You are on page 1of 34

Articles

Safety, efficacy, and tolerability of efgartigimod in patients


with generalised myasthenia gravis (ADAPT):
a multicentre, randomised, placebo-controlled, phase 3 trial
James F Howard Jr, Vera Bril, Tuan Vu, Chafic Karam, Stojan Peric, Temur Margania, Hiroyuki Murai, Malgorzata Bilinska, Roman Shakarishvili,
Marek Smilowski, Antonio Guglietta, Peter Ulrichts, Tony Vangeneugden, Kimiaki Utsugisawa, Jan Verschuuren, Renato Mantegazza,
and the ADAPT Investigator Study Group*

Summary
Lancet Neurol 2021; 20: 526–36 Background There is an unmet need for treatment options for generalised myasthenia gravis that are effective,
See Comment page 499 targeted, well tolerated, and can be used in a broad population of patients. We aimed to assess the safety and efficacy
*Study group members are in the of efgartigimod (ARGX-113), a human IgG1 antibody Fc fragment engineered to reduce pathogenic IgG autoantibody
appendix (pp 15–23) levels, in patients with generalised myasthenia gravis.
Department of Neurology,
The University of Methods ADAPT was a randomised, double-blind, placebo-controlled, phase 3 trial done at 56 neuromuscular
North Carolina at Chapel Hill,
Chapel Hill, NC, USA
academic and community centres in 15 countries in North America, Europe, and Japan. Patients aged at least 18 years
(Prof J F Howard Jr MD); Ellen & with generalised myasthenia gravis were eligible to participate in the study, regardless of anti-acetylcholine receptor
Martin Prosserman Centre for antibody status, if they had a Myasthenia Gravis Activities of Daily Living (MG-ADL) score of at least 5
Neuromuscular Diseases, (>50% non-ocular), and were on a stable dose of at least one treatment for generalised myasthenia gravis. Patients
University Health Network,
University of Toronto, Toronto,
were randomly assigned by interactive response technology (1:1) to efgartigimod (10 mg/kg) or matching placebo,
ON, Canada (V Bril MD); administered as four infusions per cycle (one infusion per week), repeated as needed depending on clinical response
Department of Neurology, no sooner than 8 weeks after initiation of the previous cycle. Patients, investigators, and clinical site staff were all
University of South Florida masked to treatment allocation. The primary endpoint was proportion of acetylcholine receptor antibody-positive
Morsani College of Medicine,
Tampa, FL, USA (Prof T Vu MD);
patients who were MG-ADL responders (≥2-point MG-ADL improvement sustained for ≥4 weeks) in the first
Penn Neuroscience treatment cycle. The primary analysis was done in the modified intention-to-treat population of all acetylcholine
Center-Neurology, Hospital of receptor antibody-positive patients who had a valid baseline MG-ADL assessment and at least one post-baseline
the University of Pennsylvania,
MG-ADL assessment. The safety analysis included all randomly assigned patients who received at least one dose or
Philadelphia, PA, USA
(C Karam MD); Neurology Clinic, part dose of efgartigimod or placebo. This trial is registered at ClinicalTrials.gov (NCT03669588); an open-label
Clinical Center of Serbia, extension is ongoing (ADAPT+, NCT03770403).
Faculty of Medicine, University
of Belgrade, Belgrade, Serbia
Findings Between Sept 5, 2018, and Nov 26, 2019, 167 patients (84 in the efgartigimod group and 83 in the placebo
(S Peric MD); Department of
Neurology and group) were enrolled, randomly assigned, and treated. 129 (77%) were acetylcholine receptor antibody-positive. Of
Neurorehabilitation, these patients, more of those in the efgartigimod group were MG-ADL responders (44 [68%] of 65) in cycle 1 than in
New Hospitals, Tbilisi, Georgia the placebo group (19 [30%] of 64), with an odds ratio of 4·95 (95% CI 2·21–11·53, p<0·0001). 65 (77%) of 84 patients
(T Margania MD); Department
in the efgartigimod group and 70 (84%) of 83 in the placebo group had treatment-emergent adverse events, with the
of Neurology, School of
Medicine, International most frequent being headache (efgartigimod 24 [29%] vs placebo 23 [28%]) and nasopharyngitis (efgartigimod ten
University of Health and [12%] vs placebo 15 [18%]). Four (5%) efgartigimod-treated patients and seven (8%) patients in the placebo group had a
Welfare, Narita, Japan serious adverse event. Three patients in each treatment group (4%) discontinued treatment during the study. There
(Prof H Murai MD); Department
were no deaths.
and Clinic of Neurology,
Wroclaw Medical University,
Wroclaw, Poland Interpretation Efgartigimod was well tolerated and efficacious in patients with generalised myasthenia gravis. The
(M Bilinska MD); Sarajishvili individualised dosing based on clinical response was a unique feature of ADAPT, and translation to clinical practice
Institute of Neurology and
Neurosurgery, Tbilisi, Georgia
with longer term safety and efficacy data will be further informed by the ongoing open-label extension.
(R Shakarishvili MD);
Department of Hematology Funding argenx.
and Bone Marrow
Transplantation, Medical
University of Silesia, Katowice,
Copyright © 2021 Elsevier Ltd. All rights reserved.
Poland (M Smilowski MD);
argenx, Ghent, Belgium Introduction Most patients with generalised myasthenia gravis
(A Guglietta MD, P Ulrichts PhD, Generalised myasthenia gravis is a rare, chronic, auto­ (about 85%) have IgG autoantibodies, most often directed
T Vangeneugden PhD);
Department of Neurology,
immune disease that causes debilitating and potentially against the skeletal muscle nicotinic acetylcholine receptor
Hanamaki General Hospital, life-threatening muscle weakness affecting ocular motility, and less frequently against muscle-specific tyrosine kinase
Hanamaki, Japan swallowing, speech, mobility, and respiratory function, (MUSK) and low-density lipoprotein receptor-related
(K Utsugisawa MD); Department which can significantly impair independence and quality protein 4 (LRP4).2,3 A small proportion of patients have no
of Neurology, Leiden
of life.1 identifiable antibodies. These autoantibodies exert a direct

526 www.thelancet.com/neurology Vol 20 July 2021


Articles

University Medical Center,


Research in context Leiden, Netherlands
(Prof J Verschuuren MD);
Evidence before this study autoantibody status. During the 26-week study, efgartigimod Department of
We searched PubMed up to Nov 5, 2020, for relevant clinical was well tolerated, with most adverse events being either mild Neuroimmunology and
studies in generalised myasthenia gravis, with no language or moderate in severity. Additionally, significantly more Neuromuscular Diseases,
Fondazione Istituto
restrictions. Key search terms included “neonatal Fc receptor”, patients in the efgartigimod group than in the placebo group Neurologico Carlo Besta, Milan,
“IgG recycling”, “antibody fragment”, and “autoantibody had clinically meaningful improvements in their Myasthenia Italy (R Mantegazza MD)
reduction”. Although preclinical and early-phase studies had Gravis Activities of Daily Living and Quantitative Myasthenia Correspondence to:
been completed with some neonatal Fc receptor antagonists, Gravis scores. Prof James F Howard Jr,
we found no phase 3 studies in generalised myasthenia gravis. Department of Neurology,
Implications of all the available evidence The University of North Carolina
Additionally, although therapeutic plasma exchange had shown
The study used four validated myasthenia gravis-specific at Chapel Hill, Chapel Hill,
the effect of IgG reduction, there were no previous NC 27599, USA
outcome measures, utilising both patient-reported and
pharmacological approaches to achieving such a targeted howardj@neurology.unc.edu
physician-reported information, to assess the effects of
reduction in IgG. Finally, many of the studies in myasthenia See Online for appendix
efgartigimod in patients with generalised myasthenia gravis.
gravis had been constrained by other factors, such as only
Notably, the primary and some secondary endpoints required
including patients who have acetylcholine receptor
patients to have a clinically meaningful improvement in the
autoantibodies.
associated outcome measure, and for this improvement to
Added value of this study persist for at least 4 weeks. The data suggest that reduction of
Although there are treatment options available to patients with pathogenic IgG antibodies through inhibition of neonatal Fc
generalised myasthenia gravis, they are frequently receptor recycling might be an effective approach to the
burdensome, have substantial side-effects, do not alleviate treatment of a broad population of generalised myasthenia
symptoms, or are reserved for refractory patients. ADAPT was gravis and should encourage research in other IgG-mediated
the largest clinical trial in patients with generalised myasthenia autoimmune diseases.
gravis and the only one to include patients regardless of their

pathogenic effect in generalised myasthenia gravis and recycled by FcRn (eg, IgM or IgA).15 Following cellular
their mechanisms of action include functional blockade, uptake, the Fc region of an IgG antibody binds two FcRn
accelerated internali­ sation, and degradation of acetyl­ receptors under acidic conditions in the endosome.16 IgGs
choline receptors and activation of complement.4–7 These bound to FcRn are rescued from lysosomal degradation
actions lead to reduced density of functional acetylcholine and released at physiological pH outside the cell.17–20
receptors and damage to the neuromuscular junction, Therefore, FcRn perpetuates the availability of IgG auto­
resulting in impaired neuromuscular transmission.4–7 Most antibodies in IgG-mediated diseases such as generalised
acetylcholine recep­tor and LRP4 antibodies are of the IgG1 myasthenia gravis. The utility of removing autoantibodies
subtype, which can activate complement, whereas the in generalised myasthenia gravis has been demonstrated
IgG4 subtype, which includes MUSK antibodies, do not.2,8–10 by the effectiveness of plasma exchange and immuno­
Existing treatments, including corticosteroids and non- adsorption; however, their use is limited by availability
steroidal immunosuppressive therapies (NSISTs), broadly and requirement for specialised facilities.21 Blocking
suppress the immune system and do not selectively FcRn is a rational therapeutic approach to target the key
target IgG autoantibodies that are central to generalised pathogenic driver in generalised myasthenia gravis.
myasthenia gravis pathophysiology.7 Moreover, these Efgartigimod (ARGX-113) is a human IgG1 antibody
treat­ments frequently provide insufficient symptom relief Fc-fragment, a natural ligand of FcRn, that has been
and are associated with burdensome side-effects such as engineered for increased affinity to FcRn compared with
glucose intolerance, weight gain, arterial hypertension, endogenous IgG while retaining the characteristic pH
osteoporosis, gastrointestinal issues, bradycardia, and dependence.22 It outcompetes endogenous IgG binding,
renal dysfunction, which can limit their use.7 Another thereby reducing IgG recycling and increasing IgG
therapeutic approach has been to block complement degrada­tion.22 In phase 1 and 2 trials, efgartigimod
activation, targeting one of the downstream pathogenic significantly reduced concentrations of all IgG subtypes
pathways triggered specifically by acetylcholine receptor without decreasing levels of other immunoglobulins
antibodies.11,12 Overall, there remains a significant unmet or albumin, which is also recycled by FcRn.22,23 These
need for generalised myasthenia gravis treatment options reductions were associated with clinically meaningful and
that are effective, targeted, well tolerated, and can be used sustained improvements in generalised myasthenia gravis
in a broad population of patients.2,13,14 symptoms and activities of daily living. The phase 3
The neonatal Fc receptor (FcRn) is an MHC class I-like ADAPT study aimed to assess the safety and efficacy of
molecule that recycles IgG, extending its half-life by about efgartigimod, in patients with generalised myasthenia
four times that of other immunoglobulins that are not gravis.23

www.thelancet.com/neurology Vol 20 July 2021 527


Articles

Methods at individual centres, randomisation was done across,


Study design rather than within, the centres. Investi­gators, patients,
ADAPT was a randomised, double-blind, placebo- study person­nel, clinic staff, and the funder remained
controlled, multicentre, phase 3 trial of efgartigimod in masked to treatment assignments for the duration of
patients with generalised myasthenia gravis. Patients were the study.
recruited from 56 neuromuscular academic and com­
munity centres across Japan and 14 countries in Europe Procedures
and North America (appendix pp 2–11). Independent Patients were screened for the study during a 2-week
ethics committees and international review boards period, which was followed by a 26-week treatment period.
provided written approval for the study protocol and all Efgartigimod (10 mg/kg) or matching placebo was
amendments. The trial was conducted according to the administered as four infusions per cycle (one infusion per
principles outlined in the Declaration of Helsinki. week). After each cycle there was a period of at least
5 weeks of follow-up. All patients received an initial
Participants cycle, with subsequent cycles administered according to
Patients aged at least 18 years with generalised myasthenia individual clinical response when MG-ADL score was at
gravis, with or without acetylcholine receptor antibodies, least 5 (with >50% MG-ADL non-ocular) and, if the patient
were eligible if their disease was categorised as Myasthenia was an MG-ADL responder, when they no longer had a
Gravis Foundation of America class II to IV and they had a clinically meaningful decrease (MG-ADL clinically mean­
Myasthenia Gravis Activities of Daily Living (MG-ADL) ing­ful improvement defined as having ≥2-point improve­
score of at least 5 (with >50% of the MG-ADL score due ment in total MG-ADL score) compared with baseline.
to non-ocular symptoms). Diagnosis was supported by Subsequent cycles could commence no sooner than
a history of abnormal neuro­muscular transmission tests, a 8 weeks from initiation of the previous cycle; a maximum
positive edrophonium chloride test, or improvement with of three cycles were possible in the 26-week study. Patients
acetylcholinesterase inhibitors. Eligibility criteria also who required rescue therapy were discon­ tinued from
required patients to be on a stable dose of at least one study treatment. Patients who completed the study or
treatment for generalised myasthenia gravis (ie, acetylcho­ could not complete a cycle before study end (retreatment
linesterase inhibitors, corticosteroids, or NSISTs) before after day 126) were able to rollover to the ongoing open-label
screening and through­out the trial. There was no require­ extension study (ADAPT+; NCT03770403).
ment for specific generalised myasthenia gravis therapies. Efficacy was assessed with the MG-ADL scale
Patients were excluded if they had received rituximab or (patient-reported, physician-recorded outcome measure);24
eculizumab in the 6 months before screening, undergone Quantitative Myasthenia Gravis (QMG) score (physi­cian
thymectomy within 3 months, had intravenous immuno­ assessed, including quanti­­ta­tive measures; clinically mean­
globulin or plasma exchange within 1 month of screening, ing­ful improvement ≥3-point reduction),25 Myasthenia
had active hepatitis B, were seropositive for hepatitis C, Gravis Composite (MGC) scale (patient and physician
seropositive for HIV with low CD4 count, had serum IgG assessed; clinically meaning­ ful improvement ≥3-point
levels less than 6 g/L at screening, or were pregnant. reduction);26 the 15-item revised version of the Myasthenia
A complete list of inclusion and exclusion criteria is in the Gravis Quality of Life (MG-QOL15r)27 question­­naire
appendix (pp 11–12). Potential patients were recruited (patient completed), and EQ5D quality of life scale (patient
through the investigators’ practice or via physician referral. completed).
All patients provided written informed consent before Assessments were done weekly for 8 weeks after initia­
starting the study. tion of each cycle and then every 2 weeks up to 26 weeks.
Pharmacodynamic effects were analysed using
Randomisation and masking validated assays of total IgG, IgG subtypes (IgG1, IgG2,
Patients were randomly assigned in a 1:1 ratio to either IgG3, and IgG4), and autoantibodies (anti-acetylcholine
efgartigimod or placebo. Placebo was matched to receptor antibodies for the acetylcholine receptor
efgartigimod in appearance and supplied in identical con­ antibody-positive patients and antibodies against MUSK
tainers. Randomisation was performed centrally using for the MUSK antibody-positive patients). The validated
interactive response technology, using both web and voice assays were a radioimmunoassay (IBL International,
systems, by an independent company (SGS, Zwijnaarde, Hamburg, Germany) that used acetylcholine receptor
Belgium) that held randomisation codes until after the labelled with ¹²⁵I-α-bungarotoxin for anti-acetylcholine
final database lock. Randomisation was based on three receptor antibodies and an ELISA (IBL International) for
stratification factors: acetylcholine receptor antibody anti-MUSK antibodies.
status (positive vs negative), NSISTs (taking vs not taking),
and Japanese nationality (yes vs no). The stratification Outcomes
factors were selected to ensure con­sistency of effect across The primary efficacy endpoint was the proportion of
antibody status, concomitant medication, and ethnic­ acetylcholine receptor antibody-positive patients who were
ities. Due to the small number of patients anticipated MG-ADL responders in the first treatment cycle. An

528 www.thelancet.com/neurology Vol 20 July 2021


Articles

MG-ADL responder was defined as a patient who had at


least a 2-point improvement (reduction) in MG-ADL 216 patients screened

score, sustained for at least 4 consecutive weeks, with the


first improvement occurring by week 4 of the cycle (1 week 49 ineligible
32 did not meet inclusion criteria
after the fourth infusion). Secondary endpoints were 1 sponsor decision
assessed in hierarchical order, as follows: (1) proportion of 2 withdrew consent
QMG responders (defined as a ≥3 point improvement in 11 seronegative cap reached
3 other*
the total QMG score for ≥4 consecutive weeks with the
first improvement occurring by week 4 of cycle 1) in
the acetylcholine receptor antibody-positive population; (2) 167 patients enrolled
percentage of MG-ADL responders in cycle 1 in the overall
population (ie, acetylcholine receptor antibody-positive
and antibody-negative patients); (3) proportion of time
84 randomly assigned to efgartigimod 83 randomly assigned to placebo
patients showed a clinically meaningful improve­ment in
MG-ADL score in the acetylcholine receptor antibody-
positive population, up to day 126; (4) time from 84 received efgartigimod 83 received placebo
day 28 (1 week after the fourth infusion in cycle 1) to
not having clinically meaningful improvement in the
4 discontinued in first treatment cycle 9 discontinued in first treatment cycle
acetylcholine receptor antibody-positive population; and 2 adverse event 3 adverse event
(5) proportion of early MG-ADL responders in cycle 1 1 protocol deviation 1 prohibited medications
1 rescue therapy 2 rescue therapy
(MG-ADL responders with first MG-ADL improvement of 1 sponsor decision
≥2 points occurring by week 2) in the acetylcholine 2 patient withdrew
receptor antibody-positive population.
Safety was assessed through incidence of adverse events 80 completed first treatment cycle 74 completed first treatment cycle
and changes in clinical laboratory values and vital signs,
and on electrocardiograms. Tertiary endpoints were
pharma­co­dynamics and immunogenicity. 1 discontinued in second treatment cycle, 1 discontinued in second treatment cycle,
adverse event patient withdrew
Predefined exploratory endpoints assessed time to onset
of effect; magnitude of effect, including proportion of
patients achieving minimal symptom expression (defined 79 completed treatment 73 completed treatment
as MG-ADL score of 0 or 1) and proportion of patients
with increasing levels of MG-ADL and QMG score Figure 1: Trial profile
improve­ ment in each cycle; duration of response in *Absence of laboratory results on the day before the final day of the screening period.
MG-ADL responders; repeatability of effect with second
treatment cycle; and the change in Myasthenia Gravis overall population with a two-sided α level of 5%, allowing
Composite and MG-QOL15r scores. for a 10% dropout rate.
Efficacy analyses were done in the modified intention-
Statistical analysis to-treat population, including all randomly assigned
The proportion of MG-ADL responders in the placebo patients who had a valid baseline MG-ADL assessment
group was hypothesised to be 30%. The treatment and at least one post-baseline MG-ADL assessment.
difference was assumed to be 35% for acetylcholine Safety analyses were done in all patients who received at
receptor antibody-positive patients and 5% for acetyl­ least one dose or part of a dose. Patients were discontinued
choline receptor antibody-negative patients. A difference from treatment if they became pregnant, received rescue
of total MG-ADL responder rate of 35% between therapy (plasma exchange, intravenous immunoglobulin,
the placebo and efgartigimod primary acetylcholine immunoadsorption, any new type of corticosteroid, or
recep­tor antibody-positive population is considered increased dose of current steroid; rescue was permitted
clinically relevant. In the phase 2 ARGX-113-1602 study,23 per protocol in case of new or worsening respiratory or
the total MG-ADL responder rate was 33% for placebo bulbar symptoms or at least 2-point increase of individual
(three of 12) and 75% (nine of 12) for efgartigimod. non-ocular MG-ADL items), developed a serious adverse
Sample size was based on allowing enrolment of up to event that could jeopardise the safety of the patient, or
20% acetylcholine receptor antibody-negative patients. developed a bacterial, viral, or fungal infection (evaluated
Based on this quota, a sample size of 150 provided power on a case-by-case basis). After discontinuation, patients
of 96% in the primary population of acetylcholine who did not withdraw con­sent were followed up for safety
receptor antibody-positive patients to detect a difference and disease severity assessments through the rest of
of 35% in the proportion of responders with 120 patients. the trial.
The sample size also provided a power of 90% to detect a Statistical analyses were done using SAS, version 9.2 or
29% difference in the proportion of responders in the higher, and the software package R, where applicable. The

www.thelancet.com/neurology Vol 20 July 2021 529


Articles

Percentage of time patients showed a clinically meaningful


All patients Acetylcholine receptor
antibody-positive patients improve­ ment in MG-ADL score in the acetylcholine
receptor antibody-positive population was analysed using
Efgartigimod Placebo Efgartigimod Placebo
group group group group an ANCOVA model. In this analysis, randomised treat­
(n=84) (n=83) (n=65) (n=64) ment group and the stratifica­tion variables were included
Age, years 45·9 (14·4) 48·2 (15·0) 44·7 (15·0) 49·2 (15·5) as factors, and baseline total MG-ADL score was included
Sex as a covariate. Time from day 28 to not having clinically
Female 63 (75%) 55 (66%) 46 (71%) 40 (63%) meaningful improvement in the acetylcholine receptor
Male 21 (25%) 28 (34%) 19 (29%) 24 (38%)
antibody-positive population was estimated using Kaplan-
Race
Meier time-to-event analysis and compared by means of
Asian 9 (11%) 7 (8%) 7 (11%) 4 (6%)
a stratified log-rank test, stratified for the stratification
variables. Additional endpoints assessing efficacy, safety,
Black or African American 3 (4%) 3 (4%) 1 (2%) 3 (5%)
pharmacodynamics, and immunogenicity were analysed
White 69 (82%) 72 (87%) 54 (83%) 56 (88%)
in a descriptive manner. This study is registered at
Other* 3 (4%) 1 (1%) 3 (5%) 1 (2%)
ClinicalTrials.gov (NCT03669588).
Time since generalised myasthenia gravis 10·1 (9·0) 8·8 (7·6) 9·7 (8·3) 8·9 (8·2)
diagnosis, years
MGFA class at screening Role of the funding source
II 34 (40%) 31 (37%) 28 (43%) 25 (39%)
The funder of the study had a role in study design, data
III 47 (56%) 49 (59%) 35 (54%) 36 (56%)
collection, data interpretation, data analysis, and writing
IV 3 (4%) 3 (4%) 2 (3%) 3 (5%)
of the report.
Previous thymectomy 59 (70%) 36 (43%) 45 (69%) 30 (47%)
Acetylcholine receptor antibody-positive 65 (77%) 64 (77%) 65 (100%) 64 (100%)
Results
216 patients were screened between Sept 5, 2018, and
MUSK antibody-positive 3 (4%) 3 (4%) 0 0
Nov 26, 2019, of whom 167 (84 in the efgartigimod group
Acetylcholine receptor or MUSK antibody- 16 (19%) 16 (19%) 0 0
negative and 83 in the placebo group) were enrolled, randomly
Total MG-ADL score 9·2 (2·6) 8·8 (2·3) 9·0 (2·5) 8·6 (2·1) assigned, and treated (figure 1). 129 (77%) were acetyl­
Total Quantitative Myasthenia Gravis score 16·2 (5·0) 15·5 (4·6) 16·0 (5·1) 15·2 (4·4) choline receptor antibody-positive and 38 (23%) were
Total Myasthenia Gravis Composite score 18·8 (6·1) 18·3 (5·5) 18·6 (6·1) 18·1 (5·2)
acetylcholine receptor antibody-negative, of whom six (4%)
Total MG-QOL15r score 16·1 (6·4) 16·8 (5·7) 15·7 (6·3) 16·6 (5·5)
were MUSK antibody-positive. There were five treatment
At least one previous NSIST 62 (74%) 57 (69%) 47 (72%) 43 (67%)
discontinuations in the efgartigimod group and ten in the
Myasthenia gravis therapy at baseline
placebo group.
Patient characteristics were representative of the
Any steroid 60 (71%) 67 (81%) 46 (71%) 51 (80%)
general generalised myasthenia gravis population and
Any NSIST 51 (61%) 51 (61%) 40 (62%) 37 (58%)
were well balanced between the efgartigimod and placebo
Steroid and NSIST 43 (51%) 44 (53%) 34 (52%) 31 (48%)
groups (table 1), except more patients in the efgartigimod
No steroid or NSIST 16 (19%) 7 (8%) 13 (20%) 6 (9%)
group had previously undergone thymectomy than in the
Data are mean (SD) or n (%). MG-ADL=Myasthenia Gravis Activities of Daily Living. MGFA=Myasthenia Gravis placebo group. The mean time since thymectomy in
Foundation of America. MG-QOL15r=15-item revised version of the Myasthenia Gravis Quality of Life.
NSIST=non-steroidal immunosuppressant therapy. *Includes American Indian or Alaska Native (n=2) and multiple these patients was 10·84 (SD 9·0) years.
(n=1) for the efgartigimod group, and not reported for the placebo group (n=1). Most patients (144 [86%] of 167) were receiving
immunosuppressive treatment (steroids or NSISTs).
Table 1: Baseline demographic and clinical characteristics
48 (29%) had never previously been treated with an
NSIST. The mean baseline MG-ADL and QMG scores
primary endpoint was tested by means of a two-sided exact demonstrate substantial disease burden despite ongoing
test using a logistic regression model with baseline total generalised myasthenia gravis treatment.
score as a covariate and the three stratification factors as The number of cycles of assigned treatment received by
variables. The treatment effect was presented as an odds patients during the study is in the appendix (p 13). In
ratio (OR) with 95% CI and two-sided p value. If the efgartigimod-treated patients, the median duration of
primary endpoint met significance at the 5% two-sided cycle 1 (time from first infusion in cycle 1 until first
α level, secondary endpoints were tested at a 5% two-sided infusion in cycle 2 or final visit of study) was 10 weeks
significance level in hierarchical order using a fixed- (IQR 71–113 days) and for placebo-treated patients, the
sequence approach. The secondary endpoints of QMG median duration was 10 weeks (71–141 days).
responders in cycle 1 in the acetylcholine receptor A significantly higher proportion of acetylcholine
antibody-positive population, MG-ADL responders in receptor antibody-positive patients in the efgartigimod
cycle 1 in the overall population, and percentage of early group (44 [68%] of 65) were MG-ADL responders in cycle 1
MG-ADL responders in cycle 1 in the acetylcholine receptor (primary endpoint) than in the placebo group (19 [30%]
antibody-positive population were tested using the same of 64; OR 4·95 [95% CI 2·21–11·53], p<0·0001; table 2).
logistic regression model as for the primary endpoint. Additionally, a significantly greater proportion of patients

530 www.thelancet.com/neurology Vol 20 July 2021


Articles

Efgartigimod group Placebo group OR (95% CI) p value


MG-ADL responder in cycle 1 (primary endpoint) 44/65 (68%) 19/64 (30%) 4·95 (2·21–11·53) <0·0001
Quantitative Myasthenia Gravis responder in cycle 1 41/65 (63%) 9/64 (14%) 10·84 (4·18–31·20) <0·0001
MG-ADL responder in cycle 1 (all patients) 57/84 (68%) 31/83 (37%) 3·70 (1·85–7·58) <0·0001
Percentage of time with ≥2-point improvement in MG-ADL up to day 126 48·7% 26·6% ·· 0·0001
Median time from day 28 until no clinically meaningful improvement, days 35 (18–71) 8 (1–57) ·· 0·26
Early MG-ADL responder (cycle 1) 37/65 (57%) 16/64 (25%) ·· Not assessed*
Data are n/N (%), or median (IQR), unless stated otherwise. Analyses were done in acetylcholine receptor antibody-positive patients unless otherwise stated.
MG-ADL=Myasthenia Gravis Activities of Daily Living. *Secondary endpoints were tested in hierarchical order. The fifth secondary endpoint was not assessed because the
fourth secondary endpoint was not significant.

Table 2: Summary of primary and secondary endpoints

A B
0
Mean change in Quantitative Myasthenia Gravis score

–1 –1
Mean change in MG-ADL score

–2
–2
–3
* * *
–3 –4

–5
*
–4 * *
* *
–6
* Efgartigimod group *
*
–5 * Placebo group –7
* * *

C D
0 0
Mean change in Myasthenia Gravis Composite scale

–1 –1
–2
–2
Mean change in MG-QOL15r

–3
–3
–4
*
–5 * –4

–6 –5
–7 *
–6
–8 * *
* –7
–9 * *
*
–8 *
–10 *
* *
* *
–11 –9
0 1 2 3 4 5 6 7 8 10 0 1 2 3 4 5 6 7 8 10
Time since baseline (weeks) Time since baseline (weeks)

Figure 2: Change in MG-ADL (A), Quantitative Myasthenia Gravis score (B), Myasthenia Gravis Composite scale (C), and MG-QOL15r (D) during cycle 1,
in acetylcholine receptor antibody-positive patients
Error bars show standard error. MG-ADL=Myasthenia Gravis Activities of Daily Living. MG-QOL15r=15-item revised version of the Myasthenia Gravis Quality of Life
questionnaire. *p<0·05.

in the efgartigimod group (41 [63%] of 65) were QMG sustained through week 7 in all measures (figure 2). The
responders in cycle 1 than in the placebo group (nine [14%] maximum improvement in efgartigimod treated patients
of 64; OR 10·84 [95% CI 4·18–31·20], p<0·0001; table 2). occurred at week 5 for MG-QOL15r and week 4 for other
Patients in the efgartigimod group had greater total measures.
mean score improvements in MG-ADL, QMG, MCG, and A greater proportion of patients in the efgartigimod
MG-QOL15r in cycle 1, with statistically significant group than in the placebo group achieved higher levels of
differences from baseline observed from week 1 and improvement in MG-ADL (up to 9-point reduction) and

www.thelancet.com/neurology Vol 20 July 2021 531


Articles

(1–57) with placebo (p=0·26 log-rank test; table 2).


A
Efgartigimod group
Although a log-rank test was not significant, a Wilcoxon
9 14·3% 0% Placebo group test done post hoc was significant (p=0·013).
Among cycle 1 MG-ADL responders, the duration of
responder status was 6–7 weeks in 14 (32%) of 44 patients,
Minimum improvement in MG-ADL score

8 20·6% 1·7%
8–11 weeks in ten (23%) patients, and 12 weeks or more
7 27·0% 3·3%
in 15 (34%) patients (data not shown).
6 39·7% 8·3%
In patients who received a second cycle, a greater
proportion of patients in the efgartigimod group (36 [71%]
5 55·6% 11·7% of 51) were MG-ADL responders compared with the
placebo group (11 [26%] of 43), with similar rates to cycle 1.
4 63·5% 23·3% Similar to cycle 1, there was a greater total mean score
improvement in MG-ADL and QMG in the efgartigimod
3 73·0% 36·7%
group than in the placebo group in the second treatment
2 77·8% 48·3% cycle (data not shown). Of the 44 acetylcholine receptor
antibody-positive patients in the efgartigimod group
who were MG-ADL responders in cycle 1, 32 qualified
B for retreatment and 29 (90%) of these were MG-ADL
10 25·8% 0%
responders again in cycle 2. Among 21 patients in the
efgartigimod group who were not MG-ADL responders
9 33·9% 0% during cycle 1, 19 were retreated and seven (37%) of these
Minimum improvement in Quantitative

were MG-ADL responders in cycle 2. Six (86%) of seven


8 37·1% 1·7% patients in the efgartigimod group who received a third
Myasthenia Gravis score

cycle were MG-ADL responders (data not shown).


7 45·2% 1·7%
Predefined exploratory analyses did not show any
6 50·0% 5·2% efficacy differences based on gender, age, or baseline
MG-ADL (data not shown). Concomitant use of NSISTs
5 59·7% 12·1% also did not affect efficacy, with 18 (72%) MG-ADL
responders of the 25 acetylcholine receptor antibody-
4 64·5% 15·5%
positive patients in the efgartigimod group who were
3 74·2% 25·9%
not on NSISTs. Of the 45 acetylcholine receptor antibody-
positive efgartigimod-treated patients with previous
100 80 60 40 30 0 20 40 60 80 100 thymectomy, 27 (60%) were MG-ADL responders, com­
Cumulative percentage pared with 17 (85%) of 20 patients who had not previously
Figure 3: Minimum point improvement in MG-ADL (A) and Quantitative Myasthenia Gravis (B) score in
undergone thymectomy.
cycle 1, in acetylcholine receptor antibody-positive patients Results in the overall population were similar to those in
Minimum improvements 1 week after the last infusion of cycle 1 (week 4). MG-ADL=Myasthenia Gravis Activities the acetylcholine receptor antibody-positive population,
of Daily Living. including significantly more patients in the efgartigimod
group (57 [68%] of 84) who were MG-ADL responders in
QMG (up to 10-point reduction) score at week 4 (figure 3). cycle 1 than in the placebo group (31 [37%] of 83; OR 3·70
26 (40%) of 65 patients in the efgartigimod group attained [95% CI 1·85–7·58], p<0·0001; table 2).
an MG-ADL score of 0 or 1 (minimal symptom expres­ Predefined exploratory analyses showed that in
sion) in cycle 1 compared with seven (11%) of 63 in the acetylcholine receptor antibody-negative patients, there
placebo group (p<0·0001). was a similar number of MG-ADL responders in each
More patients in the efgartigimod group than the treatment group in cycle 1: 13 (68%) of 19 patients in the
placebo group were early MG-ADL responders (table 2). efgartigimod group versus 12 (63%) of 19 in the placebo
In the 44 acetylcholine receptor antibody-positive group. There were more QMG responders in the
MG-ADL responders in the efgartigimod group, the onset efgartigimod group than in the placebo group in cycle 1:
of response occurred by week 2 in 37 (84%) patients. ten (53%) versus seven (37%) patients. Six (32%) patients
Efgartigimod-treated patients showed a clinically in the efgartigimod group achieved minimal symptom
meaningful improvement in MG-ADL score for 48·7% of expression in cycle 1 versus three (16%) in the placebo
the time between start of study and day 126, compared group (data not shown).
with 26·6% of the same period in the placebo group A post-hoc analysis of acetylcholine receptor antibody-
(p=0·0001; table 2). negative patients assessed the proportion of patients who
The median time from day 28 to not having clinically were both MG-ADL and QMG responders in cycle 1:
meaningful improvement over the course of the study nine (47%) of 19 patients in the efgartigimod group
was 35 days (IQR 18–71) with efgartigimod and 8 days and four (21%) of 19 in the placebo group. Among the

532 www.thelancet.com/neurology Vol 20 July 2021


Articles

acetylcholine receptor antibody-negative patients, six were


Efgartigimod group Placebo group
MUSK antibody-positive, three in each treatment group. (n=84) (n=83)
All six patients were MG-ADL responders in cycle 1.
Any adverse event 65 (77%) 70 (84%)
In acetylcholine receptor antibody-positive patients,
Any serious adverse event 4 (5%) 7 (8%)
there were mean maximum reductions of 61·3% (SD 0·9)
Any adverse event leading to 3 (4%) 3 (4%)
in IgG and 57·6% (1·3) in acetylcholine receptor anti­ discontinuation of study drug
bodies, 1 week after the fourth infusion in cycle 1 Any infection 39 (46%) 31 (37%)
(appendix p 14). Levels returned to baseline by week 12 Infusion-related reaction event 3 (4%) 8 (10%)
(9 weeks after the last infusion of cycle 1). Reductions
Most common adverse events
were similar across subtypes, with mean maximum
Headache 24 (29%) 23 (28%)
reductions of 68% (1·0) for IgG1, 60% (1·7) for IgG2,
Nasopharyngitis 10 (12%) 15 (18%)
63% (1·2) for IgG3, and 52% (1·7) for IgG4. Similar
Nausea 7 (8%) 9 (11%)
reductions in IgG and acetylcholine receptor antibodies
Diarrhoea 6 (7%) 9 (11%)
were seen with each treatment cycle. However, no
Upper respiratory tract 9 (11%) 4 (5%)
reductions in albumin levels were observed. infection
No deaths occurred during the study in either the Urinary tract infection 8 (10%) 4 (5%)
efgartigimod or placebo groups. 65 (77%) of 84 patients in
Data are n (%).
the efgartigimod group and 70 (84%) of 83 in the placebo
group had adverse events, with the most frequent being Table 3: Summary of adverse events in all patients
headache, nasopharyngitis, nausea, diarrhoea, upper
respiratory tract infections, and urinary tract infections
(table 3). Incidence of headache was similar between with generalised myasthenia gravis were consistent across
groups; nausea, diarrhoea, and nasopharyngitis occurred four myasthenia gravis-specific scales, and these benefits
in more placebo patients, and upper respiratory tract were observed early and were reproducible and durable.
infections and urinary tract infections occurred in more The study enrolled a broad population of patients
efgartigimod patients. Most adverse events were mild or with generalised myasthenia gravis, including both
moderate in severity, with nine (11%) patients with severe antibody-positive and acetylcholine receptor antibody-
events in the efgartigimod group and eight (10%) in the negative patients, with no requirement for patients to
placebo group. Four (5%) efgartigimod-treated patients have had specific myasthenia gravis medication. Most
had a serious adverse event, which were thrombocytosis, patients were receiving steroids or NSISTs; however,
rectal adenocarcinoma, myasthenia gravis worsening about 30% had not previously received an NSIST.
(each leading to treatment discontinuation), and depres­ At enrolment, despite ongoing myasthenia gravis
sion. In the placebo group, seven (8%) patients had a therapy, patients still had poor scores on the myasthenia
serious adverse event, including one case each of myo­ gravis strength and function scales. Treatment with
cardial ischaemia, atrial fibrillation, and spinal ligament efgartigimod provided significant, clinically meaningful,
ossification, which all led to treatment discontinuation. and durable clinical benefit to most of these patients.
The remaining events were upper respiratory infec­ Many patients had improvement beyond the clinically
tion, spinal compression fracture, myasthenia gravis meaningful threshold, achieving up to 9-point reductions
worsening, and myasthenia gravis crisis. Adverse events in MG-ADL and 10-point reductions in QMG. Minimal
related to infections occurred in 39 (46%) patients in symptom expression was achieved by 40% of acetyl­choline
the efgartigimod group and 31 (37%) in the placebo receptor antibody-positive efgartigimod-treated patients.
group. All infections were reported as mild-to-moderate Most patients had a clinically meaningful improvement in
severity except for three severe events, which were MG-ADL within 2 weeks of starting treatment. Although
influenza and pharyngitis in the efgartigimod group and 68% of acetylcholine receptor antibody-positive patients
upper respiratory tract infections in the placebo group. were MG-ADL responders with the first treatment cycle,
Infusion-related reactions were reported in three (4%) 78% of patients achieved this status during the study with
patients in the efgartigimod group and eight (10%) in the further treatment cycles.
placebo group, all mild in severity. There were no clin­ The early onset of action, observed benefit in patients
ically meaningful changes in haematology or chemistry with or without previous NSIST exposure, and the
parameters (including no decrease in albumin levels), favourable tolerability profile suggest that efgartigimod
electrocardiograms, or vital signs in either group. might be able to be used throughout disease continuum
of patients with generalised myasthenia gravis. Acetyl­
Discussion choline receptor antibodies cause a net reduction of
The ADAPT phase 3 trial showed that efgartigimod was functional acetylcholine receptors at the postsynaptic
well tolerated and efficacious in patients with generalised membrane. However, patients with general­ ised myas­
myasthenia gravis. The reduction in disease burden and thenia gravis also have increased acetylcholine receptor
improvement in strength and quality of life in patients synthesis and repopulation, shown through mRNA and

www.thelancet.com/neurology Vol 20 July 2021 533


Articles

protein production, presumably as compen­satory mech­ with each cycle and across IgG subtypes and a similar
anisms.28,29 Because of this, the reduction of acetylcholine reduction in the antibody-negative patients. The reduction
receptor antibodies by efgartigimod after one infusion in acetylcholine receptor autoantibodies was similar to
could lead to a corresponding increase in acetylcholine that of IgG, and both paralleled the improve­ments in
receptors at the postsynaptic membrane and potentially symptoms. This showed that selective removal of IgG is
account for the early onset of effect. an effective treatment approach in generalised myasthenia
Although more patients in the efgartigimod group had gravis, which is in line with the data available from
previously undergone thymectomy, a post-hoc analysis plasma exchange, a treatment that removes auto­ anti­
showed that the proportion of patients who were MG-ADL bodies and is considered highly efficacious but is limited
responders was lower in patients with previous thymec­ by its administrative challenges.
tomy. Therefore, the increased prevalence of thymectomy Existing treatments for generalised myasthenia gravis
in the efgartigimod group did not appear to favour are associated with burdensome short-term and long-
efgartigimod. term side-effects that can limit their use. Efgartigimod
This phase 3 study tested efgartigimod adminis­ was well tolerated in this study, with most adverse events
tered in treatment cycles, with the frequency of cycles mild or moderate in severity and low incidence of
defined by the duration of clinical effect in each patient. infusion reactions. Although headache was the most
This individualised approach to treatment according to common adverse event, it occurred in equal numbers of
patient’s need proved effective, with reproducible efficacy patients in both treatment groups. Efgartigimod did not
after a second and third cycle. reduce albumin levels, demonstrating its selectivity for
A third of acetylcholine receptor antibody-positive the IgG binding site of FcRn and suggesting that it does
MG-ADL responders maintained a clinically meaningful not alter the function of FcRn.
improvement in MG-ADL score for more than 12 weeks, The rate of infections is of special interest because
suggesting that a proportion of patients have clinical patients with myasthenia gravis are predisposed to infec­
benefit beyond the reduction in IgG and acetylcholine tions, probably exacerbated by concomitant immuno­
receptor antibodies. Production of sufficient acetylcholine suppressive treatments.31,32 In the efgartigimod-treated
receptor to restore the safety factor for neurotransmission group, 46% of patients had an infection compared with
might explain the prolonged effect in some patients, with 37% in the placebo group. Most infections were mild
appropriate reserves established to maintain neurotrans­ to moderate, with only two graded as severe in the
mission even after return of acetylcholine receptor efgartigimod-treated patients. Although longer term data
antibodies to normal levels.30 are required to assess the risk of infection, these results
The secondary endpoint of time from day 28 of cycle 1 are reassuring. Additionally, the action of efgartigimod is
(1 week after the last infusion) until the patient not having selective, with transient and incomplete reduction of IgG
a clinically meaningful improvement was numerically and no effect on other immunoglobulins. Preclinical
greater in the acetylcholine receptor antibody-positive models have also shown that IgG production is not
efgartigimod group than placebo (35 days compared to impaired.33 Because of these factors, efgartigimod-treated
8 days); however, it was not significant (log-rank test, patients should retain the potential to mount an IgG
p=0·26). The log-rank test was not the most appropriate immune response.
test, because the data did not show proportional hazards. Strengths of this study include the randomised placebo-
Patients were likely to require retreatment at some point controlled design, using validated scales incorporating
in the future so the chance of the event occurring was not physician and patient assessment, and endpoints requir­
equal throughout the duration of the study. ing a combination of clinically meaningful improve­ment
68% of acetylcholine receptor antibody-negative and sustained effect. The prolonged response requirement
efgartigimod-treated patients had a response, similar to aimed to reduce the placebo effect and more reliably
that in acetylcholine receptor antibody-positive patients, ascertain the treatment effect of efgartigimod. A broad
but there was an unexpectedly high response rate in the population of patients with generalised myasthenia gravis
placebo group. A post-hoc analysis of acetylcholine was recruited.
receptor antibody-negative patients who were both Study limitations included the length of follow-up,
MG-ADL and QMG responders in cycle 1 showed a treat­ which will be addressed by the ongoing open-label exten­
ment effect, suggesting efgartigimod might be effective in sion study. The retreatment criteria requiring patients’
this patient population. There were only six patients with MG-ADL score to return to less than a 2-point reduction
anti-MUSK antibodies, three in each treatment group, from baseline was a rigorous ADAPT study criterion and
and all six were MG-ADL responders in cycle 1. Further the usefulness in the real world will be established in
information regarding the efficacy in acetylcholine clinical practice. The inclusion of acetylcholine receptor
receptor antibody-negative patients will be gained in the antibody-negative patients was important due to the
ongoing open-label extension trial. limited treatment options these patients have and their
Efgartigimod reduced IgG levels in acetylcholine recep­ lack of inclusion in previous clinical trials. However, only
tor antibody-positive patients, with similar reductions a few of these patients were recruited, and the trial

534 www.thelancet.com/neurology Vol 20 July 2021


Articles

was not statistically powered to assess efficacy in this clinical trial data can be requested by qualified researchers who engage
population. in rigorous independent scientific research and will only be provided
after review and approval of a research proposal and statistical analysis
The results of the phase 3 ADAPT trial suggest that the plan and execution of a data sharing agreement. Data requests can be
novel mechanism of selective IgG reduction through submitted at any time and the data will be accessible for 12 months.
blocking of FcRn with efgartigimod is an effective and Requests can be submitted to ESR@argenx.com.
well tolerated treatment for patients with generalised Acknowledgments
myasthenia gravis. We thank all the study participants, investigators, and trial teams for
their participation in the trial. We would also like to acknowledge the
Contributors
support provided by Benjamin Van Hoorick, Patricia Crabbe,
All authors had full access to the study design information and all data
and Caroline T’joen. Medical writing assistance was provided by Cello
in the study and had final responsibility for the decision to submit for
Health Communications (funded by argenx) and Brant Hubbard
publication. JFH, VB, HM, AG, PU, TVa, JV, and RM contributed to the
(argenx). The ADAPT study was sponsored by argenx.
concept or design of the study. All authors contributed to the analysis
and interpretation of data. JFH, AG, PU, and TVa accessed and verified References
the data in the study. Cello Health Communications, a medical writing 1 Gilhus NE, Tzartos S, Evoli A, Palace J, Burns TM,
company, produced the first draft of the paper based on input and Verschuuren JJGM. Myasthenia gravis. Nat Rev Dis Primers 2019;
direction from all authors. All authors had full access to study data, 5: 30.
reviewed, edited, and provided final approval of the manuscript content, 2 Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup
and had final responsibility for the decision to submit for publication. classification and therapeutic strategies. Lancet Neurol 2015;
14: 1023–36.
Declaration of interests 3 Zisimopoulou P, Evangelakou P, Tzartos J, et al. A comprehensive
JFH has received research support from Alexion Pharmaceuticals, analysis of the epidemiology and clinical characteristics of
argenx, the Centers for Disease Control and Prevention (Atlanta, GA, anti-LRP4 in myasthenia gravis. J Autoimmun 2014; 52: 139–45.
USA), the Muscular Dystrophy Association, the National Institutes of 4 Cole RN, Ghazanfari N, Ngo ST, Gervásio OL, Reddel SW,
Health (NIH; including the National Institute of Neurological Disorders Phillips WD. Patient autoantibodies deplete postsynaptic
and Stroke and the National Institute of Arthritis and Musculoskeletal muscle-specific kinase leading to disassembly of the ACh receptor
and Skin Diseases), Patient Centered Outcomes Research Institute, scaffold and myasthenia gravis in mice. J Physiol 2010; 588: 3217–29.
and Ra Pharmaceuticals (now UCB); honoraria from Alexion 5 Drachman DB, Adams RN, Josifek LF, Self SG. Functional activities
Pharmaceuticals, argenx, Immunovant, Ra Pharmaceuticals (now UCB), of autoantibodies to acetylcholine receptors and the clinical severity
Regeneron Pharmaceuticals, and Viela Bio; and non-financial support of myasthenia gravis. N Engl J Med 1982; 307: 769–75.
from Alexion Pharmaceuticals, argenx, Ra Pharmaceuticals (now UCB), 6 Engel AG, Arahata K. The membrane attack complex of
and Toleranzia. VB has received research support from Commonwealth complement at the endplate in myasthenia gravis. Ann N Y Acad Sci
Serum Laboratories, Grifols, UCB, Bionevia, Shire, and Octapharma. 1987; 505: 326–32.
TVu has served as a speaker for Alexion; has done consulting work for 7 Skeie GO, Apostolski S, Evoli A, et al. Guidelines for treatment of
argenx and UCB; and participated in trials in myasthenia gravis autoimmune neuromuscular transmission disorders. Eur J Neurol
2010; 17: 893–902.
sponsored by NIH, Alexion Pharmaceuticals, argenx, Ra, Viela Bio,
UCB, and Grifols. CK has served on advisory boards for Acceleron, 8 Gilhus NE, Skeie GO, Romi F, Lazaridis K, Zisimopoulou P,
Tzartos S. Myasthenia gravis—autoantibody characteristics and
Akcea, Alexion, Alnylam, argenx, Biogen, CSL Behring, Cytokinetics,
their implications for therapy. Nat Rev Neurol 2016; 12: 259–68.
and Sanofi-Genzyme; and received research grants from Genzyme and
9 Rødgaard A, Nielsen FC, Djurup R, Somnier F, Gammeltoft S.
Akcea. SP reports lecture honoraria from Pfizer, Teva Actavis, Berlin
Acetylcholine receptor antibody in myasthenia gravis: predominance
Chemie Menarini, Mylan, Worwag, Adoc, and Salveo; research grants of IgG subclasses 1 and 3. Clin Exp Immunol 1987; 67: 82–88.
from Kedrion, Octapharma, and Argenx; consultant fees from argenx,
10 Shen C, Lu Y, Zhang B, et al. Antibodies against low-density
Mylan, and Roche; and travel grants from Octapharma, Kedrion, lipoprotein receptor-related protein 4 induce myasthenia gravis.
Teva Actavis, Sanofi Genzyme, Pfizer, Roche, Adoc, and Berlin Chemie J Clin Invest 2013; 123: 5190–202.
Menarini, all outside the submitted work. HM has served as a paid 11 Howard JF Jr. Myasthenia gravis: the role of complement at the
consultant for Alexion Pharmaceuticals, argenx, and Ra Pharma; neuromuscular junction. Ann N Y Acad Sci 2018; 1412: 113–28.
and has received speaker honoraria from the Japan Blood Products 12 Howard JF Jr, Utsugisawa K, Benatar M, et al. Safety and efficacy of
Organization and research support from the Ministry of Health, Labor, eculizumab in anti-acetylcholine receptor antibody-positive
and Welfare, Japan. AG, PU, and TVa are full-time employees of argenx, refractory generalised myasthenia gravis (REGAIN): a phase 3,
Ghent, Belgium. KU has served as a paid consultant for argenx, randomised, double-blind, placebo-controlled, multicentre study.
Ra Pharma, UCB Pharma, Viela Bio, and Regeneron Pharmaceuticals; Lancet Neurol 2017; 16: 976–86.
and has received speaker honoraria from Alexion Pharmaceuticals and 13 Behin A, Le Panse R. New pathways and therapeutic targets in
the Japan Blood Products Organization. JV receives research support autoimmune myasthenia gravis. J Neuromuscul Dis 2018; 5: 265–77.
from Target to B consortium, Prinses Beatrix Spierfonds, and argenx; 14 Gilhus NE. Myasthenia gravis. N Engl J Med 2016; 375: 2570–81.
has been involved in trials or consultancies for argenx, Alexion, 15 Kuo TT, Baker K, Yoshida M, et al. Neonatal Fc receptor:
and Ra pharma; JV is coinventor on patent applications based on from immunity to therapeutics. J Clin Immunol 2010; 30: 777–89.
MUSK-related research; and is a member of the European Reference 16 Jensen PF, Schoch A, Larraillet V, et al. A two-pronged binding
Network for Rare Neuromuscular Diseases; and JV’s institution received mechanism of IgG to the neonatal Fc receptor controls complex
royalties from Immuno-Biological Laboratories. RM has received stability and IgG serum half-life. Mol Cell Proteomics 2017;
personal compensation for consulting, serving on a scientific advisory 16: 451–56.
board, speaking, or other activities with BioMarin, Catalyst, Alexion 17 Ghetie V, Hubbard JG, Kim JK, Tsen MF, Lee Y, Ward ES.
Pharmaceuticals, UCB, and argenx. All other authors declare no Abnormally short serum half-lives of IgG in beta 2-microglobulin-
competing interests. deficient mice. Eur J Immunol 1996; 26: 690–96.
18 Junghans RP, Anderson CL. The protection receptor for IgG
Data sharing catabolism is the beta2-microglobulin-containing neonatal intestinal
argenx is committed to responsible data sharing regarding the clinical transport receptor. Proc Natl Acad Sci USA 1996; 93: 5512–16.
trials they fund. Included in this commitment is access to anonymised, 19 Roopenian DC, Akilesh S. FcRn: the neonatal Fc receptor comes of
individual, and trial-level data (analysis datasets), and other information age. Nat Rev Immunol 2007; 7: 715–25.
(eg, protocols and clinical study reports), as long as the trials are not part 20 Ward ES, Zhou J, Ghetie V, Ober RJ. Evidence to support the
of an ongoing or planned regulatory submission. This includes requests cellular mechanism involved in serum IgG homeostasis in
for clinical trial data for unlicensed products and indications. These humans. Int Immunol 2003; 15: 187–95.

www.thelancet.com/neurology Vol 20 July 2021 535


Articles

21 Liu JF, Wang WX, Xue J, et al. Comparing the autoantibody levels 27 Burns TM, Sadjadi R, Utsugisawa K, et al. International clinimetric
and clinical efficacy of double filtration plasmapheresis, evaluation of the MG-QOL15, resulting in slight revision and
immunoadsorption, and intravenous immunoglobulin for the subsequent validation of the MG-QOL15r. Muscle Nerve 2016;
treatment of late-onset myasthenia gravis. Ther Apher Dial 2010; 54: 1015–22.
14: 153–60. 28 Vincent A. Antibodies and receptors: from neuromuscular junction
22 Ulrichts P, Guglietta A, Dreier T, et al. Neonatal Fc receptor to central nervous system. Neuroscience 2020; 439: 48–61.
antagonist efgartigimod safely and sustainably reduces IgGs in 29 Guyon T, Wakkach A, Poea S, et al. Regulation of acetylcholine
humans. J Clin Invest 2018; 128: 4372–86. receptor gene expression in human myasthenia gravis muscles.
23 Howard JF Jr, Bril V, Burns TM, et al. Randomized phase 2 study of Evidences for a compensatory mechanism triggered by receptor
FcRn antagonist efgartigimod in generalized myasthenia gravis. loss. J Clin Invest 1998; 102: 249–63.
Neurology 2019; 92: e2661–73. 30 Ruff RL. Endplate contributions to the safety factor for
24 Wolfe GI, Herbelin L, Nations SP, Foster B, Bryan WW, Barohn RJ. neuromuscular transmission. Muscle Nerve 2011; 44: 854–61.
Myasthenia gravis activities of daily living profile. Neurology 1999; 31 Furst DE. Serum immunoglobulins and risk of infection: how low
52: 1487–89. can you go? Semin Arthritis Rheum 2009; 39: 18–29.
25 Barohn RJ, McIntire D, Herbelin L, Wolfe GI, Nations S, Bryan WW. 32 Kassardjian CD, Widdifield J, Paterson JM, et al. Serious infections
Reliability testing of the quantitative myasthenia gravis score. in patients with myasthenia gravis: population-based cohort study.
Ann N Y Acad Sci 1998; 841: 769–72. Eur J Neurol 2020; 27: 702–08.
26 Burns TM, Conaway M, Sanders DB. The MG composite: a valid 33 Nixon AE, Chen J, Sexton DJ, et al. Fully human monoclonal
and reliable outcome measure for myasthenia gravis. Neurology antibody inhibitors of the neonatal fc receptor reduce circulating
2010; 74: 1434–40. IgG in non-human primates. Front Immunol 2015; 6: 176.

536 www.thelancet.com/neurology Vol 20 July 2021


1

Table of Contents

Table 1. List of Study Investigators and Sites……………………………………...pages 2 - 11

Table 2. Inclusion and Exclusion Criteria……………………………………...…pages 11 - 12

Table 3. Number of cycles received by all patients……………………………………..page 13

Figure 1. Change in antibody levels…………………………………………………….page 14

Table 4. ADAPT Investigator Study Group…………………………………………….page 15

1
2

Supplemental Table 1. List of Investigators and Sites

Investigator Site Country


AZ Sint-Lucas & Volkskliniek, Campus Sint-Lucas,
Jan De Bleecker Belgium
Neurology
AZ Sint-Lucas & Volkskliniek, Campus Sint-Lucas,
Belgium
Kathy De Koning Neurology
AZ Sint-Lucas & Volkskliniek, Campus Sint-Lucas,
Belgium
Katrien De Mey Neurology
AZ Sint-Lucas & Volkskliniek, Campus Sint-Lucas,
Belgium
Annelien De Pue Neurology
Rudolf Mercelis Antwerp University Hospital, Department of Neurology Belgium

Antwerp University Hospital, Department of Neurology Belgium


Maren Wyckmans
Antwerp University Hospital, Department of Neurology Belgium
Caroline Vinck
Antwerp University Hospital, Department of Neurology Belgium
Linda Wagemaekers
Antwerp University Hospital, Department of Neurology Belgium
Jonathan Baets
Vera Bril Toronto General Hospital, University Health Network Canada

Toronto General Hospital, University Health Network Canada


Eduardo Ng
Toronto General Hospital, University Health Network Canada
Jafar Shabanpour
Toronto General Hospital, University Health Network Canada
Lubna Daniyal
Toronto General Hospital, University Health Network Canada
Shabber Mannan
Toronto General Hospital, University Health Network Canada
Hans Katzberg
Angela Genge Montreal Neurological Institute & Hospital Canada

Zaeem Siddiqi University of Alberta Hospital Canada

Czech
Jana Junkerová Neurology Clinic, Faculty Hospital Ostrava
Republic
Czech
Neurology Clinic, Faculty Hospital Ostrava
Jana Horakova Republic
Czech
Neurology Clinic, Faculty Hospital Ostrava
Katerina Reguliova Republic
General Faculty Hospital Prague, Neurology Clinic - Czech
Michaela Tyblova
Centre for Myastenia Gravis Republic

2
3

General Faculty Hospital Prague, Neurology Clinic - Czech


Ivana Jurajdova Centre for Myastenia Gravis Republic
General Faculty Hospital Prague, Neurology Clinic - Czech
Iveta Novakova Centre for Myastenia Gravis Republic
General Faculty Hospital Prague, Neurology Clinic - Czech
Michala Jakubikova Centre for Myastenia Gravis Republic
General Faculty Hospital Prague, Neurology Clinic - Czech
Jiri Pitha Centre for Myastenia Gravis Republic
Czech
Stanislav Vohanka Fakultní nemocnice Brno
Republic
Czech
Fakultní nemocnice Brno
Katerina Havelkova Republic
Czech
Fakultní nemocnice Brno
Tomas Horak Republic
Czech
Fakultní nemocnice Brno
Josef Bednarik Republic
Czech
Fakultní nemocnice Brno
Mageda Horakova Republic
Charité - Universitätsmedizin Berlin, Department of
Andreas Meisel Germany
Neurology, NeuroCure Clinical Research Center
Charité - Universitätsmedizin Berlin, Department of
Germany
Dike Remstedt Neurology, NeuroCure Clinical Research Center
Charité - Universitätsmedizin Berlin, Department of
Germany
Claudia Heibutzki Neurology, NeuroCure Clinical Research Center
Charité - Universitätsmedizin Berlin, Department of
Germany
Siegfried Kohler Neurology, NeuroCure Clinical Research Center
Charité - Universitätsmedizin Berlin, Department of
Germany
Sarah Hoffman Neurology, NeuroCure Clinical Research Center
Charité - Universitätsmedizin Berlin, Department of
Germany
Frauke Stascheit Neurology, NeuroCure Clinical Research Center
Copenhagen Neuromuscular Center, 8077,
John Vissing Denmark
Rigshospitalet, Universitu of Copenhagen
Copenhagen Neuromuscular Center, 8077,
Denmark
Lizzie Zafirakos Rigshospitalet, Universitu of Copenhagen
Copenhagen Neuromuscular Center, 8077,
Denmark
Kuldeep Kumar Khatri Rigshospitalet, Universitu of Copenhagen
Copenhagen Neuromuscular Center, 8077,
Denmark
Anne Autzen Rigshospitalet, Universitu of Copenhagen
Mads Peter Godtfeldt Copenhagen Neuromuscular Center, 8077,
Denmark
Stemmerik Rigshospitalet, Universitu of Copenhagen

Henning Andersen Aarhus University Hospital Denmark


Hôpital de la Timone, Service de Neurologie et
Shahram Attarian France
Maladies Neuromusculaires
Temur Margania Ltd. "New Hospitals", Department of Neurology Georgia

3
4

Roman Sakarishvili Ltd Petre Sarajishvili Institute of Neurology Georgia

Pineo Medical Ecosystem Ltd., Department of


Alexander Tsiskaridze Georgia
Neurology Service
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Csilla Rózsa Hungaray
Neurológiai Osztály,
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Hungaray
Gedeonne Margo Jakab Neurológiai Osztály,
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Hungaray
Szilvia Toth Neurológiai Osztály,
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Hungaray
Gyorgyi Szabo Neurológiai Osztály,
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Hungaray
David Bors Neurológiai Osztály,
Jahn Ferenc Dél-pesti Kórház és Rendelőintézet,
Hungaray
Eniko Szabo Neurológiai Osztály,
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Renato Mantegazza Italy
Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Angela Campanella Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Fiammetta Vanoli Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Rita Frangiamore Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Carlo Antozzi Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Silvia Bonanno Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Lorenzo Maggi Neuroimmunologia e malattie neuromuscolari
lstituto Neurologico Carlo Besta, I.R.C.C.S U.O.
Italy
Riccardo Giossi Neuroimmunologia e malattie neuromuscolari
Azienda Ospedaliera Universitaria FEDERICO II,
Francesco Saccà Dipartimento di Neuroscienze, Scienze Riproduttive E Italy
Odontostomatologia
Azienda Ospedaliera Universitaria FEDERICO II,
Dipartimento di Neuroscienze, Scienze Riproduttive E Italy
Angela Marsili Odontostomatologia
Azienda Ospedaliera Universitaria FEDERICO II,
Dipartimento di Neuroscienze, Scienze Riproduttive E Italy
Tiziana Imbriglio Odontostomatologia
Neurology Unit, Sant'Andrea Hospital-University of
Giovanni Antonini Italy
Rome La Sapienza
Neurology Unit, Sant'Andrea Hospital-University of
Girolamo Alfieri Italy
Rome La Sapienza
Neurology Unit, Sant'Andrea Hospital-University of
Italy
Stefania Morino Rome La Sapienza

4
5

Neurology Unit, Sant'Andrea Hospital-University of


Italy
Matteo Garibaldi Rome La Sapienza
Neurology Unit, Sant'Andrea Hospital-University of
Italy
Laura Fionda Rome La Sapienza
Neurology Unit, Sant'Andrea Hospital-University of
Italy
Luca Leonardi Rome La Sapienza
Toho University Hospital Ohashi Medical Center,
Shingo Konno Japan
Department of Neurology
Akiyuki Uzawa Chiba University Hospital, Department of Neurology Japan

Chiba University Hospital, Department of Neurology Japan


Kaoru Sakuma
Chiba University Hospital, Department of Neurology Japan
Chiho Watanabe
Chiba University Hospital, Department of Neurology Japan
Yukiko Ozawa
Chiba University Hospital, Department of Neurology Japan
Manato Yasuda
Chiba University Hospital, Department of Neurology Japan
Yosuke Onishi
Makoto Samukawa Kindai University Hospital, Department of Neurology Japan

Kimiaki Utsugisawa General Hanamaki Hospital Japan

Tomoko Tsuda
General Hanamaki Hospital Japan

Yasushi Suzuki Sendai Medical Center, Department of Neurology Japan

Sendai Medical Center, Department of Neurology Japan


Sayaka Ishida
Sendai Medical Center, Department of Neurology Japan
Genya Watanabe
International University of Health and Welfare, Mita
Hiroyuki Murai Japan
Hospital, Department of Neurology

Masanori Takahashi Osaka University Hospital, Department of Neurology Japan

Osaka University Hospital, Department of Neurology Japan


Hiroko Nakamura
Osaka University Hospital, Department of Neurology Japan
Erina Sugano
Osaka University Hospital, Department of Neurology Japan
Tomoya Kubota
Sapporo Medical University Hospital, Division of
Tomihiro Imai Japan
Neurology
Sapporo Medical University Hospital, Division of
Japan
Mari Suzuki. Neurology

5
6

Sapporo Medical University Hospital, Division of


Japan
Ayako Mori Neurology
Sapporo Medical University Hospital, Division of
Japan
Daisuke Yamamoto Neurology
Sapporo Medical University Hospital, Division of
Japan
Kazuna Ikeda Neurology
Sapporo Medical University Hospital, Division of
Japan
Shin Hisahara Neurology
Tokyo Medical University Hospital, Department of
Masayuki Masuda Japan
Neurology
Tokyo Medical University Hospital, Department of
Japan
Miki Takaki Neurology
Tokyo Medical University Hospital, Department of
Japan
Kanako Minemoto Neurology
Tokyo Medical University Hospital, Department of
Japan
Nobuhiro Ido Neurology
Tokyo Medical University Hospital, Department of
Japan
Makiko Naito Neurology
Tokyo Medical University Hospital, Department of
Japan
Yoshihiko Okubo Neurology

Takamichi Sugimoto Hiroshima City Hiroshima Citizens Hospital Japan

Hiroshima City Hiroshima Citizens Hospital Japan


Yuka Takematsu
Hiroshima City Hiroshima Citizens Hospital Japan
Ayumi Kamei
Hiroshima City Hiroshima Citizens Hospital Japan
Mihiro Shimizu
Hiroshima City Hiroshima Citizens Hospital Japan
Hiroyuki Naito
Hiroshima City Hiroshima Citizens Hospital Japan
Eiichi Nomura
Johannes Verschuuren Leids Universitair Medisch Centrum (LUMC) Netherlands

Leids Universitair Medisch Centrum (LUMC) Netherlands


Marjolein Van Heur
Leids Universitair Medisch Centrum (LUMC) Netherlands
Anne-Marie Peters
Leids Universitair Medisch Centrum (LUMC) Netherlands
Martijn Tannemaat
Leids Universitair Medisch Centrum (LUMC) Netherlands
Annabel Ruiter
Leids Universitair Medisch Centrum (LUMC) Netherlands
Kevin Keene
University Clinical Center, Department of Adult
Malgorzata Bilinska Poland
Neurology

6
7

Marek Halas University Clinical Center, Department of Adult


Poland
Neurology
Krakow Academy of Neurology, Center of Clinical
Andrzej Szczudlik Poland
Neurology
Krakow Academy of Neurology, Center of Clinical
Poland
Marta Pinkosz Neurology
Krakow Academy of Neurology, Center of Clinical
Poland
Monika Frasinska Neurology
Krakow Academy of Neurology, Center of Clinical
Poland
Grazyna Zwolinska Neurology
Independent Public Central Hospital in Warsaw,
Anna Kostera-Pruszczyk Poland
Neurology Clinic
Independent Public Central Hospital in Warsaw,
Poland
Aleksandra Golenia Neurology Clinic
Independent Public Central Hospital in Warsaw,
Poland
Piotr Szczudlik Neurology Clinic
NZOZ Wielospecjalistyczna Poradnia Lekarska
Lech Szczechowski Poland
"Synapsis"
Ewelina Marek NZOZ Wielospecjalistyczna Poradnia Lekarska
Poland
"Synapsis"
Samara Regional Clinical Hospital, Department of
Irina Poverennova Russia
Neurosurgery
Samara Regional Clinical Hospital, Department of
Russia
Lubov Urtaeva Neurosurgery
Samara Regional Clinical Hospital, Department of
Russia
Nadezhda Kuznetsova Neurosurgery
Samara Regional Clinical Hospital, Department of
Russia
Tatiana Romanova Neurosurgery
Novosibirsk Regional Hospital, Regional Center of
Malkova Nadezhda Multiple Sclerosis and other Autoimmune Disorders of Russia
Nervous System
Novosibirsk Regional Hospital, Regional Center of
Multiple Sclerosis and other Autoimmune Disorders of Russia
Elena Lapochka Nervous System
Novosibirsk Regional Hospital, Regional Center of
Multiple Sclerosis and other Autoimmune Disorders of Russia
Denis Korobko Nervous System
Novosibirsk Regional Hospital, Regional Center of
Multiple Sclerosis and other Autoimmune Disorders of Russia
Ilona Vergunova Nervous System
Novosibirsk Regional Hospital, Regional Center of
Multiple Sclerosis and other Autoimmune Disorders of Russia
Anna Melnikova Nervous System
Novosibirsk Regional Hospital, Regional Center of
Multiple Sclerosis and other Autoimmune Disorders of Russia
Ekaterina Bulatova Nervous System
Nizhegorodsky Regional Clinical Hospital named after
Elena Antipenko Russia
NA Semashko

7
8

Stojan Peric Clinical Center of Serbia, Clinic for Neurology Serbia

Clinical Center of Serbia, Clinic for Neurology Serbia


Ivo Bozovic
Clinical Center of Serbia, Clinic for Neurology Serbia
Dragana Lavrnic
Clinical Center of Serbia, Clinic for Neurology Serbia
Vidosava Rakocevic Stojanovic
University of Southern California, Healthcare United
Said Beydoun
Consultation Center II States
University of Southern California, Healthcare United
Salma Akhter Consultation Center II States
University of Southern California, Healthcare United
Ali Malekniazi Consultation Center II States
University of Southern California, Healthcare United
Leila Darki Consultation Center II States
University of Southern California, Healthcare United
Norianne Pimentel Consultation Center II States
University of Southern California, Healthcare United
Victoria Cannon Consultation Center II States
United
James F. Howard Jr. The University of North Carolina
States
United
The University of North Carolina
Manisha Chopra States
United
The University of North Carolina
Rebecca Traub States
UCI ALS & Neuromuscular Center, Neuromuscular United
Tahseen Mozaffar
Clinical Trials Unit States
UCI ALS & Neuromuscular Center, Neuromuscular United
Ivonne Turner Clinical Trials Unit States
UCI ALS & Neuromuscular Center, Neuromuscular United
Ali Habib Clinical Trials Unit States
UCI ALS & Neuromuscular Center, Neuromuscular United
Namita Goyal Clinical Trials Unit States
UCI ALS & Neuromuscular Center, Neuromuscular United
Manisha Kak Clinical Trials Unit States
United
Tuan Vu University of South Florida
States
United
University of South Florida
Erik Velasquez States
United
University of South Florida
Lucy Lam States
United
University of South Florida
Niraja Suresh States
United
University of South Florida
Jerrica Farias States

8
9

United
Sarah Jones University of Virginia Health System
States
United
University of Virginia Health System
Mary Wagoner States
United
University of Virginia Health System
Debbie Eggleston States
United
Tulio Bertorini Wesley Neurology Clinic
States
United
Wesley Neurology Clinic
Cindy Benzel States
United
Wesley Neurology Clinic
Robert Henegar States
United
Wesley Neurology Clinic
Rekha Pillai States
University of Texas Health Science Center at San United
Ratna Bharavaju-Sanka
Antonio States
University of Texas Health Science Center at San United
Carolyn Paiz Antonio States
University of Texas Health Science Center at San United
Carlayne Jackson Antonio States
Medical University of South Carolina, Department of United
Katherine Ruzhansky
Neurology States
United
Chafic Karam Oregon Health & Science University
States
United
Oregon Health & Science University
Diana Dimitrova States
United
Oregon Health & Science University
Amy Visser States
United
Oregon Health & Science University
Nizar Chahin States
United
Todd Levine HonorHealth Neurology, 3501 N. Scottsdale Road
States
United
Robert Lisak Wayne State University, Department of Neurology
States
United
Wayne State University, Department of Neurology
Kelly Jia States
United
Wayne State University, Department of Neurology
Flicia Mada States
United
Wayne State University, Department of Neurology
Evanthia Bernitsas States
University of Kansas Medical Center, Department of United
Mamatha Pasnoor
Neuromuscular Research States
University of Kansas Medical Center, Department of United
Katherine Roath Neuromuscular Research States
University of Kansas Medical Center, Department of United
Samantha Colgan Neuromuscular Research States

9
10

University of Kansas Medical Center, Department of United


Melissa Currence Neuromuscular Research States
University of Kansas Medical Center, Department of United
Andrew Heim Neuromuscular Research States
University of Kansas Medical Center, Department of United
Richard Barohn Neuromuscular Research States
University of Kansas Medical Center, Department of United
Mazen Dimachkie Neuromuscular Research States
University of Kansas Medical Center, Department of United
Jeffrey Statland Neuromuscular Research States
University of Kansas Medical Center, Department of United
Omar Jawdat Neuromuscular Research States
University of Kansas Medical Center, Department of United
Duaa Jabari Neuromuscular Research States
University of Kansas Medical Center, Department of United
Constantine Farmakidis Neuromuscular Research States
Southern Illinois University School of Medicine, United
James Gilchrist
Department of Neurology States
United
Yuebing Li Cleveland Clinic Foundation, Neuromuscular Center
States
United
Cleveland Clinic Foundation, Neuromuscular Center
Irys Caristo States
United
Cleveland Clinic Foundation, Neuromuscular Center
Debbie Hastings States
United
Cleveland Clinic Foundation, Neuromuscular Center
John Anthony Morren States
United
Michael Weiss University of Washington, Department of Neurology
States
United
Srikanth Muppidi Stanford Neuromuscular Research
States
United
Stanford Neuromuscular Research
Tia Nguyen States
United
Stanford Neuromuscular Research
Lesly Welsh States
United
Stanford Neuromuscular Research
Yuen So States
United
Stanford Neuromuscular Research
Neelam Goyal States
United
Michael Pulley University of Florida Health Science Center Jacksonville
States
United
University of Florida Health Science Center Jacksonville
Cathy Bailey States
United
University of Florida Health Science Center Jacksonville
Lisa Smith States
United
University of Florida Health Science Center Jacksonville
Alan Berger States

10
11

North County Neurology Associates, Research United


Gregory Sahagian
Department States
North County Neurology Associates, Research United
Yasmin Camberos Department States
North County Neurology Associates, Research United
Benjamin Frishberg Department States

Table 2. Inclusion and Exclusion Criteria.

Inclusions Criteria
• Patients with the ability to understand the requirements of the trial, provide written
informed consent, and comply with the trial protocol procedures.
• Adult patients with gMG.
• All serotypes, regardless of Ab status and including MuSK, LRP4, and AChR-Ab- in
addition to AChR-Ab+.
• MGFA Class II-IV gMG. The confirmation of the diagnosis should be documented
and supported by ≥1 of the following 3 tests:
• MGFA Class II-IV gMG. The confirmation of the diagnosis should be documented
and supported by ≥1 of the following 3 tests:
o History of abnormal neuromuscular transmission demonstrated by single-fiber
electromyography or repetitive nerve stimulation
o History of positive edrophonium chloride test
o Patient has demonstrated improvement in MG signs on oral
acetylcholinesterase inhibitors as assessed by the treating physician
• MG-ADL score ≥5, with ≥50% of the total score due to non-ocular symptoms
• Receiving a stable dose of ≥1 of the following gMG treatments prior to randomization:
acetylcholinesterase inhibitors (no dose change for 2 weeks prior to screening),
steroids (at least 3 months of treatment, no dose change for 1 month) or NSIST (at
least 6 months of treatment, no dose change for 3 months)

Exclusion Criteria:
• Have been treated with IVIg/plasma exchange in the past month, with rituximab or
eculizumab in the past 6 months, with an investigational drug within 3 months or 5
half-lives of the drug (whichever is longer) prior to screening, or had a thymectomy in
the past 3 months or a planned thymectomy during the trial period.
• MGFA Class I and V patients
• Have hepatitis B or C, HIV, severe infections, or malignancies
• Have low IgG serum levels (<6 g/L)
• Female patients who are pregnant or lactating, or are intending to become pregnant
• Male patients who are sexually active and do not intend to use effective methods of
contraception during the trial or within 90 days after the last dosing or male patients
who plan to donate sperm during the trial or within 90 days after the last dosing
• History of any known bacterial, viral, or fungal infection or any major episode of
infection that required hospitalization or injectable antimicrobial therapy in the last 8
weeks prior to screening
• History of autoimmune disease other than MG (e.g. thyroiditis, rheumatoid arthritis,
etc.) that would interfere with an accurate assessment of clinical symptoms
• Documented lack of clinical response to PLEX

11
12

• Patients who previously participated in a clinical trial with efgartigimod


• Patients who received a vaccination (e.g., influenza vaccine) within the last 4 weeks
prior to screening
• Patients with clinical evidence of other significant serious disease or patients who
underwent a recent major surgery, which could confound the results of the trial or put
the patient at undue risk. Patients with renal/hepatic function impairment could be
included
• Patients who have a history of malignancy, including malignant thymoma, or
myeloproliferative or lymphoproliferative disorders, unless deemed cured by adequate
treatment with no evidence of recurrence for ≥3 years before screening. Patients with
completely excised non-melanoma skin cancer (such as basal cell carcinoma or
squamous cell carcinoma) or cervical carcinoma in situ would be permitted at any time
• Patients with worsening muscle weakness secondary to concurrent infections or
medications (aminoglycosides, fluoroquinolones, beta-blockers, etc.)
AChR-Ab+, acetylcholine receptor autoantibody positive; gMG, generalized myasthenia gravis; HIV, human
immunodeficiency virus; IgG, immunoglobulin G; IVIg, intravenous immunoglobulin G; LRP4, lipoprotein
receptor-related protein-4; MG, myasthenia gravis; MG-ADL, Myasthenia Gravis Activities of Daily Living;
MuSK, muscle-specific kinase; NSIT, non-steroidal immunosuppressive therapy; PLEX, plasma exchange

12
13

Table 2: Number of cycles received by all patients

Efgartigimod (n=84) Placebo (n=83)

Maximum Patients, n (%) Discontinued Patients, n (%) Discontinued

number of treatment during treatment during

cycles cycle cycle

1 21 (25%) 4 26 (31⸱3%) 9

2 56 (66⸱7%) 1 54 (65⸱1%) 1

3 7 (8⸱3%) 0 3 (3⸱6%) 0

13
14

Figure 4

Figure 1: Change in antibody levels: Mean change over time in C1 of IgG and AChR-Ab
levels in AChR-Ab+ patients.

14
15

Table 4. ADAPT Investigator Study Group

Jan De Bleeker

Kathy De Koning

Katrien De Mey

Annelien De Pue

Rudolf Mercelis

Maren Wyckmans

Caroline Vinck

Linda Wagemaekers

Jonathan Baets

Eduardo Ng

Jafar Shabanpour

Lubna Daniyal

Shabber Mannan

Hans Katzberg

Angela Genge

Zaeem Siddiqi

Jana Junkerová

Jana Horakova

Katerina Reguliova

Michaela Tyblova

Ivana Jurajdova

Iveta Novakova

15
16

Michala Jakubikova

Jiri Pitha

Stanislav Vohanka

Katerina Havelkova

Tomas Horak

Josef Bednarik

Mageda Horakova

Andreas Meisel

Dike Remstedt

Claudia Heibutzki

Siegfried Kohler

Sarah Hoffman

Frauke Stascheit

John Vissing

Lizzie Zafirakos

Kuldeep Kumar Khatri

Anne Autzen

Mads Peter Godtfeldt Stemmerik

Henning Andersen

Shahram Attarian

Alexander Tsiskaridze

Csilla Rózsa

Gedeonne Margo Jakab

16
17

Szilvia Toth

Gyorgyi Szabo

David Bors

Eniko Szabo

Angela Campanella

Fiammetta Vanoli

Rita Frangiamore

Carlo Antozzi

Silvia Bonanno

Lorenzo Maggi

Riccardo Giossi

Francesco Saccà

Angela Marsili

Tiziana Imbriglio

Giovanni Antonini

Girolamo Alfieri

Stefania Morino

Matteo Garibaldi

Laura Fionda

Luca Leonardi

Shingo Konno

Akiyuki Uzawa

Kaoru Sakuma

17
18

Chiho Watanabe

Yukiko Ozawa

Manato Yasuda

Yosuke Onishi

Makoto Samukawa

Tomoko Tsuda

Yasushi Suzuki

Sayaka Ishida

Genya Watanabe

Masanori Takahashi

Hiroko Nakamura

Erina Sugano

Tomoya Kubota

Tomihiro Imai

Mari Suzuki.

Ayako Mori

Daisuke Yamamoto

Kazuna Ikeda

Shin Hisahara

Masayuki Masuda

Miki Takaki

Kanako Minemoto

Nobuhiro Ido

18
19

Makiko Naito

Yoshihiko Okubo

Takamichi Sugimoto

Yuka Takematsu

Ayumi Kamei

Mihiro Shimizu

Hiroyuki Naito

Eiichi Nomura

Marjolein Van Heur

Anne-Marie Peters

Martijn Tannemaat

Annabel Ruiter

Kevin Keene

Marek Halas

Andrzej Szczudlik

Marta Pinkosz

Monika Frasinska

Grazyna Zwolinska

Anna Kostera-Pruszczyk

Aleksandra Golenia

Piotr Szczudlik

Lech Szczechowski

Ewelina Marek

19
20

Irina Poverennova

Lubov Urtaeva

Nadezhda Kuznetsova

Tatiana Romanova

Malkova Nadezhda

Elena Lapochka

Denis Korobko

Ilona Vergunova

Anna Melnikova

Ekaterina Bulatova

Elena Antipenko

Ivo Bozovic

Dragana Lavrnic

Vidosava Rakocevic Stojanovic

Said Beydoun

Salma Akhter

Ali Malekniazi

Leila Darki

Norianne Pimentel

Victoria Cannon

Manisha Chopra

Rebecca Traub

Tahseen Mozaffar

20
21

Ivonne Turner

Ali Habib

Namita Goyal

Manisha Kak

Erik Velasquez

Lucy Lam

Niraja Suresh

Jerrica Farias

Sarah Jones

Mary Wagoner

Debbie Eggleston

Tulio Bertorini

Cindy Benzel

Robert Henegar

Rekha Pillai

Ratna Bharavaju-Sanka

Carolyn Paiz

Carlayne Jackson

Katherine Ruzhansky

Diana Dimitrova

Amy Visser

Nizar Chahin

Todd Levine

21
22

Robert Lisak

Kelly Jia

Flicia Mada

Evanthia Bernitsas

Mamatha Pasnoor

Katherine Roath

Samantha Colgan

Melissa Currence

Andrew Heim

Richard Barohn

Mazen Dimachkie

Jeffrey Statland

Omar Jawdat

Duaa Jabari

Constantine Farmakidis

James Gilchrist

Yuebing Li

Irys Caristo

Debbie Hastings

John Anthony Morren

Michael Weiss

Srikanth Muppidi

Tia Nguyen

22
23

Lesly Welsh

Yuen So

Neelam Goyal

Michael Pulley

Cathy Bailey

Lisa Smith

Alan Berger

Gregory Sahagian

Yasmin Camberos

Benjamin Frishberg

23

You might also like