Open Access
Review  |   June 2024
An Overview of Optic Pathway Glioma With Neurofibromatosis Type 1: Pathogenesis, Risk Factors, and Therapeutic Strategies
Author Affiliations & Notes
  • 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
  • Correspondence: Xianqun Fan, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, P.R. China; [email protected]
  • Yefei Wang, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, P.R. China; [email protected]
  • Footnotes
     YC and JY contributed equally to this report.
Investigative Ophthalmology & Visual Science June 2024, Vol.65, 8. doi:https://doi.org/10.1167/iovs.65.6.8
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Ying Chen, Jie Yu, Shengfang Ge, Renbing Jia, Xin Song, Yefei Wang, Xianqun Fan; An Overview of Optic Pathway Glioma With Neurofibromatosis Type 1: Pathogenesis, Risk Factors, and Therapeutic Strategies. Invest. Ophthalmol. Vis. Sci. 2024;65(6):8. https://doi.org/10.1167/iovs.65.6.8.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
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.

Optic pathway gliomas (OPGs) are the most predominant pilocytic astrocytomas (PAs), classified as World Health Organization (WHO) grade I, constituting up to 7.0% of all gliomas.1,2 OPGs are most commonly found in patients with neurofibromatosis type 1 (NF1), the most common tumor predisposition syndrome. According to a retrospective study, 59% of the 133 patients with OPGs are diagnosed as NF1.3 Meanwhile, among children with NF1, OPGs are also the most predominant central nervous system (CNS) tumors, with an estimated prevalence of 15% to 20%.47 Generally, NF1-associated OPGs (NF1-OPGs) are typically diagnosed in children younger than 7 years old (mean of 4.5 years old) and affect the optic pathway from the optic nerves (ONs), optic chiasm, and optic tracts to optic radiations.811 Specifically, 75% to 85% of OPGs in individuals with NF1 occur in the ONs and chiasm (anterior optic pathway).12 A retrospective study showed that approximately 34.8% of patients with NF1-OPGs have bilateral gliomas.10 There are various distinctions between NF1-OPGs and sporadic OPGs despite their common pathologic classification. Sporadic OPGs are more likely to affect chiasmal and postchiasmal lesions, which exhibit more severe clinical symptoms and require more clinical intervention.1317 Without NF1 loss, most sporadic OPGs are characterized by activation of BRAF via fusion with KIAA1549, which activates downstream pathways.1821 
Although many NF1-OPGs tend to be indolent low-grade gliomas (LGGs), 30% to 50% of patients have visual disturbance owing to compression of the optic nerve5,22,23 or precocious puberty caused by hypothalamic involvement.24,25 The deterioration of vision usually manifests as a progressive visual decline or normal visual symptoms with anomalous ophthalmological tests.26 Ophthalmological signs of OPGs range from optic nerve edema, optic nerve pallor, optic nerve atrophy, and/or proptosis.4 Notably, although toddlers may suffer from deteriorative vision loss, they seldom complain about it.26 Hence, early screening and risk assessments are essential. A series of clinical studies have found that in patients who are younger than 2 years old2729 and girls27,30 as well, as having tumors located in the postchiasmal optic pathway9,10,22,27,29 or hypothalamus,25 NF1-OPGs may produce more aggressive symptoms and have worse prognoses. However, more accurate prognostic risk factors are needed for precision medicine strategies for related patients. 
As mentioned above, most of NF1-OPGs are indolent and asymptomatic. Hence, the main objective of therapy is to reduce the risk of a permanent, clinically significant visual impairment.31 Therapy intervention is frequently provided for symptomatic children who have worsening vision loss (a 2-line reduction in visual acuity).4,32 The role of surgical resection is limited due to the danger of irreversible nerve damage. Secondary neoplasms33 and radiotherapy-related vasculopathy34 also prevent radiotherapy from being used. Therefore, chemotherapies, such as carboplatin and vincristine combined,3537 are the most common effective treatments in symptomatic children. Although chemotherapy can successfully attenuate tumor progression, it is estimated that nearly 30% of affected children remain visually impaired after treatment.27,38 Therefore, there is an urgent need to design new therapeutic strategies to prevent vision deterioration and improve visual outcomes in patients with NF1-OPGs before irreversible vision deficits occur. In this review, we summarized the latest advances in NF1-OPGs in terms of activated Ras effector pathways in optic glioma stem cells (o-GSCs), astrocytes and retinal ganglion cells (RGCs) as well as complex interactions within the tumor microenvironment, aiming to identify potential risk assessment algorithms and therapeutic modalities for affected children. 
Activated Ras Effector Pathways Mediate Glioma Pathogenesis
Located on chromosome 17q11.2, NF1 is a major tumor suppressor gene with 57 exons encoding a protein product of 220 to 250 kDa called neurofibromin.3942 Neurofibromin acts as a negative operator of the p21-Ras proto-oncogene by converting the active GTP-bound Ras proto-oncogene into an inactive GDP-bound state.26,4245 Based on the two-hit hypothesis of tumorigenesis,46 young patients with NF1 are born with one nonfunctional NF1 allele (germline NF1 gene mutation) in cells throughout the body that are heterozygous for NF1 (NF1+/−) but form gliomas only upon somatic mutation of another functional allele in stem cells.26,47,48 Whole-genome sequencing (WGS) analysis of NF1-PA samples revealed that loss of heterozygosity (LOH), frameshift mutations, and methylation were all possible mechanisms of somatic NF1 gene silencing.49 Deprivation of neurofibromin in NF1-related gliomas leads to increased Ras activation,5052 which plays a critical role in promoting cell development and survival in various tissues, including the developing brain of mammals.53,54 
There are 3 distinct Ras effector pathways: the phosphatidylinositol-3 kinase (PI3K)/protein kinase-B (AKT)/mechanistic target of rapamycin (mTOR) pathway,51,52,55 the mitogen activated protein kinase (MEK)/mitogen activated protein kinase (ERK) pathway,5557 and the cyclic adenosine monophosphate (cAMP) pathway.58,59 Upregulated Ras downstream pathways have distinct effects on diverse cells, such as o-GSCs, astrocytes, and RGCs, to determine tumorigenesis (Fig. 1). 
Figure 1.
 
Ras signaling pathway. Deprivation of neurofibromin in NF1-related OPGs led to activate Ras and its 3 effector pathways. Nf1−/− astrocytes displayed both upregulated MEK-ERK and PI3K-AKT manners, which individually contributed to activating mTOR activity and promoting tumor proliferation. Specifically, PI3K/AKT manner promoted the proliferation of o-GSCs and MEK/ERK pathway determined the progenitors multilineage differentiation. Besides, neurofibromin decreased the levels of cAMP in Nf1−/− astrocytes to positively accelerate tumor development while downregulated cAMP levels attenuated Nf1+/− RGC growth through PKCζ, which was related with visual dysfunction. This figure was created using the BioRender software.
Figure 1.
 
Ras signaling pathway. Deprivation of neurofibromin in NF1-related OPGs led to activate Ras and its 3 effector pathways. Nf1−/− astrocytes displayed both upregulated MEK-ERK and PI3K-AKT manners, which individually contributed to activating mTOR activity and promoting tumor proliferation. Specifically, PI3K/AKT manner promoted the proliferation of o-GSCs and MEK/ERK pathway determined the progenitors multilineage differentiation. Besides, neurofibromin decreased the levels of cAMP in Nf1−/− astrocytes to positively accelerate tumor development while downregulated cAMP levels attenuated Nf1+/− RGC growth through PKCζ, which was related with visual dysfunction. This figure was created using the BioRender software.
Optic Glioma Stem Cells
A widespread mechanism behind pediatric brain cancers is stalled developmental programs, resulting in the development of stem or progenitor populations to abnormally maintain their early stem-like properties.60 The rarity of NF1-OPGs in adults suggests that there might be transient neural progenitor cells (NPCs) susceptible to tumorigenesis through loss of neurofibromin during specific developmental periods.48 Researchers have discovered that o-GSCs are likely to reside in the third ventricular zone (TVZ) rather than the lateral ventricular subventricular zone (lv-SVZ).61,62 There may be a potential correlation between the specific anatomic location of putative o-GSCs and their impact on normal ONs growth. During embryonic development, astrocytes are produced locally from astrocyte precursor cells (APCs).63,64 Oligodendrocytes are derived from brain-derived oligodendrocyte precursor cells (OPCs) that arrive at the distal ONs at birth.64 Migrating OPCs are also referred to as migrating glial progenitors (GPs), owing to their role in differentiating into type 2 astrocytes.63 As early as the late embryonic stages, migratory GPs were thought to originate in radial glia (RG) in the TVZ.6568 
Following Nf1 loss, overactive Ras effector pathways interrupt the equilibrium of original cell maintenance in the TVZ and promote abnormal persistence of NPCs.65,69 Specifically, the PI3K/AKT pathway promoted the proliferation of progenitors, and the MEK/ERK pathway regulated progenitor multilineage differentiation.70 When ERK was hyperactivated, NPCs from the hypothalamic ventricular zone (hVZ) migrated into the hypothalamic mantle zone (hMZ) and failed to differentiate normally.62 Under MEK/SMAD family member 3 (Smad3)-dependent signaling, hairy and enhancer of split 1 (Hes1) mediate neuron and astrocyte differentiation, whereas oligodendrocyte lineage transcription factor 2 (Olig2) induces oligodendrocyte differentiation.70 In contrast, APCs in the ONs were independent of Nf1 loss, so they might not be the source of NF1-OPGs.62 In addition, migrating GPs induced the formation of OPGs in the neonatal ONs, leading to RGC apoptosis in a BCL-2-associated X protein (Bax)-dependent manner.62 Transient neonatal pharmacological inhibition with MEK or genetic loss of Mek1/Mek2 attenuated RGC loss and axon degeneration, which inhibited OPGs formation before irreversible neurological damage occurred.62 
Various Cre driver lines have been applied to target somatic Nf1 loss in different populations of NPCs in genetically engineered mice (GEM) to identify the specific o-GSCs of NF1-OPGs. Previous studies incorporated Cre-loxp strategies and revealed that Nf1+/− mice with conditional Nf1 mutations in Gfap+,71 CD133+ (Prom1),72 or Blbp+58 progenitors all formed OPGs by 3 months of age. In contrast, OPGs were not generated by somatic Nf1 loss in Ng2+ (nerve/glial antigen 2) cells during the embryogenic period73 or in GFAP+ astrocytes after birth.65 Consistent with the above results, further investigations identified 3 NPCs within the TVZ and revealed that Nestin+ Blbp+ CD133+ cells are the most likely source of murine Nf1 optic gliomas. They are the most clonogenic, proliferative, and abundant population during embryogenesis and then dramatically decline during early postnatal life.61 When multiple original cells have unique differentiation pathways to generate gliomas, transforming events often produce different tumor latencies and partially determine glioma biology. For example, Nf1 loss in Olig2+ OPCs postponed optic glioma latency to 6 months.72 Whole-tumor RNA-sequencing analyses also confirmed that FMC (Nf1flox/mut; GFAP-Cre) and FMOC (Nf1flox/mut; Olig2-cre) optic gliomas were completely distinct entities.74 
Somatic Nf1 mutation during a restricted developmental window was also critical to OPG gliomagenesis in children. Using the Nf1flox/null, Prom1-cre mouse model,75 studies have shown that murine Nf1 optic glioma formation requires somatic Nf1 loss during late embryonic development because postnatal loss within the same o-GSCs does not cause tumorigenesis.72 It was also mentioned above that progenitors within the TVZ were most plentiful during embryogenesis and sharply disappeared by postnatal day 1. In human TVZ, proliferation of progenitors also decreased with age,69 which might explain the NF1-OPG development in young children. There was a combination of factors that contributed to the unique patterning and penetrance of Nf1 optic gliomas, including the regulation of overactivated Ras signaling, composition of o-GSCs in TVZ, changes in progenitor proliferation over time, and the requirement for somatic Nf1 gene inactivation during embryonic development. 
Astrocytes
Nf1 −/− astrocytes displayed both upregulated MEK/ERK and PI3K/AKT signaling, which individually contributed to the activation of mTOR and promotion of tumor proliferation.55 In this regard, MEK/ERK activates mTOR by p90RSK, independent of the tuberous sclerosis complex (TSC)/Ras homolog enriched in the brain (Rheb).55,76 The inhibition of PI3K, MEK, or mTOR prevented Nf1 optic glioma development and decreased tumor volumes in vivo.55,77 Unfortunately, inhibition of these pathways was only effective during treatment, and tumors proliferated to pretreatment levels after the treatment had ended.77 Therefore, it is possible that combining treatments targeting mTOR with different effector arms may yield longer lasting improvements in OPGs outcomes.55 
Compared to the above two signaling pathways, neurofibromin decreased the level of cAMP in Nf1−/− astrocytes to positively accelerate tumor development.7880 A reduction in tumor proliferation could be observed by increasing intracellular cAMP levels with the phosphodiesterase-4A1 (PDE4A1) inhibitor rolipram.59 Furthermore, when PDE4A1 was ectopically expressed in the cortex, it produced glioma formation in the brain regions where OPGs did not grow naturally.59 Glioma progression can also be dramatically blocked by rolipram in Nf1 GEM in vivo. Therefore, it is possible to explain the pattern of NF1 gliomagenesis by differences in cAMP levels between brain regions. 
Retinal Ganglion Cells
In contrast to its positive role in Nf1-deficient astrocytes, impaired neurofibromin function downregulates cAMP levels to attenuate Nf1+/− RGC growth through the atypical protein kinase C-zeta (PKCζ), which is related to visual dysfunction.81,82 In Nf1­mutant mice, it was shown that a combination of progressively damaged RGC axons,83 accumulated RGC death,82 and reduced visual acuity30 led to vision loss. There was evidence that inhibition of Ras activity (lovastatin) or upregulation of cAMP levels (rolipram) can inhibit RGC apoptosis in mice with optic gliomas.82,84 
Tumor Microenvironment Permissiveness Contributes to Gliomagenesis
The tumor microenvironment plays a significant role in the formation and development of tumors, holding both positive and negative impacts on tumor cell growth.8587 The relevant non-neoplastic cells include microglia, T cells, neurons, endothelial cells, Müller cells, and so on, except fibroblasts and mast cells in both human NF1-related OPGs and Nf1 GEMs.47,88 Previous studies have shown that homozygous inactivation of Nf1 in o-GSCs alone was insufficient for tumor formation. In contrast, in the context of the Nf1 heterozygous brain environment, Nf1 inactivation in o-GSCs led to the growth of OPGs by 3 months of age.71,89,90 These observations suggested that nonneoplastic Nf1+/− cells may have an essential impact on Nf1-null astrocyte growth, which provides the supportive environment that is needed for tumor growth. 
Microglia
Microglia have been commonly identified as major stromal constituents in gliomas, including PAs.9195 In a mouse model (Nf1flox/mut; GFAP-Cre), increased Nf1+/− microglia were identified as early as 3 weeks of age (WOA), before the formation of obvious tumors, which was consistent with a period of fast astrocyte proliferation in vivo.89 The critical role of microglia in glioma formation has been identified by several independent approaches in Nf1 GEMs. First, Nf1+/− microglia could promote the proliferation of Nf1−/− astrocytes in vitro, and treatment with minocycline (a microglia inhibitor) dramatically inhibited mouse OPGs growth in vivo.96 Second, neurofibromin mediated microglial properties by the c-Jun NH2-kinase (JNK)-relevant pathway involving mixed lineage kinase (MLK) and Ras-related C3 botulinum toxin substrate 1 (Rac1) manner, so that SP600125 (a JNK inhibitor) treatment could reduce optic glioma proliferation in vivo.97 Further observation used CD11b-TK mice98 to ablate resident brain microglia genetically by ganciclovir administration, which resulted in attenuated OPG development in vivo.99 
Further evidence revealed that o-GSCs expressed chemokine C-X3-C-motif ligand 1 (CX3CL1) to recruit microglia,100 and microglial chemokine C-X3-C-motif receptor 1 (CX3CR1) was a chemokine receptor that regulated microglia migration.101,102 Genetic CX3CR1 reduction can impair microglia dispersal, resulting in an accumulation of microglia in ONs99 and postponed OPG formation103 in vivo. In turn, to affect astrocyte proliferation and tumor progression, microglia incorporate additional molecules, including stromal cell-derived factor 1 (CXCL12), chemokine ligand 5 (Ccl5), meningioma-expressed antigen-5 (MGEA5), and interleukin-1β (IL-1β).80,96 
CXCL12 could boost the proliferation of Nf1−/− astrocytes and the apoptosis of wild-type astrocytes by sustaining the suppression of cAMP levels.80 Whereas the injection of lentiviral-encoded CXCL12 into the cerebral cortex of mice with Nf1 optic gliomas resulted in a decreased tumor growth rate, AMD3100 (a CXCR4 inhibitor) treatment did not inhibit OPG growth.104 This may be due to differences in the bioavailability of the CXCL12 used in that study and the counterregulatory mechanisms of CXCR4 between the optic pathway and the cortex. The existence of other cAMP-regulatory mechanisms might also neutralize the effects of CXCL12.104 Ccl5 was uniquely expressed by microglia in 3 to 6 WOA for OPG maintenance.105107 Culminated Ccl5 acted in glioma cells through the AKT/glycogen synthase kinase-3 beta (GSK3β)/cyclic-AMP response binding protein (CREB) pathway to restrain stem cell apoptosis and boost tumor proliferation.105,107,108 Administration of Ccl5 neutralizing antibodies prevented murine optic glioma proliferation and ameliorated retinal defects in vivo.105 Importantly, accumulated Ccl5 expression had a negative correlation with survival in patients with gliomas.108 In addition, the expression of MGEA5 by Nf1+/− microglia in vivo was also enhanced, but future studies are needed to clarify its specific impact on the body. 
In Nf1 GEMs, female subjects displayed greater RGC reduction, a thinner retinal nerve fiber layer (RNFL; composed of RGC axons) and more decreased visual acuity than male subjects, despite identical tumor proliferation in both sexes.30,109 It was demonstrated that estrogen induced microglia by estrogen receptor-β (ERβ), which was exclusively expressed in microglia. ERβ-activated microglia produce neurotoxins, such as IL-1β and other inflammatory cytokines, resulting in vision deterioration.84,109 Pharmacologic inhibition of microglial ERβ reversed visual defects in female Nf1 GEMs.109 There may be a potential model in which microglia play a positive role in glioma growth in both male and female Nf1 GEMs by producing nonsexually dimorphic paracrine factors, such as Ccl5,107 whereas female gonadal sex hormones activate microglia, creating a neurotoxic microenvironment resulting in increased axonal damage and retinal dysfunction.109 
T Cells
It was found in 59 NF1 glioma samples that, in comparison with high-grade gliomas, NF1-LGGs exhibited more strongly enriched immune signatures, such as cytolytic T lymphocyte infiltration and mutation-derived neoantigens with enhanced HLA binding.47 In addition, murine o-GSCs could not generate tumors in T-cell-deprived mice, indicating that the activation of T cells was necessary for Nf1 optic glioma proliferation in vivo.106,110 Herein, CD8+ lymphocytes constituted the majority of T cells, and deprivation of CD8+ T cells inhibited OPGs growth in vivo.108 
Previous investigations have found that o-GSCs secrete Ccl2 to activate T cells by Ras-dependent signaling. T cells infiltrate glioma from the meningeal space in an integrin-dependent mechanism.105,108 Then, activated T cells produce Ccl4 by the low-density lipoprotein receptor-related protein 1 (Lrp1)/calcineurin pathway,108 promoting microglia to excrete Ccl5 by nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), which provides supportive microenvironment for o-GSC integration.106,107 Notably, treatments with anti-Ccl2 antibody and anti-Ccl5 antibody abrogate OPG growth but make no difference in microglial contents and ON volumes.105 It is likely that the interruption of the stromal axis hinders Nf1 optic glioma development but does not eliminate the initiation of glioma. 
Furthermore, there is an inverse epidemiological risk relationship between asthma and OPGs, in which T-cell-mediated disorders reduce glioma morbidity.111 After asthma induction in Nf1 GEMs at 4 to 6 WOA, no tumor had grown at 12 to 24 WOA, and ON volumes decreased.112 The potential mechanism was elucidated: upregulated expression of decorin (a member of the small leucine-rich proteoglycans) in T cells attenuated Ccl5 expression in microglia by inhibiting the NFκB pathway.112 Hence, treatments with decorin or NFκB inhibitor in GEM at 4 to 6 WOA could hinder tumor formation at 12 WOA.112 
Neurons
In recent studies, it has been demonstrated that as constituents of the tumor microenvironment, hyperexcitable neurons contribute greatly to tumor growth by the paracrine pathway and electrochemical communication.113,114 There is evidence that neurofibromin modulates hyperpolarization-activated cyclic nucleotide-gated (HCN) channels,115 which can affect the activity of neurons.116,117 Further study has shown that tumor-related NF1-mutant RGCs exhibit increased excitability, controlled by HCN channel activity.118 Then, RGCs trigger upregulated expression of midkine to activate T-cell and microglial signaling cascades by the nuclear factor of activated T-cell-1 (NFAT-1) pathway in both human and murine OPGs.108 Therefore, targeting the HCN channel with the antiseizure medication lamotrigine prevented the progression of Nf1-OPG in vivo.118 In addition, optic gliomas grow around the axons of RGCs, that is, ONs, which are responsible for transmitting light-induced signals from the retina to the cerebral cortex. The upregulation of ON activity induced by visual experience (light) was shown to initiate and promote optic glioma progression during a developmental period, whereas light deprivation inhibited tumor formation and rescued RGCs from death via the neuronal activity-dependent neuroligin 3 (NLGN3) signaling axis.119,120 Similarly, the secretion of NLGN3 has also been shown to promote the proliferation of high-grade gliomas by active neurons.121,122 
Other Non-Neoplastic Cells
Additionally, other cells within the tumor microenvironment may also contribute to the development of OPGs. There were elevated numbers of endothelial cells and overexpression of vascular endothelial growth factor (VEGF) in both human and murine OPGs, which were negatively correlated with the progression-free survival rate.89,123125 In the inner retinal layer, there are retina-specific glia named Müller cells, which secrete various trophic factors and cytokines into the surrounding environment via glia-glia (e.g. microglia) and glia-neuron interactions in the absence of NF1.90 There is still much to discover about how the microenvironment and paracrine cells interact in tumor formation in future preclinical studies. 
Risk Factors for Glioma Formation and Progression
There has been an increasing focus on the early detection of NF1-OPGs by developing a scientific risk assessment algorithm to stratify patients based on their risk levels. Potential risk factors may include germline gene mutations, co-existing gene mutations, genomic alterations, and sexually dimorphic differences (Fig. 2). 
Figure 2.
 
The risk factors of NF1-OPGs. *A series of clinical and preclinical trials have found some potential risk factors of NF1-OPGs, including age, sex, tumor location, germline gene mutations, co-existent gene mutations, and genomic alterations. Further investigation is needed to develop a scientific risk assessment algorithm to stratify patients based on their risk levels. This figure was created using the BioRender software.
Figure 2.
 
The risk factors of NF1-OPGs. *A series of clinical and preclinical trials have found some potential risk factors of NF1-OPGs, including age, sex, tumor location, germline gene mutations, co-existent gene mutations, and genomic alterations. Further investigation is needed to develop a scientific risk assessment algorithm to stratify patients based on their risk levels. This figure was created using the BioRender software.
Germline Gene Mutations
It is generally known that different germline Nf1 gene mutations give rise to various expression levels of neurofibromin and distinct biological properties.126,127 There are GEMs with two typical NF1-patient-originating Nf1 gene mutations (Gly848Arg128 and Arg681X129). However, it was only the mice with the Arg681X mutation that generated optic gliomas with upregulated proliferative rates, glial fibrillary acidic protein (GFAP) immunoreactivity, and RGC loss.100,130 In addition, genetic mutations also affect the recruitment of non-neoplastic cells and the paracrine effects of the tumor microenvironment.100 There was more Ccl5 secretion and JNKThr183/Tyr185 activation in accumulated microglia infiltration, as well as increased Ccl5-regulated glial AKTThr308 in neoplastic astrocytes in Arg681XCKO mice.130 It appeared that germline mutations of the Nf1 gene had both effects intrinsic to the cell and effects from the surrounding tissue (stromal effects) on optic glioma phenotypes.130 
There were also potential genotype-phenotype relations in NF1-OPGs in clinical trials, which suggested that the germline mutations of NF1 may be useful as a biomarker for OPGs risk prediction. Studies have considered that patients with NF1-OPGs were more likely to have mutations in the 5′-tertile of the NF1 gene,131133 whereas other studies did not confirm this.134 Given the small samples in the above studies, a combined cohort study was implemented to investigate the predictive role of NF1 mutations in 215 patients with NF1 (100 of whom had OPGs),135 and the results showed that those who had mutations in the cysteine/serine-rich domain (CSRD; located in the fifth tertile) were more likely to develop OPGs, whereas those with mutations in the HEAT-like repeat region (HLR; amino acids 1825-2428) had a lower risk of forming OPGs. However, because there are many known variants of the large NF1 gene without definite genotype-phenotype correlations, genetic screening has proven to be challenging.136,137 
Co-Existent Gene Mutations
It is generally accepted that NF1-related gliomas have complete NF1 gene inactivation, but recent studies have revealed that NF1-OPGs may also contain additional heterozygous deletions of PTEN, duplications of KIAA1549: BRAF19,138,139 or FGFR1 alteration (needs further evidence).88 Based on an integrated analysis of 1000 pediatric LGGs (pLGGs), genetic rearrangements were associated with a younger age of occurrence and less aggressive clinical symptoms; therefore, they were classified as low risk.88 Single-nucleotide variant (SNV)-driven pLGGs, on the other hand, were more likely to be diagnosed later in life and necessitated multiple therapies and longer follow-ups.88 
GEM experiments indicated that OPGs expressing KIAA1549: BRAF exhibited no extra biological characteristics, whereas PTEN deletion and the loss of Nf1 may cooperate genetically to increase tumor aggressiveness.140 PTEN counteracts the action of PI3K/AKT and inhibits the development of OPGs.140 In Nf1flox/mut, Ptenflox/wt, and GFAP-Cre (FMPC) mice, there was a greater proliferative index and microglial infiltration in larger optic gliomas, as well as a greater thinning of the RNFL by AKT activation, independent of mTOR.140 According to whole-tumor RNA sequencing and deconvolution, FMPC mice displayed differentially expressed genes caused by downregulation of PTEN, which was normalized by PI3K inhibition (BKM120).74 PTEN-enriched tumors showed significantly reduced progression-free survival compared to those depleted in BKM120 treatment signatures in low- and high-grade gliomas.74 
Genomic Alterations
There is a hypothesis that the progression of pLGGs is also affected by epigenetic alteration and specific miRNA expression differences, which might enhance stroma-induced MEK activation.141,142 In this regard, epigenetic characteristics contribute greatly to the risk identification of glioma subgroups.143 For example, a subset of PAs with H3K27 alteration are more likely to have worse clinical progression. In a sex-specific manner, a modification gene (Arlm1) on mouse chromosome 12 conferred astrocytoma resistance.144 Although the modifier genes are not totally clear, potential candidates include those that regulate neurofibromin progression, o-GSC ability, and chemokine or microglial activation.145 In addition, genetic backgrounds also had dramatic influences on gliomagenesis in Nf1 GEMs. For example, heterozygous inactivation of Nf1 and Trp53 in mice resulted in astrocytoma progression at a greater incidence on the B6/C57BL/6J (B6) genetic background than on the 129S4/SvJae (129S4) background.146,147 To determine how epigenetic events play a crucial role in NF1-OPG pathogenesis, future research is needed. 
Sexually Dimorphic Differences
Currently, a sexually dimorphic interpretation of NF1 clinical abnormalities is intriguing. According to clinical investigation, girls with NF1-OPGs were 5 to 10 times more likely to experience visual dysfunction and require therapies than boys.30,148 Female GEMs, as mentioned earlier, also displayed decreased visual capacity due to neurotoxins produced by ERβ-activated microglia.109 In addition, two clinical studies demonstrated that boys were more likely to suffer from specific learning problems,149,150 and only male Nf1 GEMs exhibited spatial learning/memory difficulties related to higher Ras activity and lower dopamine levels in the hippocampus.30 The above findings suggested that sex, as a genomic determinant, was a key prognostic factor responsible for neuronal malfunction in NF1 through epigenetic mechanisms and that future preclinical or clinical studies should take this into account. Currently, a prospective observational multicenter study is underway to determine prognostic factors for visual outcomes. 
Targeted Therapies and Vision Recovery Strategies
Considering that tumor growth is slow in young patients with still developing brains, it is imperative that treatments can halt further tumor progression and neurologic degeneration without causing long-term neurocognitive consequences. Currently, promising targeted therapies for neoplastic cells or stromal cells have attracted much attention, and vision conservation treatments have flourished as well. Aside from functional outcomes, it is equally important to understand the best initial timing and length of therapy, the duration of efficacy, and the late outcomes.151,152 
Targeting Neoplastic Cells
With chemotherapies having both short- and long-term complications,153,154 therapeutic strategies targeting Nf1-null neoplastic cells by Ras pathways have emerged. As an inhibitor of B-RAF, sorafenib was used in a phase II clinical trial to treat children with low-grade astrocytomas.155 The trial was halted due to an unprecedented promotion of tumor development by paradoxical ERK activation in patients.156,157 To bypass this unexpected effect, selumetinib, an MEK inhibitor, is currently being tested in children with NF1-related LGGs.158,159 These results indicated that selumetinib had an encouraging antitumor impact on patients with recurrent or refractory LGGs and could become an alternative for chemotherapy. At the time of this writing, there are several ongoing clinical studies to assess the treatment outcome or maximum tolerated dose of selumetinib (NCT03871257, NCT03326388, and NCT01089101), the effectiveness and correct dose of an orally selective MEK1/2 inhibitor called binimetinib (NCT02285439) and the efficacy of an oral pan-RAF inhibitor tovorafenib (NCT05566795; see the Table). In addition, treatment with the mTOR inhibitor everolimus also demonstrated considerable disease stability/shrinkage with stabilized VA in individuals with recurrent/progressive NF1-related LGG.160,161 Rapamycin was combined with erlotinib, an EGFR inhibitor, to treat children with recurrent LGGs, which revealed prolonged disease stabilization in some patients.162 
Table.
 
The Ongoing Clinical Trials for LGG or OPG
Table.
 
The Ongoing Clinical Trials for LGG or OPG
However, partial NF1-OPGs continued to grow after common therapies and needed alternative second-line treatments on account of the resistance mechanisms. For example, rapamycin treatment did not have a durable curative effect in OPGs because of its decreased sensitivity observed in o-GSCs through high-level expression of ATP binding cassette subfamily G member 1 (ABCG1).110 Given that mTOR activity in o-GSCs could be increased by p90RSK hyperactivation, combined p90RSK and mTOR inhibitors are needed to ameliorate rapamycin resistance and improve clinical outcomes.110 
Targeting Non-Neoplastic Cells
There has been a growing recognition of the importance of the tumor microenvironment in disease pathogenesis, leading to new therapies targeting non-neoplastic cells and related signals from the tumor stroma.163 Preclinical studies have demonstrated that inhibiting microglial function could mitigate tumor growth, but clinical trials have not been conducted.96,97,99 In contrast, bevacizumab, an anti-VEGF receptor agent, combined with irinotecan or traditional chemotherapy regimens has been successfully used in recurrent LGGs.164167 Despite the small sample sizes, in clinical trials, treatment with bevacizumab resulted in disease control and even improved vision function with no significant toxicity.168170 Recently, in a large-scale clinical trial in the United Kingdom, 88 children with LGG received bevacizumab-based treatment.171 In addition to inducing a better overall visual response, bevacizumab treatment significantly controlled LGGs in the short term and prevented further deterioration. An ongoing trial (NCT02840409) is assessing the efficacy of bevacizumab and vinblastine combination in patients with NF1-LGG. In addition, a phase I trial of the anti-angiogenic agent lenalidomide has exhibited good tolerability and promising treatment results in pediatric LGGs.172 In addition, using IL-12, interferon beta/gamma gene delivery and immunomodulatory gene therapies against high-grade tumors enhanced the T-cell-mediated immune response,173 providing new directions and reference significance for the treatment of OPGs. There have also been some vaccine- and interferon-based immunotherapeutic clinical trials conducted on patients with NF1-LGG (NCT02358187 and NCT02343224). 
Therapeutic Strategies for Visual Recovery
Previous studies considered tumor volume and vision dysfunction to be the main outcome measures of optic glioma evolution in children with NF1. However, it should be noted that the size of NF1-OPGs may not be related to visual function.27 Reductions in visual function are often misinterpreted as an indication of tumor progression, which can lead to unnecessary interventions. There are some OPGs that can develop dramatically but cause no discernible deterioration of vision acuity or function.14 Meanwhile, smaller gliomas with irregular growth may result in severe visual problems. Additionally, spontaneous regression or shrinkage of gliomas may accompany vision loss partially owing to pressure gradients and kinking of axons.174 A conference report also outlined that visual acuity should be considered as a primary outcome measure in future clinical studies.152 Given that current successful chemotherapies and molecularly targeted treatments of NF1-OPGs could lead to decreased tumor volume without satisfactory improvement in visual function,175 it is necessary to cautiously assess the effectiveness of future therapies in clinical trials for NF1-OPG and develop feasible strategies to enhance vision function. 
There was a vulnerable window between the onset of RGC death and the loss of more than 30% of RGCs (predicted by RNFL thickness of 7.5 microns), according to promising preclinical studies.84 During this window of susceptibility, administration of rolipram (increasing cAMP levels),82 lovastatin (inhibiting Ras activation),81,84 or PHTPP (antagonizing ERβ)109 can reduce RGC loss and reverse vision impairment. Future clinical trials should focus on these therapeutic directions and identify optimal dosing intervals for the greatest benefits in visual outcomes. 
Furthermore, neuroprotective strategies may be promising treatments for visual impairment. In a randomized phase II clinical trial investigating treatment with nerve growth factor (NGF) eye drops, 13 patients with NF1-OPGs exhibited improved RGC function and enlarged visual fields with few systemic side effects.176 Currently, a randomized trial is being conducted to determine the safety and effectiveness of painless NGF CHF6467 eye drops on patients with OPGs (NCT05733572). There are various approaches to regenerate RGCs and improve retinal function in mice with other RGC-degenerated or -injured diseases, such as altering RGC gene expression,177,178 reprogramming Müller cells into RGCs,179,180 and transplanting induced pluripotent stem cell (iPSC)-derived RGCs.181 Treatments for NF1-OPGs involving vision restoration can benefit from the above approaches in the future.12 
Conclusions and Perspectives
These observations illustrate that the gliomagenesis of OPGs demands complex interplay between neoplastic and non-neoplastic cells (Fig. 3).145 Susceptible o-GSCs differentiate and grow into astrocytes following tumor-initiating genetic mutations during a specific developmental window. Then, o-GSCs and astrocytes both secrete molecules to recruit nonneoplastic cells, which form a permissive tumor microenvironment. In turn, the non-neoplastic cells in the tumor stroma support neoplastic cell proliferation and tumor progression by a paracrine mechanism. 
Figure 3.
 
The complex communication between neoplastic and non-neoplastic cells. *O-GSCs expressed CX3CL1 to recruit microglia which elaborated molecules to support neoplastic cells proliferation, including Ccl5, CXCL12, and MGEA5, and, in turn, neoplastic cells also secreted molecules (“stromagens”) to recruit microglia. Activated by Ccl2 secreted from o-GSCs, T cells then produced Ccl4 to promotes microglia to excrete Ccl5. As immune system regulators, RGCs held upregulated expression of midkine to activate T cells and initiated optic gliomas progression by NLGN3 signaling axis. By contrast, in female GEMs, estrogen induced microglia to produce neurotoxins, like IL-1β and other inflammatory cytokines, resulting in vision deterioration. Besides, other cells within the tumor microenvironment, like endothelial cells and Müller cells, might also contribute to the development of OPGs. This figure was created using the BioRender software.
Figure 3.
 
The complex communication between neoplastic and non-neoplastic cells. *O-GSCs expressed CX3CL1 to recruit microglia which elaborated molecules to support neoplastic cells proliferation, including Ccl5, CXCL12, and MGEA5, and, in turn, neoplastic cells also secreted molecules (“stromagens”) to recruit microglia. Activated by Ccl2 secreted from o-GSCs, T cells then produced Ccl4 to promotes microglia to excrete Ccl5. As immune system regulators, RGCs held upregulated expression of midkine to activate T cells and initiated optic gliomas progression by NLGN3 signaling axis. By contrast, in female GEMs, estrogen induced microglia to produce neurotoxins, like IL-1β and other inflammatory cytokines, resulting in vision deterioration. Besides, other cells within the tumor microenvironment, like endothelial cells and Müller cells, might also contribute to the development of OPGs. This figure was created using the BioRender software.
In addition, there is temporal and spatial heterogeneity among NF1-OPGs. Studies on o-GSCs have shown that the original cells determine the timing of murine OPG formation and somatic Nf1 loss given the temporal window.61,62,72 The tumor microenvironment also contributes to this effect, such as a greater increase in Nf1+/− microglia than WT microglia during 5 to 6 WOA99 and a decrease in the expression of CXCL12 with increasing age.80 Astrocytes from various regions of the CNS have unique gene expression patterns and biological characteristics. Astrocytes from the ON and brainstem expressed higher levels of the Nf1 gene and had more increased numbers following Nf1 gene inactivation than neocortical astrocytes, in agreement with the rarity of astrocytomas in the cortex.182 Cues from the surrounding stroma have also suggested that low cAMP levels accompanied by high CXCL12 expression along the optic pathway could account for the preferential localization of NF1-related gliomas.59,80 
Although our understanding of NF1-OPGs is constantly evolving from a molecular and immunological perspective, there are many unsolved mysteries hidden in the tumor microenvironment. First, further research is needed to determine how stromal cell paracrine factors influence the activated Ras signaling axis in neoplastic cells. In addition, scientists observed that T-cell-mediated microglial Ccl5 expression could be hindered by decorin expression from T cells in asthma.112 Understanding the molecular mechanisms that underlie OPGs with T-cell-mediated systemic diseases, such as eczema, rheumatoid arthritis, and diabetes, may provide insights into potential immunomodulatory therapies for neurological conditions. Immunological therapies, such as checkpoint inhibitors, may be uniquely beneficial to patients with NF1-LGG in a high-immune subgroup. In future studies, it will be necessary to establish immunocompetent mouse models and investigate the molecular and clinical implications of the immune microenvironment of patients at the signal cell level.152 Furthermore, previous studies have confirmed that in sporadic PAs, BRAF activation in astrocytes, or neural stem cells results in oncogene-induced senescence (OIS) for growth arrest caused by MAPK/ERK hyperactivation.183,184 It was demonstrated that OIS expressed a senescence-associated secretory phenotype (SASP), which constituted a complex inflammatory network to further promote OIS.185 It may be connected to the negative function of the tumor microenvironment in NF1-OPGs if this OIS/SASP circuit is present. The mechanism might explain the intriguing phenomenon that some children with NF1-OPGs experienced spontaneous regression or even complete disappearance documented by MRI.186 As a result, therapies that mediate senescence and promote OIS may also prove promising in gliomas in the future.187 
Because human specimens are rarely available and the difficulty of maintaining in vitro or developing as xenografts in vivo, mouse optic glioma models have been widely recognized as classic research tools to investigate the molecular and cellular mechanisms of human NF1-OPGs. Murine models have many advantages,71,89,124,188 but they still have a large gap in comparison to human NF1-OPGs. First, GEMs have lower microglia levels and lack several classical features, such as eosinophilic granular bodies and Rosenthal fibers.189,190 Second, gliomas appear later than normally seen in patients with NF1-OPGs (according to equivalent species ages) and do not affect the optic tracts or radiations.189 Furthermore, mice depend more on olfaction for survival than vision and have a smaller vision system with fewer RGC subtypes.12,189 Hence, due to their tractability and accessibility, minipigs have been successfully used as complementary NF1-OPG models.191 There are striking similarities between swine and humans in terms of visual anatomy and physiological functions (such as drug metabolism), which can provide a unique opportunity to research the natural history and drug efficacy of tumors.192,193 In addition, considering species differences, NF1 individual-derived iPSCs also have the potential to serve as models.194 Currently, NF1 iPSCs can be reprogrammed to generate human microglia-like cells195 or engineered with 7 different patient-derived NF1 mutations to identify mutational equivalency in tumors.126 
Acknowledgments
Supported by Innovative Research Team of High-level Local Universities in Shanghai (SHSMU-ZDCX20210900 and SHSMU-ZDCX20210902). These figures were created using the BioRender software. 
Disclosure: Y. Chen, None; J. Yu, None; S. Ge, None; R. Jia, None; X. Song, None; Y. Wang, None; X. Fan, None 
References
Dutton J . Gliomas of the anterior visual pathway. Surv Ophthalmol. 1994; 38(5): 427–452. [CrossRef] [PubMed]
Louis DN, Perry A, Reifenberger G, et al. The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol (Berl). 2016; 131(6): 803–820. [CrossRef]
Nicolin G, Parkin P, Mabbott D, et al. Natural history and outcome of optic pathway gliomas in children. Pediatr Blood Cancer. 2009; 53(7): 1231–1237. [CrossRef] [PubMed]
Listernick R, Ferner RE, Liu GT, Gutmann DH. Optic pathway gliomas in neurofibromatosis-1: controversies and recommendations. Ann Neurol. 2007; 61(3): 189–198. [CrossRef] [PubMed]
Listernick R, Louis D, Packer R, Gutmann D. Optic pathway gliomas in children with neurofibromatosis. 1. Consensus statement from the NF1 optic pathway glioma task force. Ann Neurol. 1997; 41(2): 143–149. [CrossRef] [PubMed]
Listernick R, Charrow J, Greenwald M, Mets M. Natural history of optic pathway tumors in children with neurofibromatosis type I: a longitudinal study. J Pediatr. 1994; 125(1): 63–66. [CrossRef] [PubMed]
Listernick R, Charrow J, Greenwald MJ, Esterly NB. Optic gliomas in children with neurofibromatosis type 1. J Pediatr. 1989; 114(5): 788–792. [CrossRef] [PubMed]
Guillamo JS, Créange A, Kalifa C, et al. Prognostic factors of CNS tumours in neurofibromatosis 1 (NF1): a retrospective study of 104 patients. Brain J Neurol. 2003; 126(Pt 1): 152–160.
Liu G, Brodsky M, Phillips P, et al. Optic radiation involvement in optic pathway gliomas in neurofibromatosis. Am J Ophthalmol. 2004; 137(3): 407–414. [CrossRef] [PubMed]
Prada CE, Hufnagel RB, Hummel TR, et al. The use of magnetic resonance imaging screening for optic pathway gliomas in children with neurofibromatosis type 1. J Pediatr. 2015; 167(4): 851–856. [CrossRef] [PubMed]
Sellmer L, Farschtschi S, Marangoni M, et al. Serial MRIs provide novel insight into natural history of optic pathway gliomas in patients with neurofibromatosis 1. Orphanet J Rare Dis. 2018; 13:62.
Freret ME, Gutmann DH. Insights into optic pathway glioma vision loss from mouse models of neurofibromatosis type 1. J Neurosci Res. 2019; 97(1): 45–56. [CrossRef] [PubMed]
Hamideh D, Hoehn ME, Harreld JH, Klimo PD, Gajjar A, Qaddoumi I. Isolated optic nerve glioma in children with and without neurofibromatosis: retrospective characterization and analysis of outcomes. J Child Neurol. 2018; 33(6): 375–382. [CrossRef] [PubMed]
Czyzyk E, Jóźwiak S, Roszkowski M, Schwartz RA. Optic pathway gliomas in children with and without neurofibromatosis 1. J Child Neurol. 2003; 18(7): 471–478. [CrossRef] [PubMed]
Listernick R, Darling C, Greenwald M, Strauss L, Charrow J. Optic pathway tumors in children: the effect of neurofibromatosis type 1 on clinical manifestations and natural history. J Pediatr. 1995; 127(5): 718–722. [CrossRef] [PubMed]
Gil Margolis M, Yackobovitz-Gavan M, Toledano H, et al. Optic pathway glioma and endocrine disorders in patients with and without NF1. Pediatr Res. 2023; 93(1): 233–241. [CrossRef] [PubMed]
Shlobin NA, Montgomery EY, Mohammad LM, et al. Visual outcomes after treatment for sporadic optic pathway gliomas in pediatric patients: a systematic review. World Neurosurg. 2022; 164: 436–449.e2. [CrossRef] [PubMed]
Helfferich J, Nijmeijer R, Brouwer OF, et al. Neurofibromatosis type 1 associated low grade gliomas: a comparison with sporadic low grade gliomas. Crit Rev Oncol Hematol. 2016; 104: 30–41. [CrossRef] [PubMed]
Jones DTW, Kocialkowski S, Liu L, et al. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008; 68(21): 8673–8677. [CrossRef] [PubMed]
Jones DTW, Kocialkowski S, Liu L, Pearson DM, Ichimura K, Collins VP. Oncogenic RAF1 rearrangement and a novel BRAF mutation as alternatives to KIAA1549:BRAF fusion in activating the MAPK pathway in pilocytic astrocytoma. Oncogene. 2009; 28(20): 2119–2123. [CrossRef] [PubMed]
Kaul A, Chen YH, Emnett RJ, Dahiya S, Gutmann DH. Pediatric glioma-associated KIAA1549:BRAF expression regulates neuroglial cell growth in a cell type-specific and mTOR-dependent manner. Genes Dev. 2012; 26(23): 2561–2566. [CrossRef] [PubMed]
Balcer L, Liu G, Heller G, et al. Visual loss in children with neurofibromatosis type 1 and optic pathway gliomas: relation to tumor location by magnetic resonance imaging. Am J Ophthalmol. 2001; 131(4): 442–445. [CrossRef] [PubMed]
Kinori M, Armarnik S, Listernick R, Charrow J, Zeid JL. Neurofibromatosis type 1-associated optic pathway glioma in children: a follow-up of 10 years or more. Am J Ophthalmol. 2021; 221: 91–96. [CrossRef] [PubMed]
Habiby R, Silverman B, Listernick R, Charrow J. Precocious puberty in children with neurofibromatosis type 1. J Pediatr. 1995; 126(3): 364–367. [CrossRef] [PubMed]
Picariello S, Cerbone M, D'Arco F, et al. A 40-year cohort study of evolving hypothalamic dysfunction in infants and young children (<3 years) with optic pathway gliomas. Cancers. 2022; 14(3): 747. [CrossRef] [PubMed]
Gutmann DH, Ferner RE, Listernick RH, Korf BR, Wolters PL, Johnson KJ. Neurofibromatosis type 1. Nat Rev Dis Primer. 2017; 3(1): 1–7.
Fisher MJ, Loguidice M, Gutmann DH, et al. Visual outcomes in children with neurofibromatosis type 1-associated optic pathway glioma following chemotherapy: a multicenter retrospective analysis. Neuro-Oncol. 2012; 14(6): 790–797. [CrossRef] [PubMed]
Opocher E, Kremer LCM, Da Dalt L, et al. Prognostic factors for progression of childhood optic pathway glioma: a systematic review. Eur J Cancer Oxf Engl 1990. 2006; 42(12): 1807–1816.
Kotch C, Avery R, Getz KD, et al. Risk factors for treatment-refractory and relapsed optic pathway glioma in children with neurofibromatosis type 1. Neuro-Oncol. 2022; 24(8): 1377–1386. [CrossRef] [PubMed]
Diggs-Andrews KA, Brown JA, Gianino SM, Rubin JB, Wozniak DF, Gutmann DH. Sex is a major determinant of neuronal dysfunction in neurofibromatosis type 1. Ann Neurol. 2014; 75(2): 309–316. [CrossRef] [PubMed]
de Blank PMK, Fisher MJ, Liu GT, et al. Optic pathway gliomas in neurofibromatosis type 1: an update: surveillance, treatment indications, and biomarkers of vision. J Neuroophthalmol. 2017; 37: S23–S32. [CrossRef] [PubMed]
Avery RA, Ferner RE, Listernick R, Fisher MJ, Gutmann DH, Liu GT. Visual acuity in children with low grade gliomas of the visual pathway: implications for patient care and clinical research. J Neurooncol. 2012; 110(1): 1–7. [CrossRef] [PubMed]
Sharif S, Ferner R, Birch JM, et al. Second primary tumors in neurofibromatosis I patients treated for optic glioma: substantial risks after radiotherapy. J Clin Oncol. 2006; 24(16): 2570–2575. [CrossRef] [PubMed]
Grill J, Couanet D, Cappelli C, et al. Radiation-induced cerebral vasculopathy in children with neurofibromatosis and optic pathway glioma. Ann Neurol. 1999; 45(3): 393–396. [CrossRef] [PubMed]
Demaerel P, de Ruyter N, Casteels I, Renard M, Uyttebroeck A, van Gool S. Visual pathway glioma in children treated with chemotherapy. Eur J Paediatr Neurol EJPN Off J Eur Paediatr Neurol Soc. 2002; 6(4): 207–212. [CrossRef]
Kato T, Sawamura Y, Tada M, Ikeda J, Ishii N, Abe H. Cisplatin/vincristine chemotherapy for hypothalamic/visual pathway astrocytomas in young children. J Neurooncol. 1998; 37(3): 263–270. [CrossRef] [PubMed]
Mahoney D, Cohen M, Friedman H, et al. Carboplatin is effective therapy for young children with progressive optic pathway tumors: a Pediatric Oncology Group phase II study. Neuro-Oncol. 2000; 2(4): 213–220. [CrossRef] [PubMed]
José P, Couceiro R, Passos J, Jorge Teixeira F. Visual outcomes of optic pathway glioma treated with chemotherapy in neurofibromatosis type 1. J Pediatr Ophthalmol Strabismus. 2022; 59(2): 128–135. [CrossRef] [PubMed]
Anastasaki C, Le LQ, Kesterson RA, Gutmann DH. Updated nomenclature for human and mouse neurofibromatosis type 1 genes. Neurol Genet. 2017; 3(4): e169. [CrossRef] [PubMed]
Viskochil D, Buchberg AM, Xu G, et al. Deletions and a translocation interrupt a cloned gene at the neurofibromatosis type 1 locus. Cell. 1990; 62(1): 187–192. [CrossRef] [PubMed]
Wallace MR, Marchuk DA, Andersen LB, et al. Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science. 1990; 249(4965): 181–186. [CrossRef] [PubMed]
Xu GF, Lin B, Tanaka K, et al. The catalytic domain of the neurofibromatosis type 1 gene product stimulates Ras GTPase and complements IRA mutants of S. cerevisiae. Cell. 1990; 63(4): 835–841. [CrossRef] [PubMed]
Ballester R, Marchuk D, Boguski M, et al. The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell. 1990; 63(4): 851–859. [CrossRef] [PubMed]
Martin GA, Viskochil D, Bollag G, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with Ras p21. Cell. 1990; 63(4): 843–849. [CrossRef] [PubMed]
Nix JS, Blakeley J, Rodriguez FJ. An update on the central nervous system manifestations of neurofibromatosis type 1. Acta Neuropathol (Berl). 2020; 139(4): 625–641. [CrossRef]
Ag K . Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA. 1971; 68(4): 820–823. [PubMed]
D'Angelo F, Ceccarelli M, Tala null, et al. The molecular landscape of glioma in patients with neurofibromatosis 1. Nat Med. 2019; 25(1): 176–187. [CrossRef] [PubMed]
Ryall S, Zapotocky M, Fukuoka K, et al. Integrated molecular and clinical analysis of 1,000 pediatric low-grade gliomas. Cancer Cell. 2020; 37(4): 569–583.e5. [CrossRef] [PubMed]
Gutmann DH, McLellan MD, Hussain I, et al. Somatic neurofibromatosis type 1 (NF1) inactivation characterizes NF1-associated pilocytic astrocytoma. Genome Res. 2013; 23(3): 431–439. [CrossRef] [PubMed]
Dasgupta B, Li W, Perry A, Gutmann DH. Glioma formation in neurofibromatosis 1 reflects preferential activation of K-Ras in astrocytes. Cancer Res. 2005; 65(1): 236–245. [CrossRef] [PubMed]
Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res. 2005; 65(7): 2755–2760. [CrossRef] [PubMed]
Lau N, Feldkamp MM, Roncari L, et al. Loss of neurofibromin is associated with activation of Ras/MAPK and PI3-K/AKT signaling in a neurofibromatosis 1 astrocytoma. J Neuropathol Exp Neurol. 2000; 59(9): 759–767. [CrossRef] [PubMed]
Fernández-Medarde A, Santos E. Ras in cancer and developmental diseases. Genes Cancer. 2011; 2(3): 344–358. [CrossRef] [PubMed]
Simanshu DK, Nissley DV, McCormick F. Ras proteins and their regulators in human disease. Cell. 2017; 170(1): 17–33. [CrossRef] [PubMed]
Kaul A, Toonen JA, Cimino PJ, Gianino SM, Gutmann DH. Akt- or MEK-mediated mTOR inhibition suppresses Nf1 optic glioma growth. Neuro-Oncol. 2015; 17(6): 843–853. [CrossRef] [PubMed]
Jessen WJ, Miller SJ, Jousma E, et al. MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors. J Clin Invest. 2013; 123(1): 340–347. [CrossRef] [PubMed]
Chang T, Krisman K, Theobald EH, et al. Sustained MEK inhibition abrogates myeloproliferative disease in Nf1 mutant mice. J Clin Invest. 2013; 123(1): 335–339. [CrossRef] [PubMed]
Hegedus B, Dasgupta B, Shin JE, et al. Neurofibromatosis-1 regulates neuronal and glial cell differentiation from neuroglial progenitors in vivo by both cAMP- and Ras-dependent mechanisms. Cell Stem Cell. 2007; 1(4): 443–457. [CrossRef] [PubMed]
Warrington NM, Gianino SM, Jackson E, et al. Cyclic AMP suppression is sufficient to induce gliomagenesis in a mouse model of neurofibromatosis-1. Cancer Res. 2010; 70(14): 5717–5727. [CrossRef] [PubMed]
Jessa S, Blanchet-Cohen A, Krug B, et al. Stalled developmental programs at the root of pediatric brain tumors. Nat Genet. 2019; 51(12): 1702–1713. [CrossRef] [PubMed]
Brossier NM, Thondapu S, Cobb OM, Dahiya S, Gutmann DH. Temporal, spatial, and genetic constraints contribute to the patterning and penetrance of murine neurofibromatosis-1 optic glioma. Neuro-Oncol. 2021; 23(4): 625–637. [CrossRef] [PubMed]
Jecrois ES, Zheng W, Bornhorst M, et al. Treatment during a developmental window prevents NF1-associated optic pathway gliomas by targeting Erk-dependent migrating glial progenitors. Dev Cell. 2021; 56(20): 2871–2885. [CrossRef] [PubMed]
Miller RH, Ffrench-Constant C, Raff MC. The macroglial cells of the rat optic nerve. Annu Rev Neurosci. 1989; 12: 517–534. [CrossRef] [PubMed]
Tao C, Zhang X. Development of astrocytes in the vertebrate eye. Dev Dyn Off Publ Am Assoc Anat. 2014; 243(12): 1501–1510.
Lee DY, Gianino SM, Gutmann DH. Innate neural stem cell heterogeneity determines the patterning of glioma formation in children. Cancer Cell. 2012; 22(1): 131–138. [CrossRef] [PubMed]
Marsters CM, Rosin JM, Thornton HF, et al. Oligodendrocyte development in the embryonic tuberal hypothalamus and the influence of Ascl1. Neural Develop. 2016; 11(1): 20. [CrossRef]
Ono K, Hirahara Y, Gotoh H, et al. Origin of oligodendrocytes in the vertebrate optic nerve: a review. Neurochem Res. 2018; 43(1): 3–11. [CrossRef] [PubMed]
Tchoghandjian A, Fernandez C, Colin C, et al. Pilocytic astrocytoma of the optic pathway: a tumour deriving from radial glia cells with a specific gene signature. Brain J Neurol. 2009; 132(Pt 6): 1523–1535.
Dahiya S, Lee DY, Gutmann DH. Comparative characterization of the human and mouse third ventricle germinal zones. J Neuropathol Exp Neurol. 2011; 70(7): 622–633. [CrossRef] [PubMed]
Chen YH, Gianino SM, Gutmann DH. Neurofibromatosis-1 regulation of neural stem cell proliferation and multilineage differentiation operates through distinct Ras effector pathways. Genes Dev. 2015; 29(16): 1677–1682. [CrossRef] [PubMed]
Bajenaru ML, Hernandez MR, Perry A, et al. Optic nerve glioma in mice requires astrocyte Nf1 gene inactivation and Nf1 brain heterozygosity. Cancer Res. 2003; 63(24): 8573–8577. [PubMed]
Solga AC, Toonen JA, Pan Y, et al. The cell of origin dictates the temporal course of neurofibromatosis-1 (Nf1) low-grade glioma formation. Oncotarget. 2017; 8(29): 47206–47215. [CrossRef] [PubMed]
Solga AC, Gianino SM, Gutmann DH. NG2-cells are not the cell of origin for murine neurofibromatosis-1 (Nf1) optic glioma. Oncogene. 2014; 33(3): 289–299. [CrossRef] [PubMed]
Pan Y, Bush EC, Toonen JA, et al. Whole tumor RNA-sequencing and deconvolution reveal a clinically-prognostic PTEN/PI3K-regulated glioma transcriptional signature. Oncotarget. 2017; 8(32): 52474–52487. [CrossRef] [PubMed]
Zhu L, Gibson P, Currle DS, et al. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature. 2009; 457(7229): 603–607. [CrossRef] [PubMed]
Banerjee S, Crouse NR, Emnett RJ, Gianino SM, Gutmann DH. Neurofibromatosis-1 regulates mTOR-mediated astrocyte growth and glioma formation in a TSC/Rheb-independent manner. Proc Natl Acad Sci USA. 2011; 108(38): 15996–16001. [CrossRef] [PubMed]
Hegedus B, Banerjee D, Yeh TH, et al. Preclinical cancer therapy in a mouse model of neurofibromatosis-1 optic glioma. Cancer Res. 2008; 68(5): 1520–1528. [CrossRef] [PubMed]
Tong J, Hannan F, Zhu Y, Bernards A, Zhong Y. Neurofibromin regulates G protein-stimulated adenylyl cyclase activity. Nat Neurosci. 2002; 5(2): 95–96. [CrossRef] [PubMed]
Dasgupta B, Dugan LL, Gutmann DH. The neurofibromatosis 1 gene product neurofibromin regulates pituitary adenylate cyclase-activating polypeptide-mediated signaling in astrocytes. J Neurosci Off J Soc Neurosci. 2003; 23(26): 8949–8954. [CrossRef]
Warrington NM, Woerner BM, Daginakatte GC, et al. Spatiotemporal differences in CXCL12 expression and cyclic AMP underlie the unique pattern of optic glioma growth in neurofibromatosis type 1. Cancer Res. 2007; 67(18): 8588–8595. [CrossRef] [PubMed]
Anastasaki C, Gutmann DH. Neuronal NF1/Ras regulation of cyclic AMP requires atypical PKC activation. Hum Mol Genet. 2014; 23(25): 6712–6721. [CrossRef] [PubMed]
Brown JA, Gianino SM, Gutmann DH. Defective cAMP generation underlies the sensitivity of CNS neurons to neurofibromatosis-1 heterozygosity. J Neurosci. 2010; 30(16): 5579–5589. [CrossRef] [PubMed]
Hegedus B, Hughes FW, Garbow JR, et al. Optic nerve dysfunction in a mouse model of neurofibromatosis-1 optic glioma. J Neuropathol Exp Neurol. 2009; 68(5): 542–551. [CrossRef] [PubMed]
Toonen JA, Ma Y, Gutmann DH. Defining the temporal course of murine neurofibromatosis-1 optic gliomagenesis reveals a therapeutic window to attenuate retinal dysfunction. Neuro-Oncol. 2017; 19(6): 808–819. [PubMed]
Polyak K, Haviv I, Campbell IG. Co-evolution of tumor cells and their microenvironment. Trends Genet TIG. 2009; 25(1): 30–38. [CrossRef] [PubMed]
Rubin J, Gutmann D. Neurofibromatosis type 1 - a model for nervous system tumour formation? Nat Rev Cancer. 2005; 5(7): 557–564. [CrossRef] [PubMed]
Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol. 2006; 1: 119–150. [CrossRef] [PubMed]
Fisher MJ, Jones DTW, Li Y, et al. Integrated molecular and clinical analysis of low-grade gliomas in children with neurofibromatosis type 1 (NF1). Acta Neuropathol (Berl). 2021; 141(4): 605–617. [CrossRef]
Bajenaru ML, Garbow JR, Perry A, Hernandez MR, Gutmann DH. Natural history of neurofibromatosis 1-associated optic nerve glioma in mice. Ann Neurol. 2005; 57(1): 119–127. [CrossRef] [PubMed]
Zhu Y, Harada T, Liu L, et al. Inactivation of NF1 in CNS causes increased glial progenitor proliferation and optic glioma formation. Development. 2005; 132(24): 5577–5588. [CrossRef] [PubMed]
Badie B, Schartner JM. Flow cytometric characterization of tumor-associated macrophages in experimental gliomas. Neurosurgery. 2000; 46(4): 957–961; discussion 961-962. [PubMed]
Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors. Glia. 2002; 40(2): 252–259. [CrossRef] [PubMed]
Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007; 10(11): 1387–1394. [CrossRef] [PubMed]
Roggendorf W, Strupp S, Paulus W. Distribution and characterization of microglia/macrophages in human brain tumors. Acta Neuropathol (Berl). 1996; 92(3): 288–293. [CrossRef]
Streit WJ, Conde JR, Fendrick SE, Flanary BE, Mariani CL. Role of microglia in the central nervous system's immune response. Neurol Res. 2005; 27(7): 685–691. [CrossRef] [PubMed]
Daginakatte GC, Gutmann DH. Neurofibromatosis-1 (Nf1) heterozygous brain microglia elaborate paracrine factors that promote Nf1-deficient astrocyte and glioma growth. Hum Mol Genet. 2007; 16(9): 1098–1112. [CrossRef] [PubMed]
Gc D, Sm G, Nw Z, As P, Dh G. Increased c-Jun-NH2-kinase signaling in neurofibromatosis-1 heterozygous microglia drives microglia activation and promotes optic glioma proliferation. Cancer Res. 2008; 68(24): 10358–10366. [PubMed]
Gowing G, Vallières L, Julien JP. Mouse model for ablation of proliferating microglia in acute CNS injuries. Glia. 2006; 53(3): 331–337. [CrossRef] [PubMed]
Simmons GW, Pong WW, Emnett RJ, et al. Neurofibromatosis-1 heterozygosity increases microglia in a spatially and temporally restricted pattern relevant to mouse optic glioma formation and growth. J Neuropathol Exp Neurol. 2011; 70(1): 51–62. [CrossRef] [PubMed]
Guo X, Pan Y, Gutmann DH. Genetic and genomic alterations differentially dictate low-grade glioma growth through cancer stem cell-specific chemokine recruitment of T cells and microglia. Neuro-Oncol. 2019; 21(10): 1250–1262. [CrossRef] [PubMed]
Liang KJ, Lee JE, Wang YD, et al. Regulation of dynamic behavior of retinal microglia by CX3CR1 signaling. Invest Ophthalmol Vis Sci. 2009; 50(9): 4444–4451. [CrossRef] [PubMed]
Zhu J, Zhou Z, Liu Y, Zheng J. Fractalkine and CX3CR1 are involved in the migration of intravenously grafted human bone marrow stromal cells toward ischemic brain lesion in rats. Brain Res. 2009; 1287: 173–183. [CrossRef] [PubMed]
Pong WW, Higer SB, Gianino SM, Emnett RJ, Gutmann DH. Reduced microglial CX3CR1 expression delays neurofibromatosis-1 glioma formation. Ann Neurol. 2013; 73(2): 303–308. [CrossRef] [PubMed]
Sun T, Gianino SM, Jackson E, Piwnica-Worms D, Gutmann DH, Rubin JB. CXCL12 alone is insufficient for gliomagenesis in Nf1 mutant mice. J Neuroimmunol. 2010; 224(1-2): 108–113. [CrossRef] [PubMed]
de Andrade Costa A, Chatterjee J, Cobb O, et al. Immune deconvolution and temporal mapping identifies stromal targets and developmental intervals for abrogating murine low-grade optic glioma formation. Neuro-Oncol Adv. 2022; 4(1): vdab194–vdab194. [CrossRef]
Pan Y, Xiong M, Chen R, et al. Athymic mice reveal a requirement for T-cell-microglia interactions in establishing a microenvironment supportive of Nf1 low-grade glioma growth. Genes Dev. 2018; 32(7-8): 491–496. [CrossRef] [PubMed]
Solga AC, Pong WW, Kim KY, et al. RNA sequencing of tumor-associated microglia reveals Ccl5 as a stromal chemokine critical for neurofibromatosis-1 glioma growth. Neoplasia. 2015; 17(10): 776–788. [CrossRef] [PubMed]
Guo X, Pan Y, Xiong M, et al. Midkine activation of CD8+ T cells establishes a neuron-immune-cancer axis responsible for low-grade glioma growth. Nat Commun. 2020; 11(1): 2177. [CrossRef] [PubMed]
Toonen JA, Solga AC, Ma Y, Gutmann DH. Estrogen activation of microglia underlies the sexually dimorphic differences in Nf1 optic glioma-induced retinal pathology. J Exp Med. 2017; 214(1): 17–25. [CrossRef] [PubMed]
Chen YH, McGowan LD, Cimino PJ, et al. Mouse low-grade gliomas contain cancer stem cells with unique molecular and functional properties. Cell Rep. 2015; 10(11): 1899–1912. [CrossRef] [PubMed]
Porcelli B, Zoellner NL, Abadin SS, Gutmann DH, Johnson KJ. Associations between allergic conditions and pediatric brain tumors in Neurofibromatosis type 1. Fam Cancer. 2016; 15(2): 301–308. [CrossRef] [PubMed]
Chatterjee J, Sanapala S, Cobb O, et al. Asthma reduces glioma formation by T cell decorin-mediated inhibition of microglia. Nat Commun. 2021; 12(1): 7122. [CrossRef] [PubMed]
Monje M, Borniger JC, D'Silva NJ, et al. Roadmap for the emerging field of cancer neuroscience. Cell. 2020; 181(2): 219–222. [CrossRef] [PubMed]
Venkatesh HS, Morishita W, Geraghty AC, et al. Electrical and synaptic integration of glioma into neural circuits. Nature. 2019; 573(7775): 539–545. [CrossRef] [PubMed]
Omrani A, van der Vaart T, Mientjes E, et al. HCN channels are a novel therapeutic target for cognitive dysfunction in neurofibromatosis type 1. Mol Psychiatry. 2015; 20(11): 1311–1321. [CrossRef] [PubMed]
Herrmann S, Schnorr S, Ludwig A. HCN channels–modulators of cardiac and neuronal excitability. Int J Mol Sci. 2015; 16(1): 1429–1447. [CrossRef] [PubMed]
Santoro B, Shah MM. Hyperpolarization-activated cyclic nucleotide-gated channels as drug targets for neurological disorders. Annu Rev Pharmacol Toxicol. 2020; 60: 109–131. [CrossRef] [PubMed]
Anastasaki C, Mo J, Chen JK, et al. Neuronal hyperexcitability drives central and peripheral nervous system tumor progression in models of neurofibromatosis-1. Nat Commun. 2022; 13(1): 2785. [CrossRef] [PubMed]
Pan Y, Hysinger JD, Barron T, et al. NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature. 2021; 594(7862): 277-282. [CrossRef] [PubMed]
Tian N, Copenhagen DR. Visual deprivation alters development of synaptic function in inner retina after eye opening. Neuron. 2001; 32(3): 439–449. [CrossRef] [PubMed]
Venkatesh HS, Johung TB, Caretti V, et al. Neuronal activity promotes glioma growth through neuroligin-3 secretion. Cell. 2015; 161(4): 803–816. [CrossRef] [PubMed]
Venkatesh HS, Tam LT, Woo PJ, et al. Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature. 2017; 549(7673): 533–537. [CrossRef] [PubMed]
Bartels U, Hawkins C, Ma J, et al. Vascularity and angiogenesis as predictors of growth in optic pathway/hypothalamic gliomas. J Neurosurg. 2006; 104(5): 314–320. [PubMed]
Kim KY, Ju WK, Hegedus B, Gutmann DH, Ellisman MH. Ultrastructural characterization of the optic pathway in a mouse model of neurofibromatosis-1 optic glioma. Neuroscience. 2010; 170(1): 178–188. [CrossRef] [PubMed]
Sikkema AH, de Bont ESJM, Molema G, et al. Vascular endothelial growth factor receptor 2 (VEGFR-2) signalling activity in paediatric pilocytic astrocytoma is restricted to tumour endothelial cells. Neuropathol Appl Neurobiol. 2011; 37(5): 538–548. [CrossRef] [PubMed]
Anastasaki C, Wegscheid ML, Hartigan K, et al. Human iPSC-derived neurons and cerebral organoids establish differential effects of germline NF1 gene mutations. Stem Cell Rep. 2020; 14(4): 541–550. [CrossRef]
Anastasaki C, Woo AS, Messiaen LM, Gutmann DH. Elucidating the impact of neurofibromatosis-1 germline mutations on neurofibromin function and dopamine-based learning. Hum Mol Genet. 2015; 24(12): 3518–3528. [CrossRef] [PubMed]
Pascual-Castroviejo I, Pascual-Pascual SI, Velazquez-Fragua R, Botella P, Viaño J. Familial spinal neurofibromatosis. Neuropediatrics. 2007; 38(2): 105–108. [PubMed]
Kocova M, Kochova E, Sukarova-Angelovska E. Optic glioma and precocious puberty in a girl with neurofibromatosis type 1 carrying an R681X mutation of NF1: case report and review of the literature. BMC Endocr Disord. 2015; 15: 82. [CrossRef] [PubMed]
Toonen JA, Anastasaki C, Smithson LJ, et al. NF1 germline mutation differentially dictates optic glioma formation and growth in neurofibromatosis-1. Hum Mol Genet. 2016; 25(9): 1703–1713. [CrossRef] [PubMed]
Anastasaki C, Morris SM, Gao F, Gutmann DH. Children with 5′-end NF1 gene mutations are more likely to have glioma. Neurol Genet. 2017; 3(5): e192. [CrossRef] [PubMed]
Bolcekova A, Nemethova M, Zatkova A, et al. Clustering of mutations in the 5′ tertile of the NF1 gene in Slovakia patients with optic pathway glioma. Neoplasma. 2013; 60(6): 655–665. [CrossRef] [PubMed]
Sharif S, Upadhyaya M, Ferner R, et al. A molecular analysis of individuals with neurofibromatosis type 1 (NF1) and optic pathway gliomas (OPGs), and an assessment of genotype-phenotype correlations. J Med Genet. 2011; 48(4): 256–260. [CrossRef] [PubMed]
Hutter S, Piro RM, Waszak SM, et al. No correlation between NF1 mutation position and risk of optic pathway glioma in 77 unrelated NF1 patients. Hum Genet. 2016; 135(5): 469–475. [CrossRef] [PubMed]
Xu M, Xiong H, Han Y, et al. Identification of mutation regions on NF1 responsible for high- and low-risk development of optic pathway glioma in neurofibromatosis type I. Front Genet. 2018; 9: 270. [CrossRef] [PubMed]
Tsipi M, Poulou M, Fylaktou I, et al. Phenotypic expression of a spectrum of neurofibromatosis type 1 (NF1) mutations identified through NGS and MLPA. J Neurol Sci. 2018; 65(2): 95–105.
Sabbagh A, Pasmant E, Imbard A, et al. NF1 molecular characterization and neurofibromatosis type I genotype-phenotype correlation: the French Experience. Hum Mutat. 2013; 34(11): 1510–1518. [CrossRef] [PubMed]
Rodriguez EF, Scheithauer BW, Giannini C, et al. PI3K/AKT pathway alterations are associated with clinically aggressive and histologically anaplastic subsets of pilocytic astrocytoma. Acta Neuropathol (Berl). 2011; 121(3): 407–420. [CrossRef]
Rodriguez FJ, Ligon AH, Horkayne-Szakaly I, et al. BRAF duplications and MAPK pathway activation are frequent in gliomas of the optic nerve proper. J Neuropathol Exp Neurol. 2012; 71(9): 789–794. [CrossRef] [PubMed]
Kaul A, Toonen JA, Gianino SM, Gutmann DH. The impact of coexisting genetic mutations on murine optic glioma biology. Neuro-Oncol. 2015; 17(5): 670–677. [CrossRef] [PubMed]
Milde T, Rodriguez FJ, Barnholtz-Sloan JS, Patil N, Eberhart CG, Gutmann DH. Reimagining pilocytic astrocytomas in the context of pediatric low-grade gliomas. Neuro-Oncol. 2021; 23(10): 1634–1646. [CrossRef] [PubMed]
Yuan M, Da Silva ACAL, Arnold A, et al. MicroRNA (miR) 125b regulates cell growth and invasion in pediatric low grade glioma. Sci Rep. 2018; 8(1): 12506. [CrossRef] [PubMed]
Lambert SR, Witt H, Hovestadt V, et al. Differential expression and methylation of brain developmental genes define location-specific subsets of pilocytic astrocytoma. Acta Neuropathol (Berl). 2013; 126(2): 291–301. [CrossRef]
Amlin-Van Schaick JC, Kim S, DiFabio C, Lee MH, Broman KW, Reilly KM. Arlm1 is a male-specific modifier of astrocytoma resistance on mouse Chr 12. Neuro-Oncol. 2012; 14(2): 160–174. [CrossRef] [PubMed]
Thangarajh M, Gutmann DH. Low-grade gliomas as neurodevelopmental disorders: insights from mouse models of neurofibromatosis-1. Neuropathol Appl Neurobiol. 2012; 38(3): 241–253. [CrossRef] [PubMed]
Hawes JJ, Tuskan RG, Reilly KM. Nf1 expression is dependent on strain background: implications for tumor suppressor haploinsufficiency studies. Neurogenetics. 2007; 8(2): 121–130. [CrossRef] [PubMed]
Reilly KM, Tuskan RG, Christy E, et al. Susceptibility to astrocytoma in mice mutant for Nf1 and Trp53 is linked to chromosome 11 and subject to epigenetic effects. Proc Natl Acad Sci USA. 2004; 101(35): 13008–13013. [CrossRef] [PubMed]
Fisher MJ, Loguidice M, Gutmann DH, et al. Gender as a disease modifier in neurofibromatosis type 1 optic pathway glioma. Ann Neurol. 2014; 75(5): 799–800. [CrossRef] [PubMed]
Coudé FX, Mignot C, Lyonnet S, Munnich A. Academic impairment is the most frequent complication of neurofibromatosis type-1 (NF1) in children. Behav Genet. 2006; 36(5): 660–664. [CrossRef] [PubMed]
Hyman SL, Shores A, North KN. The nature and frequency of cognitive deficits in children with neurofibromatosis type 1. Neurology. 2005; 65(7): 1037–1044. [CrossRef] [PubMed]
Hill CS, Devesa SC, Ince W, Borg A, Aquilina K. A systematic review of ongoing clinical trials in optic pathway gliomas. Childs Nerv Syst. 2020; 36(9): 1869–1886. [CrossRef] [PubMed]
Packer RJ, Iavarone A, Jones DTW, et al. Implications of new understandings of gliomas in children and adults with NF1: report of a consensus conference. Neuro-Oncol. 2020; 22(6): 773–784. [CrossRef] [PubMed]
Packer RJ, Lange B, Ater J, et al. Carboplatin and vincristine for recurrent and newly diagnosed low-grade gliomas of childhood. J Clin Oncol Off J Am Soc Clin Oncol. 1993; 11(5): 850–856. [CrossRef]
Verstappen CCP, Heimans JJ, Hoekman K, Postma TJ. Neurotoxic complications of chemotherapy in patients with cancer: clinical signs and optimal management. Drugs. 2003; 63(15): 1549–1563. [CrossRef] [PubMed]
Karajannis MA, Legault G, Fisher MJ, et al. Phase II study of sorafenib in children with recurrent or progressive low-grade astrocytomas. Neuro-Oncol. 2014; 16(10): 1408–1416. [CrossRef] [PubMed]
Hatzivassiliou G, Song K, Yen I, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010; 464(7287): 431–435. [CrossRef] [PubMed]
Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010; 464(7287): 427–430. [CrossRef] [PubMed]
Fangusaro J, Onar-Thomas A, Young Poussaint T, et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol. 2019; 20(7): 1011–1022. [CrossRef] [PubMed]
Banerjee A, Jakacki RI, Onar-Thomas A, et al. A phase I trial of the MEK inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or refractory low-grade glioma: a Pediatric Brain Tumor Consortium (PBTC) study. Neuro-Oncol. 2017; 19(8): 1135–1144. [CrossRef] [PubMed]
Ullrich NJ, Prabhu SP, Packer RJ, et al. Visual outcomes following everolimus targeted therapy for neurofibromatosis type 1-associated optic pathway gliomas in children. Pediatr Blood Cancer. 2021; 68(4): e28833. [CrossRef] [PubMed]
Ullrich NJ, Prabhu SP, Reddy AT, et al. A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1-associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study. Neuro-Oncol. 2020; 22(10): 1527–1535. [CrossRef] [PubMed]
Yalon M, Rood B, MacDonald TJ, et al. A feasibility and efficacy study of rapamycin and erlotinib for recurrent pediatric low-grade glioma (LGG). Pediatr Blood Cancer. 2013; 60(1): 71–76. [CrossRef] [PubMed]
Pienta KJ, McGregor N, Axelrod R, Axelrod DE. Ecological therapy for cancer: defining tumors using an ecosystem paradigm suggests new opportunities for novel cancer treatments. Transl Oncol. 2008; 1(4): 158–164. [CrossRef] [PubMed]
Gururangan S, Fangusaro J, Poussaint TY, et al. Efficacy of bevacizumab plus irinotecan in children with recurrent low-grade gliomas–a Pediatric Brain Tumor Consortium study. Neuro-Oncol. 2014; 16(2): 310–317. [CrossRef] [PubMed]
Kalra M, Heath JA, Kellie SJ, et al. Confirmation of bevacizumab activity, and maintenance of efficacy in retreatment after subsequent relapse, in pediatric low-grade glioma. J Pediatr Hematol Oncol. 2015; 37(6): e341–346. [CrossRef] [PubMed]
Packer RJ, Jakacki R, Horn M, et al. Objective response of multiply recurrent low-grade gliomas to bevacizumab and irinotecan. Pediatr Blood Cancer. 2009; 52(7): 791–795. [CrossRef] [PubMed]
Zhukova N, Rajagopal R, Lam A, et al. Use of bevacizumab as a single agent or in adjunct with traditional chemotherapy regimens in children with unresectable or progressive low-grade glioma. Cancer Med. 2019; 8(1): 40–50. [CrossRef] [PubMed]
Avery RA, Hwang EI, Jakacki RI, Packer RJ. Marked recovery of vision in children with optic pathway gliomas treated with bevacizumab. JAMA Ophthalmol. 2014; 132(1): 111–114. [CrossRef] [PubMed]
Hwang EI, Jakacki RI, Fisher MJ, et al. Long-term efficacy and toxicity of bevacizumab-based therapy in children with recurrent low-grade gliomas. Pediatr Blood Cancer. 2013; 60(5): 776–782. [CrossRef] [PubMed]
Yamasaki F, Takano M, Yonezawa U, et al. Bevacizumab for optic pathway glioma with worsening visual field in absence of imaging progression: 2 case reports and literature review. Childs Nerv Syst ChNS Off J Int Soc Pediatr Neurosurg. 2020; 36(3): 635–639. [CrossRef]
Green K, Panagopoulou P, D'Arco F, et al. A nationwide evaluation of bevacizumab-based treatments in pediatric low-grade glioma in the UK: safety, efficacy, visual morbidity, and outcomes. Neuro-Oncol. 2023; 25(4): 774–785. [CrossRef] [PubMed]
Warren KE, Goldman S, Pollack IF, et al. Phase I trial of lenalidomide in pediatric patients with recurrent, refractory, or progressive primary CNS tumors: pediatric brain tumor consortium study PBTC-018. J Clin Oncol Off J Am Soc Clin Oncol. 2011; 29(3): 324–329. [CrossRef]
Giotta Lucifero A, Luzzi S. Against the resilience of high-grade gliomas: gene therapies (part II). Brain Sci. 2021; 11(8): 976. [CrossRef] [PubMed]
Parsa CF, Hoyt CS, Lesser RL, et al. Spontaneous regression of optic gliomas: thirteen cases documented by serial neuroimaging. Arch Ophthalmol Chic Ill 1960. 2001; 119(4): 516–529. [CrossRef]
Dalla Via P, Opocher E, Pinello ML, et al. Visual outcome of a cohort of children with neurofibromatosis type 1 and optic pathway glioma followed by a pediatric neuro-oncology program. Neuro-Oncol. 2007; 9(4): 430–437. [PubMed]
Falsini B, Chiaretti A, Rizzo D, et al. Nerve growth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. Brain J Neurol. 2016; 139(Pt 2): 404–414.
Ishikawa H, Takano M, Matsumoto N, et al. Effect of GDNF gene transfer into axotomized retinal ganglion cells using in vivo electroporation with a contact lens-type electrode. Gene Ther. 2005; 12(4): 289–298. [CrossRef] [PubMed]
Wilson AM, Di Polo A. Gene therapy for retinal ganglion cell neuroprotection in glaucoma. Gene Ther. 2012; 19(2): 127–136. [CrossRef] [PubMed]
Jorstad NL, Wilken MS, Grimes WN, et al. Stimulation of functional neuronal regeneration from Müller glia in adult mice. Nature. 2017; 548(7665): 103–107. [CrossRef] [PubMed]
Sanges D, Simonte G, Di Vicino U, et al. Reprogramming Müller glia via in vivo cell fusion regenerates murine photoreceptors. J Clin Invest. 2016; 126(8): 3104–3116. [CrossRef] [PubMed]
Venugopalan P, Wang Y, Nguyen T, Huang A, Muller KJ, Goldberg JL. Transplanted neurons integrate into adult retinas and respond to light. Nat Commun. 2016; 7: 10472. [CrossRef] [PubMed]
Yeh TH, Lee DY, Gianino SM, Gutmann DH. Microarray analyses reveal regional astrocyte heterogeneity with implications for neurofibromatosis type 1 (NF1)-regulated glial proliferation. Glia. 2009; 57(11): 1239–1249. [CrossRef] [PubMed]
Jacob K, Quang-Khuong DA, Jones DTW, et al. Genetic aberrations leading to MAPK pathway activation mediate oncogene-induced senescence in sporadic pilocytic astrocytomas. Clin Cancer Res Off J Am Assoc Cancer Res. 2011; 17(14): 4650–4660. [CrossRef]
Raabe EH, Lim KS, Kim JM, et al. BRAF activation induces transformation and then senescence in human neural stem cells: a pilocytic astrocytoma model. Clin Cancer Res Off J Am Assoc Cancer Res. 2011; 17(11): 3590–3599. [CrossRef]
Buhl JL, Selt F, Hielscher T, et al. The Senescence-associated Secretory Phenotype Mediates oncogene-induced senescence in pediatric pilocytic astrocytoma. Clin Cancer Res Off J Am Assoc Cancer Res. 2019; 25(6): 1851–1866. [CrossRef]
Piccirilli M, Lenzi J, Delfinis C, Trasimeni G, Salvati M, Raco A. Spontaneous regression of optic pathways gliomas in three patients with neurofibromatosis type I and critical review of the literature. Childs Nerv Syst. 2006; 22(10): 1332–1337. [CrossRef] [PubMed]
Short S, Fielder E, Miwa S, von Zglinicki T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine. 2019; 41: 683–692. [CrossRef] [PubMed]
Gutmann DH, Hunter-Schaedle K, Shannon KM. Harnessing preclinical mouse models to inform human clinical cancer trials. J Clin Invest. 2006; 116(4): 847–852. [CrossRef] [PubMed]
Khatua S, Gutmann DH, Packer RJ. Neurofibromatosis type 1 and optic pathway glioma: molecular interplay and therapeutic insights. Pediatr Blood Cancer. 2018; 65(3): e26838. [CrossRef]
Cassina M, Frizziero L, Opocher E, et al. Optic pathway glioma in type 1 neurofibromatosis: review of its pathogenesis, diagnostic assessment, and treatment recommendations. Cancers. 2019; 11(11): 1790. [CrossRef] [PubMed]
Isakson SH, Rizzardi AE, Coutts AW, et al. Genetically engineered minipigs model the major clinical features of human neurofibromatosis type 1. Commun Biol. 2018; 1(1): 158. [CrossRef] [PubMed]
Swindle MM, Makin A, Herron AJ, Clubb FJ, Frazier KS. Swine as models in biomedical research and toxicology testing. Vet Pathol. 2012; 49(2): 344–356. [CrossRef] [PubMed]
Walters EM, Wells KD, Bryda EC, Schommer S, Prather RS. Swine models, genomic tools and services to enhance our understanding of human health and diseases. Lab Anim. 2017; 46(4): 167–172. [CrossRef]
Wegscheid ML, Anastasaki C, Gutmann DH. Human stem cell modeling in neurofibromatosis type 1 (NF1). Exp Neurol. 2018; 299(Pt B): 270–280. [PubMed]
Muffat J, Li Y, Yuan B, et al. Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med. 2016; 22(11): 1358–1367. [CrossRef] [PubMed]
Figure 1.
 
Ras signaling pathway. Deprivation of neurofibromin in NF1-related OPGs led to activate Ras and its 3 effector pathways. Nf1−/− astrocytes displayed both upregulated MEK-ERK and PI3K-AKT manners, which individually contributed to activating mTOR activity and promoting tumor proliferation. Specifically, PI3K/AKT manner promoted the proliferation of o-GSCs and MEK/ERK pathway determined the progenitors multilineage differentiation. Besides, neurofibromin decreased the levels of cAMP in Nf1−/− astrocytes to positively accelerate tumor development while downregulated cAMP levels attenuated Nf1+/− RGC growth through PKCζ, which was related with visual dysfunction. This figure was created using the BioRender software.
Figure 1.
 
Ras signaling pathway. Deprivation of neurofibromin in NF1-related OPGs led to activate Ras and its 3 effector pathways. Nf1−/− astrocytes displayed both upregulated MEK-ERK and PI3K-AKT manners, which individually contributed to activating mTOR activity and promoting tumor proliferation. Specifically, PI3K/AKT manner promoted the proliferation of o-GSCs and MEK/ERK pathway determined the progenitors multilineage differentiation. Besides, neurofibromin decreased the levels of cAMP in Nf1−/− astrocytes to positively accelerate tumor development while downregulated cAMP levels attenuated Nf1+/− RGC growth through PKCζ, which was related with visual dysfunction. This figure was created using the BioRender software.
Figure 2.
 
The risk factors of NF1-OPGs. *A series of clinical and preclinical trials have found some potential risk factors of NF1-OPGs, including age, sex, tumor location, germline gene mutations, co-existent gene mutations, and genomic alterations. Further investigation is needed to develop a scientific risk assessment algorithm to stratify patients based on their risk levels. This figure was created using the BioRender software.
Figure 2.
 
The risk factors of NF1-OPGs. *A series of clinical and preclinical trials have found some potential risk factors of NF1-OPGs, including age, sex, tumor location, germline gene mutations, co-existent gene mutations, and genomic alterations. Further investigation is needed to develop a scientific risk assessment algorithm to stratify patients based on their risk levels. This figure was created using the BioRender software.
Figure 3.
 
The complex communication between neoplastic and non-neoplastic cells. *O-GSCs expressed CX3CL1 to recruit microglia which elaborated molecules to support neoplastic cells proliferation, including Ccl5, CXCL12, and MGEA5, and, in turn, neoplastic cells also secreted molecules (“stromagens”) to recruit microglia. Activated by Ccl2 secreted from o-GSCs, T cells then produced Ccl4 to promotes microglia to excrete Ccl5. As immune system regulators, RGCs held upregulated expression of midkine to activate T cells and initiated optic gliomas progression by NLGN3 signaling axis. By contrast, in female GEMs, estrogen induced microglia to produce neurotoxins, like IL-1β and other inflammatory cytokines, resulting in vision deterioration. Besides, other cells within the tumor microenvironment, like endothelial cells and Müller cells, might also contribute to the development of OPGs. This figure was created using the BioRender software.
Figure 3.
 
The complex communication between neoplastic and non-neoplastic cells. *O-GSCs expressed CX3CL1 to recruit microglia which elaborated molecules to support neoplastic cells proliferation, including Ccl5, CXCL12, and MGEA5, and, in turn, neoplastic cells also secreted molecules (“stromagens”) to recruit microglia. Activated by Ccl2 secreted from o-GSCs, T cells then produced Ccl4 to promotes microglia to excrete Ccl5. As immune system regulators, RGCs held upregulated expression of midkine to activate T cells and initiated optic gliomas progression by NLGN3 signaling axis. By contrast, in female GEMs, estrogen induced microglia to produce neurotoxins, like IL-1β and other inflammatory cytokines, resulting in vision deterioration. Besides, other cells within the tumor microenvironment, like endothelial cells and Müller cells, might also contribute to the development of OPGs. This figure was created using the BioRender software.
Table.
 
The Ongoing Clinical Trials for LGG or OPG
Table.
 
The Ongoing Clinical Trials for LGG or OPG
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×