first discovered member of a family of neurotrophic factors, collectively indicated as neurotrophins, (which include brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin 4/5). NGF was discovered for its action on the survival and differentiation of selected populations of peripheral neurons. Since then, an enormous number of basic and human studies were undertaken to explore the role of purified NGF to prevent the death of NGF-receptive cells. These studies revealed that NGF possesses important therapeutic properties, after topical administration, on human cutaneous pressure ulcer, corneal ulcers, glaucoma, retinal maculopathy, Retinitis Pigmentosa and in pediatric optic gliomas and brain traumas. The aim of this review is to present our previous, recent and ongoing clinical studies on the therapeutic properties of NGF.
Collapse
Affiliation(s)
| | | | | | - Luigi Aloe
- Address correspondence to this author at the Fondazione IRET ONLUS, Via Tolara di Sopra 41/E, 40064 Ozzano Emilia (BO), Italy; Tel: +39-051-798776; Fax: +39-051-799673; E-mail:
| |
Collapse
|
42
|
Tirassa P, Rosso P, Iannitelli A. Ocular Nerve Growth Factor (NGF) and NGF Eye Drop Application as Paradigms to Investigate NGF Neuroprotective and Reparative Actions. Methods Mol Biol 2018; 1727:19-38. [PMID: 29222770 DOI: 10.1007/978-1-4939-7571-6_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The eye is a central nervous system structure that is uniquely accessible to local treatment. Through the ocular surface, it is possible to access the retina, optic nerve, and brain. Animal models of retina degeneration or optic nerve crush could thus serve as tools to investigate whether and how factors, which are anterogradely or retrogradely transported through the optic nerve, might contribute to activate neuroprotection and eventually regeneration. Among these factors, nerve growth factor (NGF) plays a crucial role during development of the visual system, as well as during the entire life span, and in pathological conditions. The ability of NGF to exert survival and trophic actions on the retina and brain cells when applied intraocularly and topically as eye drops is critically reviewed here, together with the effects of ocular neurotrophins on neuronal pathways influencing body rhythm, cognitions, and behavioral functions. The latest data from animal models and humans are presented, and the mechanism of action of ocularly administered NGF is discussed. NGF eye drops are proposed as an experimental strategy to investigate the role and cellular targets of neurotrophins in the mechanism(s) underlying neurodegeneration/regeneration and their involvement in the regulation of neurological and behavioral dysfunctions.
Collapse
Affiliation(s)
- Paola Tirassa
- National Research Council (CNR), Institute of Cell Biology & Neurobiology, Rome, Italy.
| | - Pamela Rosso
- National Research Council (CNR), Institute of Cell Biology & Neurobiology, Rome, Italy.,Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Angela Iannitelli
- Department of Human Sciences, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
43
|
Affiliation(s)
- Neil R Miller
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Huang D, Lin C, Wen X, Gu S, Zhao P. A Potential Nanofiber Membrane Device for Filling Surgical Residual Cavity to Prevent Glioma Recurrence and Improve Local Neural Tissue Reconstruction. PLoS One 2016; 11:e0161435. [PMID: 27548322 PMCID: PMC4993477 DOI: 10.1371/journal.pone.0161435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 08/05/2016] [Indexed: 12/20/2022] Open
Abstract
This study aims to develop a novel device with nanofiber membrane capable of sustained release of temozolomide (TMZ) and neuron growth factor (NGF). An improved bio-availability of TMZ and NGF in surroundings proximal to the device was expected to be attained for a prolonged period of time. The device was developed by integrating TMZ-doped polycaprolactone (PCL) nanofiber (TP) membrane and NGF-coated PCL (NGFP) membrane using sodium alginate hydrogel. TP was prepared by direct electrospinning of TMZ/PCL. NGFP membrane was developed by layer-by-layer assembling technology. The incorporation of TMZ-doped nanofiber and NGFP nanofiber in the device was confirmed by scanning electron microscopy. The number of NGF layer in NGF-coated PCL membrane could be readily measured with energy spectrum analysis. The in vitro release study showed that TP-NGFP-TP membrane could efficiently liberate TMZ to inhibit the growth of C6 glioma cells, and sufficient NGF to induce the differentiation of PC12 neuron cells over four weeks. Such TP-NGFP-TP membrane device can be employed as a tampon to fill up surgical residual cavity and afford residual glioma removal, structural support, hemostasis, and local neural tissue reconstruction in the surgical treatment of glioma. The study opens a horizon to develop multifunctional biomaterial device for maximized glioma treatment efficacy.
Collapse
Affiliation(s)
- Daoxiang Huang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Institute of Biomedical Engineering and Nanoscience, Tongji University School of Medicine, Shanghai, People's Republic of China
- Institute of Nano and Bio-Polymeric Materials, School of Material Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Chao Lin
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Institute of Biomedical Engineering and Nanoscience, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xuejun Wen
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Institute of Biomedical Engineering and Nanoscience, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shuying Gu
- Key Laboratory of Advanced Civil Engineering Materials Ministry of Education, School of Material Science and Engineering, Tongji University, Shanghai, People's Republic of China
- Institute of Nano and Bio-Polymeric Materials, School of Material Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Peng Zhao
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Institute of Biomedical Engineering and Nanoscience, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
45
|
Editorial. Brain 2016; 139:1001. [DOI: 10.1093/brain/aww058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
46
|
Freret ME, Gutmann DH. Optic pathway gliomas in neurofibromatosis-1: controversies and recommendations. Ann Neurol 2007; 61:189-98. [PMID: 17387725 PMCID: PMC5908242 DOI: 10.1002/ana.21107] [Citation(s) in RCA: 373] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Optic pathway glioma (OPG), seen in 15% to 20% of individuals with neurofibromatosis type 1 (NF1), account for significant morbidity in young children with NF1. Overwhelmingly a tumor of children younger than 7 years, OPG may present in individuals with NF1 at any age. Although many OPG may remain indolent and never cause signs or symptoms, others lead to vision loss, proptosis, or precocious puberty. Because the natural history and treatment of NF1-associated OPG is different from that of sporadic OPG in individuals without NF1, a task force composed of basic scientists and clinical researchers was assembled in 1997 to propose a set of guidelines for the diagnosis and management of NF1-associated OPG. This new review highlights advances in our understanding of the pathophysiology and clinical behavior of these tumors made over the last 10 years. Controversies in both the diagnosis and management of these tumors are examined. Finally, specific evidence-based recommendations are proposed for clinicians caring for children with NF1.
Collapse
Affiliation(s)
| | - David H. Gutmann
- To whom correspondence should be addressed: Telephone: 314-362-7379, Facsimile: 314-362-2388,
| |
Collapse
|