Rationale:Multifocal motor neuropathy (MMN) is a rare neuropathy characterized by progressive asymmetric weakness and atrophy without sensory abnormalities. MMN is considered a chronic immune-mediated neuropathy driven by the classical complement…
ID
Source
Brief title
Condition
- Autoimmune disorders
- Neuromuscular disorders
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
Primary Objectives:
To evaluate the safety and tolerability of ARGX-117 compared to placebo in
adult participants previously stabilized with IVIg •
Primary Endpoints:
Safety outcomes based on adverse event (AE) monitoring and other safety
assessments
Secondary outcome
Secondary Objectives:
• To evaluate the efficacy of ARGX-117 compared to placebo on muscle strength
and/or motor function in adult participants previously stabilized with IVIg
• To evaluate the efficacy of ARGX-117 on functional ability, arm and hand
function, quality of life, and fatigue in adult participants with MMN
• To evaluate the effect of ARGX-117 on health-related productivity and work
productivity
• To evaluate medication treatment satisfaction
• To assess the PK, PD, and immunogenicity of ARGX-117
Secondary Endpoints:
A. Time to the first retreatment with IVIg since the final IVIg treatment of
the IVIg monitoring period
B. Time-to-relapse
C. AUC of the change from baseline in mMRC-10 sum score
D. Value and change from baseline in the average score of the 2 most important
muscle groups as assessed by the mMRC-14 sum score
E. Value and change from baseline in the mMRC-14 sum score
F. Proportion of participants showing a deterioration of 1 or more points in at
least 2 muscle groups as assessed by the mMRC-14 sum score
G. Proportion of participants with no deterioration in 2 or more muscle groups
as assessed by mMRC-14 sum score
H. AUC of the change from baseline in GS
I. Proportion of participants with a GS decrease of 8 kilopascal (kPa) or more
over 3 consecutive days
J. Values, change, and percent change from baseline in GS
K. Values and change from baseline in the Rasch-built overall disability scale
for MMN (MMN*RODS©)
L. Values and change from baseline in the average time for the upper extremity
(arm and hand) function (9-Hole Peg Test [9-HPT], or timed Peg Board Test)
M. Proportion of participants by level of severity on each dimension of the
EQ-5D-5L scale
N. Value and change from baseline in EQ-5D-5L visual analog scale (VAS)
O. Values and change from baseline in the chronic acquired polyneuropathy
patient reported index (CAP-PRI)
P. Values of the Patient Global Impression Change (PGIC) scale
Q. Values and change from baseline in the 9-item Fatigue Severity Scale (FSS)
R. Values and change for work-related and household core activities of the
Heatlh-Related Productivity Questionnaire (HRPQ) at each visit:
* Hours lost because of absenteeism
* Hours lost because of presenteeism
* Total hours lost (absenteeism + presenteeism)
* Percentage of scheduled hours lost because of absenteeism
* Percentage of scheduled hours lost because of presenteeism
* Percentage of scheduled hours lost in total (absenteeism +
presenteeism)
S. Effectiveness, side effects, convenience, and overall satisfaction scores as
assessed by the Treatment Satisfaction 14-Item Questionnaire for Medication
(TSQM)
T. Serum concentrations and PK parameters for ARGX-117
U. Values and change from baseline in free C2, total C2, functional complement
activity (CH50)
V. Incidence and prevalence of ADA against ARGX-117
Background summary
MMN is considered a rare chronic immune-mediated neuropathy involving
progressive muscle weakness of mainly the hands, forearms, and lower legs. It
is clinically characterized by progressive asymmetric weakness involving 2 or
more nerves and partial motor conduction block. The estimated prevalence of MMN
is between 0.6 to 2 per 100,000 people and typically presents as an
asymmetrical upper limb pure motor neuropathy. The hallmark of the disease is
the presence of multifocal motor conduction blocks, ie. impaired propagation of
action potentials
along the axon, and patients often show high serum levels of immunoglobulin M
(IgM) antibodies against the ganglioside GM1
(monosialotetrahexosylganglioside). GM1 is widely expressed in the nervous
system by neurons, particularly around the node of Ranvier, and
Schwann cells. The current prevailing view is that GM1 antibodies target the
axolemma at the node of Ranvier. This is thought to interfere with axon-Schwann
cell interactions, causing widening of the node, and direct damage to the axon.
The presence and titers of IgM anti-ganglioside GM1 (anti-GM1) antibodies and
their complement activating properties, correlate with clinical features such
as weakness and axonal damage. Moreover, the binding and subsequent classical
complement pathway activation of
anti-GM1 IgM from patients with MMN to motor neurons derived from induced
pluripotent stem cells causes disturbed calcium homeostasis and structural
damage to these pluripotent cells in vitro, resembling the changes that occur
in MMN.
Anti-GM1 (and other gangliosides) IgM antibodies are produced by a limited
number of activated B cells; however, the mechanism of this B cell activation
has not yet been established. Binding of these anti-GM1 antibodies to GM1 leads
to activation of the classical complement
pathway, and subsequent MAC deposition. Consequently, this MAC deposition leads
to disruption of Schwann cell-axolemma junctions, displacement of ion-channel
clustering, and the disturbance of membrane integrity at the (para)nodal
regions resulting in demyelination.
Anti-GM1 IgM antibodies are identified in at least 40% of patients with MMN.
These findings suggest that complements play an important role in the
pathogenesis of MMN, therefore, the inhibition of complement activation may
provide a new therapeutic option in this disease.
High dose IV immunoglobulin (IVIg) treatment is the only approved treatment for
MMN. IVIg treatment often improves muscle strength; however, the efficacy of
IVIg in reducing MMN symptoms declines after several years and many patients
report progressive neurological deficits. The mechanism of action of IVIg in
MMN is not well understood; however, IVIg may have effects on humoral immunity
beneficial to those with MMN. IVIg partially reduced complement activity in
sera13 and may interfere with anti-GM1 IgM-mediated complement nerve
deposition. Despite treatment with IVIg, MMN related disabilities will continue
to progress due to ongoing axonal degeneration. Complement-modulating treatment
was previously evaluated using eculizumab, a monoclonal antibody against
complement factor 5 (C5) preventing
formation of MAC. Results showed that eculizumab was well tolerated, however,
only a small improvement was seen in selected motor performance measurements.
Considering the high levels of CD59 expression, iPSC-derived motor neurons show
a high innate ability to inhibit the
terminal portion of the complement cascade. Thus, the use of a C5 inhibitor to
preserve motor neuron function may be limited Moreover, C5 is downstream of C3,
whereas the presence of C3aR on MNs suggest that complement activation upstream
of C5 may have functional
consequences for MNs.
There is an unmet medical need for an efficacious treatment option with a more
favorable safety and tolerability profile and lower duration of administration
than the current standard of care.
Study objective
Rationale:
Multifocal motor neuropathy (MMN) is a rare neuropathy characterized by
progressive asymmetric weakness and atrophy without sensory abnormalities. MMN
is considered a chronic immune-mediated neuropathy driven by the classical
complement pathway related to the presence of anti-ganglioside GM1
(monosialotetrahexosylganglioside [anti-GM1]) (and other gangliosides)
immunoglobulin M (IgM) antibodies produced by a limited number of B cell
clones. These antibodies activate the complement system's classical pathway and
subsequently yield membrane attack complex (MAC) deposition leading to
disruption of Schwann cell-axolemma junctions, displacement of ion-channel
clustering, and disturbance of membrane integrity at the (para)nodal regions
that result in demyelination and motor nerve conduction block.
Patients with MMN initially respond to the standard of care, intravenous
immunoglobulin (IVIg); however, the disease will continue to progress despite
treatment. There is an unmet medical need for an efficacious treatment option
with a more favorable safety and tolerability profile and a lower duration of
administration than the current standard of care.
ARGX-117, a therapeutic complement-inhibiting antibody that targets complement
factor 2 (C2), is being developed to reduce tissue inflammation and attenuate
the adaptive immune response by blocking both the lectin and classical
complement pathways. Inhibition of C2 in complement-mediated neuronal damage is
a promising mechanism for preventing axonal and glial injury. In an ex vivo
mouse model of acute neuropathy, the use of a C2 blocking antibody (Bro2) in
GalNAcT-/--Tg (neuronal) mice prevented the loss of neurofilament staining at
the nerve terminal, thereby maintaining axonal integrity. Additionally, C2
inhibition by Bro2 in GalNAcT-/--Tg(glial) mice resulted in the preservation of
ankyrin B at the distal paranode. Using an in vitro model for MMN, ARGX-117
blocked IgM-mediated classical pathway complement activation on both motor
neurons and Schwann cells, providing further support for developing ARGX-117 in
patients with MMN.
The safety and tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and
immunogenicity of single and multiple doses of ARGX-117 administered
intravenously (IV), and ARGX-117 comixed with recombinant human hyaluronidase
PH20 (rHuPH20) administered subcutaneously (SC), are being evaluated in the
ongoing, first-in-human (FIH), phase 1 study, ARGX-117-1901.
This phase 2 clinical trial serves to evaluate the safety and efficacy of
different dose regimens of ARGX-117 versus placebo, in participants with MMN
previously stabilized with IVIg.
Study design
ARGX-117-2002 is a randomized, double-blinded, placebo-controlled,
parallel-group, multicenter trial to evaluate the safety and tolerability,
efficacy, PK, PD, and immunogenicity of different dose regimens of ARGX-117 in
adults with MMN. Two cohorts of at least 24 participants each are planned for
enrollment.
This trial consists of a screening period, an IVIg dependency period (if
applicable), an IVIg monitoring period, a double-blinded treatment period
(DBTP), and a safety follow-up period.
All participants will begin with a screening period and the diagnosis of MMN
will be assessed by the MMN Confirmation Committee (MCC). The MCC will also
assess IVIg dependency. Participants whose IVIg dependency is uncertain will
enter an IVIg dependency period to assess the impact of a delayed IVIg
administration on grip strength (GS) and/or motor function. The IVIg criterion
is considered fulfilled in an individual stabilized to IVIg for longer than 3
months if a clinically meaningful deterioration from any of these 2 parameters
is established. The IVIg criterion is also considered fulfilled if an
individual stabilized to IVIg for less than 3 months demonstrates a clinical
improvement following the initiation of IVIg therapy.
After completing the screening period (including the IVIg dependency period, if
applicable), all participants will begin the IVIg monitoring period.
Participants will receive IVIg during the IVIg monitoring period at a
frequency, duration, and dose established by their medical history. The IVIg
monitoring period includes 3 IVIg treatment cycles, and will establish baseline
values for all clinical endpoints assessed during the DBTP.
Participants will be randomized on day 1 of the DBTP in:
- a 2:1 ratio to ARGX-117 or placebo in cohort 1, and for option 1 (dose
regimen 2: high dose ARGX-117) or option 2 (dose regimen 3: lower dose
ARGX-117) in cohort 2
- in a 2:2:1:1 ratio to ARGX-117 (high dose), ARGX-117 (lower dose), placebo
(high dose), or placebo (lower dose) for option 3 in cohort 2
Randomization will be stratified based on an individual*s IVIg dose frequency:
1. IVIg dosed every 2 or 3 weeks
2. IVIg dosed every 4 or 5 weeks
The dosing of ARGX-117 or placebo will begin on day 1 of the DBTP, 7 days after
the final IVIg administration of the IVIg monitoring period.
Participants will be retreated with IVIg during the DBTP if there is a
clinically meaningful deterioration in muscle strength and/or motor function.
Investigational medicinal product (IMP) administration will continue throughout
the DBTP. A clinically meaningful deterioration is defined as a >30% decline of
the GS of either hand observed for at least 2 consecutive days (based on the
3-day averaged calculations) and/or a decline of at least 2 points on the
modified Medical Research Council (mMRC)-10 sum score. Based on their clinical
judgment, the investigator may choose to not re-treat the participant with
IVIg. All trial participants can request IVIg retreatment anytime during the
DBTP.
End of Trial/Rollover
After completing the 16-week DBTP, participants may enroll in a long-term
extension (LTE) study and receive ARGX-117; otherwise, participants will enter
the 9-month safety follow-up period.
The following interim database locks will occur:
- After the completion of the 16-week DBTP by all participants included in
cohort 1.
- After the completion of the 16-week DBTP by all participants included in
cohort 2.
A final database lock will occur when all participants have completed the
safety follow-up period, or have rolled over to the LTE study, if applicable.
Intervention
The first IMP administration will begin 7 days after the final IVIg
administration at the end of the IVIg monitoring period.
IMP includes:
• ARGX-117 administered by IV infusion
• Placebo administered by IV infusion
ARGX-117 and placebo will be administered by an IV infusion over approximately
2 hours at visit 1. ARGX-117 and placebo will be administered by an IV infusion
over approximately 1 hour for all subsequent administrations.
Dose regimen 1 will be assessed in cohort 1. Dose regimen 1 will include an
ARGX-117 dose of 30 mg/kg on day 1, followed by a dose of 10 mg/kg every 7 days
for 4 weeks (4 infusions total), and a dose of 10 mg/kg every 14 days (5
infusions total) until the end of the DBTP.
Three options are available as the dose regimen for cohort 2, of which 1 will
be assessed per the decision of the EDRT:
• Option 1*Dose regimen 2 (high dose): a single dose of 60 mg/kg ARGX--117 or
placebo on day 1, followed by 4 weekly doses of 30 mg/kg
ARGX--117 or placebo on days 8, 15, 22, and 29 (4 total infusions), and a dose
of 30 mg/kg ARGX-117 or placebo every 2 weeks until the end of the DBTP,
starting from day 43 (5 total infusions in total).).
• Option 2*Dose regimen 3 (lower dose): a single dose of 15 mg/kg ARGX-117 or
placebo on day 1, followed by 4 weekly doses of 5 mg/kg ARGX-117 or placebo on
days 8, 15, 22, and 29 (4 total infusions), and a dose of 5 mg/kg ARGX-117 or
placebo every 4 weeks, on days 57 and 85, until the end of the DBTP (2 total
infusions). Additionally, placebo will be given every 4 weeks, on days 43, 71,
and 99.
• Option 3*Dose regimens 2 and 3
Study burden and risks
There was 1 reported serious adverse event (SAE) of *abscess* which was
considered not related to IMP (ARGX-117 or placebo).
No SAEs considered related to blinded treatment have been reported.
No deaths or life-threatening events have been reported.
Industriepark Zwijnaarde 7
Zwijnaarde (Ghent) B-9052
BE
Industriepark Zwijnaarde 7
Zwijnaarde (Ghent) B-9052
BE
Listed location countries
Age
Inclusion criteria
Participants are eligible to be included in the trial only if all of the
following criteria apply:
1. Capable of giving signed informed consent as described in Appendix 1,
Section 10.1.3 which includes compliance with the requirements and restrictions
listed in the informed consent form (ICF) and in this protocol (including
consent for the use and disclosure of research-related health information).
Participants must be able to read and write and be willing and able to comply
with the trial protocol procedures (including attending the required trial
visits).
2. Male/female at least 18 years of age at the time the ICF is signed
3. Probable or definite MMN according to the EFNS/PNS 2010 guidelines (Appendix
8: Section 10.8) at screening confirmed by the MCC
4. Receiving a stable IVIg regimen for at least 3 months before screening or
recently initiated IVIg treatment (refer to inclusion criterion 5.1a) and both
of the following:
a. IVIg treatment interval of 2 to 5 weeks
b. IVIg dose of 0.4 to 2.0 grams per kg body weight and infusion
5.1 IVIg treatment dependency confirmation by the MCC at screening or after
IVDP when applicable, based on 1 of the following:
a. Recently initiated IVIg treatment (less than 3 months):
- Clinical improvement following IVIg initiation documented in the
participant*s medical record
b. Maintenance therapy with IVIg (longer than 3 months), based on 1 of the
following:
- Clinical deterioration following IVIg withdrawal, IVIg dose reduction,
or IVIg delayed administration within 12 months prior to screening (documented
in the participant*s medical record)
- Clinical deterioration following IVIg delayed administration during the
IVDP
6. Immunization with the first meningococcal vaccine and pneumococcal vaccine,
and the single Haemophilus influenza type B vaccine must be performed at least
14 days before IMP administration at V1 according to local country-specific
immunization schedules. A documented history of vaccination against Neisseria
meningitides, Haemophilus influenza type B, and streptococcus pneumonia will be
permitted.
7. Contraceptive use by men and women should be consistent with local
regulations regarding the methods of contraception for those participating in
clinical studies
a. Male participants must agree to not donate sperm from the time the ICF is
signed until 15 months after the last IMP administration
b. Women of childbearing potential (WOCBP) (defined in Appendix 4, Section
10.4.1.1) must have a negative serum pregnancy test at screening and a negative
urine pregnancy test at baseline before IMP can be administered
The contraceptive requirements for WOCBP are described in Appendix 4, Section
10.4.2).
Exclusion criteria
Participants are excluded from the trial if any of the following criteria apply:
1. Any coexisting condition which may interfere with the outcome assessments
(eg, diabetic neuropathy, CIDP, inflammatory arthritis, or osteoarthritis
affecting the hand)
2. Clinical signs or symptoms suggestive for neuropathies other than MMN such
as motor neuron disease (eg, bulbar signs or brisk reflexes) or other
inflammatory neuropathies (eg, sensory neuropathy)
3. Severe psychiatric disorder (such as severe depression, psychosis, bipolar
disorder), history of suicide attempt, or current suicidal ideation that in the
opinion of the investigator could create undue risk to the participant or could
affect adherence with the trial protocol. See Section 8.2.6.
Note: At screening, suicidality will be assessed using the Columbia-suicide
severity rating scale (C SSRS) (see Section 8.2.6.1); participants with a high
suicide risk will be excluded from the trial (ie, participants will be excluded
with a positive answer to questions #4 and/or #5 of the suicidal ideation
subscale [over the past 3 months]; and/or any positive answer to the suicidal
behavior subscale [over the past year]). Any positive answer to the these
questions under *Lifetime/time he/she felt most suicidal* should be carefully
evaluated for any current risk of suicide by the investigator prior to trial
entry.
4. Clinically significant uncontrolled active or chronic bacterial, viral, or
fungal infection during the screening and/or IVMP.
5. Any other known autoimmune disease that, in the opinion of the investigator,
would interfere with an accurate assessment of clinical symptoms of MMN or put
the participant at undue risk (eg, SLE).
6. History of malignancy unless resolved by adequate treatment with no evidence
of recurrence for >=3 years before the first administration of the IMP.
Participants with the following carcinomas will be eligible:
a. Adequately treated basal cell or squamous cell skin cancer
b. Carcinoma in situ of the cervix
c. Carcinoma in situ of the breast or
d. Incidental histological finding of prostate cancer (TNM stage T1a or T1b)
7. Clinical evidence of other significant serious diseases, (including
splenectomy at any time), have had a recent major surgery, or who have any
other condition in the opinion of the investigator, that could confound the
results of the trial or put the participant at undue risk
8. Prior/concomitant therapy
a. Cyclophosphamide and/or rituximab and/or eculizumab and/or mycophenolate
mofetil within 3 months prior to screening
b. Use of an investigational product within 3 months or 5 half-lives (whichever
is longer) before the first dose of the IMP.
9. Positive serum test at screening for an active viral infection with any of
the following conditions:
a. Hepatitis B virus (HBV) that is indicative of an acute or chronic infection
(https://www.cdc.gov/hepatitis/HBV/PDFs/SerologicChartv8.pdf)
b. Hepatitis C virus (HCV) based on HCV antibody assay
c. HIV based on test results that are associated with an AIDS-defining
condition or a CD4 count <200 cells/mm3
10. Current or history of (ie, within 12 months of screening) alcohol, drug, or
medication abuse
11. Known hypersensitivity reaction to 1 of the components of the IMP or any of
its excipients
12. Female participants with a positive serum or urine pregnancy test,
lactating females, and those who intend to become pregnant during the trial or
within 12
months after last dose of the IMP
13. ALT or AST >=2 × upper limit of normal and total bilirubin >=1.5 × upper
limit of normal of the central laboratory reference range, or any other
clinically significant
laboratory abnormality. These tests will be performed by the central
laboratory
14. An estimated glomerular filtration rate of <=60 mL/min/1.73m2 calculated
by the central laboratory using the 4-variable Modification of Diet in the
Renal-Disease
(MDRD) equation
Design
Recruitment
Medical products/devices used
Followed up by the following (possibly more current) registration
No registrations found.
Other (possibly less up-to-date) registrations in this register
No registrations found.
In other registers
Register | ID |
---|---|
EudraCT | EUCTR2021-003302-50-NL |
ClinicalTrials.gov | NCT05225675 |
CCMO | NL79411.028.21 |