Optic Neuropathy and Immune Checkpoint Inhibitors

From EyeWiki

All content on Eyewiki is protected by copyright law and the Terms of Service. This content may not be reproduced, copied, or put into any artificial intelligence program, including large language and generative AI models, without permission from the Academy.


Overview

Immune checkpoint inhibitors (ICI) are a novel treatment against malignancies. ICI work by activating the patient’s immune system to attack cancer cells[1]. Currently, there are three commonly used classes of ICI’s: 1) cytotoxic T-lymphocyte antigen-4 (CTLA-4), 2) programmed death-1 (PD-1), and 3) the PD ligand (PD-L1)[2]. Inhibition of the immune checkpoint however can produce unintended immune-related adverse events (irAEs) and secondary autoimmune disease. We review the epidemiology, pathogenesis, evaluation, management, and prognosis of optic neuropathy as an irAE secondary to ICI.

Epidemiology

The overall incidence of neuro ophthalmic irAE after ICI therapy is low but may occur in up to 0.46% of patients with cutaneous melanoma being the most common ICI treatment indication[1][3]. The frequency of ophthalmic immune-related adverse events (OirAEs) after ICI treatment for melanoma was 341.8/100,000 and 369.6/100,000 in patients with nonmelanoma cancer[4]. Another comparative study of 2,911 patients on ICI for melanoma reported an increased 1-year incidence rate of OirAEs compared to patients with non-melanoma cancer receiving ICI therapy[4]. A 2021 study reported 31 patients receiving one or more ICI including PD-1 inhibitors (e.g., pembrolizumab, nivolumab, or cemiplimab), PD-L1 inhibitors (e.g., atezolizumab, avelumab, or durvalumab), or the CTLA-4 inhibitor ipilimumab[5]. In this study, pembrolizumab was most commonly used ICI followed by combined ipilimumab and nivolumab, and then nivolumab alone[5].

Mechanisms of Action

Cytotoxic T-lymphocyte antigen-4 (CTLA-4) Inhibitors

CTLA-4 molecule is present on T lymphocytes and inhibits T cell activation in response to an antigen[6]. Normally, when presented with an antigen, the major histocompatibility complex (MHC) on dendritic cells interacts with a T cell receptor (TCR) on a T lymphocyte. This receptor interaction serves as an activating signal for an immune response. Another activating signal is initiated when the B7 complex on dendritic cells interacts with the CD28 complex on T cells[6]. With both activating signals, T lymphocytes can be activated to mount an immune response. However, when CTLA-4 binds B7, T lymphocyte activation is inhibited (i.e., immune checkpoint)[6]. Mutations in CTLA-4 genes contribute to the pathogenesis of both autoimmune disease and tumor progression[7]. Thus, CTLA-4 inhibitors block the immune checkpoint and can result in antitumor immunity or an OirAE by potentiating the T lymphocyte response[8].

Programmed death-1 (PD-1) and Programmed death ligand-1 (PDL-1) Inhibitors

The programmed death-1 (PD-1) molecule is an inhibitory receptor present on T lymphocytes that serves as an immune response regulator (i.e., “immune checkpoint”) like CTLA-42[9]. PD-1 on T lymphocytes interacts with programmed death ligand-1 (PDL-1) on hematopoietic and non-hematopoietic cells throughout the body, and this interaction prevents the activation of self-reactive T cells and mediates peripheral immune tolerance[10][11]. However during the development of a tumor, the PD-1 and PDL-1 pathway can be upregulated by malignancies and lead to tumor immune escape by downregulating host immune responses[10]. PD-1 and PDL-1 inhibitors act to disrupt this regulatory mechanism and promote robust host immune responses[10][12].

Lymphocyte activation gene-3 (LAG-3) Inhibitors

The lymphocyte activation gene-3 (LAG-3) molecule is a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells that limits the expansion of activated T cells and the size of the immune memory pool[13]. LAG-3 binds MHC class II on antigen presenting cells, inhibits the activation of its host cell, and promotes a more suppressive immune response[13][14]. Specifically, LAG-3 inhibits cytokine and granzyme production and proliferation and encourages differentiation from cytotoxic T cells to T regulatory cells. LAG-3 inhibitors interfere with another immune regulatory mechanism and are emerging as an important target for immunotherapy in combination with other ICI’s[13].

Currently approved immune checkpoint inhibitors (ICI)[1]

CTLA-4 Inhibitors - Cytotoxic T-Lymphocyte Antigen-4 Inhibitors PD-1 Inhibitors - Programmed Death-1 Inhibitor PD-L1 Inhibitors - Programmed Death-1 Ligand Inhibitor LAG-3 Inhibitors - Lymphocyte Activation Gene-3 Inhibitor
Drug Class Drug Year First Approved
CTLA-4 Inhibitors Ipilimumab 2011[15]
Tremelimumab 2022[16]
PD-1 Inhibitors Pembrolizumab 2014[17]
Nivolumab 2014[18]
Cemiplimab 2018[19]
PD-L1 Inhibitors Atezolizumab 2016[20]
Avelumab 2017[21]
Durvalumab 2017[22]
LAG-3 Inhibitors Relatlimab 2022[23]

Immune check point inhibitors associated Optic Neuropathy

An autoimmune optic neuropathy can occur as an OirAE from ICI. The clinical findings of an optic neuropathy include loss of visual acuity, visual field, and color vision, an ipsilateral relative afferent pupillary defect (RAPD) in unilateral or bilateral but asymmetric, and a swollen, pale, or normal (retrobulbar) optic nerve. A complete eye examination including testing of visual acuity and optic nerve functions is recommended for all patients suspected of having an optic neuropathy.

In a previous systematic review, there were 11 articles out of 331 that identified optic neuropathy due to OirAE from ICI therapy[24]. Four (57%) were men and 3 (43%) were female. The average patient age was 60 years old with a standard deviation of 17.4 years. Of the 7 patients, 2 were treated for malignant melanoma; the other five were treated for metastatic nonsmall cell lung cancer, hodgkin’s lymphoma, metastatic renal cell carcinoma, metastatic prostate cancer, and squamous cell carcinoma of the head and neck . The most common indication for ICI was malignant melanoma[1][3][25]. Of 7 patients, 5 had optic disc edema and not enough information was provided for the other 2 cases. Magnetic resonance imaging (MRI) of the head and orbit with and without contrast was performed in 4 patients, of which 2 had optic nerve enhancement. All patients were treated with corticosteroids. The final visual acuity in affected patients was improved in all 7 patients. Of 7 patients, 4 had complete discontinuation of the ICI, 1 had treatment momentarily halted, and 2 had no change in their ICI treatment despite the development of optic neuropathy.

Case Series for Different ICI-associated Optic Neuropathy

F - Female, M - Male, L - Left, R - Right, OD - Oculus Dexter, OS - Oculus Sinister, ICI Therapy - Immune Checkpoint Inhibitor Therapy, MRI - Magnetic Resonance Imaging, IV - Intravenous, CSF - Cerebrospinal Fluid
Drug Name Patient Info Indication for ICI Therapy Chief Complaint Physical Exam Neuroimaging Treatment
Atezolizumab 82 yr old M Metastatic non-

small cell lung

cancer

- sudden-onset

painless vision

loss

- fundoscopic

exam: R eye pale

macula with a

cherry-red spot,

cotton wool spots,

and a pale and

swollen optic

nerve head with

obscuration of

vessels

- visual acuity: no

light perception in

the R eye

- MRI: not

performed

- IV methylprednisolone followed

by oral prednisolone

Pembrolizumab 67 yr old F Hodgkin’s

Lymphoma

- blurred vision

- visual field

impairment

- altered color

perception

Initial:

- fundoscopic

exam: R eye

papilledema in the

temporal optic

disc,

- visual acuity:

0.05 in R eye

- Visual field

testing: horizontal

visual field loss


Recurrence:

- fundoscopic

exam: L eye

sectorial

papilledema-

visual acuity: 0.4

in L eye

- Visual field

testing: horizontal

visual field loss

Initial:

- MRI: no

abnormalities


Recurrence:

- MRI: increased

CSF signal along

L optic nerve

Initial:

- IV methylprednisolone

followed by oral prednisone

taper over four months


Recurrence:

- IV methylprednisolone,

plasmapheresis,

immunoglobulin treatment,

retrobulbar injection of steroids

and mycophenolate mofetil

followed by six month long steroid taper

Ipilimumab 67 yr old M Metastatic

melanoma

- bilateral blurred

vision

- bilateral

photopsias of

temporal visual

fields

- fundoscopic

exam: mild

bilateral optic disc

edema

- Slit lamp

examination:1+

anterior chamber

cell and posterior

synechia

bilaterally

- Visual acuity:

20/20

- MRI: small

vessel ischemic

changes

- topical

prednisolone

every 2 hours and

atropine drops

27 yr old F Metastatic renal

cell carcinoma

- bilateral vision

loss

- fundoscopic

exam: severe

bilateral optic disc

swelling

- visual acuity:

20/80

- visual field

testing: bilateral

large paracentral

scotoma

- MRI: bilateral T2

hyperintensity of

optic discs and

nerves and 1 small

enhancing

periventricular

lesion

- IV methylprednisolone followed

by oral steroid taper

- plasma

exchange

Durvalumab 65 yr old M Metastatic

prostate cancer

-L inferior

scotoma and

discomfort with

extraocular

movements

-fundoscopic

exam: 4+ disc

edema in L eye

- visual field

testing: near

complete central

sparing inferior

defect in L eye

- MRI: no

abnormalities

- IV high dose

corticosteroids

Nivolumab 64 yr old M Malignant

melanoma

-visual field

impairment

- intermediate

uveitis with

bilateral papillitis

and acute anterior

ischemic optic

neuropathy of L

eye

-information not

included in review

- no treatment

administered

Cemiplimab 50 yr old F Squamous cell

carcinoma of the

head and neck

-visual impairment -visual field

testing: OD:

20/30

OS: 20/100

-information not

included in review

- Methylprednisolon e and prednisone

Atezolizumab-associated arteritic anterior ischemic optic neuropathy (AION)

An 82 year old male with metastatic non-small cell lung cancer was treated with the PD-L1 agent, atezolizumab and developed an optic neuropathy consistent with arteritic AION[26]. The patient initially presented to the emergency department with a 1-week history of sudden-onset painless vision loss associated with frontal headaches and jaw claudication[26]. On exam, there was a diffusely swollen optic disc in the right eye (OD), and RAPD OD, and delayed choroidal and retinal arterial filling on fundus fluorescein angiography OD.

Pembrolizumab-associated optic neuropathy

A patient with Hodgkin lymphoma was treated with pembrolizumab and developed a unilateral optic neuropathy OD 19 weeks after initiation of treatment[27]. Despite cessation of pembrolizumab contralateral involvement occurred in the left eye (OS), 7 months later[27].

Ipilimumab-associated optic neuropathy

There are multiple reported cases of ipilimumab-associated optic neuropathy[25]. A 67-year-old man with stage III metastatic melanoma treated with ipilimumab monotherapy developed bilateral optic disc edema after his third ICI infusion[28]. Another case reported a 27-year-old woman treated with ipilimumab for metastatic renal cell carcinoma who also developed bilateral optic neuropathy 5 weeks after her last infusionu[29].

Durvalumab-associated optic neuropathy

A 65-year-old male with metastatic prostate cancer was treated with durvalumab and developed unilateral optic neuropathy of his left eye[30]. He developed acute scotoma in the left eye with grade 4 optic nerve edema and near complete inferior altitudinal defect seen on visual field testing[30].

Nivolumab-associated optic neuropathy

A 64-year-old male was treated with nivolumab for metastatic melanoma and consequently developed intermediate uveitis with bilateral papillitis and acute anterior ischemic optic neuropathy of his left eye[31]. He also had concurrent hypertensive meningitis, another irAE[31].

Cemiplimab-associated optic neuropathy

A 50-year-old female diagnosed with squamous cell carcinoma of the head and neck was treated with cemiplimab[5]. She subsequently developed bilateral optic neuropathy with visual field deficits[5].

Diagnosis & Management

The Common Terminology Criteria for Adverse Events (CTCAE) Version 5, published by the US Department of Health and Human Services has four grades for patients with ocular toxicity from medications[1][9]. There are currently no formal treatment recommendations for neuro-ophthalmic or OirAE associated with ICIs. The Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group has suggested general guidelines for management of immune related adverse events by grade[1][32].

Although corticosteroids are the mainstay of presumed OirAE after ICI[25][29][30][33][34] the decision for discontinuation or dose reduction of the ICI treatment should be decided on a case by case basis in a multidisciplinary team approach to weigh the risks and benefits of ICI treatment[30].

Diagnosis and Management Guidelines[1]

ICI -Immune Checkpoint Inhibitor, ADL - Activities of Daily Living
Grade Description Management
Grade 1 Mild toxicity (patients may be asymptomatic,

but have clinically detectable findings)

Typically, do not require corticosteroid

administration or cessation of ICI

Grade 2 Moderately symptomatic, which may interfere

with ADL’s and with visual acuity of 20/40 or

better (or loss of 3 lines or fewer from

baseline)

Might consider temporary suspension of ICI

and initiation of systemic corticosteroids

(either intravenous or oral). Consider resuming

ICI once symptoms have improved to grade 1

Grade 3 Decrease in vision (worse than 20/40, or more

than 3 lines decreased from baseline, but

better than 20/200), limiting activities of daily

living, severe pain, and visual field defects

Warrant consideration of suspension of ICI

and cessation if symptoms haven’t resolved

within 4 – 6 weeks, as well as systemic

steroids

Grade 4 Visual acuity equivalent to or worse than

20/200

Typically discontinue ICI and give systemic

steroids

Rituximab

Rituximab is a chimeric CD-20 monoclonal antibody classically used for relapsed or chemorefractory low-grade or follicular non-Hodgkin's lymphoma[35]. Rituximab direct mechanisms of action include complement-mediated cytotoxicity and antibody-dependent cell-mediated cytotoxicity[35]. Indirectly, rituximab can induce apoptosis and sensitize cancer cells to chemotherapy[35]. As a monoclonal antibody, Rituximab has also shown significant improvement in the management of ICI related cutaneous, musculocutaneous, neurological, and hematologic adverse events[35][36][37].

Cutaneous adverse effects presenting in patients treated with ICI therapy bullous dermatoses like bullous pemphigoid, other autoimmune bullous dermatoses or bullous drug reaction[35][36]. One study showed that in seven patients with corticosteroid-refractory immune related cutaneous adverse events (ircAEs), rituximab lead to significant improvement of ircAEs[38]. Another study showed that rituximab was associated with ircAE improvement of ≥2 grades in every patient treated[36]. Additionally, rituximab can provide significant relief for musculoskeletal irAEs like primary myositis, however treatment must be carefully monitored due to rituximab’s long duration of action[35]. Furthermore, for treatment refractory neurologic irAEs including autoimmune encephalopathy and myasthenia gravis, rituximab may be considered alongside plasmapharesis[35]. Finally, many dangerous hematologic irAEs including CNS hemorrhage, thrombosis/embolism, or renal failure may be treated with rituximab if there is no improvement or worsening while on corticosteroids[35].

Atezolizumab-associated Arteritic anterior ischemic optic neuropathy

For this specific case study, after the initial diagnosis of AAION was made, high-dose corticosteroid therapy was initiated (intravenous methylprednisolone followed by oral prednisolone[26] and atezolizumab was discontinued.

Pembrolizumab-associated optic neuropathy

In this report, when the right eye was affected, pembrolizumab treatment was halted and high-dose steroid treatment was initiated (intravenous methylprednisolone for five days), followed by a prednisone taper over four months[27]. 7 months later when the left eye was affected, high-dose steroids (intravenous methylprednisolone), plasmapheresis, immunoglobulin treatment, retrobulbar injection of steroids and mycophenolate mofetil helped treat symptoms[27]. All of these treatment modalities were then followed with a six month long steroid taper[27].

Ipilimumab-associated bilateral optic neuropathy

For the 67-year-old patient, initial symptoms were treated with topical prednisolone and atropine drops in both eyes, and treatment with ipilimumab was continued[28]. Further exacerbations in optic neuropathy were treated with topical prednisolone every 2 hours in both eyes. Systemic corticosteroids were considered but administered due to patient preference[28]. Another 27-year-old woman with metastatic renal cell carcinoma was given intravenous methylprednisolone (1g per day for 3 days), followed by an oral steroid taper (1mg per kg per day) and plasma exchange (10 sessions)[29]. Ipilimumab treatment was halted for six months and resumed with addition of nivolumab.

Durvalumab-associated optic neuropathy

After the diagnosis of ICI related optic neuropathy was made, the patient was treated with IV high dose corticosteroids and durvalumab treatments were not terminated[30]. The patient endorsed significant improvement of his visual deficits after steroid treatment[30].

Nivolumab-associated optic neuropathy

After the patient was diagnosed with unilateral uveitis and optic neuropathy, nivolumab treatment was discontinued, and no other treatments were administered[31]. The patients had transient worsening of symptoms with eventual partial remission[31].

Cemiplimab-associated optic neuropathy

Once the patient was diagnosed with bilateral optic neuropathy, cemiplimab was held and high dose IV methylprednisolone at up to 1 gm/day for 1–7 days was administered followed by oral tapering over a period of several months[5]. Significant improvement of visual symptoms was noted[5].

Prognosis

Since OirAEs have a relatively low frequency, long term complications and prognosis are still unknown[1][39]. However, sources agree that most OirAEs can be well controlled or resolved with topical or systemic corticosteroids[33][40].

Conclusion

Clinicians should be aware that optic neuropathy can occur unilaterally or bilaterally in patients treated with ICI as an OirAE. Neuroimaging studies are recommended to exclude alternative etiologies for the optic neuropathy including metastatic, radiation induced, or paraneoplastic etiologies. Unfortunately, there is no current biomarker for OirAE and the condition is a diagnosis of exclusion. Discontinuation or dose reduction of the ICI should be decided via a multidisciplinary approach including ophthalmology and oncology. Treatment with corticosteroids is recommended but the dose, route, and duration remain ill defined. The use of rituximab or other B-cell depletion strategy as an immune countermeasure to OirAE due to ICI likewise remains controversial. Further research is needed to determine the precise treatment and prognosis for these patients.

References

  1. 1.0 1.1 1.2 1.3 1.4 1.5 1.6 1.7 Neuro Ophthalmic Complications of Immune Checkpoint Inhibitors. EyeWiki. https://eyewiki.org/Neuro_Ophthalmic_Complications_of_Immune_Checkpoint_Inhibitors
  2. Lentz RW, Colton MD, Mitra SS, Messersmith WA. Innate Immune Checkpoint Inhibitors: The Next Breakthrough in Medical Oncology?. Mol Cancer Ther. 2021;20(6):961-974. doi:10.1158/1535-7163.MCT-21-0041
  3. 3.0 3.1 Yu CW, Yau M, Mezey N, Joarder I, Micieli JA. Neuro-ophthalmic Complications of Immune Checkpoint Inhibitors: A Systematic Review. Eye Brain. 2020;12:139-167. Published 2020 Nov 3. doi:10.2147/EB.S277760
  4. 4.0 4.1 Braun D, Getahun D, Chiu VY, et al. Population-Based Frequency of Ophthalmic Adverse Events in Melanoma, Other Cancers, and After Immune Checkpoint Inhibitor Treatment. Am J Ophthalmol. 2021;224:282-291. doi:10.1016/j.ajo.2020.12.013
  5. 5.0 5.1 5.2 5.3 5.4 5.5 Sun MM, Seleme N, Chen JJ, et al. Neuro-Ophthalmic Complications in Patients Treated With CTLA-4 and PD-1/PD-L1 Checkpoint Blockade. J Neuroophthalmol. 2021;41(4):519-530. doi:10.1097/WNO.0000000000001148
  6. 6.0 6.1 6.2 Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood. 2018;131(1):58-67. doi:10.1182/blood-2017-06-741033
  7. Hosseini A, Gharibi T, Marofi F, Babaloo Z, Baradaran B. CTLA-4: From mechanism to autoimmune therapy. Int Immunopharmacol. 2020;80:106221. doi:10.1016/j.intimp.2020.106221
  8. Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J Clin Oncol. 2015;33(17):1974-1982. doi:10.1200/JCO.2014.59.4358
  9. 9.0 9.1 Common Terminology Criteria for Adverse Events (CTCAE). Version 5.; 2017
  10. 10.0 10.1 10.2 Jiang Y, Chen M, Nie H, Yuan Y. PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations. Hum Vaccin Immunother. 2019;15(5):1111-1122. doi:10.1080/21645515.2019.1571892
  11. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18(3):153-167. doi:10.1038/nri.2017.108
  12. Zhang N, Tu J, Wang X, Chu Q. Programmed cell death-1/programmed cell death ligand-1 checkpoint inhibitors: differences in mechanism of action. Immunotherapy. 2019;11(5):429-441. doi:10.2217/imt-2018-0110
  13. 13.0 13.1 13.2 Sauer N, Szlasa W, Jonderko L, et al. LAG-3 as a Potent Target for Novel Anticancer Therapies of a Wide Range of Tumors. Int J Mol Sci. 2022;23(17):9958. Published 2022 Sep 1. doi:10.3390/ijms23179958
  14. Li Y, Ju M, Miao Y, Zhao L, Xing L, Wei M. Advancement of anti-LAG-3 in cancer therapy. FASEB J. 2023;37(11):e23236. doi:10.1096/fj.202301018R
  15. Yervoy (Ipilimumab) [Package Insert]. Bristol Myers Squibb; 2020
  16. IMJUDO® (tremelimumab-actl) [Prescribing Information]. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2023
  17. Keytruda (pembrolizumab) [package insert]. Merck Sharp Dohme; 2021
  18. Opdivo (nivolumab) [package insert]. Bristol Myers Squibb; 2015
  19. Libtayo (cemiplimab) [package insert]. Regeneron Pharmaceuticals; 2021
  20. Tecentriq (atezolizumab) [package insert]. Genentech Inc; 2016
  21. Bavencio (avelumab) [package insert]. EMD Serono Inc; 2017
  22. Imfinzi (durvalumab) [package insert]. Astrazeneca UK Ltd; 2021
  23. Opdualag [package insert]. Princeton, NJ: Bristol-Myers Squibb Company; 2022
  24. Pietris, J., Santhosh, S., Ferdinando Cirocco, G., Lam, A., Bacchi, S., Tan, Y., Gupta, A. K., Kovoor, J. G., & Chan, W. (2023). Immune Checkpoint Inhibitors and Optic Neuropathy: A Systematic Review. Seminars in ophthalmology, 38(6), 547–558. https://doi.org/10.1080/08820538.2023.2168494
  25. 25.0 25.1 25.2 Dalvin LA, Shields CL, Orloff M, Sato T, Shields JA. CHECKPOINT INHIBITOR IMMUNE THERAPY: Systemic Indications and Ophthalmic Side Effects. Retina. 2018;38(6):1063-1078. doi:10.1097/IAE.0000000000002181
  26. 26.0 26.1 26.2 Berry EC, Mullany S, Quinlivan A, et al. Eosinophilic Vasculitis and Arteritic Anterior Ischemic Optic Neuropathy Associated With Anti-PD-L1 Therapy. J Immunother. 2022;45(1):51-55. doi:10.1097/CJI.0000000000000394
  27. 27.0 27.1 27.2 27.3 27.4 Daetwyler E, Zippelius A, Meyer P, Läubli H. Pembrolizumab-induced optic neuropathy - a case report. Front Immunol. 2023;14:1171981. Published 2023 May 9. doi:10.3389/fimmu.2023.1171981
  28. 28.0 28.1 28.2 Yeh OL, Francis CE. Ipilimumab-associated bilateral optic neuropathy. J Neuroophthalmol. 2015;35(2):144-147. doi:10.1097/WNO.0000000000000217
  29. 29.0 29.1 29.2 Boisseau W, Touat M, Berzero G, et al. Safety of treatment with nivolumab after ipilimumab-related meningoradiculitis and bilateral optic neuropathy. Eur J Cancer. 2017;83:28-31. doi:10.1016/j.ejca.2017.05.036
  30. 30.0 30.1 30.2 30.3 30.4 30.5 Noble CW, Gangaputra SS, Thompson IA, et al. Ocular Adverse Events following Use of Immune Checkpoint Inhibitors for Metastatic Malignancies. Ocul Immunol Inflamm. 2020;28(6):854-859. doi:10.1080/09273948.2019.1583347
  31. 31.0 31.1 31.2 31.3 Chaudot F, Sève P, Rousseau A, et al. Ocular Inflammation Induced by Immune Checkpoint Inhibitors. J Clin Med. 2022;11(17):4993. Published 2022 Aug 25. doi:10.3390/jcm11174993
  32. Puzanov I, Diab A, Abdallah K, et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. Journal for ImmunoTherapy of Cancer. 2017;5(1). doi:10.1186/s40425-017-0300-z
  33. 33.0 33.1 Fortes BH, Liou H, Dalvin LA. Ophthalmic adverse effects of immune checkpoint inhibitors: the Mayo Clinic experience. Br J Ophthalmol. 2021;105(9):1263-1271. doi:10.1136/bjophthalmol-2020-316970
  34. Wladis EJ, Kambam ML. Ophthalmic complications of immune checkpoint inhibitors. Orbit. 2022;41(1):28-33. doi:10.1080/01676830.2020.1867192
  35. 35.0 35.1 35.2 35.3 35.4 35.5 35.6 35.7 Cerny, T., Borisch, B., Introna, M., Johnson, P., & Rose, A. L. (2002). Mechanism of action of rituximab. Anti-cancer drugs, 13 Suppl 2, S3–S10. https://doi.org/10.1097/00001813-200211002-00002
  36. 36.0 36.1 36.2 Mital, R., Otto, T. S., Savu, A., Baumrin, E., Cardones, A. R., Carlesimo, M., Caro, G., Freites-Martinez, A., Hirner, J. P., Markova, A., McLellan, B. N., Rossi, A., Sauder, M. B., Seminario-Vidal, L., Sibaud, V., Owen, D. H., Dulmage, B. O., Chen, S. T., & Kaffenberger, B. H. (2023). Detection of novel therapies using a multi-national, multi-institutional registry of cutaneous immune-related adverse events and management. International journal of dermatology, 62(8), 1020–1025. https://doi.org/10.1111/ijd.16714
  37. Perdigoto, A. L., Kluger, H., & Herold, K. C. (2021). Adverse events induced by immune checkpoint inhibitors. Current opinion in immunology, 69, 29–38. https://doi.org/10.1016/j.coi.2021.02.002
  38. Phillips, G. S., Wu, J., Hellmann, M. D., Postow, M. A., Rizvi, N. A., Freites-Martinez, A., Chan, D., Dusza, S., Motzer, R. J., Rosenberg, J. E., Callahan, M. K., Chapman, P. B., Geskin, L., Lopez, A. T., Reed, V. A., Fabbrocini, G., Annunziata, M. C., Kukoyi, O., Pabani, A., Yang, C. H., … Lacouture, M. E. (2019). Treatment Outcomes of Immune-Related Cutaneous Adverse Events. Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 37(30), 2746–2758. https://doi.org/10.1200/JCO.18.02141
  39. Vishnevskia-Dai V, Rozner L, Berger R, et al. Ocular side effects of novel anti-cancer biological therapies. Sci Rep. 2021;11(1):787. Published 2021 Jan 12. doi:10.1038/s41598-020-80898-7
  40. Mazharuddin AA, Whyte AT, Gombos DS, et al. Highlights on Ocular Toxicity of Immune Checkpoint Inhibitors at a US Tertiary Cancer Center. J Immunother Precis Oncol. 2022;5(4):98-104. Published 2022 Nov 30. doi:10.36401/JIPO-22-14
The Academy uses cookies to analyze performance and provide relevant personalized content to users of our website.