Heterogeneity of acetylcholine receptor autoantibody-mediated
complement activity in patients with myasthenia gravis.
Abeer H. Obaid
1,2,3
, Chryssa Zografou
1,2
, Douangsone D. Vadysirisack
4
, Bailey Munro-Sheldon
1
, Miriam
L. Fichtner
1,2
, Bhaskar Roy
1
, William M. Philbrick
2
, Jeffrey L. Bennett
5
, Richard J. Nowak
1
, and Kevin C.
O'Connor
1,2
1. Department of Neurology, Yale School of Medicine, New Haven, CT 06511 USA.
2. Department of Immunobiology, Yale School of Medicine, New Haven, CT 06511 USA.
3. Institute of Biomedical Studies, Baylor University, Waco, TX 76706 USA.
4. UCB Pharma, Cambridge, MA 02140, USA
5. Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology,
University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA.
Supplemental Data: eTable 1, eTable 2, eFigure 1, eFigure 2, eFigure 3, eFigure 4, eFigure 5, eFigure 6,
eFigure 7.
Search Terms: [132] Autoimmune diseases, [179] Myasthenia, Complement, Autoantibodies.
Submission Type: Article
Title Character Count (with spaces): 117
Number of Tables: 0
Number of Figures: 4
Word Count of Abstract: 350
Word Count of Paper: 4158
Number of References: 49
Corresponding Author:
Kevin C. O’Connor, PhD
Yale School of Medicine
300 George Street Room 353J
New Haven, CT 06511
Ph: 203-737-3321; email: [email protected]
Study funding: KCO was supported by the National Institute of Allergy and Infectious Diseases of the
NIH under award numbers R01-AI114780 and R21 AI164590; through an MGNet pilot grant awarded
through the Rare Diseases Clinical Research Consortia of the NIH (award number U54-NS115054) and
by a High Impact Clinical Research and Scientific Pilot Project award from the Myasthenia Gravis
Foundation of American (MGFA). This project was also supported, in part, by grants to KCO and RJN,
from Ra Pharmaceuticals, now a part of UCB. JLB was supported by the National Eye Institute R01-
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.
2
EY022936 and R21-032399. MLF was supported in part by the SPIN award through Grifols and has
further been supported by a DFG Research fellowship (FI 2471/1-1).
Financial disclosures:
Abeer H. Obaid reports no disclosures.
Dr. Chryssa Zografou reports no disclosures.
Dr. Douangsone D. Vadysirisack is an employee of UCB Ra Pharma.
Dr. Bailey Munro-Sheldon
reports no disclosures.
Dr. Miriam L. Fichtner
reports no disclosures.
Dr. Roy is a consultant for Alexion Pharmaceuticals, (now part of AstraZeneca), and Takeda
Pharmaceuticals.
Dr. William M. Philbrick reports no disclosures.
Dr. Jeffrey L. Bennett reports payment consultation fees for study design/consultation from Viela Bio,
now part of Horizon Therapeutics; and personal fees from Mitsubishi Tanabe Pharma Corporation;
Reistone Bio, AbbVie, Alexion; Chugai, Clene Nanomedicine, Genentech, Genzyme, Mitsubishi-Tanabe,
and Roche; grants from Novartis, Mallinckrodt; and has a patent for Aquaporumab issued.
Dr. Richard J. Nowak has received research support from the National Institutes of Health (NIH),
Genentech, Alexion Pharmaceuticals, argenx, Annexon Biosciences, UCB Ra Pharmaceuticals,
Myasthenia Gravis Foundation of America, Momenta, Immunovant, Grifols, and Viela Bio, now part of
Horizon Therapeutics.
RJN has served as consultant/advisor for Alexion Pharmaceuticals, argenx,
Cabaletta Bio, CSL Behring, Grifols, Ra Pharmaceuticals, now a part of UCB Pharma, Immunovant,
Momenta, and Viela Bio, now part of Horizon Therapeutics.
Dr. Kevin C. O’Connor has received research support from Ra Pharma, now a part of UCB Pharma, and
Alexion, now part of AstraZeneca, and Viela Bio, now part of Horizon Therapeutics. KCO is a consultant
and equity shareholder of Cabaletta Bio. KCO has served as consultant/advisor for Alexion
Pharmaceuticals, now part of AstraZeneca, and for Roche, and he has received speaking fees from
Alexion, Roche, Genentech, and Viela Bio, now part of Horizon Therapeutics.
Statistical Analysis:
Statistical analyses were conducted by Abeer H. Obaid and Bhaskar Roy, MBBS, MHS (Yale School of
Medicine, Department of Neurology).
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
3
Abstract
Background Autoantibodies targeting the acetylcholine receptor (AChR), found in patients with
myasthenia gravis (MG), mediate pathology through three mechanisms: complement-directed tissue
damage, blocking of the acetylcholine binding site, and internalization of the AChR. Clinical assays, used
to diagnose and monitor patients, measure only autoantibody binding. Consequently, they are limited in
providing association with disease burden, understanding of mechanistic heterogeneity, and monitoring
therapeutic response. Objective Develop a cell-based assay that measures AChR autoantibody-mediated
complement membrane attack complex (MAC) formation. Methods An HEK293T cell linemodified
using CRISPR/Cas9 genome editing to disrupt expression of the complement regulator genes (CD46,
CD55 and CD59)was used to measure AChR autoantibody-mediated MAC formation via flow
cytometry. Results Serum samples (n=155) from 96 clinically confirmed AChR MG patients,
representing a wide range of disease burden and autoantibody titer, were tested along with 32 healthy
donor (HD) samples. AChR autoantibodies were detected in 139 of the 155 (89.7%) MG samples via a
cell-based assay. Of the 139 AChR positive samples, autoantibody-mediated MAC formation was
detected in 83 (59.7%), while MAC formation was undetectable in the HD group or AChR positive
samples with low autoantibody levels. MAC formation was positively associated with autoantibody
binding in most patient samples; ratios (MFI) of MAC formation to AChR autoantibody binding ranged
between 0.2748, with a median of 0.79 and interquartile range of 0.43 (0.581.1). However, the
distribution of ratios was asymmetric and included extreme values; 16 samples were beyond the 1090
percentile, with high-MAC to low-AChR autoantibody binding ratio or the reverse. Correlation between
MAC formation and clinical disease scores suggested a modest positive association (rho=0.34,
p=0.0023), which included a subset of outliers that did not follow this pattern. MAC formation did not
associate with exposure to immunotherapy, thymectomy, or MG subtypes defined by age-of-onset.
Conclusions A novel assay for evaluating AChR autoantibody-mediated complement activity was
developed. A subset of patients that lack association between MAC formation and autoantibody binding
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
4
or disease burden was identified. The assay may provide a better understanding of the heterogeneous
autoantibody molecular pathology and identify patients expected to benefit from complement inhibitor
therapy.
Introduction
The impairment in autoimmune myasthenia gravis (MG) is caused by autoantibodies that target
components of the neuromuscular junction (NMJ)
1-3
. The most common MG subtype is characterized by
autoantibodies targeting the acetylcholine receptor (AChR)
1
. Detection of AChR autoantibodies confirms
a diagnosis of MG, however titers (serum concentration) vary widely among patients and within
individuals during their disease, and it is generally recognized that cross-sectional measurements of titer
do not correlate well with disease severity among different patients
4-7
.
AChR autoantibodies are broadly polyclonal and heterogeneous in their specificity; MG serum can
include autoantibodies that can recognize any of the four different AChR subunits (a, b, e/g, or d)
8
. AChR
autoantibodies can use three distinct mechanisms
9-15
to effect pathology: (i) complement-directed tissue
damage, (ii) blocking the binding site for acetylcholine on the receptor, and (iii) modulation
(internalization) of the AChR. Furthermore, there may also be a subgroup of binding-only antibodies that
recognize AChR in clinical assays but do not have any pathogenic properties.
In addition to the lack of understanding concerning the disconnect between autoantibody titer and disease
severity, gaps in our knowledge have recently become more consequential because new therapeutics
specifically targeting the pathogenic mechanisms of AChR autoantibodiesincluding complement
inhibitorshave been introduced
16
. Eculizumab is a recently approved biological therapeutic for the
treatment of generalized AChR MG
17
; it inhibits autoantibody-mediated activity of the terminal
complement cascade. The phase III clinical trial showed efficacy, but approximately 40% of AChR
autoantibody-positive patients did not meet the trial endpoint without longer-term treatment, and some
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
5
experienced exacerbation events requiring rescue therapy
17-19
. These data are challenging to reconcile as
the poor responders had measurable circulating AChR autoantibodies. This trial also highlights the
limitations of AChR autoantibody titer as a biomarker and importantly underscores the need for further
understanding of AChR autoantibody-mediated pathogenic mechanisms so that response to treatments
can be better predicted. To that end, we sought to develop an in vitro assay that measures AChR
autoantibody-mediated complement activation.
Methods
Standard Protocol Approvals, Registrations, and Patient Consent
This study was approved by Yale Universitys Institutional Review Board (clinicaltrials.gov ||
NCT03792659). Informed written consent was received from all participating patients prior to inclusion
in this study. Serum was collected from AChR MG patients at the Yale Medicine Myasthenia Gravis
Clinic
20
. Demographics and clinical data are presented in Table e1.
CRISPR/Cas9 genome editing of HEK293T cells
CD46, CD55, and CD59 CRISPR sgRNAs (Table e2) were cloned into the pSpCas9(BB) -2A-Puro
(PX459) V2.0
21
vector (a gift from Dr. Feng Zhang (Addgene plasmid # 62988)) and were transfected
into human embryonic kidney (HEK)293T cells (ATCC CRL3216). The cells were harvested after 48
hours, and dilutions were used to derive single cell clonal cell lines. These clones were then tested
phenotypically for the deletions of CD46, CD55, and CD59 via PCR, Sanger sequencing, and flow
cytometry. Clones that exhibited deletions underwent FACS sorting to ensure the selection of cells with
the three deletions.
Autoantibody binding live cell-based assay (CBA)
Wild type (WT), triple KO HEK293T, or Chinese hamster ovary (CHO) cells (CCL-61) were transfected
with adult AChR (2α, β, δ, and ε) and rapsyn-GFP plasmids (generous gifts from Drs. Angela Vincent,
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
6
David Beeson, and Patrick Waters) using branched polyethylenimine or Lipofectamine 2000 (CHO only).
After a 24-hour incubation, the media was refreshed, and the CBA was conducted the following day.
Cells were exposed to serum (1:20 dilution) from healthy donors (HD) or MG patients or monoclonal
antibodies (mAbs) (10 µg/ml). Bound antibodies were detected with an anti-human IgG Fcγ (309-605-
008; Jackson ImmunoResearch) with a LSR Fortessa (BD Biosciences). FlowJo software (v10.6.2) was
used for analysis. In serial dilution testing, some serum samples can provide aberrantly low CBA D mean
florescence intensity (MFI) values when tested at 1:20, due to assay saturation and/or interference.
Accordingly, these data points were not considered in the analyses. For testing AQP4 binding, cells were
transiently transfected with an AQP4-GFP (M1 isoform) plasmid
22
, using the CBA conditions described
above.
Complement cell-based assay
Complement competent normal human serum (NHS) and factor B-depleted human serum were both
purchased from Complement Technologies (Tyler, TX). The same lot was used within experiments.
Human recombinant mAbs were expressed as we have previously described
23
.
CD46, CD55, CD59 KO HEK293T cells or CHO cells were prepared as outlined in the live CBA. Cells
were washed twice and resuspended in complete DMEM. Cells were exposed to AChR MG patient
serum, HD serum (both heat-inactivated), or control mAbs suspended in 25% NHS or Factor B depleted
serum. MAC formation was allowed to proceed for three hours at 37°C, then fixed with 2% PFA at 4°C
for 15 minutes. The cells were then stained with anti-C9 neoantigen (Hycult, Cat# HM2264), followed by
washing and secondary staining (APC (Cat # 407109) or PE IgG2a clone RMG2a-62 (Cat# 407109)).
FACS analysis was conducted as described for the binding CBA.
Statistical analysis
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
7
Threshold values for the CBA and complement CBA positivity were determined by the application of a
cutoff value, determined by calculating the mean +4 SD of HD subject samples. For descriptive statistics,
mean, median, or percentage were used as appropriate. Comparison between two groups was performed
using the Mann-Whitney U test, and the Spearman correlation was used to examine the relationship
between two parameters. A box-whisker plot (whisker representing 1090 percentile) was used to
graphically represent the distribution of the ratios to examine the relationship between MAC and AChR
autoantibody binding. P-values below 0.05 were considered significant. All statistical analyses were
performed on Graphpad Prism software (v9.1.2). STROBE cohort reporting guidelines were used.
24
Data Availability
Anonymized data will be shared upon request from qualified investigators and completion of materials
transfer agreements.
Results
Measuring autoantibody-mediated MAC formation using a CBA platform
AChR autoantibodies can be measured with live CBAs
25
. In this assay format, recombinant human AChR
subunits and the clustering protein, rapsyn, are expressed in human embryonic kidney (HEK) cells and
tested for binding of autoantibodies. We sought to measure AChR autoantibody-mediated complement
activity by modifying the clustered AChR CBA. To that end, the assay was performed by using
autoantibodies (recombinant mAbs or autoimmune patient-derived serum, in which the complement was
heat-inactivated) and normal human serum (NHS) as a consistent complement source.
We first evaluated antibody-dependent MAC formation, by detecting a C9 neo-epitope, with the modified
CBA using an established positive control; specifically, human aquaporin (AQP4) autoantibodies from
patients with neuromyelitis optica spectrum disorder (NMOSD), which have been demonstrated to
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
8
efficiently fix complement in vitro
26
. AQP4 was expressed in HEK293T cells, then a human AQP4-
specific recombinant IgG1 subclass mAb (mAb-58
27
) or serum from an AQP4 autoantibody positive
NMOSD patient was added along with NHS. MAC formation was detected when the AQP4-specific mAb
or NMOSD serum and NHS were present. MAC formation was not detected in the absence of a AQP4-
specific antibody source, absence of NHS, or when heat-inactivated NHS was included (eFigure 1A,
eFigure 1B). We next tested AChR autoantibody-mediated MAC formation under the same conditions,
but with AChR-rapsyn expressed in HEK293T cells. An AChR-specific human recombinant IgG1
subclass mAb (mAb-637
28
) and serum from an AChR autoantibody positive patient (MG-87) were tested.
In contrast to the results obtained in the AQP4 system, no AChR-autoantibody-dependent MAC
formation was detected (eFigure 1C).
Most human cells express the complement regulators CD46, CD55, and CD59, which can interfere with
complement deposition. Therefore, we tested the complement CBA with CHO cells, which do not express
these regulators
29
. An AChR-specific cell-based binding assay with CHO cells was established, but
nonspecific background binding by human IgG prevented its use for complement detection (eFigure 2A-
D). A previous study demonstrated that the human HAP1 cell line, in which the expression of
complement regulators was disrupted through gene editing, provided increased complement component
deposition
30
. Thus, we reasoned that a genome-edited HEK293T cell line, in which the complement
regulators CD46, CD55, and CD59 were not expressed, would be an effective tool in evaluating AChR
autoantibody-mediated complement activity. Accordingly, we produced a modified HEK293T cell line,
using CRISPR/Cas9 genome editing to target these genes (eFigure 3A-C, eTable 2). We then confirmed
that autoantibody binding was uncompromised in the modified HEK293T cells (eFigure 4A) and that
AChR was consistently expressed (eFigure 4B).
AChR autoantibody-mediated MAC formation in the absence of complement regulators
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
9
The CD46/55/59 KO HEK293T engineered cells were then tested in the complement CBA, first using the
AQP4 antigen. The AQP4-specific mAb-58 or serum from an AQP4 autoantibody positive NMOSD
patient, was added to the cells and tested for MAC formation as described for the WT HEK cells. MAC
formation was detected when the AQP4-specific mAb or serum was added together with NHS.
Conversely, MAC formation was not detected in the absence of either the AQP4 autoantibody source or
NHS, or when the mAb or serum was added with heat-inactivated NHS (Figure 1A). The AChR-specific
mAb-637, serum from a patient with MG, and healthy controls, which were tested under the same
conditions, did not initiate MAC formation in the AQP4 complement CBA (Figure 1A).
To test the complement CBA with the modified cells for AChR autoantibody-mediated MAC formation,
AChR and rapsyn were co-expressed, and then the 637-mAb was added to the cells and tested for MAC
formation. MAC formation was detected when the AChR-specific mAb and NHS were present (Figure
1B). We next tested MAC formation with serum samples from four AChR autoantibody positive MG
patients and four non-MG serum samples. Two were derived from MG patients (eTable 1) with AChR
autoantibody titersas measured by the clinical diagnostic radioimmunoassay (RIA)—that were
exceptionally high (MG-87 and MG-56, 43nM and 88nM respectively) and two had mid-low range titers
(MG-43 and MG-44, 14.3nM and 1.47nM respectively). Serum from MG-87 mediated robust MAC
formation (Figure 1C). Serum from MG-56 and MG-43 mediated weak MAC formation and MG-44
showed none. HD serum, NMOSD serum, and the AQP4-specific mAb-58 did not mediate MAC
formation on the AChR-expressing cells (Figure 1C).
To confirm that the antibody-mediated classical complement pathway was generating the MAC, we tested
the contribution of the alternative pathway. This was achieved through inhibiting the alternative pathway
by using NHS, in which factor Brequired for the alternative pathwaywas depleted. The MAC was
detected when AQP4-specific mAb-58 and factor B depleted serum were present but not in the absence of
either the autoantibody or factor B depleted serum, or when the mAb was added with heat-inactivated
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
10
factor B depleted serum (Figure 1D). To confirm that the self-antigen was driving autoantibody binding
and subsequent MAC formation, non-transfected cells were tested in the complement CBA using the
AQP4-specific mAb-58, AChR-specific mAb-637, HD serum, and an MG patient serum sample (MG-
87). MAC formation was below or at background levels using the non-transfected cells, indicating the
requirement of antigen recognition in initiating antibody-mediated MAC formation (Figure 1E).
Testing MG patient serum for autoantibody-mediated MAC formation.
Having demonstrated that the complement CBA could measure AChR autoantibody-mediated classical
pathway MAC formation, we next tested MG serum samples to explore how AChR autoantibody-
mediated complement activity was represented in a heterogeneous population of MG patients. MAC
formation was tested using heat inactivated serum from 30 HD subjects (n=32 samples) and from 96
patients (n=155 samples) with a confirmed diagnosis of AChR MG (eTable 1). While all the MG patients
were diagnosed with the AChR disease subtype, we specifically included patient samples that were AChR
autoantibody positive and others that were negative at the time of collection (eTable 1). An assay
positivity cutoff was established using the MAC MFI from the HD cohort (156
±
14 MFI); samples with
an MFI > mean + 4SD of the HD cohort were considered positive. Serum from the HD cohort did not
show any measurable AChR autoantibody-mediated MAC formation (Figure 2A). A subset (83/155,
53.5%) of the MG patient-derived samples, demonstrated AChR autoantibody-mediated MAC formation
(Figure 2A). To determine whether those samples, which did not mediate MAC formation, included
autoantibodies that were detectable by CBA, we next evaluated the association between MAC formation
and AChR autoantibody binding by comparing the complement CBA results to those of the antibody-
binding CBA. There was a significant correlation (rho=0.897; p<0.0001) observed between MAC
formation and AChR autoantibody binding (Figure 2B). Samples for which there was no detectable
binding in the CBA did not mediate detectable MAC formation in the complement CBA (MAC formation
was detected in 83/139 (59.7%) AChR positive samples). Additionally, samples that provided a positive,
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
11
yet low, signal in the binding CBA did not mediate detectable MAC formation. Samples that were
collected longitudinally presented a similar association pattern (eFigure 5).
Despite a strong correlation between MAC formation and AChR autoantibody binding (Figure 2B), we
noted some samples with high MAC values despite relatively low AChR autoantibody binding, and some
with high AChR autoantibody binding with relatively low MAC values, which suggests a degree of
heterogeneity among this population. To further explore the disassociation between MAC formation and
AChR autoantibody binding, we examined the distribution of the ratio of MAC formation to AChR
autoantibody binding. The ratios ranged between 0.2748, with a median of 0.79 and interquartile range
of 0.43 (0.581.1), with an asymmetric, non-normal distribution with extreme values (skewness 5.1 and
kurtosis 26 (leptokurtic)). The box-whisker plot (Figure 2C) shows relatively higher or lower ratios
between MAC and AChR autoantibody binding beyond 1090 percentiles, graphically representing the
aforementioned heterogeneity among this population. These collective results suggest that, for the
majority of MG patient serum AChR autoantibodies, CBA binding positively associates with
autoantibody-mediated CBA MAC formation, but a subset did not follow this pattern.
Comparing titers of MG patient serum autoantibody binding and autoantibody-mediated MAC formation
While CBA-based binding and MAC formation were closely associated for many samples, the results also
suggest that a subset of patients may harbor AChR binding autoantibodies that range broadly in their
ability to mediate MAC formation. Accordingly, 14 samples were selected for further study (Figure 2B),
based on their observed association/disassociation between the MAC formation and AChR autoantibody
binding. Each sample was tested for AChR autoantibody binding and MAC formation at various serum
dilutions (Figure 3, eFigure 6). The binding CBA, which tested serum over three-fold dilutions from
1:201:1350, showed a range of binding titers. The complement CBA, which tested serum over two-fold
dilutions from 1:201:320, showed a wide range of MAC formation. Of interest was that several samples
showed similarly strong AChR autoantibody binding profiles but differed considerably in their MAC
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
12
formation capability. For example, samples MG-20 and MG-34 have comparable binding profiles but
considerably differ in their MAC formation capability; MG-20 mediated robust MAC formation at three
of the serum dilutions tested, while MG-34 did not provide a signal above the cutoff at any serum
dilution. Similarly, MG-49, MG-51, and MG-89 provided strong binding signals, but only MG-89
provided a strong MAC formation signal. These collective results imply that the autoantibodies harbored
by these patients have differing pathogenic capacities irrespective of binding capability.
Association between AChR autoantibody assay data and disease severity, treatment, or MG subtypes.
We first sought to explore the relationship between disease severity and autoantibody binding and
autoantibody-mediated MAC formation. We first categorized the MG patients into two subgroups using
the MGFA clinical classification: asymptomatic/ocular disease (MGFA = 0/I) and mild-to-severe
generalized disease (MGFA ³ II) severity (eTable 1). Patients with higher disease severity showed both
higher CBA binding and MAC formation when compared to those with minimal disease burden as
measured by the MGFA clinical classification (p <0.0001 and p <0.0001 respectively) (Figure 4A and
B). We next assessed the relationship between AChR autoantibody binding and MAC formation with the
MG composite score (MGC)—a weighted disease severity score scaleby Spearman correlation. Both
binding and MAC formation showed mild-to-moderate correlation with MGC (rho = 0.437, p = 0.0001;
rho = 0.3512, p = 0.0023 respectively) (Figure 4C and 4D). Samples that were collected longitudinally
presented a similar qualitative association between MAC formation, binding, and disease severity
(eFigure 5). However, several samples in both the cross sectional (Figure 4) and longitudinal (eFigure 5)
analyses stood out because of their lack of a positive association, reflecting a similar disassociation to that
which we observed between binding and MAC formation. For example, MG37 had a moderate to high
disease score, yet low antibody binding and MAC formation values. The opposite case was also found
(MG-52) where both binding and MAC formation values were high relative to other samples, but the
disease scores were low (eFigure 5). We also examined if there were differences in autoantibody binding
and autoantibody-complement mediated MAC formation in specific MG patient subsets. Differences
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
13
between treatment naïve and patients receiving immunotherapy, early-onset MG (EOMG) and late-onset
MG (LOMG), and patients with or without thymectomy (eFigure 7) were evaluated. None of the subsets
showed any significant differences in terms of MAC formation and CBA binding. Collectively, these
findings suggest that antibody-mediated complement activity moderately associates with disease burden,
but some patients with mild and others with more severe disease have respectively high or low
autoantibody-mediated MAC formation activity.
Discussion
Detection of AChR autoantibodies confirms a diagnosis of MG, however the discordance between titer
and disease severity may be, in part, due to how AChR autoantibody titer is measured. Specifically, the
clinical assays used to diagnose patients and provide titer values are unable to discriminate between the
detection of the autoantibodies and their pathogenic properties (effector functions). Importantly,
nonpathogenic AChR binding-only autoantibodies, which would be detected in clinical assays, may also
exist, further complicating the disconnect between disease severity and autoantibody titer. Such
nonpathogenic binding autoantibodies have been identified in other autoimmune conditions including
pemphigus
31
and NMOSD
32
. The nonpathogenic NMOSD autoantibodies, which are thought to be poor at
activating complement due to altered Fc regions, compete with pathogenic autoantibodies for antigen
binding
32
. Our data revealed a positive association between autoantibody binding and MAC formation,
suggesting that a considerable fraction of circulating AChR autoantibodies can initiate the complement
cascade. However, binding alone did not account for the variation observed in complement activity. We
reason that heterogeneous autoantibody characteristics, among other factors, may play an integral role in
determining MAC formation efficiency. Such characteristics would minimally include subunit and
epitope specificity, affinity, IgG subclass usage, Fc structure, and post-translation modifications (PTM),
all of which can alter complement activating capability in addition to structure and stability
33, 34
. Although
it is currently unclear how AChR autoantibody heterogeneity affects MAC formation in MG patients,
parallels to the mechanisms governing AQP4 autoantibody-mediated pathology in NMOSD provide
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
14
insight. AQP4-binding autoantibodies display broad efficiency in inducing complement-mediated cell
death
26
. Here, a combination of distinct properties, including particular epitope binding and the assembly
of multimeric antigen platforms, are required for ideal activation of autoantibody complement-mediated
cell death
26
. Specifically, AQP4 autoantibodies that target a distinct epitope on the AQP4 extracellular
loop C demonstrate remarkably enhanced complement activity when compared to autoantibodies
recognizing other epitopes. Furthermore, AQP4 can form supramolecular orthogonal arrays that arrange
these epitopes in a manner that benefits autoantibody multimeric complexes, which support autoantibody
Fc-Fc interactions that are critical for efficient C1q binding and subsequent complement activity
26
.
Whether or not similar mechanisms occur in the context of AChR-specific MG has yet to be explored.
However, given the variable complement activity shown here and that AChR is tightly clustered by the
intracellular scaffolding protein, rapsyn, such organized formations of self-antigen may support Fc-Fc
interactions facilitating efficient complement activation, by AChR autoantibodies recognizing
particularbut not yet identifiedepitopes. Thus, we propose that circulating AChR autoantibodies
include subtypes that contribute to heterogeneous pathogenetic mechanisms and show variable efficiency
in affecting these mechanisms, and finally that their composition varies among patients and perhaps
longitudinally within a patient. We further suggest that AChR autoantibody subunit specificity and
epitope specificity is linked to these effector functions, including activation of the classical complement
pathway.
We recognize that this study includes limitations. First, measuring complement formation on live cells
with flow cytometry presents challenges given that the cells may be actively lysing or dying and therefore
not detected. We empirically arrived at a single timepoint after the addition of NHS that provided
measurable differences in MAC formation in the AQP4 and AChR complement CBAs when comparing
patient samples and controls. This approach was chosen in consideration of assay ease-of-use.
Longitudinal measurements after the addition of NHS or different approaches that measure cumulative
cell death, such as Cr
52
release, may provide additional information on AChR autoantibody-mediated
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
15
activity, but were outside the scope of our study. Second, autoantibody-mediated MAC formation was not
detected in a number of samples, most of which showed lower, but still positive, CBA binding. It is
reasonable to conclude that these samples do indeed include AChR autoantibodies capable of mediating
MAC formation. However, the sensitivity of the complement CBA may require further optimization to
detect their activity. Finally, we recognize that the in vitro autoantibody-mediated complement activity
may not fully reflect an in vivo complement response. We are using an in vitro system with diminished
expression of complement inhibitory proteins. Our system reflects the capability of the autoantibody to
initiate complement but does not consider the varying levels of complement inhibitory protein expression
on various muscle cell surfaces. In this regard, the assay only simulates more complex activity at the
NMJ; however, the clinical binding CBA also lacks any representation of the NMJ other than antigen
expression, but it is recognized as being highly reliable for diagnosis of MG. Further emphasizing this
point, it is challenging to understand why it was required to remove the complement regulators, as they
are present in the NMJ. It may be related to in vitro assay sensitivity, but further study is required to
understand this observation. Interestingly, this requirement has been consistently observed in other
studies that investigated pathophysiologic properties of AChR autoantibodies
35
using in vitro assays and
in vivo models; specifically, CD55 knockout mice, which are more susceptible to the effects of
pathogenic MG autoantibodies
36-38
. These regulators have also been implicated in ocular MG, where
reduced expression levels of CD55 and CD59 in extraocular muscles offer a potential explanation for why
these muscle subgroups are highly associated with MG
36
. To reflect the NMJ-specific conditions more
accurately, our assay could be complemented by other approaches. For example, motor neuronsthe
target cells in patients with multifocal motor neuropathy (MMN)have been used for investigating the
pathogenicity of MMN autoantibodies
39
. In similar fashion, functional consequences of complement
damage could be assessed with NMJ-specific cell types.
We observed a modest correlation between autoantibody-mediated complement activity and disease
burden. While additional mechanisms must be considered, this suggests that in some patients
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
16
complement activity may make major contributions to pathology. Conversely, patients who did not fit this
pattern, for example those with high disease burden and low complement activity, may harbor
autoantibodies that mediate pathology through other mechanisms (blocking and/or modulating).
Furthermore, we did not observe differences in autoantibody-mediated complement activity between
EOMG and LOMG, which suggests that the properties of the autoantibodies may be similar in these
major MG subsets. Immunomodulatory therapy and thymectomy did not reveal diminished complement
activity. Autoantibody titer often persists following such treatments
40, 41
, thus our findings suggest that
these treatments may not alter either the properties or relative frequency of complement activating
autoantibodies.
It is anticipated that measuring AChR autoantibody-mediated pathogenesis will have clinical utility. The
last decade has seen a considerable number of new and existing therapeutics become available for the
treatment of MG
16
. Included are biologics that directly target the autoantibody effector function
(eculizumab and others)
46
. Clinical trials of these treatments have provided outcomes demonstrating
benefit in some patients but unexpected failure in others, thus intensifying challenges to the field,
including the need to target the correct therapy to patients from the growing number of options, and the
need to inform treatment decisions through understanding the immunopathology. Therapeutic
complement inhibition provides one such example of our limited understanding of AChR MG
immunomechanisms: it is unclear why eculizumab (approved for the treatment of generalized AChR
MG
17
) does not yield clinical benefit in some AChR autoantibody-positive patients
17, 18
, even with longer-
term treatment
19
. The AChR autoantibody titers of the patients who responded to eculizumab treatment
neither predicted treatment efficacy nor correlated with its progression. In addition to emphasizing the
limitations of AChR autoantibody titer as a biomarker, these studies also underscore the need to further
understand AChR autoantibody mechanisms so that response to treatments can be better predicted. Future
experiments, which include testing samples from complement inhibitor trials, could be performed to
investigate whether the complement CBA is valuable in forecasting response. To this end, we suggest a
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
17
suite of clinical assays that measure the composition of AChR autoantibodies with variable pathogenic
mechanisms will be valuable. These assays would ideally measure binding-only, classical complement
activation as shown here, and modulating and blocking functions, which have been demonstrated with
similar CBA platforms
48, 49
. These collective measurements may be useful for assessing disease
progression and serve as an improved MG biomarker when compared to autoantibody binding. Thus,
patients would be provided with the opportunity for a more individualized treatment plan that targets their
unique autoantibody-mediated pathogenic pathways.
Acknowledgements
The authors thank Drs. Soumya Yandamuri, Gianvito Masi, Philip Coish, and Ms. Sarah Ohashi for
critical reading of the manuscript.
References
1. Vincent A. Unravelling the pathogenesis of myasthenia gravis. Nat Rev Immunol 2002;2:797-
804.
2. Gilhus NE, Skeie GO, Romi F, Lazaridis K, Zisimopoulou P, Tzartos S. Myasthenia gravis -
autoantibody characteristics and their implications for therapy. Nat Rev Neurol 2016;12:259-268.
3. Yi JS, Guptill JT, Stathopoulos P, Nowak RJ, O'Connor KC. B cells in the pathophysiology of
myasthenia gravis. Muscle Nerve 2018;57:172-184.
4. Vincent A, Newsom Davis J. Anti-acetylcholine receptor antibodies. J Neurol Neurosurg
Psychiatry 1980;43:590-600.
5. Lefvert AK, Bergstrom K, Matell G, Osterman PO, Pirskanen R. Determination of acetylcholine
receptor antibody in myasthenia gravis: clinical usefulness and pathogenetic implications. J Neurol
Neurosurg Psychiatry 1978;41:394-403.
6. Lindstrom JM, Seybold ME, Lennon VA, Whittingham S, Duane DD. Antibody to acetylcholine
receptor in myasthenia gravis. Prevalence, clinical correlates, and diagnostic value. Neurology
1976;26:1054-1059.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
18
7. Tindall RS. Humoral immunity in myasthenia gravis: clinical correlations of anti-receptor
antibody avidity and titer. Ann N Y Acad Sci 1981;377:316-331.
8. Tzartos SJ, Barkas T, Cung MT, et al. Anatomy of the antigenic structure of a large membrane
autoantigen, the muscle-type nicotinic acetylcholine receptor. Immunol Rev 1998;163:89-120.
9. Lindstrom JM, Engel AG, Seybold ME, Lennon VA, Lambert EH. Pathological mechanisms in
experimental autoimmune myasthenia gravis. II. Passive transfer of experimental autoimmune myasthenia
gravis in rats with anti-acetylcholine recepotr antibodies. J Exp Med 1976;144:739-753.
10. Sterz R, Hohlfeld R, Rajki K, et al. Effector mechanisms in myasthenia gravis: end-plate function
after passive transfer of IgG, Fab, and F(ab')2 hybrid molecules. Muscle Nerve 1986;9:306-312.
11. Howard FM, Jr., Lennon VA, Finley J, Matsumoto J, Elveback LR. Clinical correlations of
antibodies that bind, block, or modulate human acetylcholine receptors in myasthenia gravis. Ann N Y
Acad Sci 1987;505:526-538.
12. Drachman DB, Angus CW, Adams RN, Michelson JD, Hoffman GJ. Myasthenic antibodies
cross-link acetylcholine receptors to accelerate degradation. N Engl J Med 1978;298:1116-1122.
13. Drachman DB, Adams RN, Josifek LF, Self SG. Functional activities of autoantibodies to
acetylcholine receptors and the clinical severity of myasthenia gravis. N Engl J Med 1982;307:769-775.
14. Kao I, Drachman DB. Myasthenic immunoglobulin accelerates acetylcholine receptor
degradation. Science 1977;196:527-529.
15. Gomez CM, Richman DP. Anti-acetylcholine receptor antibodies directed against the alpha-
bungarotoxin binding site induce a unique form of experimental myasthenia. Proc Natl Acad Sci U S A
1983;80:4089-4093.
16. Nguyen-Cao TM, Gelinas D, Griffin R, Mondou E. Myasthenia gravis: Historical achievements
and the "golden age" of clinical trials. J Neurol Sci 2019;406:116428.
17. Howard JF, Jr., Utsugisawa K, Benatar M, et al. Safety and efficacy of eculizumab in anti-
acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3,
randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol 2017;16:976-986.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
19
18. Muppidi S, Utsugisawa K, Benatar M, et al. Long-term safety and efficacy of eculizumab in
generalized myasthenia gravis. Muscle Nerve 2019;60:14-24.
19. Howard JF, Jr., Karam C, Yountz M, O'Brien FL, Mozaffar T, Group RS. Long-term efficacy of
eculizumab in refractory generalized myasthenia gravis: responder analyses. Ann Clin Transl Neurol
2021;8:1398-1407.
20. Anil R, Kumar A, Alaparthi S, et al. Exploring outcomes and characteristics of myasthenia
gravis: Rationale, aims and design of registry - The EXPLORE-MG registry. J Neurol Sci
2020;414:116830.
21. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the
CRISPR-Cas9 system. Nat Protoc 2013;8:2281-2308.
22. Stathopoulos P, Chastre A, Waters P, et al. Autoantibodies against Neurologic Antigens in
Nonneurologic Autoimmunity. J Immunol 2019;202:2210-2219.
23. Takata K, Stathopoulos P, Cao M, et al. Characterization of pathogenic monoclonal
autoantibodies derived from muscle-specific kinase myasthenia gravis patients. JCI Insight
2019;4:e127167.
24. von Elm E AD, Egger M, Pocock SJ, Gotzsche PC, Vandenbroucke JP. . The Strengthening the
Reporting of Observational Studies in Epidemiology (STROBE) Statement: guidelines for reporting
observational studies.
25. Rodriguez Cruz PM, Huda S, Lopez-Ruiz P, Vincent A. Use of cell-based assays in myasthenia
gravis and other antibody-mediated diseases. Exp Neurol 2015;270:66-71.
26. Soltys J, Liu Y, Ritchie A, et al. Membrane assembly of aquaporin-4 autoantibodies regulates
classical complement activation in neuromyelitis optica. J Clin Invest 2019;130:2000-2013.
27. Bennett JL, Lam C, Kalluri SR, et al. Intrathecal pathogenic anti-aquaporin-4 antibodies in early
neuromyelitis optica. Ann Neurol 2009;66:617-629.
28. Graus YF, de Baets MH, Parren PW, et al. Human anti-nicotinic acetylcholine receptor
recombinant Fab fragments isolated from thymus-derived phage display libraries from myasthenia gravis
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
20
patients reflect predominant specificities in serum and block the action of pathogenic serum antibodies. J
Immunol 1997;158:1919-1929.
29. Saifuddin M, Hedayati T, Atkinson JP, Holguin MH, Parker CJ, Spear GT. Human
immunodeficiency virus type 1 incorporates both glycosyl phosphatidylinositol-anchored CD55 and
CD59 and integral membrane CD46 at levels that protect from complement-mediated destruction. J Gen
Virol 1997;78 ( Pt 8):1907-1911.
30. Thielen AJF, van Baarsen IM, Jongsma ML, Zeerleder S, Spaapen RM, Wouters D.
CRISPR/Cas9 generated human CD46, CD55 and CD59 knockout cell lines as a tool for complement
research. J Immunol Methods 2018;456:15-22.
31. Payne AS, Ishii K, Kacir S, et al. Genetic and functional characterization of human pemphigus
vulgaris monoclonal autoantibodies isolated by phage display. J Clin Invest 2005;115:888-899.
32. Tradtrantip L, Yeaman MR, Verkman AS. Cytoprotective IgG antibodies in sera from a subset of
patients with AQP4-IgG seropositive neuromyelitis optica spectrum disorder. Sci Rep 2021;11:21962.
33. Alter G, Ottenhoff THM, Joosten SA. Antibody glycosylation in inflammation, disease and
vaccination. Semin Immunol 2018;39:102-110.
34. Quast I, Keller CW, Maurer MA, et al. Sialylation of IgG Fc domain impairs complement-
dependent cytotoxicity. J Clin Invest 2015;125:4160-4170.
35. Jacob S, Viegas S, Leite MI, et al. Presence and pathogenic relevance of antibodies to clustered
acetylcholine receptor in ocular and generalized myasthenia gravis. Arch Neurol 2012;69:994-1001.
36. Kaminski HJ, Li Z, Richmonds C, Lin F, Medof ME. Complement regulators in extraocular
muscle and experimental autoimmune myasthenia gravis. Exp Neurol 2004;189:333-342.
37. Lin F, Kaminski HJ, Conti-Fine BM, Wang W, Richmonds C, Medof ME. Markedly enhanced
susceptibility to experimental autoimmune myasthenia gravis in the absence of decay-accelerating factor
protection. J Clin Invest 2002;110:1269-1274.
38. Kaminski HJ, Kusner LL, Richmonds C, Medof ME, Lin F. Deficiency of decay accelerating
factor and CD59 leads to crisis in experimental myasthenia. Exp Neurol 2006;202:287-293.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
21
39. Harschnitz O, van den Berg LH, Johansen LE, et al. Autoantibody pathogenicity in a multifocal
motor neuropathy induced pluripotent stem cell-derived model. Ann Neurol 2016;80:71-88.
40. Nowak RJ, Coffey CS, Goldstein JM, et al. Phase 2 Trial of Rituximab in Acetylcholine Receptor
Antibody-Positive Generalized Myasthenia Gravis: The BeatMG Study. Neurology 2021.
41. Jiang R, Hoehn KB, Lee CS, et al. Thymus-derived B cell clones persist in the circulation after
thymectomy in myasthenia gravis. Proc Natl Acad Sci U S A 2020;117:30649-30660.
42. Diaz-Manera J, Martinez-Hernandez E, Querol L, et al. Long-lasting treatment effect of rituximab
in MuSK myasthenia. Neurology 2012;78:189-193.
43. Robeson KR, Kumar A, Keung B, et al. Durability of the Rituximab Response in Acetylcholine
Receptor Autoantibody-Positive Myasthenia Gravis. JAMA Neurol 2016.
44. Cree BAC, Bennett JL, Kim HJ, et al. Inebilizumab for the treatment of neuromyelitis optica
spectrum disorder (N-MOmentum): a double-blind, randomised placebo-controlled phase 2/3 trial. Lancet
2019.
45. Hewett K, Sanders DB, Grove RA, et al. Randomized study of adjunctive belimumab in
participants with generalized myasthenia gravis. Neurology 2018;90:e1425-e1434.
46. Howard JF, Jr., Barohn RJ, Cutter GR, et al. A randomized, double-blind, placebo-controlled
phase II study of eculizumab in patients with refractory generalized myasthenia gravis. Muscle Nerve
2013;48:76-84.
47. Howard JF, Jr., Bril V, Burns TM, et al. Randomized phase 2 study of FcRn antagonist
efgartigimod in generalized myasthenia gravis. Neurology 2019;92:e2661-e2673.
48. Lozier BK, Haven TR, Astill ME, Hill HR. Detection of acetylcholine receptor modulating
antibodies by flow cytometry. American journal of clinical pathology 2015;143:186-192; quiz 305.
49. Cromar A, Lozier BK, Haven TR, Hill HR. Detection of Acetylcholine Receptor Blocking
Antibodies by Flow Cytometry. American journal of clinical pathology 2016;145:81-85.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
22
Figure Legends
Figure 1. Autoantibody-mediated complement fixation assay with CD46/55/59 KO HEK293T cells.
Autoantibodies were tested for their ability to mediate complement activation through measuring MAC
formation via FACS on live cells, transfected with self-antigens, in which the complement regulators
(CD46, CD55, and CD59) were knocked out. Normal human serum (NHS) was added as consistent
complement source. Control conditions included omitted NHS or heat-inactivated NHS. All tested patient
or HD serum samples were heat-inactivated. A. Measurement of MAC formation using an in vitro
complement CBA with AQP4 transfected CD46/55/59 KO HEK293T cells. Complement fixation was
tested with AQP4-specific autoantibody sources (anti-AQP4 monoclonal mAb-58 IgG1 and NMOSD
serum) and negative control autoantibody sources (anti-AChR mAb-637 IgG1 and serum from MG
patients and HDs). NHS and HI NHS bars represents the mean of duplicate experimental conditions while
No NHS bars represent singlets. B. Representative FACS analysis of AChR transfected CD46/55/59 KO
HEK293T cells using an in vitro complement CBA. AChR transfected cells were tested without (top row)
or with the anti-AChR mAb-637 IgG1 (bottom row). C. Measurement of MAC formation using the
complement CBA with AChR transfected CD46/55/59 KO HEK293T. Complement fixation was tested
with AChR-specific autoantibody sources (anti-AChR mAb-637 IgG1 and serum from MG patients) and
negative control autoantibody sources (anti-AQP4 monoclonal mAb-58 IgG1 and serum from NMOSD
patients and HDs). NHS and HI NHS bars represents the mean of duplicate experimental conditions while
No NHS bars represent singlets. D. Factor B depleted NHS was used in the complement CBA to test
whether the alternative pathway contributed to autoantibody-mediated MAC formation. AQP4 transfected
cells were tested without (top row) or with the anti-AQP4 monoclonal mAb-58 IgG1 (bottom row). E.
Non-transfected CD46/55/59 KO HEK293T cells were tested in the complement CBA using antigen-
specific monoclonal antibodies (mAb-637 or mAb-58) or serum from MG patients and HDs to test for the
requirement of autoantigens in MAC formation. Samples used in panels A–E: mAb-58 IgG1 (anti-AQP4
monoclonal), NMOSD patient (NMOSD-1), mAb-637 IgG1 (anti-AChR monoclonal), MG patient serum
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
23
(MG-87, MG-43, MG-56, MG-44), and healthy donors (HD-9, HD-29, HD-30). Blankno serum,
NHSnormal human serum, HI NHSheat-inactivated normal human serum, Factor B-dHSfactor B
depleted human serum, HI Factor B-dHSheat-inactivated factor B depleted human serum.
Figure 2. AChR autoantibody-mediated complement formation in MG patients. AChR
autoantibodies in serum were tested for their ability to mediate complement activation through measuring
MAC formation on live AChR-expressing CD46/55/59 KO HEK293T cells. A. Comparison of MAC
formation using serum samples (n=32) from HDs and serum samples (n=155) from AChR MG patients.
The cutoff for positivity was set at the mean MFI + 4 STD (210.9 MFI) of the HD samples. Each data
point represents the mean of triplicate experimental conditions. B. Correlation between MAC formation
and AChR autoantibody binding by CBA. Spearman correlation was used to calculate the relationship.
The binding CBA cutoff for positivity was set at the mean MFI +4STD (21.91 DMFI) of the HD samples.
Each data point represents the mean of triplicate experimental conditions. Colored (gold) points represent
samples selected for further evaluation (see Figure 3). C. Graphical representation of heterogeneity in
MAC formation and AChR autoantibody binding of MG samples. Cross sectional samples (n=89) were
analyzed through a ratio of MAC formation and autoantibody binding, to observe disassociation between
AChR autoantibody binding and complement formation. The box-whisker plot shows the median (0.79)
and 1090 percentile (outlined by the whiskers) of the MAC to AChR autoantibody binding ratios. The
dots represent data outside of the 1090 percentile. The Y-axis is not to scale and was divided in three
sections for better visual representation. All samples were measured in triplicate, and those with negative
ΔMFI binding values (n=5) were excluded from the analysis.
Figure 3. Heterogeneous AChR autoantibody-mediated complement formation. Selected samples
(indicated by golden points in Figure 2B) were examined side-by-side for both MAC formation and CBA
binding. Each graph shows the data collected from individual serum samples. The X-axis represents the
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
24
serum dilution of the sample tested for complement CBA (MAC) formation (blue dots) or CBA binding
(golden dots). For AChR autoantibody binding (left y-axis), samples were tested at serum dilutions of 1:20
plus 4 additional 3-fold dilutions (1:50, 1:150, 1:450, and 1:1350). For MAC formation (right y-axis),
samples were tested at 2-fold serial dilutions (1:20, 1:40, 1:80, 1:160, and 1:320). Each data point represents
the mean of experimental triplicate experimental conditions. Dotted horizonal lines mark the positive
reactivity cutoff for complement (blue) and CBA binding (golden). Cutoffs were calculated using the mean
MFI + 4STD of the HD samples (complement and CBA 96.01 MFI and 75.35 DMFI respectively). Data
from additional samples are shown in eFigure 6.
Figure 4. Correlation between clinical disease measurements and autoantibody-mediated
complement formation. Cross-sectional samples were assessed for correlation between MGFA
classification or MG composite (MGC) score and the binding CBA or complement CBA data. A–B.
Samples with low disease severity (MGFA 0/I) were compared to samples with higher disease severity
(MGFA II-V) for differences in (A) autoantibody binding (median MGFA (0/I): 327.3 (II-V): 748.2; p-
value <0.0001) or (B) MAC formation (median MGFA (0/I): 190.3 (II-V): 468.3; p-value <0.0001). C–D.
Spearman correlation was used to calculate the relationship between cross-sectional sample MGC scores
and (C) autoantibody binding (rho 0.437, p-value = 0.0001) or (D) MAC formation (rho 0.351, p-value =
0.0023). Each data point in A–D represents the mean of triplicate experimental conditions. White and
grey lines in A and B represent the median and quartiles respectively.
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted March 6, 2022. ; https://doi.org/10.1101/2021.10.05.21264566doi: medRxiv preprint