- 1Xiamen Eye Center and Eye Institute of Xiamen University, School of Medicine, Xiamen, China
- 2Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China
- 3Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China
- 4Fujian Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- 5Xiamen Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, Fujian, China
- 6Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
Cancer is a global health threat, and its incidence and mortality are increasing annually. Cancer-related glaucoma, a severe complication caused by primary or metastatic tumors and their treatments, has complex pathogenic mechanisms. This review aims to clarify the risk factors, classification, diagnosis, and treatment progress of this glaucoma type. Mechanisms include mechanical obstruction, secondary angle closure, neovascularization, inflammation and cytokine release, alterations in aqueous humor dynamics, and secondary hemosiderosis. Clinical manifestations are diverse, such as rapid intraocular pressure increase, neovascular changes, and tumor-related characteristic signs. Treatment requires multidisciplinary cooperation, with primary tumor control as the core, combined with drugs like anti-VEGF agents and targeted therapies, and modified surgeries. Future research should focus on personalized therapeutic strategies, gene therapy applications, integration of multimodal imaging, and optimization of AI models to optimize early intervention and reduce the risk of irreversible optic nerve damage.
1 Introduction
Cancer is one of the diseases that seriously threatens human health worldwide, with its incidence and mortality rates increasing year by year. According to data from the World Health Organization (WHO), in 2020, there were over 19 million new cancer cases globally, and nearly 10 million deaths. Ocular and central nervous system tumors, though rare, cause visual dysfunction and systemic complications that severely affect quality of life (1). Advances in cancer diagnosis and treatment have prolonged patient survival, but both tumors and their therapies (such as radiotherapy, chemotherapy, targeted therapy, etc.) can induce ocular complications. Secondary glaucoma, in particular, is concerning due to its risk of irreversible optic nerve damage (2).
Tumors can lead to the development of glaucoma through multiple mechanisms. For example, ocular tumors (such as iris melanoma and retinoblastoma) can directly invade the angle structure or induce neovascular glaucoma (NVG). NVG is one of the most common secondary glaucomas, accounting for 44.9% of cases, with some of those arising from tumors (3, 4). Epidemiological studies show that among pediatric glaucoma cases, secondary glaucoma accounts for as high as 53.4%, and 28% of them are associated with acquired systemic diseases (5, 6). In addition, tumor treatment drugs (such as corticosteroids and immune checkpoint inhibitors) may also cause secondary glaucoma by increasing intraocular pressure or inducing uveitis (7, 8), highlighting the complexity of the relationship between tumors and glaucoma.
With the development of comprehensive cancer treatment, the clinical identification and management of glaucoma secondary to tumors have gradually become an important topic for multidisciplinary collaboration. However, currently, there is a lack of systematic reviews on the epidemiological characteristics, pathological mechanisms, and treatment strategies of such glaucoma (9, 10). Some studies have pointed out that tumor-related glaucoma is often misdiagnosed due to its atypical clinical manifestations. Therefore, we conducted a rigorous review, integrated existing evidence, clarified the risk factors, classification, and progress in diagnosis and treatment of glaucoma secondary to tumors, to provide a reference for clinical practice and call for strengthened ophthalmic follow-up of cancer patients to reduce the risk of irreversible vision loss (2, 11).
2 Methods
To comprehensively synthesize the latest advancements in cancer-related glaucoma, we employed a hybrid approach that combines systematic literature retrieval with flexible expansion to ensure both rigor and innovation.
2.1 Literature retrieval strategy
A structured Boolean search was initially conducted across major databases (PubMed, Web of Science, Cochrane Library, and Embase) to identify foundational literature. The core search terms were:
(“cancer-related glaucoma” OR “tumor-associated glaucoma” OR “neoplastic glaucoma” OR “secondary glaucoma AND cancer”) AND (mechanism OR pathogenesis OR “clinical manifestation” OR “treatment strategy” OR “neovascularization” OR “intraocular pressure” OR “uveal melanoma” OR “retinoblastoma”) AND [“2020/01/01” (PDAT): “2025/05/01” (PDAT)].
2.2 Inclusion and exclusion criteria
2.2.1 Inclusion criteria
Study types: original research (clinical studies, basic experiments), systematic reviews, and meta-analyses.
Content: studies focusing on pathogenesis, clinical manifestations, diagnostic methods, or therapeutic strategies of cancer-related glaucoma; those involving core mechanisms such as the HIF-1α/VEGF pathway and ferroptosis, or emerging technologies including AI-assisted diagnosis and gene editing, as cited in this review.
Publication time: January 2020 to July 2025.
2.2.2 Exclusion criteria
Study types: letters, editorials, and case reports (except for milestone rare cases).
Language: non-English language articles.
Research focus: primary glaucoma or secondary glaucoma unrelated to cancer, such as diabetic glaucoma.
Relevance: literature not explicitly addressing the “cancer-glaucoma” association, merely discussing cancer or glaucoma independently.
Duplication: duplicate publications or studies with overlapping data (the latest or larger-sample studies were retained).
Quality: low-quality literature, including those without clear methodology or statistical analysis.
2.3 Literature synthesis approach
After an in-depth review of the retrieved literature, supplementary searches were conducted to incorporate relevant studies that enhance structural completeness, hierarchical richness, and content diversity. This non-traditional approach breaks the “retrieve-then-integrate” framework, prioritizing the divergence of mechanistic logic and inclusiveness of cutting-edge research to better reflect the cross-disciplinary advancements in this field.
2.4 Pathogenesis
The pathogenesis of cancer-related glaucoma is complex and multifactorial, involving several key mechanisms. These include mechanical obstruction by tumor masses, secondary angle closure induced by various tumor-related factors, neovascularization driven by angiogenic mediators, and alterations in aqueous humor dynamics caused by tumor infiltration or debris. The essential features of these mechanisms are visually depicted in Figure 1.

Figure 1. Common pathogenic mechanisms of tumor-related secondary glaucoma. The blue arrows in the figure simulate the production and drainage pathways of intraocular aqueous humor. Part A illustrates that under the influence of iris tumors, the posterior chamber space becomes narrow, the anterior chamber angle is narrowed, and the aqueous humor produced by the ciliary body is difficult to reach the anterior chamber due to pupillary block. At the same time, the fluid in the anterior chamber also struggles to get through the trabecular meshwork through the narrowed angle. Part B shows that secondary angle closure prevents aqueous humor from being drained through the trabecular meshwork. Part C illustrates the neovascular changes in the iris and anterior chamber angle, with the disease progression depicted in this static image encompassing three distinct phases: In the first phase, new vessels emerge on the trabecular meshwork without elevated IOP; in the second phase, fibrous tissue surrounds these vessels, impairing filtration and increasing IOP; in the third phase, fibrovascular contraction causes irreversible angle closure. Responses to anti-VEGF therapy vary across these stages. Part D illustrates that the ciliary body, stimulated by inflammatory factors, leads to increased concentrations of inflammatory factors and proteins in the aqueous humor (indicated by brown dots), resulting in blockage and fibrosis of the trabecular meshwork.
2.4.1 Mechanical obstruction
Mechanical obstruction is a key pathogenic mechanism. Tumors (such as iris/ciliary body tumors, melanoma cells, metastatic lesions) can impede aqueous humor outflow through three pathways:
I. Anatomical compression: as the tumor grows in size, it can push the root of the iris, narrowing or even closing the anterior chamber angle, and directly blocking the traditional aqueous humor outflow pathway composed of the trabecular meshwork (TM) - Schlemm’s canal (SC) (12, 13). For example, iris melanoma can cause pupillary block, increasing the resistance to aqueous humor outflow (12).
II. Tissue fibrosis: factors such as TGF-β2 released by tumors can promote the fibrosis of the TM, further increasing the outflow resistance (14).
III. Kinetic changes: mechanical compression can alter the biomechanical properties of the angle tissues, reducing the viscoelasticity of the TM and the wall of Schlemm’s canal, leading to disordered aqueous humor flow (15). Studies have shown that tumor mass can cause abnormal stress distribution in the TM/JCT region and can reduce the aqueous humor flow rate by up to 9% (16).
2.4.2 Secondary angle closure
Tumor-related exudation (such as serous retinal detachment in uveal melanoma) or vitreous hemorrhage can cause posterior synechiae of the iris through mechanical obstruction or inflammatory reactions, leading to pupillary block and acute angle closure (17, 18). Studies show that approximately 7.47% of secondary glaucoma cases are associated with such complications (19). In clinical reports, patients developed refractory angle closure one month after being diagnosed with metastatic lung adenocarcinoma, which was identified to occur through choroidal metastasis. Traditional drug and laser treatments were ineffective, and the condition was finally relieved by radiotherapy (17). In addition, radiation can also damage the microvasculature of the TM, resulting in obstruction of aqueous humor outflow (20). It is worth noting that in patients with chronic primary angle-closure glaucoma (CPACG) complicated by radiation-induced maculopathy, the risk of ciliary body edema is significantly increased (21, 22).
2.4.3 Neovascularization
Retinoblastoma and choroidal melanoma secrete angiogenic factors such as VEGF, which can directly stimulate the formation of new blood vessels in the iris and angle. At the same time, it activates the hypoxia-inducible factor (HIF-1α) signaling pathway, jointly promoting pathological angiogenesis (4, 23, 24). The regulation of HIF-1α further aggravates the microcirculation disorder of the retina, forming a “hypoxia-angiogenesis” vicious cycle (25–27). These neovascular changes progress through three distinct phases. In the first phase, new blood vessels begin to grow on the trabecular meshwork, yet IOP remains unchanged; anti-VEGF injections at this stage may prevent the development of NVG and subsequent IOP elevation. In the second phase, these new vessels become surrounded by fibrous tissue that covers the trabecular meshwork, impairing aqueous humor filtration and leading to increased IOP, where anti-VEGF injections combined with IOP-lowering medications may still help control IOP. In the third phase, the fibrovascular tissue contracts, pulling the iris forward and causing irreversible closure of the anterior chamber angle, such that anti-VEGF injections primarily serve to reduce the risk of bleeding during tube implant surgery. Ultimately, these new blood vessels can block the aqueous humor outflow channels, leading to increased intraocular pressure and NVG (23, 28, 29). Studies have shown that dual-target inhibition of VEGF/HIF-1α is more effective in reducing the neovascular area than single-target intervention (30), and anti-VEGF drugs have been proven to treat NVG by blocking this pathway (24). Retinal ischemia (such as in diabetic retinopathy and retinal vein occlusion) is the most common cause of NVG (28, 31).
2.4.4 Inflammation and cytokine release
Tumor-associated inflammatory mediators disrupt ocular homeostasis through multiple pathways. For instance, in the case of blood-aqueous barrier disruption, IL-6 and TNF-α can upregulate vascular permeability, resulting in protein exudation, such as the deposition of IgG and complement C3. This also activates the macrophage-mediated inflammatory cascade (32–34). These inflammatory factors can further induce the expression of endothelial cell adhesion molecules, such as ICAM - 1/VCAM - 1, exacerbating the barrier dysfunction (35). Meanwhile, the exuded proteins and inflammatory mediators (such as IL-1β and MCP-1) co-deposit in the TM. They promote the abnormal accumulation of extracellular matrix (ECM) via the TLR4 signaling pathway, increasing the resistance to aqueous humor outflow (36, 37). Additionally, infections like HTLV - 1, a causative factor for leukemia/lymphoma, can cause TM cells to secrete excessive TNF-α and IL-6, directly impairing their contractile function (38, 39). In paraneoplastic syndromes, autoantibodies (such as anti-dsDNA antibodies) may target the surface antigens of TM cells, triggering complement activation and local inflammation (32, 40). Such autoimmune responses can act synergistically with pro-inflammatory factors released by tumors (such as IL-17 and TGF-β), accelerating the fibrosis of the TM (41).
2.4.5 Alterations in aqueous humor dynamics
Tumor cells or necrotic debris blocking the TM spaces (such as intraocular infiltration in leukemia) can lead to secondary glaucoma by mechanically obstructing the aqueous humor outflow channels. Studies have shown that after tumor cells invade the TM spaces, they directly impede the outflow of aqueous humor, and the unphagocytosed melanin granules of pigmented tumors, such a melanocytoma, further exacerbate the obstruction (42).
Ciliary body tumors can cause elevated intraocular pressure through two mechanisms: First, when the ciliary body is compressed or infiltrated by a tumor, mechanical stretching stimuli may activate the Transient Receptor Potential Vanilloid 4-hemichannel mechanism in non-pigmented ciliary epithelial (NPE) cells. If the mechanical stimulation persists, it may cause aqueous humor secretion disorders through this mechanism. It should be noted that this also depends on the regulatory direction of signaling molecules such as Adenosine Triphosphate (ATP) (43). Second, the mass effect of the tumor alters the anatomical structure of the anterior chamber, leading to impaired aqueous humor drainage. Clinical observations have shown that for glaucoma associated with active malignant tumors, filtration surgery should be avoided, and giving priority to treating the primary tumor may normalize the intraocular pressure (44, 45).
2.4.6 Secondary hemosiderosis
Secondary hemosiderosis is a pathological process in which hemosiderin abnormally deposits in ocular tissues such as the TM after long-term intraocular hemorrhage (such as retinal hemangioma or traumatic hemorrhage). Iron ions generate reactive oxygen species through the Fenton reaction, triggering oxidative stress and lipid peroxidation. This leads to dysfunction of TM cells and increased resistance to aqueous humor outflow, ultimately developing into open-angle glaucoma (46, 47). The clinical manifestations include heterochromia iridis, brownish - black deposition in the TM, and elevated intraocular pressure (48). Histopathological examination shows that iron deposition directly damages retinal ganglion cells, accelerating glaucomatous optic neuropathy (46, 49).
Table 1 summarizes the key pathogenesis mechanisms, clinical manifestations, and associated references across tumor types causing cancer-related glaucoma, facilitating a clear overview of their distinct features and links.

Table 1. Pathogenesis, clinical manifestations, and prevalence of cancer-related glaucoma by tumor type.
3 Clinical manifestation
3.1 Rapid increase in intraocular pressure
Glaucoma secondary to tumors can present as acute angle-closure glaucoma, characterized by sudden onset of headache, nausea, corneal edema, and a sharp decline in vision. Iris tumors, such as melanomas, can cause acute intraocular pressure elevation through pupillary block or direct invasion of the anterior chamber angle. Among patients with uveal melanoma, 22% develop secondary glaucoma due to neovascularization in the anterior chamber, while 76% have it due to angle narrowing or closure, and some of these patients have direct tumor invasion of the anterior chamber angle (50).
3.2 Neovascular changes
Prominent neovascularization on the iris surface, often extending radially toward the iris periphery, and fibrovascular membranes in the anterior chamber angle are typical manifestations of NVG, commonly encountered in tumor-related ischemia, including retinoblastoma and metastatic tumors (4). Approximately 71% of cases of glaucoma secondary to uveal melanoma have neovascularization as the main mechanism (50).
3.3 Tumor-related characteristic signs
I. Iris tumors: iris nodules should be studied with UBM to differentiate cystic from solid lesions. most solid lesions are melanoma, which is the most common primary uveal malignancy. Iris melanoma can present as a localized iris elevation or a space-occupying lesion of the ciliary body. clinical studies have found that approximately 43% of cases are associated with ciliary body involvement, which is significantly higher than that in patients without glaucoma (21%) (50). Iris tumors are usually pigmented (about 86%), but 14% of patients present with an amelanotic variant (51).
II. Hyphema: children with retinoblastoma may develop hyphema and vitreous seeding, accompanied by the characteristic leukocoria (2, 59). This phenomenon is caused by blood vessel rupture due to tumor necrosis or bleeding from new blood vessels. multimodal imaging shows that the hyphema is often accompanied by the deposition of tumor cells on the surface of the iris, resembling “pseudo-hypopyon” (52). It is worth noting that approximately 22% of cases develop refractory glaucoma secondary to tumor cell blockage of the anterior chamber angle or iris neovascularization. Such patients often require urgent treatment to preserve the eyeball (60). Differential diagnosis should include Coats’ disease, persistent hyperplastic primary vitreous (PHPV), etc., (61).
III. Choroidal thickening/detachment: in metastatic intraocular tumors (accounting for more than 90% of adult intraocular tumors), breast cancer and lung cancer metastases account for 75%–85% (53). these metastases most commonly involve the choroid (88%), presenting as rapidly progressive choroidal thickening (with an average thickness of 2.3 ± 0.7 mm) and serous retinal detachment (54). optical coherence tomography angiography (OCTA) can detect the characteristic “vascular void” phenomenon, which is in contrast to the “vascular arcade” structure of primary choroidal melanoma (62). approximately 35% of cases develop secondary elevation of intraocular pressure due to mechanical compression or inflammatory reactions. the incidence is significantly higher in breast cancer metastases (41%) than in lung cancer metastases (29%) (55).
3.4 Visual field and optic nerve damage
Tumor compression or ischemia can lead to early asymmetric visual field defects. Intracranial hypertension secondary to tumors is a common cause of optic disk edema (56). Acute angle-closure glaucoma may be accompanied by optic disk edema, while chronic cases show thinning of the nerve fiber layer (57, 63). Among patients with glaucoma secondary to uveal melanoma, 53% have retinal detachment, which is significantly higher than that in the non-glaucoma group (30%) (50).
3.5 Involvement of the contralateral eye
Involvement of the contralateral eye is a prominent feature of paraneoplastic ophthalmic syndromes, and it is particularly evident in cancer-associated retinopathy (CAR) and anti-Hu antibody-related optic neuritis (64). In CAR, both retinas are often affected simultaneously, presenting with visual acuity decline, visual field defects, and abnormal electroretinogram, which is caused by photoreceptor dysfunction. Approximately 60%–70% of CAR cases are associated with small-cell lung cancer (SCLC), and the anti-Hu antibody is a common paraneoplastic antibody in SCLC (65, 66). Anti-Hu antibody-positive optic neuritis leads to rapid bilateral visual loss, often accompanied by eye pain and pupillary afferent defects. Approximately 80% of patients are diagnosed with SCLC, and optic neuritis may precede the diagnosis of the tumor by several months (67, 68). MRI can show optic nerve enhancement, and a negative cerebrospinal fluid antibody test does not rule out a paraneoplastic etiology (69). When the intraocular pressure is elevated, it needs to be differentiated from primary glaucoma (70, 71).
3.6 Systemic associated symptoms
Approximately 11% of patients with uveal melanoma and newly diagnosed secondary glaucoma have synchronous metastases, which is significantly higher than 1.2% in the non-glaucoma group (50). In some cases, ocular symptoms precede the diagnosis of the primary tumor. For example, breast cancer metastases may present as vitreous opacities or choroidal masses (2, 72).
4 Treatment strategies
4.1 Supplementary information on primary tumor treatment
I. In terms of targeted therapy, the combined use of FGFR4 and EZH2 inhibitors offers a new strategy for hepatic cell carcinoma with eye metastases (73). Similar approaches may be extended to primary ocular tumors. Besides controlling neovascularization, anti-VEGF drugs, when combined with gene therapy, have shown a synergistic effect in intraocular malignancies (54, 74).
II. For uveal melanoma, although traditional radiotherapy and surgery remain the mainstream, inhibitors targeting ERRs (estrogen-related receptors) may be a breakthrough point by reversing tumor drug resistance (75). The latest clinical data show that immune checkpoint inhibitors have an objective response rate of 35%–40% for conjunctival melanoma and squamous cell carcinoma (76).
4.2 Surgical innovations and contraindications
I. For tumors with orbital invasion, modified orbital exenteration combined with intraoperative radiotherapy can increase the negative margin rate (60). As this procedure removes the eye and orbital contents, it inherently eliminates secondary glaucoma in the affected eye, rendering additional glaucoma surgery unnecessary. For tumors without orbital invasion and requiring glaucoma management, filtration surgery alternatives such as glaucoma drainage valve implantation should be preceded by ultrasound biomicroscopy (UBM) to confirm the absence of anterior chamber tumor invasion (77). Notably, invasive procedures like intravitreal injections, trabeculectomies, or tube-shunts in eyes with primary or metastatic tumors carry risks of tumor seeding, spread, or exacerbating inflammation; these are relatively contraindicated unless strictly necessary, with priority given to controlling the primary tumor while using conservative IOP-lowering strategies. However, anti-VEGF agents may be administered systemically in such cases, in close collaboration with the treating oncologist, to balance anti-angiogenic effects and tumor management.
II. Eye drops to reduce intraocular pressure, combined with intraocular anti-VEGF agent injections or glucocorticoid injections, based on the specific mechanisms of secondary glaucoma, may be considered (78, 79). Additionally, ultrasound cycloplasty, an emerging non-invasive procedure, or other cylodestructive/cyclomodulation procedures, can be employed when necessary (80).
III. Local excision of eyelid tumors combined with intraoperative cryotherapy can achieve a success rate of 82% in small tumors (< 8mm) while preserving the eyeball (81).
4.3 New progress in multidisciplinary collaboration
I. Remote consultation systems (such as EE - Explorer) can accelerate the initial diagnosis of emergency ocular tumors, especially applicable to cases like retinoblastoma that require urgent intervention (82, 83).
II. Pediatric neuro-ocular tumors require joint assessment by neuro-ophthalmology, as 54% of cases with optic pathway gliomas develop secondary optic nerve atrophy (58).
III. Special precautions: α2 adrenergic agonists (such as brimonidine) can be used as an alternative to prostaglandin analogs in patients with uveal melanoma (UM), and they may be safer (84). Studies have shown that α2 agonists have anti-angiogenic effects (85) and can modulate the tumor microenvironment (86). In contrast, prostaglandin analogs are significantly associated with an increased risk of UM metastasis (87). Additionally, α2-agonists have a comparable effect on intraocular pressure control to other drugs (88). However, it should be noted that there may be neurocognitive risks associated with long-term use (89).
IV. Future directions: ferroptosis-based nanophotosensitizers (such as Mn-doped porphyrin frameworks) have shown precise killing effects on intraocular tumors in animal models (90, 91).
V. Artificial intelligence-assisted pathological diagnosis systems are optimizing the decision-making of targeted therapy for ocular tumors (92).
5 Future prospects
5.1 Tailor strategies to individuals
The therapeutic prospects of tumor-related secondary glaucoma require strategy formulation based on etiologies and risk factors. Studies have shown that the main mechanisms of glaucoma secondary to uveal melanoma are anterior chamber angle narrowing (76% at diagnosis) and neovascularization (71% during follow-up) (50). For high-risk populations, enhanced monitoring of intraocular pressure and neovascularization is necessary. Future research may explore preventive anti-vascular endothelial growth factor injections while optimizing radiotherapy regimens to reduce complications such as angle synechia. Treatment requires multidisciplinary collaboration, integrating tumor control with glaucoma management to balance vision protection and quality of life (93).
5.2 Gene therapy
The prospects of gene therapy for tumor-related secondary glaucoma primarily lie in intervening in the disease process by targeting the regulation of genes related to intraocular pressure or providing neuroprotective effects on retinal ganglion cells. Adeno-associated virus vectors have become a commonly used delivery tool due to their high targeting and low immunogenicity, with preclinical studies showing their ability to reduce intraocular pressure and delay optic nerve degeneration (94, 95). Additionally, the combination of nanomedicine and gene therapy, such as stem cell membrane-coated nanoparticles, can improve drug delivery efficiency, offering new strategies for regulating the tumor microenvironment (96, 97). Although the existing research does not directly focus on therapeutic studies of tumor-related secondary glaucoma, it provides valuable insights into the therapeutic prospects of this condition.
5.3 Integration of multimodal imaging
Optical coherence tomography (OCT) angiography, can be combined with fluorescein angiography to improve the detection rate of neovascularization in the anterior chamber angle, but the limitation of the resolution of deep vessel imaging needs to be addressed (98). Combining liquid biopsy (such as the detection of exosomes in aqueous humor) may enhance the specificity of early diagnosis (50, 99).
5.4 Optimization of AI models
Current deep learning models are mostly based on single-modal data (98). In the future, clinical parameters (such as the intraocular pressure fluctuation curve and tumor volume) and omics data (such as mitochondrial DNA, mtDNA mutations) need to be incorporated to construct a multi-dimensional prediction system (100, 101), and its generalization ability needs to be verified through prospective cohorts.
6 Conclusion
Glaucoma secondary to tumors is a severe complication triggered by primary or metastatic tumors and their treatments, with complex and diverse pathogenic mechanisms. Mechanical obstruction (such as tumor compression of the anterior chamber angle), neovascularization, fibrosis of the TM mediated by inflammatory factors (IL-6, TNF-α), abnormal aqueous humor dynamics, and angle closure induced by either pupillary block or iris/cilliary body invasion are the main pathological mechanisms. The clinical manifestations are heterogeneous, including acute elevation of intraocular pressure, iris neovascularization, choroidal thickening, and characteristic tumor signs. Treatment requires multidisciplinary collaboration, with the control of the primary tumor as the core, combined with anti-VEGF drugs, targeted therapies (such as FGFR4/EZH2 inhibitors), and modified surgeries (such as cryotherapy combined with local excision), while avoiding the use of filtration surgery in active tumors. Future research will focus on personalized therapeutic strategies, gene therapy applications, integration of multimodal imaging, and optimization of AI models, to improve early intervention strategies and reduce the risk of irreversible optic nerve damage.
Author contributions
BL: Software, Methodology, Writing – original draft. D-kL: Methodology, Supervision, Writing – review & editing.
Funding
The author(s) declare that no financial support was received for the research and/or publication of this article.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The authors declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Fang Z, Chen L, Moser M, Zhang W, Qin Z, Zhang B. Electroporation-based therapy for brain tumors: a review. J Biomech Eng. (2021) 143:100802. doi: 10.1115/1.4051184
3. Talaat K, Fathi O, Alamoudi S, Alzahrani M, Mukhtar R, Khan M. Types of glaucoma and associated comorbidities among patients at King Abdulaziz medical city, Jeddah. Cureus. (2021) 13:e15574. doi: 10.7759/cureus.15574
4. Calugaru D, Calugaru M. Etiology, pathogenesis, and diagnosis of neovascular glaucoma. Int J Ophthalmol. (2022) 15:1005–10. doi: 10.18240/ijo.2022.06.20
5. Karaconji T, Zagora S, Grigg J. Approach to childhood glaucoma: a review. Clin Exp Ophthalmol. (2022) 50:232–46. doi: 10.1111/ceo.14039
6. Kaushik S, Senthil S, Gupta V, Balekudaru S, Dubey S, Ali H, et al. Profile of newly diagnosed childhood glaucoma in India: Indian childhood glaucoma study (ICGS) group 1. Ophthalmol Glaucoma. (2024) 7:54–65. doi: 10.1016/j.ogla.2023.07.004
7. Ahmad F, Deshmukh N, Webel A, Johnson S, Suleiman A, Mohan R, et al. Viral infections and pathogenesis of glaucoma: a comprehensive review. Clin Microbiol Rev. (2023) 36:e0005723. doi: 10.1128/cmr.00057-23
8. Hallaj S, Mirza-Aghazadeh-Attari M, Arasteh A, Ghorbani A, Lee D, Jadidi-Niaragh F. Adenosine: the common target between cancer immunotherapy and glaucoma in the eye. Life Sci. (2021) 282:119796. doi: 10.1016/j.lfs.2021.119796
9. Calugaru D, Calugaru M. Intraocular pressure modifications in patients with acute central/hemicentral retinal vein occlusions. Int J Ophthalmol. (2021) 14:931–5. doi: 10.18240/ijo.2021.06.20
10. Venkataraman P, Kisthurmal C, Ramanathan A, Ramamoorthy D, Mohan N, Shroff A. The many faces of secondary angle closure glaucoma: a practical approach to unveil. Indian J Ophthalmol. (2024) 72:1535–6. doi: 10.4103/ijo.Ijo_2905_23
11. Stingl J, Hoffmann E. [Childhood glaucoma]. Klin Monbl Augenheilkd. (2022) 239:929–43. doi: 10.1055/a-1838-5018
12. Rouse B, Le J, Gazzard G. Iridotomy to slow progression of visual field loss in angle-closure glaucoma. Cochrane Database Syst Rev. (2023) 1:Cd012270. doi: 10.1002/14651858.CD012270.pub3
13. Krzyzanowska I, Toteberg-Harms M. [Angle-closure glaucoma]. Ophthalmologie. (2022) 119:1167–79. doi: 10.1007/s00347-022-01745-w
14. Fujimoto T, Inoue-Mochita M, Iraha S, Tanihara H, Inoue T. Suberoylanilide hydroxamic acid (SAHA) inhibits transforming growth factor-beta 2-induced increases in aqueous humor outflow resistance. J Biol Chem. (2021) 297:101070. doi: 10.1016/j.jbc.2021.101070
15. Karimi A, Razaghi R, Rahmati S, Downs J, Acott T, Wang R, et al. Modeling the biomechanics of the conventional aqueous outflow pathway microstructure in the human eye. Comput Methods Programs Biomed. (2022) 221:106922. doi: 10.1016/j.cmpb.2022.106922
16. Karimi A, Halabian M, Razaghi R, Downs J, Kelley M, Acott T. Modeling the endothelial glycocalyx layer in the human conventional aqueous outflow pathway. Cells. (2022) 11:3925. doi: 10.3390/cells11233925
17. Kuo C, Hung C, Tu P, Chen Y. Acute angle-closure related with choroidal metastasis from lung adenocarcinoma: a case report and literature review. Eur J Ophthalmol. (2024) 34:N33–7. doi: 10.1177/11206721231187425
18. Tseng V, Kitayama K, Yu F, Pan D, Coleman A. Social vulnerability, prevalence of glaucoma, and incidence of glaucoma surgery in the california medicare population. Ophthalmol Glaucoma. (2023) 6:616–25. doi: 10.1016/j.ogla.2023.05.005
19. Rao G, Kumar S, Sinha N, Rath B, Pal A. Outcomes of three-piece rigid scleral fixated intraocular lens implantation in subjects with deficient posterior capsule following complications in manual small incision cataract surgery. Heliyon. (2023) 9:e20345. doi: 10.1016/j.heliyon.2023.e20345
20. Midena G, Parrozzani R, Frizziero L, Midena E. Chorioretinal side effects of therapeutic ocular irradiation: a multimodal imaging approach. J Clin Med. (2020) 9:3496. doi: 10.3390/jcm9113496
21. Xiao H, Liu Y, Guo N, Jin L, Wang Z, Lin S, et al. Clinical characteristics of microcystic macular edema in chronic primary angle-closure glaucoma and primary open-angle glaucoma patients. Ophthalmic Res. (2024) 67:145–53. doi: 10.1159/000535900
22. Kose H, Tekeli O. Comparison of microvascular parameters and diagnostic ability of optical coherence tomography angiography between eyes with primary angle closure glaucoma and primary open angle glaucoma. Photodiagnosis Photodyn Ther. (2022) 40:103114. doi: 10.1016/j.pdpdt.2022.103114
23. Senthil S, Dada T, Das T, Kaushik S, Puthuran G, Philip R, et al. Neovascular glaucoma - a review. Indian J Ophthalmol. (2021) 69:525–34. doi: 10.4103/ijo.IJO_1591_20
24. Simha A, Aziz K, Braganza A, Abraham L, Samuel P, Lindsley K. Anti-vascular endothelial growth factor for neovascular glaucoma. Cochrane Database Syst Rev. (2020) 2:Cd007920. doi: 10.1002/14651858.CD007920.pub3
25. Sun, Y, Liu L, Cai N, Liu N. Anti-angiogenic effect of rapamycin in mouse oxygen-induced retinopathy is mediated through suppression of HIF-1alpha/VEGF pathway. Int J Clin Exp Pathol. (2017) 10:10167–75.
26. Lee M, Leskova W, Eshaq R, Harris N. Retinal hypoxia and angiogenesis with methamphetamine. Exp Eye Res. (2021) 206:108540. doi: 10.1016/j.exer.2021.108540
27. Rodrigues M, Kashiwabuchi F, Deshpande M, Jee K, Goldberg M, Lutty G, et al. Expression pattern of HIF-1alpha and VEGF supports circumferential application of scatter laser for proliferative sickle retinopathy. Invest Ophthalmol Vis Sci. (2016) 57:6739–46. doi: 10.1167/iovs.16-19513
28. Rodrigues G, Abe R, Zangalli C, Sodre S, Donini F, Costa D, et al. Neovascular glaucoma: a review. Int J Retina Vitreous. (2016) 2:26. doi: 10.1186/s40942-016-0051-x
29. Yun J, Santina A, Tseng V. Medical and surgical management of neovascular glaucoma. Curr Opin Ophthalmol. (2025): doi: 10.1097/icu.0000000000001151 Online ahead of print.
30. Fu Y, Xin Z. Inhibited corneal neovascularization in rabbits following corneal alkali burn by double-target interference for VEGF and HIF-1alpha. Biosci Rep. (2019) 39:BSR20180552. doi: 10.1042/bsr20180552
31. Subhani S, Vavilala D, Mukherji M. HIF inhibitors for ischemic retinopathies and cancers: options beyond anti-VEGF therapies. Angiogenesis. (2016) 19:257–73. doi: 10.1007/s10456-016-9510-0
32. Aqel S, Hampton J, Bruss M, Jones K, Valiente G, Wu L, et al. Daily moderate exercise is beneficial and social stress is detrimental to disease pathology in murine lupus Nephritis. Front Physiol. (2017) 8:236. doi: 10.3389/fphys.2017.00236
33. Chen J, Tang Y, Wang Y, Cui Q, Inam M, Kong L, et al. Serine protease inhibitor MDSPI16 ameliorates LPS-induced acute lung injury through its anti-inflammatory activity. Int Immunopharmacol. (2020) 88:107015. doi: 10.1016/j.intimp.2020.107015
34. Lin B, Li D. The pivotal role of inflammatory factors in glaucoma: a systematic review. Front Immunol. (2025) 16:1577200. doi: 10.3389/fimmu.2025.1577200
35. Xu S, Zhang F, Wang L, Hao M, Yang X, Li N, et al. Flavonoids of rosa roxburghii tratt offers protection against radiation induced apoptosis and inflammation in mouse thymus. Apoptosis. (2018) 23:470–83. doi: 10.1007/s10495-018-1466-7
36. Mzyk P, Hernandez H, Le T, Ramirez J, McDowell C. Toll-like receptor 4 signaling in the trabecular meshwork. Front Cell Dev Biol. (2022) 10:936115. doi: 10.3389/fcell.2022.936115
37. Hernandez H, Medina-Ortiz W, Luan T, Clark A, McDowell C. Crosstalk between transforming growth factor Beta-2 and toll-like receptor 4 in the trabecular meshwork. Invest Ophthalmol Vis Sci. (2017) 58:1811–23. doi: 10.1167/iovs.16-21331
38. Zong Y, Kamoi K, Ando N, Kurozumi-Karube H, Ohno-Matsui K. Mechanism of secondary glaucoma development in HTLV-1 uveitis. Front Microbiol. (2022) 13:738742. doi: 10.3389/fmicb.2022.738742
39. Patel P, Chen Y, Kasetti R, Maddineni P, Mayhew W, Millar J, et al. Impaired TRPV4-eNOS signaling in trabecular meshwork elevates intraocular pressure in glaucoma. Proc Natl Acad Sci U S A. (2021) 118:e2022461118. doi: 10.1073/pnas.2022461118
40. Song L, Wang Y, Sui Y, Sun J, Li D, Li G, et al. High Interleukin-37 (IL-37) expression and increased mucin-domain containing-3 (TIM-3) on peripheral T cells in patients with rheumatoid arthritis. Med Sci Monit. (2018) 24:5660–7. doi: 10.12659/msm.909254
41. Callaghan B, Lester K, Lane B, Fan X, Goljanek-Whysall K, Simpson D, et al. Genome-wide transcriptome profiling of human trabecular meshwork cells treated with TGF-beta2. Sci Rep. (2022) 12:9564. doi: 10.1038/s41598-022-13573-8
42. Kusunose M, Sakino Y, Noda Y, Daa T, Kubota T. A case of iris melanocytoma demonstrating diffuse melanocytic proliferation with uncontrolled intraocular pressure. Case Rep Ophthalmol. (2017) 8:190–4. doi: 10.1159/000464349
43. Shahidullah M, Delamere N. Mechanical stretch activates TRPV4 and hemichannel responses in the nonpigmented ciliary epithelium. Int J Mol Sci. (2023) 24:1673. doi: 10.3390/ijms24021673
44. Camp D, Yadav P, Dalvin L, Shields C. Glaucoma secondary to intraocular tumors: mechanisms and management. Curr Opin Ophthalmol. (2019) 30:71–81. doi: 10.1097/icu.0000000000000550
45. Swampillai A, Booth A, Cohen V. Ciliary body and iris metastases with anterior chamber angle infiltration: a rare complication from invasive ductal breast cancer. J Glaucoma. (2020) 29:e12–5. doi: 10.1097/ijg.0000000000001445
46. Yao F, Peng J, Zhang E, Ji D, Gao Z, Tang Y, et al. Pathologically high intraocular pressure disturbs normal iron homeostasis and leads to retinal ganglion cell ferroptosis in glaucoma. Cell Death Differ. (2023) 30:69–81. doi: 10.1038/s41418-022-01046-4
47. Feng Y, Wang X, Li P, Shi X, Prokosch V, Liu H. Exogenous hydrogen sulfide and NOX2 inhibition mitigate ferroptosis in pressure-induced retinal ganglion cell damage. Biochim Biophys Acta Mol Basis Dis. (2025) 1871:167705. doi: 10.1016/j.bbadis.2025.167705
48. Huang Y, Ye Z, Li Z. Siderotic glaucoma without detectable intraocular foreign body in a pseudophakic eye: a case report. BMC Ophthalmol. (2020) 20:417. doi: 10.1186/s12886-020-01691-8
49. Zhu J, Chen H, Wu J, Li S, Lin W, Wang N, et al. Ferroptosis in glaucoma: a promising avenue for therapy. Adv Biol. (2024) 8:e2300530. doi: 10.1002/adbi.202300530
50. Stadigh A, Puska P, Kivela T. Incidence and risk factors for secondary glaucoma in eyes with uveal melanoma. Ophthalmol Glaucoma. (2023) 6:29–41. doi: 10.1016/j.ogla.2022.08.002
51. Bowen R, Raval V, Soto H, Singh A. Intraocular tumors: angiographic patterns. Asia Pac J Ophthalmol. (2020) 9:449–60. doi: 10.1097/apo.0000000000000323
52. Huang Y, Lin C, Hsia N, Lai C, Bair H, Tsai Y. Intravitreal aflibercept combined with transpupillary thermotherapy in the treatment of refractory macular edema due to primary uveal melanoma. Taiwan J Ophthalmol. (2022) 12:482–6. doi: 10.4103/tjo.tjo_44_21
53. Gerba-Gorecka K, Romanowska-Dixon B, Karska-Basta I, Cieplinska-Kechner E, Nowak M. Clinical characteristics and management of ocular metastases. Cancers. (2025) 17:1041. doi: 10.3390/cancers17061041
54. Cui Y, Yin S, Qin X, Jiao W, Ren A, Wang F, et al. Advances in the treatment of intraocular malignancies: a literature review. Front Med. (2022) 9:975565. doi: 10.3389/fmed.2022.975565
55. Babst N, Gniesmer S, Sonntag S, Furashova O, Krestanova G, Heindl L, et al. [Clinical approach for solid intraocular metastases]. Ophthalmologie. (2024) 121:352–65. doi: 10.1007/s00347-024-02031-7
56. Yousef Y, Sid Ahmed I, Kanj Ahmad D, Mohammad M, Makahleh H, AlJabari R, et al. Optic disc swelling in cancer patients: etiology and implications. J Clin Med. (2023) 12:7140. doi: 10.3390/jcm12227140
57. Kong J, Park S, Na K. Differences in optic nerve head structure between acute angle-closure glaucoma and open-angle glaucoma. Sci Rep. (2023) 13:7935. doi: 10.1038/s41598-023-35020-y
58. Ivakhnitskaia E, Bhagat D. Neuro-ophthalmic sequelae of pediatric brain tumors. Semin Pediatr Neurol. (2025) 53:101182. doi: 10.1016/j.spen.2025.101182
59. Sukkarieh G, Lejoyeux R, LeMer Y, Bonnin S, Tadayoni R. The role of near-infrared reflectance imaging in retinal disease: a systematic review. Surv Ophthalmol. (2023) 68:313–31. doi: 10.1016/j.survophthal.2022.12.003
60. Pagliara M, Tagliaferri L, Savino G, Fionda B, D’Aviero A, Lanza A, et al. High-dose-rate interstitial brachytherapy (interventional radiotherapy) for conjunctival melanoma with orbital extension. Ocul Oncol Pathol. (2021) 7:199–205. doi: 10.1159/000512344
61. Berry M, Skanchy D, Archer S, Mingardo F, Elner V, Demirci H. Unusual fundus lesion in mosaic neurofibromatosis type 2. Ophthalmic Genet. (2025) 46:211–4. doi: 10.1080/13816810.2025.2458751
62. Gonen B, Hepokur M, Guleser U, Yetik H, Sarici A. Retinal vascular patterns and capillary plexus reflectivity of intraocular tumors; an optical coherence tomography angiography study. Curr Eye Res. (2022) 47:1424–35. doi: 10.1080/02713683.2022.2101666
63. Wang Y, Guo Y, Zhang Y, Huang S, Zhong Y. Differences and similarities between primary open angle glaucoma and primary angle-closure glaucoma. Eye Brain. (2024) 16:39–54. doi: 10.2147/eb.S472920
64. Herranz-Cabarcos A, Alcubierre R, Van der Veen R. Paraneoplastic orbital myositis as a first manifestation of renal cell carcinoma. Orbit. (2024) 43:728–32. doi: 10.1080/01676830.2023.2264916
65. Przezdziecka-Dolyk J, Brzecka A, Ejma M, Misiuk-Hojlo M, Torres Solis L, Solis Herrera A, et al. Ocular paraneoplastic syndromes. Biomedicines. (2020) 8:490. doi: 10.3390/biomedicines8110490
66. Barahman M, Shamsaei G, Kashipazha D, Bahadoram M, Akade E. Paraneoplastic neurological syndromes of small cell lung cancer. Postep Psychiatr Neurol. (2024) 33:80–92. doi: 10.5114/ppn.2024.141157
67. Janssen M, van Broekhoven F, Sillevis Smitt P, Frens M, van der Geest J. The use of eye-movement recording in patients with anti-Hu antibody-associated paraneoplastic neurological syndromes to objectively determine extent and course of disease. Eur J Neurol. (2021) 28:2126–32. doi: 10.1111/ene.14799
68. Castle G, Heath G. Small cell carcinoma presenting as ocular paraneoplastic syndrome due to CRMP-5. Neurosciences. (2020) 25:403–5. doi: 10.17712/nsj.2020.5.20200095
69. Nadal Bosch J, Moya M, Serna S, Cruz R, Malcolm J. When sight and cancer collide: a rare case of paraneoplastic bilateral optic neuritis. Cureus. (2023) 15:e49923. doi: 10.7759/cureus.49923
70. Bocskei Z, Viinikka E, Dormegny L, Bigaut K, Speeg C. [Paraneoplastic ophthalmopathies]. J Fr Ophtalmol. (2022) 45:119–36. doi: 10.1016/j.jfo.2021.08.002
71. Li Y, Gulkas S, Gonzalez JE. ANCA-negative granulomatosis with polyangiitis-like intraocular and cutaneous vasculitis secondary to myelodysplastic syndrome. BMJ Case Rep. (2025) 18:e264723. doi: 10.1136/bcr-2024-264723
72. Janetos T, Volpe N, Simon S. Neuro-ophthalmic manifestations of cancer: a narrative review. Chin Clin Oncol. (2022) 11:25. doi: 10.21037/cco-21-137
73. Yang Y, Zhang Y, Cao J, Su Z, Li F, Zhang P, et al. FGFR4 and EZH2 inhibitors synergistically induce hepatocellular carcinoma apoptosis via repressing YAP signaling. J Exp Clin Cancer Res. (2023) 42:96. doi: 10.1186/s13046-023-02659-4
74. Hartnett M, Stahl A. Laser versus anti-VEGF: a paradigm shift for treatment-warranted retinopathy of prematurity. Ophthalmol Ther. (2023) 12:2241–52. doi: 10.1007/s40123-023-00744-7
75. Ren Y, Mao X, Lin W, Chen Y, Chen R, Sun P. Targeting estrogen-related receptors to mitigate tumor resistance: a comprehensive approach to bridging cellular energy metabolism. Biochim Biophys Acta Rev Cancer. (2025) 1880:189256. doi: 10.1016/j.bbcan.2024.189256
76. Serbest Ceylanoglu K, Guneri Beser B, Singalavanija T, Juntipwong S, Worden F, Demirci H. Targeted therapy and immunotherapy for advanced malignant conjunctival tumors: systematic review. Ophthalmic Plast Reconstr Surg. (2024) 40:18–29. doi: 10.1097/iop.0000000000002488
77. Loda A, Turati M, Semeraro F, Rezzola S, Ronca R. Exploring the FGF/FGFR system in ocular tumors: new insights and perspectives. Int J Mol Sci. (2022) 23:3835. doi: 10.3390/ijms23073835
78. Wang J, Guo Y, Wei J, Min J, Ye L. Comparative efficacy and safety of different anti-VEGF agents combined with different delivery methods for neovascular glaucoma: a systematic review and Bayesian network meta-analysis. BMJ Open. (2024) 14:e080103. doi: 10.1136/bmjopen-2023-080103
79. Tarim B, Kilic M. Ocular side effects of trametinib and dabrafenib: a case report. J Ophthalmic Inflamm Infect. (2023) 13:17. doi: 10.1186/s12348-023-00339-0
80. Ruixue W, Wenjun D, Le J, Fangfang F, Ning L, Xiaoya C, et al. comparative study of ultrasound cycloplasty and endoscopic cyclophotocoagulation in the treatment of secondary glaucoma. Sci Rep. (2023) 13:23073. doi: 10.1038/s41598-023-50157-6
81. Schoelles K, Auw-Haedrich C. Updates on eyelid cancers. Asia Pac J Ophthalmol. (2024) 13:100057. doi: 10.1016/j.apjo.2024.100057
82. Chen J, Wu X, Li M, Liu L, Zhong L, Xiao J, et al. EE-Explorer: a multimodal artificial intelligence system for eye emergency triage and primary diagnosis. Am J Ophthalmol. (2023) 252:253–64. doi: 10.1016/j.ajo.2023.04.007
83. Lema G, De Leacy R, Fara M, Ginsburg R, Barash A, Banashefski B, et al. A remote consult retinal artery occlusion diagnostic protocol. Ophthalmology. (2024) 131:724–30. doi: 10.1016/j.ophtha.2023.11.031
84. Kastelan S, Pavicic A, Pasalic D, Nikuseva-Martic T, Canovic S, Kovacevic P, et al. Biological characteristics and clinical management of uveal and conjunctival melanoma. Oncol Res. (2024) 32:1265–85. doi: 10.32604/or.2024.048437
85. Tanaka M, Inoue Y, Imai T, Tanida N, Takahashi K, Hara H. Guanabenz and clonidine, alpha2-adrenergic receptor agonists, inhibit choroidal neovascularization. Curr Neurovasc Res. (2021) 18:85–92. doi: 10.2174/1567202618666210518133634
86. Zhu J, Naulaerts S, Boudhan L, Martin M, Gatto L, Van den Eynde B. Tumour immune rejection triggered by activation of alpha2-adrenergic receptors. Nature. (2023) 618:607–15. doi: 10.1038/s41586-023-06110-8
87. Pals J, Mensink H, Brosens E, Verdijk R, Naus N, Paridaens D, et al. The effect of intraocular pressure-lowering medication on metastatic uveal melanomas. Cancers. (2021) 13:5657. doi: 10.3390/cancers13225657
88. Kanjee R, Popovic M, Salimi A, Hutnik C, Ahmed I, Saheb H. Prophylaxis against intraocular pressure spikes following uncomplicated phacoemulsification: a systematic-review and meta-analysis. Eye. (2024) 38:1518–28. doi: 10.1038/s41433-024-02940-6
89. Chou C, Lu Y, Weng C, Lin H, Wang I, Jou T, et al. The association between antiglaucomatous agents and Alzheimer’s disease. Eye. (2024) 38:3511–8. doi: 10.1038/s41433-024-03348-y
90. Ning S, Yao Y, Feng X, Tian Y. Recent advances in developing bioorthogonally activatable photosensitizers for photodynamic therapy. Eur J Med Chem. (2025) 291:117672. doi: 10.1016/j.ejmech.2025.117672
91. Lu X, Xue C, Dong J, Zhang Y, Gao F. Nanoplatform-based strategies for enhancing the lethality of current antitumor PDT. J Mater Chem B. (2024) 12:3209–25. doi: 10.1039/d4tb00008k
92. Schlunck G, Boneva S, Wolf J, Schlecht A, Reinhard T, Auw-Hadrich C, et al. Die RNA-Sequenzierung von formalinfixiertem und in Paraffin eingebettetem Gewebe als erganzende Methode der Ophthalmopathologie. [RNA Sequencing of Formalin-Fixed and Paraffin-Embedded Tissue as a Complementary Method in Ophthalmopathology], Klin Monbl Augenheilkd. (2020) 237:860–6. German. doi: 10.1055/a-1187-1590
93. Teixeira-Pinto T, Lima de Souza R, Grossi Marconi D, Lando L. Ophthalmic rehabilitation in oncology care. Can J Ophthalmol. (2025) 60:59–68. doi: 10.1016/j.jcjo.2024.07.004
94. Borras T, Stepankoff M, Danias J. Genes as drugs for glaucoma: latest advances. Curr Opin Ophthalmol. (2024) 35:131–7. doi: 10.1097/icu.0000000000001025
95. Chern K, Nettesheim E, Reid C, Li N, Marcoe G, Lipinski D. Prostaglandin-based rAAV-mediated glaucoma gene therapy in Brown Norway rats. Commun Biol. (2022) 5:1169. doi: 10.1038/s42003-022-04134-w
96. Tan S, Wong T. Drug delivery systems in glaucoma - Current innovations and future perspectives. Asia Pac J Ophthalmol. (2025) 14:100163. doi: 10.1016/j.apjo.2025.100163
97. Zhang W, Huang X. Stem cell membrane-camouflaged targeted delivery system in tumor. Mater Today Bio. (2022) 16:100377. doi: 10.1016/j.mtbio.2022.100377
98. Sun Y, Liu Y, Han Y, Kong F, Zhang Y, Labisi S, et al. Bibliometric analysis of glaucoma-related literature based on SCIE database: a 10-year literature analysis from 2009 to 2018. Int J Ophthalmol. (2020) 13:1998–2006. doi: 10.18240/ijo.2020.12.23
99. Liu K, Yang Y, Wu Z, Sun C, Su Y, Huang N, et al. Remyelination-oriented clemastine treatment attenuates neuropathies of optic nerve and retina in glaucoma. Glia. (2024) 72:1555–71. doi: 10.1002/glia.24543
100. Bulut E, Celebi A, Dokur M, Dayi O. Analysis of trending topics in glaucoma articles from an altmetric perspective. Int Ophthalmol. (2021) 41:2125–37. doi: 10.1007/s10792-021-01770-9
Keywords: cancer-related glaucoma, pathogenesis, clinical manifestation, treatment strategies, multidisciplinary collaboration
Citation: Lin B and Li D-k (2025) From tumor microenvironment to ocular hypertension: unraveling the pathogenesis and therapeutic strategies of cancer-related glaucoma. Front. Med. 12:1628325. doi: 10.3389/fmed.2025.1628325
Received: 30 May 2025; Accepted: 04 August 2025;
Published: 14 August 2025.
Edited by:
Karanjit Kooner, University of Texas Southwestern Medical Center, United StatesReviewed by:
Md. Rizwanullah, Jamia Hamdard University, IndiaOscar Albis-Donado, Instituto Mexicano de Oftalmología IAP, Mexico
Copyright © 2025 Lin and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Dong-kan Li, eG1lY2xka0AxNjMuY29t