Next Article in Journal
Examining the Acceptability and Feasibility of the Compassionate Mindful Resilience (CMR) Programme in Adult Patients with Chronic Kidney Disease: The COSMIC Study Protocol
Next Article in Special Issue
Interventions within the Scope of Occupational Therapy in the Hospital Discharge Process Post-Stroke: A Systematic Review
Previous Article in Journal
Improving Pharmacists’ Awareness of Inadequate Antibiotic Use for URTIs through an Educational Intervention: A Pilot Study
Previous Article in Special Issue
Using the Concept of Health Literacy to Understand How People Living with Motor Neurone Disease and Carers Engage in Healthcare: A Longitudinal Qualitative Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Optical Coherence Tomography and Optical Coherence Tomography with Angiography in Multiple Sclerosis

by
Ioannis-Nikolaos Chalkias
1,†,
Christos Bakirtzis
2,*,†,
Demetrios Pirounides
1,
Marina Kleopatra Boziki
2 and
Nikolaos Grigoriadis
2
1
Department of Ophthalmology, AHEPA University Hospital, 54626 Thessaloniki, Greece
2
Multiple Sclerosis Center, Second Department of Neurology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Healthcare 2022, 10(8), 1386; https://doi.org/10.3390/healthcare10081386
Submission received: 29 May 2022 / Revised: 14 July 2022 / Accepted: 22 July 2022 / Published: 25 July 2022
(This article belongs to the Special Issue Prevention, Intervention, and Care of Neurodegerative Diseases)

Abstract

:
Multiple sclerosis (MS) is an inflammatory and neurodegenerative, potentially disabling disease of the central nervous system. OCT (Optical Coherence Tomography) and OCT-A (Optical Coherence Tomography with Angiography) are imaging techniques for the retina and choroid that are used in the diagnosis and monitoring of ophthalmological conditions. Their use has recently expanded the study of several autoimmune disorders, including MS. Although their application in MS remains unclear, the results seem promising. This review aimed to provide insight into the most recent OCT and OCT-A findings in MS and may function as a reference point for future research. According to the current literature, the retinal nerve fibre layer (RNFL) and ganglion cell-inner plexiform complex (GC-IPL) are significantly reduced in people with MS and are inversely correlated with disease duration. The use of OCT might help distinguish between MS and neuromyelitis optica spectrum disorders (NMOSD), as the latter presents with more pronounced thinning in both the RNFL and GC-IPL. The OCT-A findings in MS include reduced vessel density in the macula, peripapillary area, or both, and the enlargement of the foveal avascular zone (FAZ) in the setting of optic neuritis. Additionally, OCT-A might be able to detect damage in the very early stages of the disease as well as disease progression in severe cases.

1. Introduction

Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system (CNS) characterised by inflammation, demyelination, and extensive axonal loss [1]. Although certain factors, such as Epstein–Barr virus (EBV) infection, smoking, childhood obesity, low levels of vitamin D, and ultraviolet B light (UVB) exposure, have been implicated in the pathogenesis of the disease, the definite underlying cause remains uncertain [2].
Visual symptoms and optic neuritis (ON) are the most common ocular manifestations, occurring in up to 25% of people with MS (pwMS) [3]. Post-mortem studies have shown that 90% of pwMS present degeneration and axonal loss in their optic nerves, regardless of ON history [4,5].
Given that the diagnosis of MS relies on a combination of clinical and magnetic resonance imaging (MRI) findings [6], an expensive and time-consuming method, the introduction of sensitive, low-cost biomarkers that can be used in the screening and monitoring of pwMS remains of vital importance.
The introduction of two commonly used ophthalmic imaging modalities, optical coherence tomography (OCT), and the more recent OCT with angiography (OCT-A) has revolutionised our understanding of the underlying pathophysiological mechanisms in MS, as they can indirectly provide information about the CNS. The imaging techniques OCT and OCT-A are fast, non-invasive, and inexpensive and provide anatomical and microvascular cross-sectional images of the retina, respectively. Their main application in ophthalmology is in the diagnosis and monitoring of diseases, such as age-related macular degeneration, retinal vascular disorders, and glaucoma. However, given that the retina is an extension of the brain, its use has expanded in the study of neurological and neuro-ophthalmological conditions [7].
The ganglion cells of the retina are located in the ganglion cell layer (GCL) and their axons form the retinal nerve fibre layer (RNFL). Both layers can be easily assessed using the aforementioned techniques, thus allowing the evaluation of potential neuronal and axonal degeneration in people.
This article aims to review the current OCT and OCT-A findings in pwMS and highlights the importance of ophthalmologic examination in the monitoring and progression of this disease.

2. Optical Coherence Tomography in Multiple Sclerosis

The imaging technique OCT was first introduced in 1991 and uses light waves to obtain cross-sectional retinal images. The RNFL and the GCL-inner plexiform layer complex (GC-IPL) are the two most commonly studied layers in MS. According to the current literature, the thickness of the RNFL and GC-IPL is found reduced in pwMS, and there is an inverse relationship with the disease duration [8,9,10]. Despite several theories, the pathophysiological mechanisms underlying these findings remain unclear [11].
Furthermore, ON is the most typical ocular manifestation, characterized by gaze-evoked retro-orbital pain and reduced visual acuity, and occurs in a significant number of pwMS [12]. Retrograde degeneration leading to axonal loss is the most probable pathological mechanism causing RNFL and GC-IPL thinning. Nevertheless, in patients with no history of ON, these findings could be either due to primary degeneration caused by the disease itself or even to retrograde degeneration after one or more subclinical episodes of ON [13].
Since MRI is the most common imaging modality used in the diagnosis of MS, many studies have focused on associating the relevant findings with those of OCT [14,15,16]. In fact, in a study by Saidha et al., GC-IPL atrophy mirrored atrophy in the white matter, brainstem, grey matter, thalamus, and the whole brain. However, a recent study by Glasner et al. suggested no correlation between white matter plaques and RNFL or GC-IPL thickness [17]. Nevertheless, grey matter atrophy seems to be the most sensitive marker of progressive MS [14]. Additionally, studies have correlated the progression of GC-IPL thinning with the development of new T2 lesions [10], damage to the optic radiations [18], and the formation of contrast-enhancing lesions [14]. One study showed that there was greater thinning in the temporal RNFL in patients with newly formed lesions in the optic radiations [19].
Several studies suggest that OCT may be used as a marker for estimating disability [20,21]. More specifically, RNFL and GC-IPL thinning were independently associated with long-term disability worsening in a recent study by Lambe et al. [22]. These findings were recently supported by Skirkova et al., who found a correlation between peripapillary RNFL values, disability, and brain MRI volumetric parameters [23]. The risk of progression can be three times higher in patients with initial RNFL thickness < 88 μm [24].
Visual acuity and contrast sensitivity are affected in patients with ON and several studies have managed to show that there are in accordance with the OCT findings [8,25,26]. GC-IPL and RNFL thinning parallel low-contrast visual acuity, and according to Lampert et al., dyschromatopsia and GC-IPL thickness are interconnected in pwMS and no history of ON [27].
Thinning of the RNFL and GC-IPL is not specific to MS; however, OCT might be helpful in differentiating it from disorders of the same spectrum. In neuromyelitis optica spectrum disorders (NMOSD), RNFL thinning tends to be more evenly distributed, whereas the temporal quadrant seems to be more affected in patients with ON MS [28]. In addition, both RNFL and GC-IPL are much more severely reduced in NMOSD than in MS. According to Brandt et al., OCT parameters may be valuable in distinguishing MS from Susac syndrome, a rare condition characterized by the triad of branch retinal artery occlusion (BRAO), encephalopathy, and hearing loss [29]. Research has shown that in patients with Susac syndrome, OCT displays scar-like pathological patterns that are strictly confined to the inner retinal layers, sparing the outer nuclear layer and the photoreceptors, thus clearly differentiating it from MS [30].
In 2008, Toledo et al. were the first to correlate OCT findings with cognitive impairment. Researchers have linked reduced RNFL thickness to cognitive disability as measured by the symbol digit modality test, a gold standard measure of information processing speed [31]. These findings were later supported by Sedighi et al., who showed that only 20% of pwMS with cognitive impairment had normal OCT findings [32]. In another study, Coric et al. noted that cognitively impaired pwMS with no history of ON had significantly reduced mean peripapillary RNFL (pRNFL) and mean macular GC-IPL (mGC-IPL) thicknesses compared to cognitively healthy pwMS [33]. Although these results may seem promising, further studies are required to support these findings.
Moreover, OCT has been used to study responses to different therapeutic options. In 2016, Pul et al. found no significant association between interferon beta (IFNβ-1b) treatment and RNFL thinning [34]. However, when Button et al. compared the effect of glatiramer acetate, natalizumab, and interferon-β-1a with the rate of GC-IPL thinning, they found that the natalizumab-treated group had a significantly lower rate of GC-IPL reduction compared to the other two [35].
Visual evoked potentials (VEP) are electrical signals generated by the visual cortex of the occipital lobe in response to visual stimuli. Klistorner et al. studied the relationship between OCT parameters and multifocal VEP (mfVEP) in patients with acute ON and found a significant reduction in RNFL thickness and mean mfVEP, particularly in the temporal quadrant [36]. Additionally, in an earlier study, researchers associated the VEP amplitude with the loss in RNFL thickness in patients with ON and noted that the main mechanism behind this loss is axonal degeneration [37].
One study examined the pupillary light response in patients with MS and found that attenuation of the melanopsin-mediated sustained pupillary constriction response was significantly associated with thinning of the GCL-IPL in pwMS with a history of ON [38].
A recent paper described peripapillary hyper-reflective ovoid mass-like structures (PHOMS) as a novel OCT finding that might be noted in people with early MS. Their presence suggests disease progression; however, more research is needed to support these results [39].
In recent years, artificial intelligence (AI) and machine learning have proven useful in medicine. In a recent study, researchers developed a system based on a convolutional neural network that can classify the disease according to the thickness of the OCT scans, thus assisting in the early diagnosis of the disorder [40]. Machine learning has also been successfully used to predict disability progression in pwMS by analysing RNFL thickness [41].

3. Optical Coherence Tomography with Angiography in Multiple Sclerosis

First introduced in 2014, the novel non-invasive imaging modality OCT-A complements its predecessor, OCT, by allowing the visualization of the retinal and choroidal microvasculature. Despite the need for further research for OCT-A to be integrated into clinical practice, current data suggest that it is more sensitive at the early stages of the disease, as well as detecting damage progression in advanced stages, compared to OCT. The role of hypoxia in MS has already been established [42,43], with studies observing decreased blood flow in the retinal microcirculation in people with relapsing-remitting MS [44,45]. Therefore, OCT-A, with its ability to document microvascular changes, may further supplement our understanding of this disorder.
Although more studies are needed to support these results, all current data suggest a reduced vessel density (VD) in the macular, peripapillary, or both areas in patients with MS [46,47,48,49,50]. In fact, in a large study by Murphy et al., OCT-A findings were correlated with the visual function and disability status of pwMS [46]. They also showed a significant reduction in the superficial vascular plexus (SVP) density in the eyes with MS but not in the deep vascular plexus (DVP). However, one study found significant DVP density reduction in MS compared to controls [47], while another study found a reduction only in pwMS with a history of ON, as compared to those without history of ON [48]. One recent study failed to find any significant differences in SVP VD between pwMS and controls but showed discrepancies between the two groups in the RNFL, GC-IPL, and radial peripapillary capillary (pRPC) VD [51]. Thinning of the SVP and enlargement of the FAZ appear to progress rapidly after an episode of ON and are associated with a persistent visual disability [52].
Research has also focused on measuring the VD around the optic nerve, with data suggesting a significant reduction in pwMS [53,54]. The quadrant that is most severely affected remains ambiguous, with one study suggesting the inferior and nasal quadrant [49], while another study points to the temporal quadrant [47].
The FAZ has also been used in the study of MS, and although Yilmaz et al. found no differences between the area and perimeter of the FAZ between pwMS and controls, its size was negatively related to the VD of the SVP and DVP [47].
Interestingly, in a longitudinal study with 50 stable pwMS and one-year follow-up, researchers found a slight increase in parafoveal VD, a finding that could potentially point to vascular regeneration in chronic MS, while the pRNFL and GC-IPL thickness remained unchanged [55]. However, further studies are required to support these findings.
Additionally, OCT-A has been used as a marker for disability. Higher levels of disability, as measured by the expanded disability status scale (EDSS), are related to a lower VD of the macular SVP and low-contrast visual acuity in pwMS [46]. It is worth mentioning that one study associated higher choriocapillary VDs with ongoing inflammatory disease activity [48]. Finally, Jiang et al. demonstrated lower volumetric VD (VVD), a newly developed metric system, in the retinal vascular network and DVP in pwMS with ON, and found a positive correlation between VVD of the macular SVP and EDSS and disease duration [50].
Promising results have recently emerged regarding the use of OCT-A for differentiating MS from NMOSD. There was a reduction in peripapillary and macular VD and FAZ size in people with NMOSD-ON compared to relapsing-remitting MS and controls in a study by Tiftikcioglu et al. [56]. An earlier study suggested that the best OCTA discriminant between NMOSD and MS is the VD reduction pattern, that is, the inferior to nasal (I/N) and I/T ratios for ON eyes and S/T and N/T ratios for non-ON eyes [57]. The combination of OCT and OCT-A parameters can also potentially differentiate the two conditions, as patients with NMOSD have a significantly lower average thickness of the pRNFL and GC-IPL and significantly smaller whole VD and perfusion density areas than those with MS [58].
Studies have also identified OCT-A as a potential tool for monitoring disease progression. Recent data suggest that marked vascular loss in the SVP, DVP, and pRPC may be observed in people with an initial demyelinating event, although no changes might occur in the RNFL, GC-IPL thickness, or clinical and radiological examinations [59].
Lastly, one study correlated OCT-A values with VEP measurements. They showed that there was significant agreement between low optic nerve head VD and RPC VD and prolonged VEP P100 [60].

4. Discussion

The OCT and OCT-A techniques are low-cost, easy-to-use, non-invasive imaging modalities that have recently expanded their use beyond their sole ophthalmologic role. As OCT has existed for more years than OCT-A, it has allowed scientists to experiment more with it and reach more conclusive results regarding the diagnosis and monitoring of MS.
In general, patients with MS exhibit RNFL and GC-IPL thinning, which are inversely related to disease duration. In ON MS, these findings are due to retrograde degeneration causing axonal loss; however, in patients with no ON history, several different theories exist. The MRI findings seem to agree with the relevant OCT values, with the grey matter being the most sensitive marker for progressive MS. Furthermore, OCT can be used to estimate the disability caused by the disease, with pwMS having thinner RNFL values initially being at an increased risk. An important distinction between MS and NMOSD is that the latter exhibited significantly lower RNFL and GC-IPL values. The OCT findings correlate with cognitive impairment, and although more data are required to support these findings, natalizumab appears to cause lower rates of GC-IPL reduction than glatiramer acetate and interferon-β-1a. Undeniably, AI and machine learning will revolutionize medicine in the upcoming years; however, currently, few studies exist, and more are needed to establish the use of OCT in predicting the risk of disability progression.
Although OCT-A is a novel imaging technique compared to OCT, few studies have been conducted, and the results still seem promising. The advantage of OCT-A is that it can detect the disease earlier than OCT and detect further damage when the disease has progressed severely. All current data point to a reduced VD in the macular or peripapillary area, or both. As to whether the SVP or DVP is affected more, the results are still vague. Enlargement of the FAZ also seems to be a feature of ON MS. High levels of disability are related to low macular SVP VD and low-contrast visual acuity. Additionally, OCT-A could help in the differential diagnosis between MS and NMOSD with the latest information suggesting a significant reduction in the peripapillary and macular VD and FAZ size in NMOSD-ON compared to MS. Finally, OCT-A may be able to detect disease progression when all other methods, such as OCT, radiological, and even clinical examination, fail. Table 1 summarises all significant findings of OCT and OCT-A in MS.

5. Conclusions

As MS is the most common demyelinating disease of the CNS, it can severely affect patients’ lives by causing a wide range of symptoms. The use of OCT and OCT-A has already been established for the diagnosis and monitoring of several different ophthalmologic conditions. In recent years, numerous studies have shed light on their potential use in various neurodegenerative conditions, including MS. Unquestionably, the existing data are still limited, especially regarding the role of OCT-A; however, various biomarkers could function as targets for future research. This review aims to highlight the most important findings available to date and to aid future research by providing a detailed overview of the current literature.

Author Contributions

Conceptualization, C.B., I.-N.C., D.P. and N.G.; Methodology, D.P., I.-N.C. and C.B.; Writing—original draft preparation, I.-N.C., D.P. and C.B.; Writing—review and editing, M.K.B. and N.G; Investigation, I.-N.C., M.K.B.; Project administration, D.P. and N.G.; Supervision, D.P. and N.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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.

Abbreviations

AIArtificial intelligence
BRAOBranch retinal artery occlusion
CNSCentral nervous system
DVPDeep vascular plexus
EBVEpstein-Barr virus
EDSSExpanded disability status scale
FAZFoveal avascular zone
GC-IPLGanglion cell inner plexiform layer complex
mGC-IPLMacular ganglion cell inner plexiform layer
FNβ-1bInterferon beta
IPLInner plexiform layer
MRIMagnetic resonance imaging
MSMultiple sclerosis
pwMSPeople with multiple sclerosis
NMOSDNeuromyelitis optica spectrum disorders
OCTOptical coherence tomography
OCT-AOptical coherence tomography with angiography
ONOptic neuritis
PHOMSPeripapillary hyper-reflective ovoid mass-like structures
RNFLRetinal nerve fibre layer
RPCRadial peripapillary capillary
pRPCPeripapillary radial peripapillary capillary
SVPSuperficial vascular plexus
UVBUltraviolet B light
VDVessel density
VEPVisual evoked potential
mVEPmultifocal visual evoked potential
VVDVolumetric vessel density

References

  1. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [Google Scholar] [CrossRef]
  2. Ascherio, A. Environmental factors in multiple sclerosis. Expert Rev. Neurother. 2013, 13, 3–9. [Google Scholar] [CrossRef]
  3. Cennamo, G.; Romano, M.R.; Vecchio, E.C.; Minervino, C.; Della Guardia, C.; Velotti, N.; Carotenuto, A.; Montella, S.; Orefice, G. Anatomical and functional retinal changes in multiple sclerosis. Eye 2016, 30, 456–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ikuta, F.; Zimmerman, H.M. Distribution of plaques in seventy autopsy cases of multiple sclerosis in the United States. Neurology 1976, 26, 26–28. [Google Scholar] [CrossRef] [PubMed]
  5. Toussaint, D.; Périer, O.; Verstappen, A.; Bervoets, S. Clinicopathological study of the visual pathways, eyes, and cerebral hemispheres in 32 cases of disseminated sclerosis. J. Clin. Neuroophthalmol. 1983, 3, 211–220. [Google Scholar]
  6. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.M.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.S.; et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  7. Wang, L.; Murphy, O.; Caldito, N.G.; Calabresi, P.A.; Saidha, S. Emerging Applications of Optical Coherence Tomography Angiography (OCTA) in neurological research. Eye Vis. 2018, 5, 11. [Google Scholar] [CrossRef]
  8. Saidha, S.; Syc, S.B.; Durbin, M.K.; Eckstein, C.; Oakley, J.D.; Meyer, S.A.; Conger, A.; Frohman, T.C.; Newsome, S.; Ratchford, J.N.; et al. Visual dysfunction in multiple sclerosis correlates better with optical coherence tomography derived estimates of macular ganglion cell layer thickness than peripapillary retinal nerve fiber layer thickness. Mult. Scler. J. 2011, 17, 1449–1463. [Google Scholar] [CrossRef]
  9. González-López, J.J.; Rebolleda, G.; Leal, M.; Oblanca, N.; Muñoz-Negrete, F.J.; Costa-Frossard, L.; Álvarez-Cermeño, J.C. Comparative Diagnostic Accuracy of Ganglion Cell-Inner Plexiform and Retinal Nerve Fiber Layer Thickness Measures by Cirrus and Spectralis Optical Coherence Tomography in Relapsing-Remitting Multiple Sclerosis. BioMed Res. Int. 2014, 2014, 128517. [Google Scholar] [CrossRef]
  10. Ratchford, J.N.; Saidha, S.; Sotirchos, E.S.; Oh, J.A.; Seigo, M.A.; Eckstein, C.; Durbin, M.K.; Oakley, J.D.; Meyer, S.A.; Conger, A.; et al. Active MS is associated with accelerated retinal ganglion cell/inner plexiform layer thinning. Neurology 2013, 80, 47–54. [Google Scholar] [CrossRef] [Green Version]
  11. Petzold, A.; Balcer, L.J.; Calabresi, P.A.; Costello, F.; Frohman, T.C.; Frohman, E.M.; Martinez-Lapiscina, E.H.; Green, A.J.; Kardon, R.; Outteryck, O.; et al. Retinal layer segmentation in multiple sclerosis: A systematic review and meta-analysis. Lancet Neurol. 2017, 16, 797–812. [Google Scholar] [CrossRef] [Green Version]
  12. Kale, N. Optic neuritis as an early sign of multiple sclerosis. Eye Brain 2016, 8, 195–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Britze, J.; Pihl-Jensen, G.; Frederiksen, J.L. Retinal ganglion cell analysis in multiple sclerosis and optic neuritis: A systematic review and meta-analysis. J. Neurol. 2017, 264, 1837–1853. [Google Scholar] [CrossRef]
  14. Saidha, S.; Al-Louzi, O.; Ratchford, J.N.; Bhargava, P.; Oh, J.; Newsome, S.D.; Prince, J.L.; Pham, D.; Roy, S.; van Zijl, P.; et al. Optical coherence tomography reflects brain atrophy in multiple sclerosis: A four-year study: Retinal Atrophy Reflects Brain Atrophy in MS. Ann. Neurol. 2015, 78, 801–813. [Google Scholar] [CrossRef] [PubMed]
  15. Dörr, J.; Wernecke, K.D.; Bock, M.; Gaede, G.; Wuerfel, J.T.; Pfueller, C.F.; Bellmann-Strobl, J.; Freing, A.; Brandt, A.U.; Friedemann, P. Association of Retinal and Macular Damage with Brain Atrophy in Multiple Sclerosis. PLoS ONE 2011, 6, e18132. [Google Scholar] [CrossRef] [Green Version]
  16. Cilingir, V.; Batur, M.; Bulut, M.D.; Milanlioglu, A.; Yılgor, A.; Yasar, T.; Tombul, T. The association between retinal nerve fibre layer thickness and corpus callosum index in different clinical subtypes of multiple sclerosis. Neurol. Sci. 2017, 38, 1223–1232. [Google Scholar] [CrossRef] [PubMed]
  17. Glasner, P.; Sabisz, A.; Chylińska, M.; Komendziński, J.; Wyszomirski, A.; Karaszewski, B. Retinal nerve fiber and ganglion cell complex layer thicknesses mirror brain atrophy in patients with relapsing-remitting multiple sclerosis. Restor. Neurol. Neurosci. 2022, 40, 35–42. [Google Scholar] [CrossRef] [PubMed]
  18. Balk, L.; Steenwijk, M.; Tewarie, P.; Daams, M.; Killestein, J.; Wattjes, M.; Vrenken, H.; Barkhof, F.; Polman, C.; Uitdehaag, B.; et al. Bidirectional trans-synaptic axonal degeneration in the visual pathway in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2015, 86, 419–424. [Google Scholar] [CrossRef]
  19. Klistorner, A.; Graham, E.C.; Yiannikas, C.; Barnett, M.; Parratt, J.; Garrick, R.; Wang, C.; You, Y.; Graham, S. Progression of retinal ganglion cell loss in multiple sclerosis is associated with new lesions in the optic radiations. Eur. J. Neurol. 2017, 24, 1392–1398. [Google Scholar] [CrossRef]
  20. Knier, B.; Leppenetier, G.; Wetzlmair, C.; Aly, L.; Hoshi, M.-M.; Pernpeintner, V.; Biberacher, V.; Berthele, A.; Mühlau, M.; Zimmer, C.; et al. Association of Retinal Architecture, Intrathecal Immunity, and Clinical Course in Multiple Sclerosis. JAMA Neurol. 2017, 74, 847–856. [Google Scholar] [CrossRef] [Green Version]
  21. Martinez-Lapiscina, E.H.; Arnow, S.; Wilson, J.A.; Saidha, S.; Preiningerova, J.L.; Oberwahrenbrock, T.; Brandt, A.U.; Pablo, L.E.; Guerrieri, S.; Gonzalez, I.; et al. Retinal thickness measured with optical coherence tomography and risk of disability worsening in multiple sclerosis: A cohort study. Lancet Neurol. 2016, 15, 574–584. [Google Scholar] [CrossRef] [Green Version]
  22. Lambe, J.; Fitzgerald, K.C.; Murphy, O.C.; Filippatou, A.G.; Sotirchos, E.S.; Kalaitzidis, G.; Vasileiou, E.; Pellegrini, N.; Ogbuokiri, E.; Toliver, B.; et al. Association of Spectral-Domain OCT with Long-term Disability Worsening in Multiple Sclerosis. Neurology 2021, 96, e2058–e2069. [Google Scholar] [CrossRef]
  23. Skirková, M.; Mikula, P.; Maretta, M.; Fedičová, M.; Vitková, M.; Frigová, L.; Szilasi, J.; Moravská, M.; Horňák, M.; Szilasiová, J. Associations of optical coherence tomography with disability and brain MRI volumetry in patients with multiple sclerosis. Neurol. Neurochir. Polska, 2022;  in press. [Google Scholar] [CrossRef] [PubMed]
  24. Bsteh, G.; Hegen, H.; Teuchner, B.; Amprosi, M.; Berek, K.; Ladstätter, F.; Wurth, S.; Auer, M.; Di Pauli, F.; Deisenhammer, F.; et al. Peripapillary retinal nerve fibre layer as measured by optical coherence tomography is a prognostic biomarker not only for physical but also for cognitive disability progression in multiple sclerosis. Mult. Scler. J. 2019, 25, 196–203. [Google Scholar] [CrossRef]
  25. Walter, S.D.; Ishikawa, H.; Galetta, K.M.; Sakai, R.E.; Feller, D.J.; Henderson, S.B.; Wilson, J.A.; Maguire, M.G.; Galetta, S.L.; Frohman, E.; et al. Ganglion Cell Loss in Relation to Visual Disability in Multiple Sclerosis. Ophthalmology 2012, 119, 1250–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sotirchos, E.; Seigo, M.A.; Calabresi, P.; Saidha, S. Comparison of Point Estimates and Average Thicknesses of Retinal Layers Measured Using Manual Optical Coherence Tomography Segmentation for Quantification of Retinal Neurodegeneration in Multiple Sclerosis. Curr. Eye Res. 2013, 38, 224–228. [Google Scholar] [CrossRef] [PubMed]
  27. Lampert, E.J.; Andorra, M.; Torres-Torres, R.; Ortiz-Pérez, S.; Llufriu, S.; Sepúlveda, M.; Sola, N.; Saiz, A.; Sánchez-Dalmau, B.; Villoslada, P.; et al. Color vision impairment in multiple sclerosis points to retinal ganglion cell damage. J. Neurol. 2015, 262, 2491–2497. [Google Scholar] [CrossRef] [PubMed]
  28. Lin, T.-Y.; Chien, C.; Lu, A.; Paul, F.; Zimmermann, H.G. Retinal optical coherence tomography and magnetic resonance imaging in neuromyelitis optica spectrum disorders and MOG-antibody associated disorders: An updated review. Expert Rev. Neurother. 2021, 21, 1101–1123. [Google Scholar] [CrossRef] [PubMed]
  29. Brandt, A.U.; Zimmermann, H.; Kaufhold, F.; Promesberger, J.; Schippling, S.; Finis, D.; Aktas, O.; Geis, C.; Ringelstein, M.; Ringelstein, E.B.; et al. Patterns of Retinal Damage Facilitate Differential Diagnosis between Susac Syndrome and MS. PLoS ONE 2012, 11, e38741. [Google Scholar] [CrossRef]
  30. Ringelstein, M.; Albrecht, P.; Kleffner, I.; Bühn, B.; Harmel, J.; Müller, A.-K.; Finis, D.; Guthoff, R.; Bergholz, R.; Duning, T.; et al. Retinal pathology in Susac syndrome detected by spectral-domain optical coherence tomography. Neurology 2015, 85, 610–618. [Google Scholar] [CrossRef]
  31. Toledo, J.; Sepulcre, J.; Salinas-Alaman, A.; García-Layana, A.; Murie-Fernandez, M.; Bejarano, B.; Villoslada, P. Retinal nerve fiber layer atrophy is associated with physical and cognitive disability in multiple sclerosis. Mult. Scler. J. 2008, 14, 906–912. [Google Scholar] [CrossRef] [PubMed]
  32. Sedighi, B.; Shafa, M.A.; Abna, Z.; Ghaseminejad, A.K.; Farahat, R.; Nakhaee, N.; Hassani, B. Association of Cognitive deficits with Optical Coherence Tomography changes in Multiple Sclerosis Patients. J. Mult. Scler. 2014, 1, 117. [Google Scholar] [CrossRef] [Green Version]
  33. Coric, D.; Balk, L.J.; Verrijp, M.; Eijlers, A.; Schoonheim, M.M.; Killestein, J.; Uitdehaag, B.M.; Petzold, A. Cognitive impairment in patients with multiple sclerosis is associated with atrophy of the inner retinal layers. Mult. Scler. J. 2018, 24, 158–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pul, R.; Saadat, M.; Morbiducci, F.; Skripuletz, T.; Pul, U.; Brockmann, D.; Sühs, K.-W.; Schwenkenbecher, P.; Kahl, K.G.; Pars, K.; et al. Longitudinal time-domain optic coherence study of retinal nerve fiber layer in IFNβ-treated and untreated multiple sclerosis patients. Exp. Ther. Med. 2016, 12, 190–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Button, J.; Al-Louzi, O.; Lang, A.; Bhargava, P.; Newsome, S.D.; Frohman, T.; Balcer, L.J.; Frohman, E.M.; Prince, J.; Calabresi, P.A.; et al. Disease-modifying therapies modulate retinal atrophy in multiple sclerosis: A retrospective study. Neurology 2017, 88, 525–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Klistorner, A.; Arvind, H.; Nguyen, T.; Garrick, R.; Paine, M.; Graham, S.; O’Day, J.; Yiannikas, C. Multifocal VEP and OCT in optic neuritis: A topographical study of the structure–function relationship. Doc. Ophthalmol. 2009, 118, 129–137. [Google Scholar] [CrossRef] [PubMed]
  37. Trip, S.A.; Schlottmann, P.G.; Jones, S.J.; Altmann, D.R.; Garway-Heath, D.F.; Thompson, A.J.; Plant, G.T.; Miller, D.H. Retinal nerve fiber layer axonal loss and visual dysfunction in optic neuritis. Ann. Neurol. 2005, 58, 383–391. [Google Scholar] [CrossRef] [PubMed]
  38. Meltzer, E.; Sguigna, P.V.; Subei, A.; Beh, S.; Kildebeck, E.; Conger, D.; Conger, A.; Lucero, M.; Frohman, B.S.; Frohman, A.N.; et al. Retinal Architecture and Melanopsin-Mediated Pupillary Response Characteristics: A Putative Pathophysiologic Signature for the Retino-Hypothalamic Tract in Multiple Sclerosis. JAMA Neurol. 2017, 74, 574–582. [Google Scholar] [CrossRef] [PubMed]
  39. Wicklein, R.; Wauschkuhn, J.; Giglhuber, K.; Kümpfel, T.; Hemmer, B.; Havla, J.; Knier, B. Association of peripapillary hyper-reflective ovoid masslike structures and disease duration in primary progressive multiple sclerosis. Eur. J. Neurol. 2021, 28, 15056. [Google Scholar] [CrossRef] [PubMed]
  40. López-Dorado, A.; Ortiz, M.; Satue, M.; Rodrigo, M.J.; Barea, R.; Sánchez-Morla, E.M.; Cavaliere, C.; Rodríguez-Ascariz, J.M.; Orduna-Hospital, E.; Boquete, L.; et al. Early Diagnosis of Multiple Sclerosis Using Swept-Source Optical Coherence Tomography and Convolutional Neural Networks Trained with Data Augmentation. Sensors 2021, 22, 167. [Google Scholar] [CrossRef] [PubMed]
  41. Montolío, A.; Cegoñino, J.; Garcia-Martin, E.; del Palomar, A.P. Comparison of Machine Learning Methods Using Spectralis OCT for Diagnosis and Disability Progression Prognosis in Multiple Sclerosis. Ann. Biomed. Eng. 2022, 50, 507–528. [Google Scholar] [CrossRef] [PubMed]
  42. Martinez Sosa, S.; Smith, K.J. Understanding a role for hypoxia in lesion formation and location in the deep and periventricular white matter in small vessel disease and multiple sclerosis. Clin. Sci. 2017, 131, 2503–2524. [Google Scholar] [CrossRef]
  43. Kleerekooper, I.; Petzold, A.; Trip, S.A. Anterior visual system imaging to investigate energy failure in multiple sclerosis. Brain 2020, 143, 1999–2008. [Google Scholar] [CrossRef]
  44. Jiang, H.; Delgado, S.; Tan, J.; Liu, C.; Rammohan, K.W.; DeBuc, D.C.; Lam, B.L.; Feuer, W.J.; Wang, J. Impaired retinal microcirculation in multiple sclerosis. Mult. Scler. J. 2016, 22, 1812–1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Modrzejewska, M.; Karczewicz, D.; Wilk, G. Assessment of blood flow velocity in eyeball arteries in multiple sclerosis patients with past retrobulbar optic neuritis in color Doppler ultrasonography. Klin. Ocz. 2007, 109, 183–186. [Google Scholar]
  46. Murphy, O.C.; Kwakyi, O.; Iftikhar, M.; Zafar, S.; Lambe, J.; Pellegrini, N.; Sotirchos, E.S.; Gonzalez-Caldito, N.; Ogbuokiri, E.; Filippatou, A.; et al. Alterations in the retinal vasculature occur in multiple sclerosis and exhibit novel correlations with disability and visual function measures. Mult. Scler. J. 2020, 26, 815–828. [Google Scholar] [CrossRef] [PubMed]
  47. Yilmaz, H.; Ersoy, A.; Icel, E. Assessments of vessel density and foveal avascular zone metrics in multiple sclerosis: An optical coherence tomography angiography study. Eye 2020, 34, 771–778. [Google Scholar] [CrossRef]
  48. Feucht, N.; Maier, M.; Lepennetier, G.; Pettenkofer, M.; Wetzlmair, C.; Daltrozzo, T.; Scherm, P.; Zimmer, C.; Hoshi, M.-M.; Hemmer, B.; et al. Optical coherence tomography angiography indicates associations of the retinal vascular network and disease activity in multiple sclerosis. Mult. Scler. J. 2019, 25, 224–234. [Google Scholar] [CrossRef] [PubMed]
  49. Ulusoy, M.O.; Horasanlı, B.; Işık-Ulusoy, S. Optical coherence tomography angiography findings of multiple sclerosis with or without optic neuritis. Neurol. Res. 2020, 42, 319–326. [Google Scholar] [CrossRef] [PubMed]
  50. Jiang, H.; Gameiro, G.R.; Liu, Y.; Lin, Y.; Hernandez, J.; Deng, Y.; Gregori, G.; Delgado, S.; Wang, J. Visual Function and Disability Are Associated with Increased Retinal Volumetric Vessel Density in Patients with Multiple Sclerosis. Am. J. Ophthalmol. 2020, 213, 34–45. [Google Scholar] [CrossRef]
  51. Balıkçı, A.; Yener, N.P.; Seferoğlu, M. Optical Coherence Tomography and Optical Coherence Tomography Angiography Findings in Multiple Sclerosis Patients. Neuro-Ophthalmology 2022, 46, 19–33. [Google Scholar] [CrossRef] [PubMed]
  52. Aly, L.; Noll, C.; Wicklein, R.; Wolf, E.; Romahn, E.F.; Wauschkuhn, J.; Hosari, S.; Mardin, C.; Berthele, A.; Hemmer, B.; et al. Dynamics of Retinal Vessel Loss After Acute Optic Neuritis in Patients with Relapsing Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2022, 9, e1159. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, X.; Jia, Y.; Spain, R.; Potsaid, B.M.; Liu, J.J.; Baumann, B.; Hornegger, J.; Fujimoto, J.G.; Wu, Q.; Huang, D. Optical coherence tomography angiography of optic nerve head and parafovea in multiple sclerosis. Br. J. Ophthalmol. 2014, 98, 1368–1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Spain, R.I.; Liu, L.; Zhang, X.; Jia, Y.; Tan, O.; Bourdette, D.; Huang, D. Optical coherence tomography angiography enhances the detection of optic nerve damage in multiple sclerosis. Br. J. Ophthalmol. 2018, 102, 520–524. [Google Scholar] [CrossRef] [PubMed]
  55. Lanzillo, R.; Cennamo, G.; Criscuolo, C.; Carotenuto, A.; Velotti, N.; Sparnelli, F.; Cianflone, A.; Moccia, M.; Morra, V.B. Optical coherence tomography angiography retinal vascular network assessment in multiple sclerosis. Mult. Scler. J. 2018, 24, 1706–1714. [Google Scholar] [CrossRef] [PubMed]
  56. Tiftikcioglu, B.I.; Emre, S.; Idiman, F.; Idiman, E. Optical coherence tomography angiography (OCTA) in differential diagnosis of aquaporin-4 antibody seronegative NMOSD and multiple sclerosis. Mult. Scler. Relat. Disord. 2022, 58, 103503. [Google Scholar] [CrossRef] [PubMed]
  57. Rogaczewska, M.; Michalak, S.; Stopa, M. Differentiation between multiple sclerosis and neuromyelitis optica spectrum disorder using optical coherence tomography angiography. Sci. Rep. 2021, 11, 10697. [Google Scholar] [CrossRef]
  58. Liu, C.; Xiao, H.; Zhang, X.; Zhao, Y.; Li, R.; Zhong, X.; Wang, Y.; Shu, Y.; Chang, Y.; Wang, J.; et al. Optical coherence tomography angiography helps distinguish multiple sclerosis from AQP4-IgG-seropositive neuromyelitis optica spectrum disorder. Brain Behav. 2021, 11, e02125. [Google Scholar] [CrossRef]
  59. Montorio, D.; Lanzillo, R.; Carotenuto, A.; Petracca, M.; Moccia, M.; Criscuolo, C.; Spiezia, A.L.; Lamberti, A.; Perrotta, F.; Pontillo, G.; et al. Retinal and Choriocapillary Vascular Changes in Early Stages of Multiple Sclerosis: A Prospective Study. J. Clin. Med. 2021, 10, 5756. [Google Scholar] [CrossRef] [PubMed]
  60. Hazar, L.; Ava, S.; Tamam, Y.; Karahan, M.; Erdem, S.; Dursun, M.; Keklikçi, U. Relationship between optical coherence tomography angiography and visual evoked potential in patients with multiple sclerosis. Indian J. Ophthalmol. 2022, 70, 873. [Google Scholar] [CrossRef]
Table 1. OCT and OCT-A findings in MS.
Table 1. OCT and OCT-A findings in MS.
OCTOCT-A
• RNFL and GC-IPL thinning inversely related to disease duration.• Earlier detection of the disease and disease-progression compared to OCT
• Lower RNFL values suggest increased disability risk• Reduced VD in both the macular and peripapillary area
• Significantly lower RNFL and GC-IPL values in NMOSD patients compared to MS• Reduced peripapillary, macular VD and FAZ size in NMOSD-ON patients compared to MS
• Natalizumab may cause lower rates of GC-IPL thinning• High disability rates relate to reduced macular SVP VD
• FAZ enlargement in ON MS patients
RNFL: retinal nerve fibre layer; GC-IPL: ganglion cell inner plexiform layer complex; NMOSD: neuromyelitis optica spectrum disorder; VD: vessel density; FAZ: foveal avascular zone, ON: optic neuritis; SVP: superficial vascular plexus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chalkias, I.-N.; Bakirtzis, C.; Pirounides, D.; Boziki, M.K.; Grigoriadis, N. Optical Coherence Tomography and Optical Coherence Tomography with Angiography in Multiple Sclerosis. Healthcare 2022, 10, 1386. https://doi.org/10.3390/healthcare10081386

AMA Style

Chalkias I-N, Bakirtzis C, Pirounides D, Boziki MK, Grigoriadis N. Optical Coherence Tomography and Optical Coherence Tomography with Angiography in Multiple Sclerosis. Healthcare. 2022; 10(8):1386. https://doi.org/10.3390/healthcare10081386

Chicago/Turabian Style

Chalkias, Ioannis-Nikolaos, Christos Bakirtzis, Demetrios Pirounides, Marina Kleopatra Boziki, and Nikolaos Grigoriadis. 2022. "Optical Coherence Tomography and Optical Coherence Tomography with Angiography in Multiple Sclerosis" Healthcare 10, no. 8: 1386. https://doi.org/10.3390/healthcare10081386

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