Next Article in Journal
The Polysomnographical Meaning of Changed Sleep Quality—A Study of Treatment with Reduced Time in Bed
Previous Article in Journal
Magnetic Resonance Imaging Biomarkers of Punding in Parkinson’s Disease
Previous Article in Special Issue
Neuro-Ophthalmologic Variability in Presentation of Genetically Confirmed Wolfram Syndrome: A Case Series and Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma

1
Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
3
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
5
Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
6
Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
7
Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Brain Sci. 2023, 13(10), 1424; https://doi.org/10.3390/brainsci13101424
Submission received: 8 September 2023 / Revised: 28 September 2023 / Accepted: 3 October 2023 / Published: 7 October 2023
(This article belongs to the Special Issue Neuro-Ophthalmology and Optic Neuropathy)

Abstract

:
Optic pathway glioma (OPG) is one of the causes of pediatric visual impairment. Unfortunately, there is as yet no cure for such a disease. Understanding the underlying mechanisms and the potential therapeutic strategies may help to delay the progression of OPG and rescue the visual morbidities. Here, we provide an overview of preclinical OPG studies and the regulatory pathways controlling OPG pathophysiology. We next discuss the role of microenvironmental cells (neurons, T cells, and tumor-associated microglia and macrophages) in OPG development. Last, we provide insight into potential therapeutic strategies for treating OPG and promoting axon regeneration.

1. Introduction

Optic pathway glioma (OPG) is a type of brain tumor that develops within the optic pathway, which connects the eye to the brain. Most OPGs (59%) arise in children younger than 10 years of age [1]. Most OPGs are histologically characterized as grade 1 pilocytic astrocytoma (2021 WHO classification), in which KIAA1549-BRAF, BRAF, and NF1 are the commonly altered genes [2]. OPGs are low-grade gliomas with features that include a low proliferation index [3,4] as well as glial fibrillary acidic protein (GFAP) and oligodendrocyte transcription factor 2 (OLIG2) immunoreactivity [5,6], and they are often slow-growing. Whereas glial markers in OPG display high positivity, they stain negatively for NeuN, indicating the glial feature exhibited by the tumor cells [5]. OPGs can develop in both anterior (prechiasmatic) and posterior (chiasmatic and postchiasmatic) visual pathways. Because the tumor is located along the optic pathway, patients may experience vision decline or vision loss in severe cases [7,8,9,10]. Some OPGs are located close to the hypothalamus and can thus induce endocrine abnormalities, including precocious puberty [10,11].
Optic pathway gliomas can be categorized into two groups: neurofibromatosis type 1 (NF1)-associated OPG and sporadic (non-NF1) OPG. As an autosomal dominant genetic disease, NF1 is a cancer predisposition syndrome that affects 1 in 2500–3000 people worldwide; 15–20% of children with NF1 develop gliomas, approximately 65–75% of which are OPGs [9,10,12,13]. Children with NF1-OPG tend to develop anterior tumors more frequently than children with sporadic OPG [14,15,16,17]. The spontaneous regression of OPG has been reported in NF1 patients [18,19,20]. In sporadic OPGs, the most common genetic event is BRAF-KIAA fusion [21], which increases BRAF activity and its downstream MAPK signaling. NF1-OPG has a better prognosis than sporadic OPG, probably because of the higher incidence of anterior tumors along the optic pathway in NF1-OPGs [10,22].
The NF1 gene encodes neurofibromin protein, a large protein comprising approximately 3000 amino acids and with partial-sequence homology to a family of proteins that inhibit RAS activity, also known as RAS GTPase-activating proteins (RAS-GAPs) [23]. Neurofibromin can inhibit several RAS proteins—namely HRAS, NRAS, and KRAS—through RAS-GTP hydrolysis (the conversion of active RAS-GTP to inactive RAS-GDP), leading to the abrogation of RAS activity [24,25]. Therefore, it is understandable that in NF1 patients, loss-of-function NF1 mutations heighten RAS activity, which in turn may be responsible for tumorigenesis [26,27]. Indeed, in both murine and human NF1-associated tumors, reduced neurofibromin expression is accompanied by RAS protein hyperactivity and the elevated activation of the RAS downstream pathways (e.g., RAF-MEK-ERK, PI3K-AKT, mTOR signaling pathways) [28,29,30]. Neurofibromin also has RAS-independent functions: for example, it positively mediates cyclic adenosine monophosphate (cAMP) levels in retinal ganglion cells (RGCs) via PKC-zeta/GRK2 [31].
The study of human OPG is difficult because OPGs are rarely resected, and even when they are, the low-grade glioma cells are rarely grown in culture or as xenografts [32,33]. To overcome this barrier and gain a deeper understanding of OPGs, researchers have generated genetically engineered mouse models of OPG [34,35,36,37,38,39]. One such model, Nf1OPG (Nf1flox/−; Gfap-Cre) mice, recapitulates the genetics of NF1 patients: the mice harbor a germline Nf1 mutation in all cells and a second-hit Nf1 loss in the glioma-initiating neural stem cells [34].

2. Methods

Following the PRISMA guidelines, authors searched the PubMed/MEDLINE and Web of Science databases and examined the relevant literature published from 1980 to 2023. Combinations of the following keywords were used to screen the topic, maximizing the specificity and sensitivity: “optic pathway glioma”, “neurofibromatosis type 1”, “cancer neuroscience”, “cancer therapy”, and “neuroinflammation”. We used these combination keywords to further screen the title and abstract.

3. Results

3.1. Effect of Optic Pathway Glioma on Vision and Retinal Ganglion Cell (RGC) Function

Approximately 30–50% of patients with NF1-OPG experience vision decline [13,40,41,42,43]. The radiographic characteristics of the OPG tumor do not seem to correlate with visual outcome [41,44,45], although a recent study suggests that larger OPG volume (measured by magnetic resonance imaging [MRI]) is associated with less retinal nerve fiber layer thickness [46]. Most NF1-OPG patients exhibit good visual outcomes [47]. Conversely, sporadic OPGs have a higher chance of progressing and producing worse visual outcomes than NF1-OPGs [10,22,48,49,50].
Although some support using MRI screening for the early diagnosis of NF1-OPG and, thus, better vision outcomes [51], some may oppose MRI scans of young patients due to the effects of the repeated sedation involved in the process [52]. Sometimes MRI is carried out only when optic nerve abnormalities or visual impairments are detected [47], and MRI may not detect a tumor or tumor progression when vision loss is observed. Optical coherence tomography (OCT) is becoming increasingly popular for monitoring vision and can detect changes in retinal nerve fiber layer thickness early on, thereby facilitating crucial decision-making processes [53,54]. Similarly, the electrophysiological monitoring of OPG patients by using electroretinography (ERG) and visual evoked potentials (VEP) may also provide early evidence of optic nerve or RGC dysfunction [55,56,57]. However, the choice of modality is still at the discretion of the treating clinician. The optimal diagnostic modality for NF1-OPG management may be better clarified in future multicenter trials.
Like patients with NF1-OPG, the Nf1OPG mice exhibit thinner retinal nerve fiber layers as measured by OCT and reduced VEP amplitudes than the non-tumor controls [58,59]. The Nf1OPG mice also display RGC death and optic nerve myelin defects [58,59]. Some NF1-associated retinal and visual deficits may potentially be intrinsic to the germline NF1 mutations, because Nf1+/− RGCs exhibit more frequent cell death, shorter neurite length, and smaller growth cone area than RGCs from wild-type counterparts [31]. The neurofibromin-cAMP axis is responsible for these phenotypes observed in primary Nf1+/− RGC culture [31].
Several risk factors for OPG-induced visual impairment have been identified, including age <2 years, postchiasmatic tumor location, and female sex [9,10,17,60]. In patients with NF1-OPG, females are more likely than males to undergo MRI and treatment for visual symptoms. Similarly, in mouse models of Nf1OPG, females have more severe RGC loss than males [42].

3.2. The Role of Immune Cells in Optic Pathway Glioma Growth

Microglia and macrophages are the immune cells found in the central nervous system (CNS). However, the origins of microglia and macrophages are different. Microglia are derived from yolk sac progenitors and are the resident immune cells in the brain, whereas macrophages are derived from circulating monocytes and can infiltrate the CNS from the bloodstream [61]. Microglia are important for maintaining brain homeostasis and responding to various types of insults, including pathogens [62,63], injury [64], and neurodegeneration (e.g., Alzheimer’s disease) [65,66]. In the context of brain tumors, microglia and macrophages can contribute to tumor progression.
Glioma cells can recruit and reprogram microglia and macrophages to adopt a pro-tumor phenotype, collectively known as glioma-associated microglia and macrophages (GAMs) [67]. GAMs can release growth factors, cytokines, and chemokines that promote tumor cell survival, invasion, and angiogenesis [68]. Targeting GAMs has been suggested as a potential therapeutic strategy for gliomas [69,70]. Overall, microglia and macrophages are critical components of the CNS immune system and have complex roles in brain tumor biology.
Although both macrophages and microglia exist in the optic nerve [71], microglia are often reported to be involved in several optic neuropathies, including optic nerve trauma [72] and OPGs [73]. Growing evidence shows that the inhibition of microglia activation by the c-Jun-NH(2)-kinase (JNK) inhibitor [74] or minocycline (a crude microglia inhibitor) [75,76] is able to delay glioma formation in the Nf1OPG mice. In addition to pharmaceutical approaches, genetically reducing the expression of CX3CR1, the receptor that modulates microglial migration [77], delays OPG formation [73]. Among the secretion molecules from GAMs, chemokine (C-C motif) ligand 5 (CCL5) is a key promoter for NF1-OPG growth. Neutralization with CCL5 antibody [78] or the deletion of Ccl5 [79] dramatically attenuated tumor growth in vivo. An interesting observation suggests that the reduced RGC numbers and thinner retinal nerve fiber layer in female Nf1OPG mice compared with male ones may be due to estrogen-induced microglial activation [80]. Thus, suppressing microglial activation, either during cell migration or mitogen secretion, is a potential therapeutic strategy against OPG.
T cells (T lymphocytes) are white blood cells that modulate adaptive immune response [81,82]. T cells are further identified as CD4+ helper T cells and CD8+ cytotoxic T cells [83]. In cancer pathology, CD8+ T cells are detected around the tumors and are thought to mediate anti-tumor responses [84]. However, CD8+ T cells fail to do so during long-period tumorigenesis because they differentiate into dysfunctional states [85]. In the case of mouse NF1-OPG, CD8+ T cells are involved in neuron-mediated T cell CCL4 production and subsequently induce microglial CCL5 secretion to support tumor growth, and the depletion of CD8+ T cells suppressed tumor growth [86], thereby suggesting that CD8+ T cells could be a therapeutic target in the optic nerve for patients with OPG.

3.3. The Role of Neurons in Optic Pathway Gliomagenesis

In the past years, ample studies have confirmed the key function of neurons in modulating cancer progression [87]. OPG grows within the optic pathway and is surrounded by neurons. Preclinical studies have shown that light-induced neuronal activity in the optic nerve is required to initiate NF1-OPG. In the Nf1OPG mice, dark-rearing inhibits tumor initiation and maintenance [88]. Molecularly, the germline Nf1 mutation aberrantly increases neuronal activity-induced secretion of ADAM10, a protease that cleaves membrane proteins, in the optic nerve. One of the substrates of ADAM10 is neuroligin-3. This Nf1/ADAM10 axis results in the increased cleavage of the ectodomain of neuroligin-3, which is sufficient for increasing OPG cell growth in vitro. Supporting the role of ADAM10/neuroligin-3 axis in the pathogenesis of NF1-OPG, genetically ablating neuroligin-3, or pharmacologically inhibiting ADAM10 in Nf1OPG mice inhibits optic gliomagenesis [88].
In addition to the light-dependent regulation of NF1 optic gliomagenesis, a light-independent neuronal pathway regulates OPG growth [89]. Certain germline Nf1 mutations in RGCs induce neuronal hyperactivity by inhibiting the hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels. This RGC hyperactivity increases neuronal production of midkine, which functions to recruit T cells that support OPG growth, as discussed in the above section. Midkine-neutralizing antibodies rescued the Nf1-mutation-induced RGC hyperactivity and inhibited tumor growth in the Nf1OPG mice [89].
The above-mentioned studies were conducted in the context of NF1-OPGs. Whether neurons are important microenvironmental contributors to sporadic OPGs that do not harbor the germline NF1 mutation remains to be determined.

3.4. Implications for Optic Pathway Glioma Treatment

3.4.1. Targeting the Neoplastic Cells

Because surgery may damage the optic pathway and radiation imposes risks for secondary malignancies and cognitive issues in NF1-OPG, the first line treatment for NF1-OPG is chemotherapy, which often involves platinum-based and vinca alkaloids-based chemotherapy agents (e.g., carboplatin and vincristine) that target the neoplastic (tumor) cells [13,90,91,92,93,94,95]. Because both NF1-associated and sporadic OPGs exhibit MAPK hyperactivation, targeted therapy is a potential strategy. In addition, NF1 loss leads to the hyperactivation of RAS and its downstream effectors (e.g., MEK/ERK, PI3K/AKT, and mTOR). Targeting these pathways inhibits tumor growth in preclinical mouse models [30]. Ongoing clinical trials primarily aim to target these aberrantly hyperactive pathways within the neoplastic cells [96,97].

3.4.2. Targeting the Microenvironment

Given the emerging evidence that the microenvironment (e.g., neurons, T cells, microglia) is critical for OPG pathogenesis, future clinical studies could consider adjuvant strategies that target microenvironmental factors, including ADAM10, neuroligin-3, midkine, CCL4, and CCL5. Environmental factors, especially light, could potentially serve as targets for preventing or treating OPG; however, because light is important for the development of visual pathway, preclinical studies should be conducted to determine the optimal light conditions that can be used to treat OPG without affecting visual pathway development. In addition, ADAM10 inhibitors and HCN activators may represent new neuron-targeting strategies to treat OPG. In malignant high-grade gliomas, neuronal activity leverages the ADAM10/neuroligin-3 axis to drive tumor growth [98,99], which has led to a clinical trial that evaluates an ADAM10 inhibitor (INCB7839) in treating recurrent or progressive pediatric high-grade gliomas (NCT04295759). Lamotrigine, an HCN agonist that is used clinically for treating seizures, rescues Nf1-mutation–induced RGC hyperactivity and inhibits tumor growth in Nf1OPG mice.
In addition to targeting the neuronal components, targeting the tumor immune microenvironment is another attractive strategy. Tumor associated microglia and macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) are tumor-promoting in many solid tumors [100,101]. Because TAMs are the most abundant immune cells in the tumor microenvironment and can be associated with poor prognosis of patients [102], several small-molecule inhibitors have been developed against TAMs and their inflammatory signaling (e.g., in prostate, breast, ovarian, colon, and skin cancers) [103]. Although successful cases of tumor inhibition were reported by small molecule treatment, liver and renal toxicities remain issues [104,105].
The development of a TAM-targeting strategy for treating OPG remains an unmet need. In preclinical models, CD8+ T cells were shown to secrete CCL4, which binds to microglial CCR5 to stimulate the microglial secretion of CCL5 via the nuclear factor-κB (NFkB) pathway [86]. Targeting these critical molecules in the immune regulation of OPG may be an effective strategy. Additionally, using a natural small molecule to inhibit aldose reductase was found to suppress microglia and macrophage migration and inflammatory cytokine secretion in mouse eyes [106,107]. Studies also showed that aldose reductase inhibition alleviated microglia activation triggered by beta-amyloid [66] and optic nerve injury [108], suggesting that aldose reductase blockade plays a protective role in the CNS and may be a therapeutic avenue for OPG. Table 1 is presented below.

4. Discussion

Several rodent models were established for OPG studies (Table 1), in which the tumor can be observed as early as two months of age. However, the study of OPG in larger animal or non-human primates remains an unmet need, despite the recent development of NF1 pig models [110,111] Whereas survival for patients with OPG is excellent, no effective treatments for OPG-induced visual impairment are currently available [8,41,49,50,112,113], likely because tumor-induced RGC loss cannot be reversed. Strategies that improve RGC survival and regeneration and that promote the remyelination of RGC axons are needed to rescue OPG-impaired vision.
Since the molecules/proteins in Table 2 are known activators for OPG pathophysiology, the pharmaceutical development of agonists/antagonists to those molecules may be beneficial for OPG therapy. Interestingly, neuroinflammation has been reported to promote axon regeneration [114], with the chemokine CCL5 seen as a critical factor [115]; however, CCL5 is also a mitogen that increases OPG growth. The balance of neuroinflammation in axon regeneration and glioma growth is another key consideration in treating OPG. In addition, the cell-intrinsic deficits induced by germline NF1 mutations (e.g., in neurons and oligodendroglial cells) will need to be considered as these strategies are tested in the context of NF1-OPG.
Glioma formation in the optic nerve causes axon degeneration [46], leading to vision impairment. Thus, promoting axon regeneration is one of the goals for treating OPG. In optic neuropathy, adeno-associated virus (AAV)-mediated gene therapy that targets RGC is promising in axon regeneration and partial visual restoration in optic nerve crush models [116,117]. Recently, the food and drug administration (FDA) approved Luxturna, an AAV-based gene therapy for treating RPE65-positive retinal dystrophy [118], which marked a clinical milestone. However, so far, no evidence shows that AAV-mediated gene therapy is able to extend regenerative axons from RGCs to go through OPG in animal models. More studies are required to warrant this therapeutic strategy in treating OPG.
In OPG patients, RGCs die followed by axon degeneration. Unfortunately, RGCs do not regenerate or are not replaced after injury in humans. RGC transplantation seems a feasible approach to replace RGCs, but the source of primary human RGC is limited. Stem cell (SC)-derived RGCs provide the opportunity for cell replacement therapy. In addition to replacing dead cells, SC-derived RGCs also improve endogenous RGC survival [119,120]. A study in non-human primates showed that axons from donor RGCs were observed in the brain [121], suggesting that SC-derived RGC transplantation may restore the axon loss in the optic nerve with OPG. Whether the microenvironment of OPG in the optic nerve would affect donor RGC survival and their axon growth would be an interesting direction for future study.
Radiation therapy is one of the most common treatments against tumors. Due to the location of the OPG, radiation therapy seems to be a better approach than the surgical procedure to eliminate a tumor in the optic nerve. However, several radiation-induced side effects are detrimental (especially for patients with NF1), including visual disturbance (7–17% of patients who received radiation therapy), vasculopathy (higher incidence of vasculopathy in patients <5 years than those >5 years after radiation therapy, 12.5% vs. 3.8%), endocrine deficiency (growth hormone deficiency is the most frequent, 59%), neurocognitive impairment, and secondary malignancy [122].
Bevacizumab (Avastin), which inhibits vascular endothelial growth factor (VEGF) from binding to its receptors, is an anti-cancer drug considered for treating glioblastoma (high-grade glioma) [123] and ovarian cancer [124]. A study showed that bevacizumab may improve vision in OPG patients [125]; yet the role of VEGF in OPG progression and OPG-induced visual impairment remains to be determined.

5. Conclusions

Combinatory therapies of chemotherapy, gene therapy, and stem cell replacement therapy may be required to reduce tumor size and rescue vision. Given that RGC rescue strategies may increase the optic nerve neuronal activity that induces OPG growth [88,89] and that some of the gene therapies are based on the deletion of an anti-tumor gene (e.g., PTEN) [126,127], the careful optimization of the treatment regimen (e.g., dose, sequence, cell-specificity, and timing) will be required for the translational study. Although curing OPG is full of challenges, several groups in the fields of visual sciences, stem cell biology, and brain tumor biology are taking on bold research initiatives to develop collaborative approaches for treating and/or slowing down the progression of OPG. Any successful preclinical and/or clinical studies will help move the field of pediatric oncology forward and give hope to patients who suffer from vision impairment caused by OPG.

Author Contributions

Writing—original draft preparation, K.I., Y.-K.H. and P.R.; writing—review and editing, J.L. and B.E.A.; supervision, Y.P. and K.-C.C.; funding acquisition, Y.P. and K.-C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Department of Defense Neurofibromatosis Research Program (HT9425-23-1-0270 and HT9425-23-1-0239 to Y.P.), Alex’s Lemonade Stand Foundation (19-16681 to Y.P.), Cancer Prevention and Research Institute of Texas (RR210085 to Y.P. as the CPRIT Scholar in Cancer Research), Gilbert Family Foundation (622030 to Y.P.), National Institutes of Health Core Grants (P30-EY008098 to K-C.C), Eye and Ear Foundation of Pittsburgh (to K-C.C), and an unrestricted grant and a career development award from Research to Prevent Blindness, New York, NY (to K-C.C).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alvord, E.C., Jr.; Lofton, S. Gliomas of the optic nerve or chiasm. Outcome by patients’ age, tumor site, and treatment. J. Neurosurg. 1988, 68, 85–98. [Google Scholar] [CrossRef] [PubMed]
  2. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  3. Cummings, T.J.; Provenzale, J.M.; Hunter, S.B.; Friedman, A.H.; Klintworth, G.K.; Bigner, S.H.; McLendon, R.E. Gliomas of the optic nerve: Histological, immunohistochemical (MIB-1 and p53), and MRI analysis. Acta Neuropathol. 2000, 99, 563–570. [Google Scholar] [CrossRef] [PubMed]
  4. Yeung, S.N.; White, V.A.; Nimmo, M.; Rootman, J. Optic nerve gliomas: Role of Ki-67 staining of tumour and margins in predicting long-term outcome. Br. J. Ophthalmol. 2011, 95, 1077–1081. [Google Scholar] [CrossRef]
  5. Reis, G.F.; Bloomer, M.M.; Perry, A.; Phillips, J.J.; Grenert, J.P.; Karnezis, A.N.; Tihan, T. Pilocytic astrocytomas of the optic nerve and their relation to pilocytic astrocytomas elsewhere in the central nervous system. Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc. 2013, 26, 1279–1287. [Google Scholar] [CrossRef]
  6. Cutarelli, P.E.; Roessmann, U.R.; Miller, R.H.; Specht, C.S.; Grossniklaus, H.E. Immunohistochemical properties of human optic nerve glioma. Evidence of type 1 astrocyte origin. Investig. Ophthalmol. Vis. Sci. 1991, 32, 2521–2524. [Google Scholar]
  7. Wan, M.J.; Ullrich, N.J.; Manley, P.E.; Kieran, M.W.; Goumnerova, L.C.; Heidary, G. Long-term visual outcomes of optic pathway gliomas in pediatric patients without neurofibromatosis type 1. J. Neuro Oncol. 2016, 129, 173–178. [Google Scholar] [CrossRef]
  8. Dalla Via, P.; Opocher, E.; Pinello, M.L.; Calderone, M.; Viscardi, E.; Clementi, M.; Battistella, P.A.; Laverda, A.M.; Da Dalt, L.; Perilongo, G. 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, 430–437. [Google Scholar] [CrossRef]
  9. Listernick, R.; Charrow, J.; Greenwald, M.; Mets, M. Natural history of optic pathway tumors in children with neurofibromatosis type 1: A longitudinal study. J. Pediatr. 1994, 125, 63–66. [Google Scholar] [CrossRef]
  10. Listernick, R.; Louis, D.N.; Packer, R.J.; Gutmann, D.H. Optic pathway gliomas in children with neurofibromatosis 1: Consensus statement from the NF1 Optic Pathway Glioma Task Force. Ann. Neurol. 1997, 41, 143–149. [Google Scholar] [CrossRef]
  11. Habiby, R.; Silverman, B.; Listernick, R.; Charrow, J. Precocious puberty in children with neurofibromatosis type 1. J. Pediatr. 1995, 126, 364–367. [Google Scholar] [CrossRef] [PubMed]
  12. Guillamo, J.S.; Creange, A.; Kalifa, C.; Grill, J.; Rodriguez, D.; Doz, F.; Barbarot, S.; Zerah, M.; Sanson, M.; Bastuji-Garin, S.; et al. Prognostic factors of CNS tumours in Neurofibromatosis 1 (NF1): A retrospective study of 104 patients. Brain 2003, 126, 152–160. [Google Scholar] [CrossRef] [PubMed]
  13. Khatua, S.; Gutmann, D.H.; Packer, R.J. Neurofibromatosis type 1 and optic pathway glioma: Molecular interplay and therapeutic insights. Pediatr. Blood Cancer 2018, 65, e26838. [Google Scholar] [CrossRef]
  14. Taylor, T.; Jaspan, T.; Milano, G.; Gregson, R.; Parker, T.; Ritzmann, T.; Benson, C.; Walker, D. Radiological classification of optic pathway gliomas: Experience of a modified functional classification system. Br. J. Radiol. 2008, 81, 761–766. [Google Scholar] [CrossRef]
  15. Yu, J.; Deshmukh, H.; Gutmann, R.J.; Emnett, R.J.; Rodriguez, F.J.; Watson, M.A.; Nagarajan, R.; Gutmann, D.H. Alterations of BRAF and HIPK2 loci predominate in sporadic pilocytic astrocytoma. Neurology 2009, 73, 1526–1531. [Google Scholar] [CrossRef]
  16. Lewis, R.A.; Gerson, L.P.; Axelson, K.A.; Riccardi, V.M.; Whitford, R.P. Von Recklinghausen neurofibromatosis. II. Incidence of optic gliomata. Ophthalmology 1984, 91, 929–935. [Google Scholar] [CrossRef] [PubMed]
  17. Listernick, R.; Charrow, J.; Greenwald, M.J.; Esterly, N.B. Optic gliomas in children with neurofibromatosis type 1. J. Pediatr. 1989, 114, 788–792. [Google Scholar] [CrossRef]
  18. Venes, J.L.; Latack, J.; Kandt, R.S. Postoperative regression of opticochiasmatic astrocytoma: A case for expectant therapy. Neurosurgery 1984, 15, 421–423. [Google Scholar] [CrossRef]
  19. Brzowski, A.E.; Bazan, C., 3rd; Mumma, J.V.; Ryan, S.G. Spontaneous regression of optic glioma in a patient with neurofibromatosis. Neurology 1992, 42, 679–681. [Google Scholar] [CrossRef]
  20. Perilongo, G.; Moras, P.; Carollo, C.; Battistella, A.; Clementi, M.; Laverda, A.; Murgia, A. Spontaneous partial regression of low-grade glioma in children with neurofibromatosis-1: A real possibility. J. Child Neurol. 1999, 14, 352–356. [Google Scholar] [CrossRef]
  21. Jacob, K.; Albrecht, S.; Sollier, C.; Faury, D.; Sader, E.; Montpetit, A.; Serre, D.; Hauser, P.; Garami, M.; Bognar, L.; et al. Duplication of 7q34 is specific to juvenile pilocytic astrocytomas and a hallmark of cerebellar and optic pathway tumours. Br. J. Cancer 2009, 101, 722–733. [Google Scholar] [CrossRef] [PubMed]
  22. Deliganis, A.V.; Geyer, J.R.; Berger, M.S. Prognostic significance of type 1 neurofibromatosis (von Recklinghausen Disease) in childhood optic glioma. Neurosurgery 1996, 38, 1114–1118, discussion 1118–1119. [Google Scholar] [CrossRef] [PubMed]
  23. Anastasaki, C.; Orozco, P.; Gutmann, D.H. RAS and beyond: The many faces of the neurofibromatosis type 1 protein. Dis. Models Mech. 2022, 15, dmm049362. [Google Scholar] [CrossRef] [PubMed]
  24. Ballester, R.; Marchuk, D.; Boguski, M.; Saulino, A.; Letcher, R.; Wigler, M.; Collins, F. The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell 1990, 63, 851–859. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, G.F.; Lin, B.; Tanaka, K.; Dunn, D.; Wood, D.; Gesteland, R.; White, R.; Weiss, R.; Tamanoi, F. The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell 1990, 63, 835–841. [Google Scholar] [CrossRef] [PubMed]
  26. Basu, T.N.; Gutmann, D.H.; Fletcher, J.A.; Glover, T.W.; Collins, F.S.; Downward, J. Aberrant regulation of ras proteins in malignant tumour cells from type 1 neurofibromatosis patients. Nature 1992, 356, 713–715. [Google Scholar] [CrossRef]
  27. DeClue, J.E.; Papageorge, A.G.; Fletcher, J.A.; Diehl, S.R.; Ratner, N.; Vass, W.C.; Lowy, D.R. Abnormal regulation of mammalian p21ras contributes to malignant tumor growth in von Recklinghausen (type 1) neurofibromatosis. Cell 1992, 69, 265–273. [Google Scholar] [CrossRef]
  28. Johannessen, C.M.; Reczek, E.E.; James, M.F.; Brems, H.; Legius, E.; Cichowski, K. The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proc. Natl. Acad. Sci. USA 2005, 102, 8573–8578. [Google Scholar] [CrossRef]
  29. Dasgupta, B.; Yi, Y.; Chen, D.Y.; Weber, J.D.; Gutmann, D.H. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res. 2005, 65, 2755–2760. [Google Scholar] [CrossRef]
  30. Kaul, A.; Toonen, J.A.; Cimino, P.J.; Gianino, S.M.; Gutmann, D.H. Akt- or MEK-mediated mTOR inhibition suppresses Nf1 optic glioma growth. Neuro Oncol. 2015, 17, 843–853. [Google Scholar] [CrossRef]
  31. Anastasaki, C.; Gutmann, D.H. Neuronal NF1/RAS regulation of cyclic AMP requires atypical PKC activation. Hum. Mol. Genet. 2014, 23, 6712–6721. [Google Scholar] [CrossRef] [PubMed]
  32. Kogiso, M.; Qi, L.; Lindsay, H.; Huang, Y.; Zhao, X.; Liu, Z.; Braun, F.K.; Du, Y.; Zhang, H.; Bae, G.; et al. Xenotransplantation of pediatric low grade gliomas confirms the enrichment of BRAF V600E mutation and preservation of CDKN2A deletion in a novel orthotopic xenograft mouse model of progressive pleomorphic xanthoastrocytoma. Oncotarget 2017, 8, 87455–87471. [Google Scholar] [CrossRef] [PubMed]
  33. Sanden, E.; Eberstal, S.; Visse, E.; Siesjo, P.; Darabi, A. A standardized and reproducible protocol for serum-free monolayer culturing of primary paediatric brain tumours to be utilized for therapeutic assays. Sci. Rep. 2015, 5, 12218. [Google Scholar] [CrossRef]
  34. Bajenaru, M.L.; Hernandez, M.R.; Perry, A.; Zhu, Y.; Parada, L.F.; Garbow, J.R.; Gutmann, D.H. Optic nerve glioma in mice requires astrocyte Nf1 gene inactivation and Nf1 brain heterozygosity. Cancer Res. 2003, 63, 8573–8577. [Google Scholar]
  35. Zhu, Y.; Harada, T.; Liu, L.; Lush, M.E.; Guignard, F.; Harada, C.; Burns, D.K.; Bajenaru, M.L.; Gutmann, D.H.; Parada, L.F. Inactivation of NF1 in CNS causes increased glial progenitor proliferation and optic glioma formation. Development 2005, 132, 5577–5588. [Google Scholar] [CrossRef]
  36. Guo, X.; Pan, Y.; Gutmann, D.H. 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, 1250–1262. [Google Scholar] [CrossRef]
  37. Kaul, A.; Toonen, J.A.; Gianino, S.M.; Gutmann, D.H. The impact of coexisting genetic mutations on murine optic glioma biology. Neuro Oncol. 2015, 17, 670–677. [Google Scholar] [CrossRef]
  38. Solga, A.C.; Gianino, S.M.; Gutmann, D.H. NG2-cells are not the cell of origin for murine neurofibromatosis-1 (Nf1) optic glioma. Oncogene 2014, 33, 289–299. [Google Scholar] [CrossRef]
  39. Toonen, J.A.; Anastasaki, C.; Smithson, L.J.; Gianino, S.M.; Li, K.; Kesterson, R.A.; Gutmann, D.H. NF1 germline mutation differentially dictates optic glioma formation and growth in neurofibromatosis-1. Hum. Mol. Genet. 2016, 25, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
  40. Listernick, R.; Ferner, R.E.; Liu, G.T.; Gutmann, D.H. Optic pathway gliomas in neurofibromatosis-1: Controversies and recommendations. Ann. Neurol. 2007, 61, 189–198. [Google Scholar] [CrossRef] [PubMed]
  41. Fisher, M.J.; Loguidice, M.; Gutmann, D.H.; Listernick, R.; Ferner, R.E.; Ullrich, N.J.; Packer, R.J.; Tabori, U.; Hoffman, R.O.; Ardern-Holmes, S.L.; et al. Visual outcomes in children with neurofibromatosis type 1-associated optic pathway glioma following chemotherapy: A multicenter retrospective analysis. Neuro Oncol. 2012, 14, 790–797. [Google Scholar] [CrossRef] [PubMed]
  42. Diggs-Andrews, K.A.; Brown, J.A.; Gianino, S.M.; Rubin, J.B.; Wozniak, D.F.; Gutmann, D.H. Sex Is a major determinant of neuronal dysfunction in neurofibromatosis type 1. Ann. Neurol. 2014, 75, 309–316. [Google Scholar] [CrossRef] [PubMed]
  43. Balcer, L.J.; Liu, G.T.; Heller, G.; Bilaniuk, L.; Volpe, N.J.; Galetta, S.L.; Molloy, P.T.; Phillips, P.C.; Janss, A.J.; Vaughn, S.; 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, 442–445. [Google Scholar] [CrossRef]
  44. Jose, 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, 128–135. [Google Scholar] [CrossRef]
  45. Kornreich, L.; Blaser, S.; Schwarz, M.; Shuper, A.; Vishne, T.H.; Cohen, I.J.; Faingold, R.; Michovitz, S.; Koplewitz, B.; Horev, G. Optic pathway glioma: Correlation of imaging findings with the presence of neurofibromatosis. AJNR Am. J. Neuroradiol. 2001, 22, 1963–1969. [Google Scholar] [PubMed]
  46. Avery, R.A.; Mansoor, A.; Idrees, R.; Trimboli-Heidler, C.; Ishikawa, H.; Packer, R.J.; Linguraru, M.G. Optic pathway glioma volume predicts retinal axon degeneration in neurofibromatosis type 1. Neurology 2016, 87, 2403–2407. [Google Scholar] [CrossRef]
  47. Zeid, J.L. Current update on the visual outcome of optic pathway glioma associated with neurofibromatosis type-1. Front. Surg. 2022, 9, 908573. [Google Scholar] [CrossRef]
  48. Varan, A.; Batu, A.; Cila, A.; Soylemezoğlu, F.; Balcı, S.; Akalan, N.; Zorlu, F.; Akyüz, C.; Kutluk, T.; Büyükpamukçu, M. Optic glioma in children: A retrospective analysis of 101 cases. Am. J. Clin. Oncol. 2013, 36, 287–292. [Google Scholar] [CrossRef]
  49. Dodgshun, A.J.; Elder, J.E.; Hansford, J.R.; Sullivan, M.J. Long-term visual outcome after chemotherapy for optic pathway glioma in children: Site and age are strongly predictive. Cancer 2015, 121, 4190–4196. [Google Scholar] [CrossRef]
  50. Kalin-Hajdu, E.; Décarie, J.C.; Marzouki, M.; Carret, A.S.; Ospina, L.H. Visual acuity of children treated with chemotherapy for optic pathway gliomas. Pediatr. Blood Cancer 2014, 61, 223–227. [Google Scholar] [CrossRef]
  51. Prada, C.E.; Hufnagel, R.B.; Hummel, T.R.; Lovell, A.M.; Hopkin, R.J.; Saal, H.M.; Schorry, E.K. The Use of Magnetic Resonance Imaging Screening for Optic Pathway Gliomas in Children with Neurofibromatosis Type 1. J. Pediatr. 2015, 167, 851–856.e1. [Google Scholar] [CrossRef]
  52. Ing, C.; DiMaggio, C.; Whitehouse, A.; Hegarty, M.K.; Brady, J.; von Ungern-Sternberg, B.S.; Davidson, A.; Wood, A.J.; Li, G.; Sun, L.S. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics 2012, 130, e476–e485. [Google Scholar] [CrossRef] [PubMed]
  53. Arnljots, U.; Nilsson, M.; Sandvik, U.; Myrberg, I.H.; Munoz, D.M.; Blomgren, K.; Hellgren, K. Optical Coherence Tomography Identifies Visual Pathway Involvement Earlier than Visual Function Tests in Children with MRI-Verified Optic Pathway Gliomas. Cancers 2022, 14, 318. [Google Scholar] [CrossRef] [PubMed]
  54. Zahavi, A.; Toledano, H.; Cohen, R.; Sella, S.; Luckman, J.; Michowiz, S.; Goldenberg-Cohen, N. Use of Optical Coherence Tomography to Detect Retinal Nerve Fiber Loss in Children With Optic Pathway Glioma. Front. Neurol. 2018, 9, 1102. [Google Scholar] [CrossRef] [PubMed]
  55. Moradi, P.; Robson, A.G.; Rose, G.E.; Holder, G.E. Electrophysiological monitoring in a patient with an optic nerve glioma. Doc. Ophthalmol. Adv. Ophthalmol. 2008, 117, 171–174. [Google Scholar] [CrossRef]
  56. Trisciuzzi, M.T.; Riccardi, R.; Piccardi, M.; Iarossi, G.; Buzzonetti, L.; Dickmann, A.; Colosimo, C., Jr.; Ruggiero, A.; Di Rocco, C.; Falsini, B. A fast visual evoked potential method for functional assessment and follow-up of childhood optic gliomas. Clin. Neurophysiol. Off. J. Int. Fed. Clin. Neurophysiol. 2004, 115, 217–226. [Google Scholar] [CrossRef]
  57. Wolsey, D.H.; Larson, S.A.; Creel, D.; Hoffman, R. Can screening for optic nerve gliomas in patients with neurofibromatosis type I be performed with visual-evoked potential testing? J. Am. Assoc. Pediatr. Ophthalmol. Strabismus 2006, 10, 307–311. [Google Scholar] [CrossRef]
  58. Toonen, J.A.; Ma, Y.; Gutmann, D.H. Defining the temporal course of murine neurofibromatosis-1 optic gliomagenesis reveals a therapeutic window to attenuate retinal dysfunction. Neuro Oncol. 2017, 19, 808–819. [Google Scholar] [CrossRef]
  59. Hegedus, B.; Hughes, F.W.; Garbow, J.R.; Gianino, S.; Banerjee, D.; Kim, K.; Ellisman, M.H.; Brantley, M.A., Jr.; Gutmann, D.H. Optic nerve dysfunction in a mouse model of neurofibromatosis-1 optic glioma. J. Neuropathol. Exp. Neurol. 2009, 68, 542–551. [Google Scholar] [CrossRef]
  60. Zeid, J.L.; Charrow, J.; Sandu, M.; Goldman, S.; Listernick, R. Orbital optic nerve gliomas in children with neurofibromatosis type 1. J. Am. Assoc. Pediatr. Ophthalmol. Strabismus 2006, 10, 534–539. [Google Scholar] [CrossRef]
  61. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef] [PubMed]
  62. Voet, S.; Prinz, M.; van Loo, G. Microglia in Central Nervous System Inflammation and Multiple Sclerosis Pathology. Trends Mol. Med. 2019, 25, 112–123. [Google Scholar] [CrossRef] [PubMed]
  63. Rock, R.B.; Gekker, G.; Hu, S.; Sheng, W.S.; Cheeran, M.; Lokensgard, J.R.; Peterson, P.K. Role of microglia in central nervous system infections. Clin. Microbiol. Rev. 2004, 17, 942–964. [Google Scholar] [CrossRef] [PubMed]
  64. Donat, C.K.; Scott, G.; Gentleman, S.M.; Sastre, M. Microglial Activation in Traumatic Brain Injury. Front. Aging Neurosci. 2017, 9, 208. [Google Scholar] [CrossRef]
  65. Hansen, D.V.; Hanson, J.E.; Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 2018, 217, 459–472. [Google Scholar] [CrossRef]
  66. Huang, Y.K.; Liu, C.C.; Wang, S.; Cheng, H.C.; Meadows, C.; Chang, K.C. The Role of Aldose Reductase in Beta-Amyloid-Induced Microglia Activation. Int. J. Mol. Sci. 2022, 23, 15088. [Google Scholar] [CrossRef]
  67. Gutmann, D.H.; Kettenmann, H. Microglia/Brain Macrophages as Central Drivers of Brain Tumor Pathobiology. Neuron 2019, 104, 442–449. [Google Scholar] [CrossRef]
  68. Xu, C.; Xiao, M.; Li, X.; Xin, L.; Song, J.; Zhan, Q.; Wang, C.; Zhang, Q.; Yuan, X.; Tan, Y.; et al. Origin, activation, and targeted therapy of glioma-associated macrophages. Front. Immunol. 2022, 13, 974996. [Google Scholar] [CrossRef]
  69. Wang, G.; Zhong, K.; Wang, Z.; Zhang, Z.; Tang, X.; Tong, A.; Zhou, L. Tumor-associated microglia and macrophages in glioblastoma: From basic insights to therapeutic opportunities. Front. Immunol. 2022, 13, 964898. [Google Scholar] [CrossRef]
  70. Poon, C.C.; Sarkar, S.; Yong, V.W.; Kelly, J.J.P. Glioblastoma-associated microglia and macrophages: Targets for therapies to improve prognosis. Brain 2017, 140, 1548–1560. [Google Scholar] [CrossRef]
  71. Cui, Q.; Yin, Y.; Benowitz, L.I. The role of macrophages in optic nerve regeneration. Neuroscience 2009, 158, 1039–1048. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, J.; He, X.; Meng, H.; Li, Y.; Dmitriev, P.; Tian, F.; Page, J.C.; Lu, Q.R.; He, Z. Robust Myelination of Regenerated Axons Induced by Combined Manipulations of GPR17 and Microglia. Neuron 2020, 108, 876–886.E4. [Google Scholar] [CrossRef] [PubMed]
  73. Pong, W.W.; Higer, S.B.; Gianino, S.M.; Emnett, R.J.; Gutmann, D.H. Reduced microglial CX3CR1 expression delays neurofibromatosis-1 glioma formation. Ann. Neurol. 2013, 73, 303–308. [Google Scholar] [CrossRef] [PubMed]
  74. Daginakatte, G.C.; Gianino, S.M.; Zhao, N.W.; Parsadanian, A.S.; Gutmann, D.H. Increased c-Jun-NH2-kinase signaling in neurofibromatosis-1 heterozygous microglia drives microglia activation and promotes optic glioma proliferation. Cancer Res. 2008, 68, 10358–10366. [Google Scholar] [CrossRef]
  75. Daginakatte, G.C.; Gutmann, D.H. Neurofibromatosis-1 (Nf1) heterozygous brain microglia elaborate paracrine factors that promote Nf1-deficient astrocyte and glioma growth. Hum. Mol. Genet. 2007, 16, 1098–1112. [Google Scholar] [CrossRef]
  76. Pan, Y.; Bush, E.C.; Toonen, J.A.; Ma, Y.; Solga, A.C.; Sims, P.A.; Gutmann, D.H. Whole tumor RNA-sequencing and deconvolution reveal a clinically-prognostic PTEN/PI3K-regulated glioma transcriptional signature. Oncotarget 2017, 8, 52474–52487. [Google Scholar] [CrossRef]
  77. Wolf, Y.; Yona, S.; Kim, K.W.; Jung, S. Microglia, seen from the CX3CR1 angle. Front. Cell Neurosci. 2013, 7, 26. [Google Scholar] [CrossRef]
  78. Solga, A.C.; Pong, W.W.; Kim, K.Y.; Cimino, P.J.; Toonen, J.A.; Walker, J.; Wylie, T.; Magrini, V.; Griffith, M.; Griffith, O.L.; et al. RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia 2015, 17, 776–788. [Google Scholar] [CrossRef]
  79. Pan, Y.; Xiong, M.; Chen, R.; Ma, Y.; Corman, C.; Maricos, M.; Kindler, U.; Semtner, M.; Chen, Y.H.; Dahiya, S.; 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, 491–496. [Google Scholar] [CrossRef]
  80. Toonen, J.A.; Solga, A.C.; Ma, Y.; Gutmann, D.H. Estrogen activation of microglia underlies the sexually dimorphic differences in Nf1 optic glioma-induced retinal pathology. J. Exp. Med. 2017, 214, 17–25. [Google Scholar] [CrossRef]
  81. Eberl, G.; Colonna, M.; Di Santo, J.P.; McKenzie, A.N. Innate lymphoid cells. Innate lymphoid cells: A new paradigm in immunology. Science 2015, 348, aaa6566. [Google Scholar] [CrossRef] [PubMed]
  82. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef]
  83. Davis, S.J.; Ikemizu, S.; Evans, E.J.; Fugger, L.; Bakker, T.R.; van der Merwe, P.A. The nature of molecular recognition by T cells. Nat. Immunol. 2003, 4, 217–224. [Google Scholar] [CrossRef] [PubMed]
  84. Romero, I.; Garrido, F.; Garcia-Lora, A.M. Metastases in immune-mediated dormancy: A new opportunity for targeting cancer. Cancer Res. 2014, 74, 6750–6757. [Google Scholar] [CrossRef]
  85. Philip, M.; Schietinger, A. CD8(+) T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 2022, 22, 209–223. [Google Scholar] [CrossRef]
  86. Guo, X.; Pan, Y.; Xiong, M.; Sanapala, S.; Anastasaki, C.; Cobb, O.; Dahiya, S.; Gutmann, D.H. Midkine activation of CD8(+) T cells establishes a neuron-immune-cancer axis responsible for low-grade glioma growth. Nat. Commun. 2020, 11, 2177. [Google Scholar] [CrossRef]
  87. Mancusi, R.; Monje, M. The neuroscience of cancer. Nature 2023, 618, 467–479. [Google Scholar] [CrossRef]
  88. Pan, Y.; Hysinger, J.D.; Barron, T.; Schindler, N.F.; Cobb, O.; Guo, X.; Yalcin, B.; Anastasaki, C.; Mulinyawe, S.B.; Ponnuswami, A.; et al. NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature 2021, 594, 277–282. [Google Scholar] [CrossRef]
  89. Anastasaki, C.; Mo, J.; Chen, J.K.; Chatterjee, J.; Pan, Y.; Scheaffer, S.M.; Cobb, O.; Monje, M.; Le, L.Q.; Gutmann, D.H. Neuronal hyperexcitability drives central and peripheral nervous system tumor progression in models of neurofibromatosis-1. Nat. Commun. 2022, 13, 2785. [Google Scholar] [CrossRef]
  90. Sharif, S.; Ferner, R.; Birch, J.M.; Gillespie, J.E.; Gattamaneni, H.R.; Baser, M.E.; Evans, D.G. Second primary tumors in neurofibromatosis 1 patients treated for optic glioma: Substantial risks after radiotherapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2006, 24, 2570–2575. [Google Scholar] [CrossRef]
  91. Armstrong, G.T.; Conklin, H.M.; Huang, S.; Srivastava, D.; Sanford, R.; Ellison, D.W.; Merchant, T.E.; Hudson, M.M.; Hoehn, M.E.; Robison, L.L.; et al. Survival and long-term health and cognitive outcomes after low-grade glioma. Neuro Oncol. 2011, 13, 223–234. [Google Scholar] [CrossRef] [PubMed]
  92. Shalitin, S.; Gal, M.; Goshen, Y.; Cohen, I.; Yaniv, I.; Phillip, M. Endocrine outcome in long-term survivors of childhood brain tumors. Horm. Res. Paediatr. 2011, 76, 113–122. [Google Scholar] [CrossRef] [PubMed]
  93. Grill, J.; Couanet, D.; Cappelli, C.; Habrand, J.L.; Rodriguez, D.; Sainte-Rose, C.; Kalifa, C. Radiation-induced cerebral vasculopathy in children with neurofibromatosis and optic pathway glioma. Ann. Neurol. 1999, 45, 393–396. [Google Scholar] [CrossRef]
  94. Ater, J.L.; Xia, C.; Mazewski, C.M.; Booth, T.N.; Freyer, D.R.; Packer, R.J.; Sposto, R.; Vezina, G.; Pollack, I.F. Nonrandomized comparison of neurofibromatosis type 1 and non-neurofibromatosis type 1 children who received carboplatin and vincristine for progressive low-grade glioma: A report from the Children’s Oncology Group. Cancer 2016, 122, 1928–1936. [Google Scholar] [CrossRef] [PubMed]
  95. Packer, R.J.; Ater, J.; Allen, J.; Phillips, P.; Geyer, R.; Nicholson, H.S.; Jakacki, R.; Kurczynski, E.; Needle, M.; Finlay, J.; et al. Carboplatin and vincristine chemotherapy for children with newly diagnosed progressive low-grade gliomas. J. Neurosurg. 1997, 86, 747–754. [Google Scholar] [CrossRef]
  96. Hill, C.S.; Devesa, S.C.; Ince, W.; Borg, A.; Aquilina, K. A systematic review of ongoing clinical trials in optic pathway gliomas. Child’s Nerv. Syst. 2020, 36, 1869–1886. [Google Scholar] [CrossRef]
  97. Tang, Y.; Gutmann, D.H. Neurofibromatosis Type 1-Associated Optic Pathway Gliomas: Current Challenges and Future Prospects. Cancer Manag. Res. 2023, 15, 667–681. [Google Scholar] [CrossRef]
  98. Venkatesh, H.S.; Johung, T.B.; Caretti, V.; Noll, A.; Tang, Y.; Nagaraja, S.; Gibson, E.M.; Mount, C.W.; Polepalli, J.; Mitra, S.S.; et al. Neuronal Activity Promotes Glioma Growth through Neuroligin-3 Secretion. Cell 2015, 161, 803–816. [Google Scholar] [CrossRef]
  99. Venkatesh, H.S.; Tam, L.T.; Woo, P.J.; Lennon, J.; Nagaraja, S.; Gillespie, S.M.; Ni, J.; Duveau, D.Y.; Morris, P.J.; Zhao, J.J.; et al. Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature 2017, 549, 533–537. [Google Scholar] [CrossRef]
  100. Luo, J.L.; Maeda, S.; Hsu, L.C.; Yagita, H.; Karin, M. Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell 2004, 6, 297–305. [Google Scholar] [CrossRef]
  101. Majety, M.; Runza, V.; Lehmann, C.; Hoves, S.; Ries, C.H. A drug development perspective on targeting tumor-associated myeloid cells. FEBS J. 2018, 285, 763–776. [Google Scholar] [CrossRef] [PubMed]
  102. Steidl, C.; Lee, T.; Shah, S.P.; Farinha, P.; Han, G.; Nayar, T.; Delaney, A.; Jones, S.J.; Iqbal, J.; Weisenburger, D.D.; et al. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N. Engl. J. Med. 2010, 362, 875–885. [Google Scholar] [CrossRef] [PubMed]
  103. Zhong, S.; Jeong, J.H.; Chen, Z.; Chen, Z.; Luo, J.L. Targeting Tumor Microenvironment by Small-Molecule Inhibitors. Transl. Oncol. 2020, 13, 57–69. [Google Scholar] [CrossRef]
  104. Mudd, T.W.; Guddati, A.K. Management of hepatotoxicity of chemotherapy and targeted agents. Am. J. Cancer Res. 2021, 11, 3461–3474. [Google Scholar]
  105. Santos, M.L.C.; de Brito, B.B.; da Silva, F.A.F.; Botelho, A.; de Melo, F.F. Nephrotoxicity in cancer treatment: An overview. World J. Clin. Oncol. 2020, 11, 190–204. [Google Scholar] [CrossRef] [PubMed]
  106. Chang, K.C.; Laffin, B.; Ponder, J.; Enzsoly, A.; Nemeth, J.; LaBarbera, D.V.; Petrash, J.M. Beta-glucogallin reduces the expression of lipopolysaccharide-induced inflammatory markers by inhibition of aldose reductase in murine macrophages and ocular tissues. Chem. Biol. Interact. 2013, 202, 283–287. [Google Scholar] [CrossRef]
  107. Chang, K.C.; Ponder, J.; Labarbera, D.V.; Petrash, J.M. Aldose reductase inhibition prevents endotoxin-induced inflammatory responses in retinal microglia. Investig. Ophthalmol. Vis. Sci. 2014, 55, 2853–2861. [Google Scholar] [CrossRef]
  108. Rao, M.; Huang, Y.K.; Liu, C.C.; Meadows, C.; Cheng, H.C.; Zhou, M.; Chen, Y.C.; Xia, X.; Goldberg, J.L.; Williams, A.M.; et al. Aldose reductase inhibition decelerates optic nerve degeneration by alleviating retinal microglia activation. Sci. Rep. 2023, 13, 5592. [Google Scholar] [CrossRef]
  109. Solga, A.C.; Toonen, J.A.; Pan, Y.; Cimino, P.J.; Ma, Y.; Castillon, G.A.; Gianino, S.M.; Ellisman, M.H.; Lee, D.Y.; Gutmann, D.H. The cell of origin dictates the temporal course of neurofibromatosis-1 (Nf1) low-grade glioma formation. Oncotarget 2017, 8, 47206–47215. [Google Scholar] [CrossRef]
  110. Isakson, S.H.; Rizzardi, A.E.; Coutts, A.W.; Carlson, D.F.; Kirstein, M.N.; Fisher, J.; Vitte, J.; Williams, K.B.; Pluhar, G.E.; Dahiya, S.; et al. Genetically engineered minipigs model the major clinical features of human neurofibromatosis type 1. Commun. Biol. 2018, 1, 158. [Google Scholar] [CrossRef]
  111. White, K.A.; Swier, V.J.; Cain, J.T.; Kohlmeyer, J.L.; Meyerholz, D.K.; Tanas, M.R.; Uthoff, J.; Hammond, E.; Li, H.; Rohret, F.A.; et al. A porcine model of neurofibromatosis type 1 that mimics the human disease. JCI Insight 2018, 3, e120402. [Google Scholar] [CrossRef] [PubMed]
  112. Moreno, L.; Bautista, F.; Ashley, S.; Duncan, C.; Zacharoulis, S. Does chemotherapy affect the visual outcome in children with optic pathway glioma? A systematic review of the evidence. Eur. J. Cancer 2010, 46, 2253–2259. [Google Scholar] [CrossRef] [PubMed]
  113. Shofty, B.; Ben-Sira, L.; Freedman, S.; Yalon, M.; Dvir, R.; Weintraub, M.; Toledano, H.; Constantini, S.; Kesler, A. Visual outcome following chemotherapy for progressive optic pathway gliomas. Pediatr. Blood Cancer 2011, 57, 481–485. [Google Scholar] [CrossRef]
  114. Wong, K.A.; Benowitz, L.I. Retinal Ganglion Cell Survival and Axon Regeneration after Optic Nerve Injury: Role of Inflammation and Other Factors. Int. J. Mol. Sci. 2022, 23, 10179. [Google Scholar] [CrossRef]
  115. Xie, L.; Yin, Y.; Benowitz, L. Chemokine CCL5 promotes robust optic nerve regeneration and mediates many of the effects of CNTF gene therapy. Proc. Natl. Acad. Sci. USA 2021, 118, e2017282118. [Google Scholar] [CrossRef]
  116. Williams, P.R.; Benowitz, L.I.; Goldberg, J.L.; He, Z. Axon Regeneration in the Mammalian Optic Nerve. Annu. Rev. Vis. Sci. 2020, 6, 195–213. [Google Scholar] [CrossRef]
  117. Fague, L.; Liu, Y.A.; Marsh-Armstrong, N. The basic science of optic nerve regeneration. Ann. Transl. Med. 2021, 9, 1276. [Google Scholar] [CrossRef]
  118. Mendell, J.R.; Al-Zaidy, S.A.; Rodino-Klapac, L.R.; Goodspeed, K.; Gray, S.J.; Kay, C.N.; Boye, S.L.; Boye, S.E.; George, L.A.; Salabarria, S.; et al. Current Clinical Applications of In Vivo Gene Therapy with AAVs. Mol. Ther. 2021, 29, 464–488. [Google Scholar] [CrossRef]
  119. Singh, R.K.; Occelli, L.M.; Binette, F.; Petersen-Jones, S.M.; Nasonkin, I.O. Transplantation of Human Embryonic Stem Cell-Derived Retinal Tissue in the Subretinal Space of the Cat Eye. Stem Cells Dev. 2019, 28, 1151–1166. [Google Scholar] [CrossRef]
  120. Luo, Z.; Chang, K.C.; Wu, S.; Sun, C.; Xia, X.; Nahmou, M.; Bian, M.; Wen, R.R.; Zhu, Y.; Shah, S.; et al. Directly induced human retinal ganglion cells mimic fetal RGCs and are neuroprotective after transplantation in vivo. Stem Cell Rep. 2022, 17, 2690–2703. [Google Scholar] [CrossRef]
  121. Luo, Z.; Xian, B.; Li, K.; Li, K.; Yang, R.; Chen, M.; Xu, C.; Tang, M.; Rong, H.; Hu, D.; et al. Biodegradable scaffolds facilitate epiretinal transplantation of hiPSC-Derived retinal neurons in nonhuman primates. Acta Biomater. 2021, 134, 289–301. [Google Scholar] [CrossRef] [PubMed]
  122. Kim, N.; Lim, D.H. Recent Updates on Radiation Therapy for Pediatric Optic Pathway Glioma. Brain Tumor Res. Treat. 2022, 10, 94–100. [Google Scholar] [CrossRef] [PubMed]
  123. Vaz-Salgado, M.A.; Villamayor, M.; Albarran, V.; Alia, V.; Sotoca, P.; Chamorro, J.; Rosero, D.; Barrill, A.M.; Martin, M.; Fernandez, E.; et al. Recurrent Glioblastoma: A Review of the Treatment Options. Cancers 2023, 15, 4279. [Google Scholar] [CrossRef] [PubMed]
  124. Lazurko, C.; Linder, R.; Pulman, K.; Lennox, G.; Feigenberg, T.; Fazelzad, R.; May, T.; Zigras, T. Bevacizumab Treatment for Low-Grade Serous Ovarian Cancer: A Systematic Review. Curr. Oncol. 2023, 30, 8159–8171. [Google Scholar] [CrossRef] [PubMed]
  125. Yamasaki, F.; Takano, M.; Yonezawa, U.; Taguchi, A.; Kolakshyapati, M.; Okumichi, H.; Kiuchi, Y.; Kurisu, K. Bevacizumab for optic pathway glioma with worsening visual field in absence of imaging progression: 2 case reports and literature review. Childs Nerv. Syst. 2020, 36, 635–639. [Google Scholar] [CrossRef] [PubMed]
  126. Park, K.K.; Liu, K.; Hu, Y.; Smith, P.D.; Wang, C.; Cai, B.; Xu, B.; Connolly, L.; Kramvis, I.; Sahin, M.; et al. Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science 2008, 322, 963–966. [Google Scholar] [CrossRef]
  127. Gicquel, J.J.; Vabres, P.; Bonneau, D.; Mercie, M.; Handiri, L.; Dighiero, P. Retinal angioma in a patient with Cowden disease. Am. J. Ophthalmol. 2003, 135, 400–402. [Google Scholar] [CrossRef]
Table 1. Genetically engineered mouse models used to study optic glioma.
Table 1. Genetically engineered mouse models used to study optic glioma.
GenotypeOptic Glioma PhenotypeRef
Nf1flox/−; hGfap-Cre
Nf1flox/flox; hGfap-Cre
tumor developed by 2 months of age[35]
Nf1flox/neo; Gfap-Cre *#tumor developed by 3 months of age[34]
Nf1flox/neo; Ptenflox/+; Gfap-Cretumor developed by 3 months of age[37]
Nf1flox/neo; Olig2-Cretumor developed by 6 months of age[109]
Nf1flox/neo; Prom1-CreERtumor developed by 3 months of age[109]
Nf1flox/R681X; Gfap-Cretumor developed by 3 months of age[39]
Nf1flox/G848R; Gfap-Creno tumor detected by 3 months of age[39]
Nf1flox/C383X; Gfap-Cre25% mice developed tumor by 3 months of age[36]
Nf1flox/R1278P; Gfap-Cretumor developed by 3 months of age[36]
* neo, engineered neomycin insertion to disrupt Nf1 function. # the activity of the Gfap promoter was detected around E15, whereas the activity of the hGfap promoter was detected around E12.
Table 2. Critical cell/tissue types and the corresponding molecules in optic glioma pathophysiology.
Table 2. Critical cell/tissue types and the corresponding molecules in optic glioma pathophysiology.
Cell/Tissue TypeMoleculeRole in Optic Glioma ProgressionRef
Retinal ganglion cellsMidkinestimulate T cell Ccl4 production by binding to LRP1[86]
Retinal ganglion cellsHCNNf1 mutation induced HCN dysfunction induces neuronal hyperactivity; modulates midkine level[89]
Optic nerveADAM10increased secretion in response to light-induced neuronal activity; cleaves neuroligin-3[88]
Optic nerveNeuroligin-3required for optic gliomagenesis[88]
CD8+ T cellsCCL4stimulate microglial Ccl5 production by binding to CCR5[86]
MicrogliaCCL5binds to CD44 on tumor cells and increases tumor cell survival[86]
MicrogliaCX3CR1required for tumorigenesis[73]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Irshad, K.; Huang, Y.-K.; Rodriguez, P.; Lo, J.; Aghoghovwia, B.E.; Pan, Y.; Chang, K.-C. The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma. Brain Sci. 2023, 13, 1424. https://doi.org/10.3390/brainsci13101424

AMA Style

Irshad K, Huang Y-K, Rodriguez P, Lo J, Aghoghovwia BE, Pan Y, Chang K-C. The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma. Brain Sciences. 2023; 13(10):1424. https://doi.org/10.3390/brainsci13101424

Chicago/Turabian Style

Irshad, Khushboo, Yu-Kai Huang, Paul Rodriguez, Jung Lo, Benjamin E. Aghoghovwia, Yuan Pan, and Kun-Che Chang. 2023. "The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma" Brain Sciences 13, no. 10: 1424. https://doi.org/10.3390/brainsci13101424

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop