Hostname: page-component-848d4c4894-8bljj Total loading time: 0 Render date: 2024-06-14T16:33:33.907Z Has data issue: false hasContentIssue false

Plasma neurofilament light chain is increased in Niemann-Pick Type C but glial fibrillary acidic protein remains normal

Published online by Cambridge University Press:  27 March 2024

Dhamidhu Eratne*
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Melbourne Neuropsychiatry Centre & Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
Courtney Lewis
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
Wendy Kelso
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia
Samantha Loi
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
Wei-Hsuan Michelle Chiu
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
Kaj Blennow
Affiliation:
Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Dahlgren’s Academy, University of Gothenburg, Gothenburg, Sweden Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
Henrik Zetterberg
Affiliation:
Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK UK Dementia Research Institute at UCL, London, UK Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
Alexander F Santillo
Affiliation:
Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Malmö, Sweden
Dennis Velakoulis
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
Mark Walterfang
Affiliation:
Neuropsychiatry Centre, Royal Melbourne Hospital, Melbourne, VIC, Australia Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
*
Corresponding author: Dhamidhu Eratne; Email: deratne@unimelb.edu.au
Rights & Permissions [Opens in a new window]

Abstract

Objective:

Niemann-Pick Type C (NPC) is a genetic neurodegenerative lysosomal storage disorder commonly associated with psychiatric symptoms and delays to accurate diagnosis and treatment. This study investigated biomarker levels and diagnostic utility of plasma neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in NPC compared to healthy controls.

Methods:

Patients with NPC were recruited from a specialist assessment and management service. Data was available from an age and sex-matched healthy control group. NfL and GFAP were measured on Quanterix Simoa HD-X analysers and groups compared using generalised linear models. NfL levels were compared to, and percentiles derived from, recently developed NfL reference ranges.

Results:

Plasma NfL was significantly elevated in 11 patients with NPC compared to 25 controls (mean 17.1 vs. 7.4 pg/ml, p < 0.001), and reference ranges (all >98th percentile). NfL distinguished NPC from controls with high accuracy. GFAP levels were not elevated in NPC (66.6 vs. 75.1 pg/ml).

Discussion:

The study adds important evidence on the potential diagnostic utility of plasma NfL in NPC, extends the literature of NfL as a diagnostic tool to differentiate neurodegenerative from primary psychiatric disorders, and adds support to the pathology in NPC primarily involving neuronal, particularly axonal, degeneration.

Type
Original Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

Significant outcomes

  • Plasma neurofilament light chain (NfL), a marker of neuronal injury, was significantly elevated in Niemann Pick Type C (NPC), compared to healthy controls

  • Plasma glial fibrillary acidic protein (GFAP), a marker of astrocytosis, was not elevated in NPC compared to healthy controls

  • NfL distinguished NPC from controls, with high accuracy

Limitations

  • This retrospective study had a small sample size and lack of many serial samples, limiting interpretations of treatment effects and changes over time

  • Our cohort only included patients with several years of symptoms, therefore limiting interpretation of diagnostic utility at earliest stages of the disease

Introduction

Niemann-Pick Type C (NPC) is a rare, severe, genetic neurodegenerative lysosomal storage disorder, associated with highly variable age at onset and symptoms. NPC is commonly associated with psychiatric symptoms, high rates of misdiagnosis and delay until accurate diagnosis and treatment (Walterfang et al., Reference Walterfang, Fietz, Fahey, Sullivan, Leane, Lubman and Velakoulis2006; Rego et al., Reference Rego, Farrand, Goh, Eratne, Kelso, Mangelsdorf, Velakoulis and Walterfang2019; Berry-Kravis, Reference Berry-Kravis2021). While there are no specific blood or cerebrospinal fluid (CSF) biomarkers which assist in the diagnosis of NPC, biomarkers of neuronal injury (e.g., neurofilament light chain protein, NfL), and astrocytosis (e.g., glial fibrillary acidic protein, GFAP) may be useful as non-specific markers. Elevated NfL and GFAP levels have been identified in neurodegenerative conditions such as Alzheimer disease and frontotemporal dementia (Khalil et al., Reference Khalil, Teunissen, Otto, Piehl, Sormani, Gattringer, Barro, Kappos, Comabella, Fazekas, Petzold, Blennow, Zetterberg and Kuhle2018; Gaetani et al., Reference Gaetani, Blennow, Calabresi, Di Filippo, Parnetti and Zetterberg2019; Ashton et al., Reference Ashton, Janelidze, Al Khleifat, Leuzy, van der Ende, Karikari, Benedet, Pascoal, Lleó, Parnetti, Galimberti, Bonanni, Pilotto, Padovani, Lycke, Novakova, Axelsson, Velayudhan, Rabinovici, Miller, Pariante, Nikkheslat, Resnick, Thambisetty, Schöll, Fernández-Eulate, Gil-Bea, López de Munain, Al-Chalabi, Rosa-Neto, Strydom, Svenningsson, Stomrud, Santillo, Aarsland, van Swieten, Palmqvist, Zetterberg, Blennow, Hye and Hansson2021), and NfL has been shown to distinguish neurodegenerative from primary psychiatric and non-neurodegenerative conditions, and potentially reduce misdiagnosis (Ashton et al., Reference Ashton, Janelidze, Al Khleifat, Leuzy, van der Ende, Karikari, Benedet, Pascoal, Lleó, Parnetti, Galimberti, Bonanni, Pilotto, Padovani, Lycke, Novakova, Axelsson, Velayudhan, Rabinovici, Miller, Pariante, Nikkheslat, Resnick, Thambisetty, Schöll, Fernández-Eulate, Gil-Bea, López de Munain, Al-Chalabi, Rosa-Neto, Strydom, Svenningsson, Stomrud, Santillo, Aarsland, van Swieten, Palmqvist, Zetterberg, Blennow, Hye and Hansson2021; Eratne et al., Reference Eratne, Kang, Malpas, Simpson-Yap, Lewis, Dang, Grewal, Coe, Dobson, Keem, Chiu, Kalincik, Ooi, Darby, Brodtmann, Hansson, Janelidze, Blennow, Zetterberg, Walker, Dean, Berk, Wannan, Pantelis, Loi, Walterfang, Berkovic, Santillo and Velakoulis2023; Eratne, Keem, et al., Reference Eratne, Keem, Lewis, Kang, Walterfang, Farrand, Loi, Kelso, Cadwallader, Berkovic, Li, Masters, Collins, Santillo and Velakoulis2022; Eratne, Loi, et al., Reference Eratne, Loi, Li, Stehmann, Malpas, Santillo, Janelidze, Cadwallader, Walia, Ney, Lewis, Senesi, Fowler, McGlade, Varghese, Ravanfar, Kelso, Farrand, Keem, Kang, Goh, Dhiman, Gupta, Watson, Yassi, Kaylor‐Hughes, Kanaan, Perucca, Dobson, Vivash, Ali, O'Brien, Hansson, Zetterberg, Blennow, Walterfang, Masters, Berkovic, Collins and Velakoulis2022; Kang et al., Reference Kang, Eratne, Dobson, Malpas, Keem, Lewis, Grewal, Tsoukra, Dang, Mocellin, Kalincik, Santillo, Zetterberg, Blennow, Stehmann, Varghese, Li, Masters, Collins, Berkovic, Evans, Kelso, Farrand, Loi, Walterfang and Velakoulis2023).

Studies have found elevated CSF and plasma NfL levels in NPC compared to primary psychiatric conditions and controls (Eratne et al., Reference Eratne, Loi, Li, Varghese, McGlade, Collins, Masters, Velakoulis and Walterfang2020; Dardis et al., Reference Dardis, Pavan, Fabris, Da Riol, Sechi, Fiumara, Santoro, Ormazabal, Milanic, Zampieri, Biasizzo and Scarpa2021), and associations with severity and miglustat treatment (Agrawal et al., Reference Agrawal, Farhat, Sinaii, Do, Xiao, Berry-Kravis, Bianconi, Masvekar, Bielekova, Solomon and Porter2023). No studies have investigated GFAP in NPC, except one that included only two people with NPC as a comparison group, not finding elevated levels (Welford et al., Reference Welford, Farine, Steiner, Garzotti, Dobrenis, Sievers, Strasser, Amraoui, Groenen, Giugliani and Mengel2022).

This study aimed to compare NfL and GFAP levels in patients with NPC to age- and sex-matched controls, and to compare NfL levels to the reference ranges we recently developed (Eratne et al., Reference Eratne, Kang, Malpas, Simpson-Yap, Lewis, Dang, Grewal, Coe, Dobson, Keem, Chiu, Kalincik, Ooi, Darby, Brodtmann, Hansson, Janelidze, Blennow, Zetterberg, Walker, Dean, Berk, Wannan, Pantelis, Loi, Walterfang, Berkovic, Santillo and Velakoulis2023). Exploratory analyses investigated associations with clinical variables and explored biomarker levels in patients who had serial bloods.

Methods

Patients were recruited from a tertiary specialist NPC assessment and management service at Neuropsychiatry Centre, Royal Melbourne Hospital, Australia. Data was available from an age-matched healthy control group with no cognitive, neurological, or psychiatric symptoms or conditions, no known renal impairment or other severe medical conditions. Plasma aliquots were collected, processed, and stored at −80°C. Plasma NfL and GFAP levels were measured on Quanterix Simoa HD-X analysers.

Statistical analyses were performed using R v4.2.2 (2022-10-31). Generalised linear models (GLMs) were used to examine relationships with log10-transformed biomarker levels, diagnostic group, age and sex as covariates, with 95% confidence intervals (nonparametric bootstrapping, 1000 replicates). Receiver operator characteristic curves were computed to estimate diagnostic performance, area under the curve (AUC), sensitivity, and specificity. For more precise exploration of NfL levels, levels were compared to and percentiles derived from reference ranges developed using generalised additive models for location, scale, and shape, previously described (Eratne et al., Reference Eratne, Kang, Malpas, Simpson-Yap, Lewis, Dang, Grewal, Coe, Dobson, Keem, Chiu, Kalincik, Ooi, Darby, Brodtmann, Hansson, Janelidze, Blennow, Zetterberg, Walker, Dean, Berk, Wannan, Pantelis, Loi, Walterfang, Berkovic, Santillo and Velakoulis2023).

This study, part of The Markers in Neuropsychiatric Disorders Study (The MiND Study, https://themindstudy.org), was approved by the Melbourne Health Human Research Ethics Committee (MH/HREC2020.142).

Results

There were 11 patients with NPC, all of whom had neurological symptoms, and 25 controls. There were no differences in age (mean age 33.9 years vs. 40.7, p = 0.098; median 42.9 (range 25–49) vs. 41.3 (17–48)) or sex (63.6% vs. 80%, p = 0.409). Four out of 11 (36%) patients had a juvenile onset; 7/11 (64%) were on treatment (details in Table 1). The mean duration of illness was 12.2 years (median 10, range 4–23) with no differences between juvenile and adult onset, or between younger age (<30 years) or older (>30 years).

Table 1. Study demographics and plasma biomarker levels

Data is mean (SD) or n (%). GFAP, glial fibrillary acidic protein; NfL, neurofilament light chain protein.

Plasma NfL and GFAP in Niemann-Pick Type C compared to controls

NfL levels were significantly higher in patients with NPC compared to controls (mean 17.1 vs. 7.4 pg/ml, GLM for logNfL: β = 1.81, 95% confidence interval: [1.50, 2.22], p < 0.001), Table 1 and Fig. 1. As demonstrated in Fig. 2, 100% of patients with Niemann-Pick Type C had significantly elevated NfL levels for their age: all were >98th percentile (z-score >2); 7/11 (64%) were >99th percentile (z-score >2.5).

Figure 1. Plasma neurofilament light chain protein (left) and glial fibrillary acidic protein (right) levels in patients with Niemann-Pick Type C and controls. + = mean level.

Figure 2. All patients with Niemann-Pick Type C had significantly elevated neurofilament light chain protein (NfL) levels for their age (all >98–99th percentile), using our previously developed interactive plasma NfL reference range app (themindstudy.org/apps).

By comparison, GFAP levels were not elevated in NPC compared to controls (66.6 vs. 75.1 pg/ml, β = −0.09 [−0.83, 0.63], p = 0.790).

Plasma NfL had high diagnostic accuracy to distinguish NPC from controls (AUC 0.96 [0.91, 1.00]). An optimal cut-off of 9.35 pg/mL was associated with 84% specificity, 100% sensitivity. A cut-off of 10.47 pg/mL resulted in better specificity (92%), but slightly reduced sensitivity (91%). GFAP did not have diagnostic utility (AUC 0.57 [0.38, 0.76]).

Exploratory analyses

Exploratory analyses investigated biomarker levels differences between treated versus untreated patients, different types of treatment, and juvenile versus adult onset. No differences were seen.

Three patients had serial bloods, as illustrated in Fig. 3. Over a two-year period, NfL levels in patient A (on miglustat and IV hydroxypropyl-beta-cyclodextrin (Cyclo Therapeutics, “cyclodextrin”) increased only slightly, by 11% (from 9.6 to 10.7 pg/ml), while GFAP increased by 36% (from 88.8 to 121 pg/ml). On the other hand, patient B (only on miglustat and acetylleucine), exhibited a 50% increase in NfL over 2 years (10.6 to 16 pg/ml), and a 76% increase in GFAP (65.3 to 114.8 pg/ml). Patient C (on acetylleucine only), had a significant neurological deterioration requiring hospitalisation at the time of their second blood sample. Their second NfL level (58 pg/ml) was markedly higher than a year prior (20 pg/ml), and GFAP increased by 28% (51.4 to 66 pg/ml).

Figure 3. Longitudinal changes in neurofilament light chain protein and glial fibrillary acidic protein in three patients.

Discussion

This study found significantly elevated plasma NfL levels in NPC compared to controls, and NfL distinguished NPC from controls with high accuracy. These findings add important evidence on the potential diagnostic utility of plasma NfL in this devastating condition that is not uncommonly misdiagnosed as a primary psychiatric condition. In addition, this study provides an important negative finding of plasma GFAP in the largest number of NPC patients to date.

The differing profile of NfL and GFAP adds weight to the pathology in NPC primarily involving neuronal, and particularly axonal, degeneration (Walterfang et al., Reference Walterfang, Fahey, Desmond, Wood, Seal, Steward, Adamson, Kokkinos, Fietz and Velakoulis2010). It is likely that varying profiles of these biomarkers will be seen in different disorders, providing important information on the underlying pathophysiological processes in various conditions and within subgroups of syndromes, and combination biomarkers could have diagnostic and wider clinical utility in various conditions and differential diagnoses.

Secondary, purely exploratory analyses did not find any differences in NfL and GFAP levels between untreated and treated patients, however the main limitation of this study is the small sample size and lack of serial levels, which limit any confident interpretations of treatment effects and changes over time. Three patients with serial levels had large increases in GFAP levels over 1–2 years. One of these two patients was on IV cyclodextrin and did not have much increase in NfL, whereas the other had a more marked increase. This may reflect a treatment effect of IV cyclodextrin on neuronal injury, however, this was not seen for intrathecal cyclodextrin in a larger study (Agrawal et al., Reference Agrawal, Farhat, Sinaii, Do, Xiao, Berry-Kravis, Bianconi, Masvekar, Bielekova, Solomon and Porter2023). The increase in GFAP over time is notable, possibly pointing to differing timelines of NfL and GFAP changes in the disease course, with NfL changing earlier, and GFAP changing later, perhaps after neuronal loss has occurred. However, the serial GFAP levels, although increased compared to baseline, were still within the control range. Patient C’s dramatic increase in serial NfL levels corresponded with severe neurological deterioration. This adds weight to NfL reflecting severe/acute deterioration and severity/prognosis/rate of deterioration, on top of being diagnostic. Our cohort included patients with several years of symptoms, therefore we cannot conclude the diagnostic utility at the earliest stages of the disease. It would be important to investigate serial biomarkers and associations with other variables in larger, well characterised cohorts.

To conclude, this study found significantly higher plasma NfL levels in NPC and demonstrated strong diagnostic utility of plasma NfL to distinguish NPC from controls, while not finding elevated GFAP levels. Other plasma biomarkers can complement the investigation in case of an elevated NfL level, as more specific testing for NPC, before genetic testing, for example: N-palmitoyl-O-phosphocholineserine, cholestane-3β,5 α,6β-triol (C-triol), 7-ketocholesterol. (Jiang and Ory, Reference Jiang and Ory2021) This extends the literature on NfL to identify neurological/neurodegenerative causes of neurological and neuropsychiatric symptoms, especially in younger people where diagnostic challenges and misdiagnosis can be higher, and adds to our understanding of the pathophysiology and utility of biomarkers in NPC for diagnosis and potentially wider clinical utility. Further studies are underway in a larger cohort, to investigate NfL, GFAP, and other biomarkers and associations with clinical and treatment variables.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/neu.2024.14.

Acknowledgements

We are grateful for funding that supported this work: NorthWestern Mental Health Research Seed Grants, the CJDSGN Memorial Award in memory of Michael Luscombe, and MACH MRFF RART 2.2 and NHMRC (1185180) funding. The role of these funding sources was to support participant recruitment, research study staff, and biosample analyses.

Finally, the authors would like to thank all the patients and their families for their participation.

The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication.

A.F.S is primarily funded by the Swedish federal government under the ALF agreement (ALF 2022 YF 0017) and The Åke Wiberg Foundation.

Author contribution

Dhamidhu Eratne — Study design and coordination, data collection, participant recruitment, data analysis and interpretation, literature search, statistical analysis, writing manuscript.

Courtney Lewis — Study coordination, data collection, participant recruitment, writing reviewing manuscript.

Wendy Kelso — Data interpretation, participant recruitment, writing and reviewing manuscript.

Samantha Loi — Data interpretation, writing and reviewing manuscript.

Wei-Hsuan Michelle Chiu — Data collection, reviewing the manuscript.

Kaj Blennow — Data interpretation, reviewing the manuscript.

Henrik Zetterberg — Data interpretation, reviewing the manuscript.

Alexander F Santillo — Data interpretation, writing and reviewing manuscript.

Dennis Velakoulis — Data interpretation, writing and reviewing manuscript.

Mark Walterfang — Study design and coordination, participant recruitment, data interpretation, writing and reviewing manuscript.

Financial support

HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2023-00356; #2022-01018 and #2019-02397), the European Union’s Horizon Europe research and innovation programme under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation, USA (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), the National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL (UKDRI-1003).

Competing interests

HZ has served at scientific advisory boards and/or as a consultant for Abbvie, Acumen, Alector, Alzinova, ALZPath, Annexon, Apellis, Artery Therapeutics, AZTherapies, Cognito Therapeutics, CogRx, Denali, Eisai, Merry Life, Nervgen, Novo Nordisk, Optoceutics, Passage Bio, Pinteon Therapeutics, Prothena, Red Abbey Labs, reMYND, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures in symposia sponsored by Alzecure, Biogen, Cellectricon, Fujirebio, Lilly, and Roche, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB, which is a part of the GU Ventures Incubator Program (outside submitted work).

None of the other authors have anything to disclose.

Statistical analysis conducted by: Dr Dhamidhu Eratne.

References

Agrawal, N, Farhat, NY, Sinaii, N, Do, AD, Xiao, C, Berry-Kravis, E, Bianconi, S, Masvekar, R, Bielekova, B, Solomon, B, Porter, FD (2023) Neurofilament light chain in cerebrospinal fluid as a novel biomarker in evaluating both clinical severity and therapeutic response in Niemann-Pick disease type C1. Genetics in Medicine 25(3), 100349.CrossRefGoogle ScholarPubMed
Ashton, NJ, Janelidze, S, Al Khleifat, A, Leuzy, A, van der Ende, EL, Karikari, TK, Benedet, AL, Pascoal, TA, Lleó, A, Parnetti, L, Galimberti, D, Bonanni, L, Pilotto, A, Padovani, A, Lycke, J, Novakova, L, Axelsson, M, Velayudhan, L, Rabinovici, GD, Miller, B, Pariante, C, Nikkheslat, N, Resnick, SM, Thambisetty, M, Schöll, M, Fernández-Eulate, G, Gil-Bea, FJ, López de Munain, A, Al-Chalabi, A, Rosa-Neto, P, Strydom, A, Svenningsson, P, Stomrud, E, Santillo, A, Aarsland, D, van Swieten, JC, Palmqvist, S, Zetterberg, H, Blennow, K, Hye, A, Hansson, O (2021) A multicentre validation study of the diagnostic value of plasma neurofilament light. Nature Communications 12(1), 3400.CrossRefGoogle ScholarPubMed
Berry-Kravis, E (2021) Niemann-Pick disease, type C: diagnosis, management and disease-targeted therapies in development. In Seminars in pediatric neurology 37. Uncommon neuropediatric diseases pp. 100879.Google Scholar
Dardis, A, Pavan, E, Fabris, M, Da Riol, RM, Sechi, A, Fiumara, A, Santoro, L, Ormazabal, M, Milanic, R, Zampieri, S, Biasizzo, J, Scarpa, M (2021) Plasma neurofilament light (NfL) in patients affected by Niemann-Pick Type C Disease (NPCD). Journal of Clinical Medicine 10(20), 4796.CrossRefGoogle ScholarPubMed
Eratne, D, Loi, SM, Li, Q-X, Varghese, S, McGlade, A, Collins, S, Masters, CL, Velakoulis, D, Walterfang, M (2020) Cerebrospinal fluid neurofilament light chain is elevated in Niemann-Pick type C compared to psychiatric disorders and healthy controls and may be a marker of treatment response. The Australian and New Zealand journal of psychiatry 54(6), 648649.CrossRefGoogle ScholarPubMed
Eratne, D, Keem, M, Lewis, C, Kang, M, Walterfang, M, Farrand, S, Loi, S, Kelso, W, Cadwallader, C, Berkovic, SF, Li, Q-X, Masters, CL, Collins, S, Santillo, A, Velakoulis, D (2022) Cerebrospinal fluid neurofilament light chain differentiates behavioural variant frontotemporal dementia progressors from non-progressors. Journal of the Neurological Sciences 442, 120439.CrossRefGoogle ScholarPubMed
Eratne, D, Loi, SM, Li, Q‐Xin, Stehmann, C, Malpas, CB, Santillo, A, Janelidze, S, Cadwallader, C, Walia, N, Ney, B, Lewis, V, Senesi, M, Fowler, C, McGlade, A, Varghese, S, Ravanfar, P, Kelso, W, Farrand, S, Keem, M, Kang, M, Goh, AMY, Dhiman, K, Gupta, V, Watson, R, Yassi, N, Kaylor‐Hughes, C, Kanaan, R, Perucca, P, Dobson, H, Vivash, L, Ali, R, O'Brien, TJ, Hansson, O, Zetterberg, H, Blennow, K, Walterfang, M, Masters, CL, Berkovic, SF, Collins, S, Velakoulis, D, The MiND Study Group (2022) Cerebrospinal fluid neurofilament light chain differentiates primary psychiatric disorders from rapidly progressive, Alzheimer’s disease and frontotemporal disorders in clinical settings. Alzheimer’s & Dementia 18(11), 22182233.CrossRefGoogle ScholarPubMed
Eratne, D, Kang, M, Malpas, C, Simpson-Yap, S, Lewis, C, Dang, C, Grewal, J, Coe, A, Dobson, H, Keem, M, Chiu, W-H, Kalincik, T, Ooi, S, Darby, D, Brodtmann, A, Hansson, O, Janelidze, S, Blennow, K, Zetterberg, H, Walker, A, Dean, O, Berk, M, Wannan, C, Pantelis, C, Loi, S, Walterfang, M, Berkovic, S, Santillo, A, Velakoulis, D, The MiND Study Group (2023) Plasma neurofilament light in behavioural variant frontotemporal dementia compared to mood and psychotic disorders. The Australian and New Zealand Journal of Psychiatry 58, 7081.CrossRefGoogle ScholarPubMed
Gaetani, L, Blennow, K, Calabresi, P, Di Filippo, M, Parnetti, L, Zetterberg, H (2019) Neurofilament light chain as a biomarker in neurological disorders. Journal of Neurology, Neurosurgery & Psychiatry 90(8), 870881.CrossRefGoogle Scholar
Jiang, X and Ory, DS (2021) Advancing diagnosis and treatment of Niemann-Pick C disease through biomarker discovery. Exploration of Neuroprotective Therapy 1(3), 146158.CrossRefGoogle ScholarPubMed
Kang, MJY, Eratne, D, Dobson, H, Malpas, CB, Keem, M, Lewis, C, Grewal, J, Tsoukra, V, Dang, C, Mocellin, R, Kalincik, T, Santillo, AF, Zetterberg, H, Blennow, K, Stehmann, C, Varghese, S, Li, Q-X, Masters, CL, Collins, S, Berkovic, SF, Evans, A, Kelso, W, Farrand, S, Loi, SM, Walterfang, M, Velakoulis, D (2023) Cerebrospinal fluid neurofilament light predicts longitudinal diagnostic change in patients with psychiatric and neurodegenerative disorders. Acta Neuropsychiatrica 36(1), 112.Google ScholarPubMed
Khalil, M, Teunissen, CE, Otto, M, Piehl, F, Sormani, MP, Gattringer, T, Barro, C, Kappos, L, Comabella, M, Fazekas, F, Petzold, A, Blennow, K, Zetterberg, H, Kuhle, J (2018) Neurofilaments as biomarkers in neurological disorders. Nature Reviews Neurology 14, 577589.CrossRefGoogle ScholarPubMed
Rego, T, Farrand, S, Goh, AMY, Eratne, D, Kelso, W, Mangelsdorf, S, Velakoulis, D, Walterfang, M (2019) Psychiatric and cognitive symptoms associated with Niemann-Pick Type C disease: Neurobiology and management. CNS Drugs 33(2), 125142.CrossRefGoogle ScholarPubMed
Walterfang, M, Fietz, M, Fahey, M, Sullivan, D, Leane, P, Lubman, DI, & Velakoulis, D (2006) The neuropsychiatry of Niemann-Pick Type C disease in adulthood. The Journal of Neuropsychiatry and Clinical Neurosciences 18(2), 158170.CrossRefGoogle ScholarPubMed
Walterfang, M, Fahey, M, Desmond, P, Wood, A, Seal, ML, Steward, C, Adamson, C, Kokkinos, C, Fietz, M, Velakoulis, D (2010) White and gray matter alterations in adults with Niemann-Pick disease type C: a cross-sectional study. Neurology 75(1), 4956.CrossRefGoogle ScholarPubMed
Welford, RWD, Farine, H, Steiner, M, Garzotti, M, Dobrenis, K, Sievers, C, Strasser, DS, Amraoui, Y, Groenen, PMA, Giugliani, R, Mengel, E (2022) Plasma neurofilament light, glial fibrillary acidic protein and lysosphingolipid biomarkers for pharmacodynamics and disease monitoring of GM2 and GM1 gangliosidoses patients. Molecular Genetics and Metabolism Reports 30, 100843.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Study demographics and plasma biomarker levels

Figure 1

Figure 1. Plasma neurofilament light chain protein (left) and glial fibrillary acidic protein (right) levels in patients with Niemann-Pick Type C and controls. + = mean level.

Figure 2

Figure 2. All patients with Niemann-Pick Type C had significantly elevated neurofilament light chain protein (NfL) levels for their age (all >98–99th percentile), using our previously developed interactive plasma NfL reference range app (themindstudy.org/apps).

Figure 3

Figure 3. Longitudinal changes in neurofilament light chain protein and glial fibrillary acidic protein in three patients.

Supplementary material: File

Eratne et al. supplementary material

Eratne et al. supplementary material
Download Eratne et al. supplementary material(File)
File 16.9 KB