The primary objective of the study is to evaluate the safety, tolerability, PK, and PD of AL002 administered in single ascending doses in healthy participants and multiple doses in participants with mild to moderate AD.
ID
Source
Brief title
Condition
- Demyelinating disorders
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
The safety endpoints of this study are:
* Incidence, nature, and severity of serious adverse event (SAE)s and adverse
events of special interest (AESI)
* Incidence of dose limiting adverse events (DLAE)
* Incidence of treatment discontinuations due to AEs
* Incidence of dose reductions due to AEs
* Mean changes in clinical laboratory tests from baseline over time; incidence
of treatment emergent abnormal laboratory values and abnormal laboratory values
reported as AEs
* Physical and neurologic examination abnormalities
* Ophthalmological examination abnormalities
* Mean change in vital signs from baseline over time and incidence of abnormal
vital sign measurements
* Suicidal ideation, suicidal behavior, and self-injurious behavior without
suicidal intent, as determined using the Sheehan-STS (for the MD participant
cohort only)
* Incidence of ADAs during the study relative to the prevalence of ADAs at
baseline (in SAD healthy adult participant cohorts and in MD participant
cohort).
AEs of special interest will be tracked and are defined as occurrences of:
* ARIA-E,
* ARIA-H,
* An AE Grade 2 or higher of Uveitis.
6.2.2. Pharmacokinetic Endpoints
Pharmacokinetic endpoints for the study are:
* Serum concentration of AL002 at specified time points,
* Relationship between serum concentration or PK parameters for AL002 and
safety endpoints, Relationship between serum, CSF concentration, or PK
parameters for AL002 and activity or PD endpoints (relationship with activity
is an endpoint only for the MD participant cohort - i.e. participants with AD).
6.2.3. Exploratory Clinical Outcomes
Exploratory clinical outcome endpoints (for the MD participant cohort only -
i.e. participants with AD) are:
* Clinical Dementia Rating Sum of Boxes (CDR-SB) score (change after dosing
relative to baseline)
* Mini-Mental State Examination (MMSE) score (change after dosing relative to
baseline)
* Repeatable Battery for the Assessment of Neuropsychological Status (RBANS)
score (change after dosing relative to baseline).
6.2.4. Exploratory Biomarker Endpoints
Analyses of exploratory biomarker endpoints for the study may include the
following, or additional exploratory analyses:
* changes in levels of sTREM2 in plasma (all cohorts) and CSF (from selected
cohorts) after dosing relative to baseline concentration,
* relationship between biomarkers at baseline, including common and rare
genetic variants, identified through whole genome sequencing (WGS) performed on
deoxyribonucleic acid extracted from blood, and safety, PK, activity,
immunogenicity, or other biomarker endpoints (relationship with activity is an
endpoint only for the MD participant cohort only - i.e. participants with AD),
* change in brain amyloid burden as assessed by Amyloid-positron emission
tomography (PET) in the MD participant cohort only, i.e. participants with AD,
* changes in markers of neuroinflammation and disease process in CSF and
plasma.
Secondary outcome
See above
Background summary
Alzheimer*s disease (AD) is a degenerative brain disease and is the most common
cause of dementia in the United States (US), affecting approximately 5.5
million Americans. Worldwide, 50 million people are living with dementia, and
this prevalence is expected to triple by 2050. Of the top 10 causes of death in
the US, AD is the only major cause of morbidity and mortality in the US without
suitable treatments for prevention, slowing or cure (2017 Alzheimer*s
Association Report).
Current therapies for AD such as acetylcholinesterase inhibitors (e.g.
donepezil) and N-methyl-D-aspartate (NMDA) receptor antagonists (e.g.
memantine) show only modest and transient benefits to cognition and behavior
parameters in AD patients but do not slow or halt the progression of the
disease (Cummings, 2004). Given the large number of Alzheimer* disease patients
and expected increases in this patient population due to an extended life
expectancy and a major worldwide growth in the population of older adults, an
effective treatment for AD remains an urgent unmet medical need.
Human genetic studies have identified inherited mutations that underlie
familial forms of AD, but these mutations are rare (occurring in less than 5%
of all cases). More recently, large human genetic association studies have
revealed genetic loci that modify the risk of common sporadic forms of AD
(Tanzi, 2012). Interestingly, many of these loci encode for proteins expressed
primarily on innate immune cells, including microglia, macrophages, and
dendritic cells. Microglia are resident macrophages of the central nervous
system and serve protective housekeeping functions such as facilitating
clearance of cellular debris through phagocytosis, as well as secretion of
growth factors. Thus, in the course of neurodegenerative disease such as AD,
these cells may serve an important protective role when activated appropriately.
The most prominent microglial gene that modifies the risk of common sporadic
forms of AD encodes for triggering receptor expressed on myeloid cells 2
(TREM2). TREM2 is an immunoglobulin-like receptor that is expressed primarily
on myeloid lineage cells, such as macrophages, dendritic cells, and microglia
(Colonna, 2016). TREM2 is thought to play a key role in modulating the innate
immune response, such as in response to bacteria in the context of infection or
to dying neurons and other debris in neurodegenerative disorders including AD
(Colonna, 2016).
Heterozygous mutations in the TREM2 gene increases the risk of AD by up to
3-fold (Guerreiro, 2013; Jonsson, 2013), and increases the rate at which brain
volume shrinks (Rajagopalan, 2013). Even individuals without AD who carry a
heterozygous TREM2 mutation show impaired cognition compared to individuals
with 2 normal TREM2 alleles.
Recent mouse genetic model studies strongly support a key role for TREM2 in AD,
with loss of TREM2 (through functional mutations and in knockout models) being
associated with increased pathology (Cheng-Hathaway, 2018). These findings are
suggestive of potential improvements in cognitive function under pathological
conditions with the application of TREM2 agonistic antibodies. Wang et al.
(Wang, 2015) showed that in the 5xFAD transgenic model of AD, deficiency in
TREM2 function exacerbated AD pathology. Subsequent studies went further to
detail the pathological changes seen in 5xFAD mice that lack 1 or both copies
of TREM2, showing a consistent defect in the ability of TREM2 mutant microglia
to effectively surround and engulf amyloid plaques (Wang, 2016; Yuan, 2016;
Jay, 2017a). In these TREM2-defective microglia, the reduction in microglia
per-plaque was associated with more diffuse plaques and more axonal damage from
surrounding plaques.
It has been shown that TREM2 expression enhances microglial cell survival,
proliferation and differentiation, and regulates microglial chemotaxis and
phagocytosis. In the context of AD pathology, TREM2 expression impacts amyloid
pathology, modulates neuritic dystrophy, tau hyperphosphorylation and
aggregation, and affects synaptic and neuronal loss (Jay, 2017b). In addition,
it has been shown that TREM2 plays a key role in limiting the development of
peri-plaque tau pathologies (Leyns, 2019).
In summary, the loss of TREM2 function is detrimental as demonstrated in human
and mouse genetic studies, while conversely, activating TREM2 on microglial
cells is protective against damage in the process of neurodegeneration.
The approach of Alector Inc. is to utilize TREM2 agonistic antibodies to
ameliorate AD pathology through activation of the innate immune system, thereby
improving the clearance and sequestration of the molecular factors causing
pathology. Alector Inc. has generated TREM2 selective agonistic antibodies that
synergize with endogenous ligands to treat AD. Alector Inc. has subsequently
developed a humanized therapeutic TREM2 antibody candidate, AL002, with
optimized agonistic activity. As demonstrated in the data from Alector Inc.*s
pre-clinical studies, activating TREM2 can effectively suppress AD pathology in
vivo, to prevent cognitive decline in animal models. To the best of Alector
Inc.*s knowledge, an anti-TREM2 agonistic antibody has not previously been
investigated in human clinical studies.
Alector Inc. proposes to evaluate the safety, tolerability, pharmacokinetic
(PK) and pharmacodynamic (PD) profiles in plasma and cerebrospinal fluid (CSF)
and anti-drug antibody (ADA) responses in normal healthy adult participants and
patients with AD following intravenous (IV) administration with AL002.
As mentioned above, defective TREM2 functions play a central role in the
pathogenesis of AD. Microglia are efficient sensors of changes in the central
nervous system microenvironment, and their neuroprotective role has been
hypothesized to be impaired during aging (Mecca, 2018), one of the strongest
risk factors for AD. It is well known that TREM2 is required to sustain
microglial trophic function in the aging brain, and animal studies showed that
an overlap exists between aged microglia phenotype and microglial molecular
signatures found in models of AD (Krasemann, 2017). This typical signature of
aged microglia includes the TREM2 pathways. In vitro experiments demonstrated
that AL002 binds to the R47H mutant form of TREM2, as illustrated in Section
4.1.2.3 of the Investigator*s Brochure, indicating that AL002 can activate the
receptor even in the presence of TREM2 coding variants that are associated with
AD risk. The rationale for including a cohort with AD patients carrying a R47H
or R62H mutation is to characterize and compare the molecular signatures of
TREM2 mutations carriers versus non-carriers and their response to treatment
with AL002 using blood (plasma and white blood cells [WBC]s), CSF, and imaging
biomarkers. This may facilitate a better understanding of the function of TREM2
in AD and will allow evaluation of whether AD patients respond differently to
treatment with AL002 based on their genetic status.
Study objective
The primary objective of the study is to evaluate the safety, tolerability, PK,
and PD of AL002 administered in single ascending doses in healthy participants
and multiple doses in participants with mild to moderate AD.
Study design
A Phase I Study Evaluating the Safety, Tolerability, Pharmacokinetics,
Pharmacodynamics, and Immunogenicity of Single and Multiple Doses of AL002 in
Healthy Participants and in Participants with Mild to Moderate Alzheimer*s
Disease.
Approximately 22 participants will be enrolled in one of two groups: L and M
and will receive multiple doses of AL002 or placebo (fake drug) directly into
the blood.
Intervention
Group L
In group L 12 participants will participate of which approximately 10
participants will receive the study drug and 2 participants placebo. Of the
first two participants in group L one will receive the study drug and the other
placebo. Randomization will determine which treatment you receive.
Group M
In group M all 10 participants will receive the study drug AL002 and no
participants will receive placebo.
We will administer study drug or placebo - at 2 visits
Study burden and risks
For the study, you have to visit study hospital 15 times in 26 weeks. A visit
will take approximately 4 hours.
- We will measure your vital signs - at 15 visits
- We will perform a physical examination - at 13 visits
- We will perform an electrocardiogram (ECG) - at 5 visits
- We will collect blood samples to:
o Asses your general health - at 10 visits, approximately 2 tubes at a time
o Measure the amount of AL002 in your blood - at 14 visits, approximately 1
tube at a time
o Measure the effect of AL002 on your body - at 14 visits, approximately 1 tube
at a time
o Perform Whole Genome Sequencing (WGS) - at 1 visit, approximately 1 tubes at
a time
o Measure Anti-drug antibodies - at 7 visits, approximately 1 tube at a time
- We will collect urine samples to:
o Test your general health - at 3 visits
o For a drug screen, the result must be negative for you to continue in the
study - at 1 visit
o Perform a urine pregnancy test, if you are able to get pregnant, the result
must be negative (not pregnant) for you to continue in the study - at 7 visits
- We will perform an alcohol breath test: when you check into the hospital on
Day 1 to see if you have been drinking any alcohol in the past 24 hours. The
result must be negative for you to continue in the study - at 1 visit
- We will perform a neurological assessment - at 13 visits
- We will perform eye exams - at 2 visits
- We will administer study drug - at 2 visits
- We will collect brain fluid samples - at 2 visits, approximately 3 tubes at a
time.
- We will have you complete a Sheehan-STS (Suicidality Tracking Scale)
questionnaire to assess suicidal thoughts and behaviors - at 13 visits. If you
are having suicidal thoughts call the study doctor at the telephone number
listed on the first page of this form
- We will perform a Mini-Mental State Examination (MMSE) - at 2 visits
- We will perform a Repeatable Battery for the Assessment of Neuropsychological
Status (RBANS) - at 2 visits
- We will perform a Clinical Dementia Rating (CDR) - at 2 visits
- We will perform a Brain MRI (magnetic resonance imaging) - at 3 visits
- We will perform an Amyloid PET (positron emission tomography) scan - at 2
visits
- We will perform a Concomitant Medication review - at 15 visits
- We will ask you about Adverse Events - at 15 visits
131 Oyster Point Blvd, Suite 600
South San Francisco CA 94080
US
131 Oyster Point Blvd, Suite 600
South San Francisco CA 94080
US
Listed location countries
Age
Inclusion criteria
1. Total body weight between 50 and 120 kg, inclusive.
2. Clinical laboratory evaluations (including chemistry panel fasted [fasted at
least 8 hours], complete blood count, and urine analysis) within the reference
range for the test laboratory, unless deemed not clinically significant by the
Investigator. A count of the segmented neutrophils and bands should be
performed when results from the white blood cells (WBCs) are not within the
reference range.
3. Negative test for selected drugs of abuse at screening (does not include
alcohol) and at admission (testing at admission does include alcohol breath
test). A positive result may be verified by re-testing (up to 1 false positive
result permitted) and may be followed up at the discretion of the Investigator.
4. Females must be non-pregnant and non-lactating, and either surgically
sterile (e.g. tubal occlusion, hysterectomy, bilateral salpingectomy, bilateral
oophorectomy), or use highly effective contraceptive method (oral
contraceptives pills [OCPs], long acting implantable hormones, injectable
hormones, a vaginal ring or an intrauterine device [IUD]) from screening until
study completion, including the follow-up period for at least 16 weeks after
the last dose of AL002, or be post-menopausal for *12 months. For healthy
volunteers, post-menopausal status will be confirmed through testing of FSH
levels (* 40 IU/mL) at screening; for participants with AD, post-menopausal
status will be assessed through medical history with assessment of potential
alternative causes of amenorrhea as clinically indicated). Females who are
abstinent from heterosexual intercourse will also be eligible.
5. Women of child-bearing potential (WOCBP) must have a negative pregnancy test
at screening and admission and be willing to have additional pregnancy tests as
required throughout the study.
6. Males must be surgically sterile (>30 days since vasectomy with no viable
sperm), abstinent, or if engaged in sexual relations with a WOCBP, the
participant and his partner must be surgically sterile (e.g. tubal occlusion,
hysterectomy, bilateral salpingectomy, bilateral oophorectomy) or using an
acceptable, highly effective contraceptive method from screening until study
completion, including the follow-up period, for at least 16 weeks after the
last dose of AL002. Acceptable methods of contraception include the use of
condoms and the use of an effective contraceptive for the female partner
(WOCBP) that includes: OCPs, long acting implantable hormones, injectable
hormones, a vaginal ring or an IUD. Male participants whose female partner is
post-menopausal, and participants who are abstinent from heterosexual
intercourse will also be eligible. Male participants must agree to refrain from
donating sperm from screening until study completion, including the follow-up
period, for at least 16 weeks after the last dose of AL002.
In addition, for the MD cohorts (i.e. participants with AD):
9. Ages 50-85 years, inclusive.
10. The participant should be capable of completing assessments either alone or
with the help of the study partner (where appropriate), per local guidelines.
11. Availability of a person (*study partner*) who, in the Investigator's
judgment, has frequent and sufficient contact with the participant and is able
to provide accurate information regarding the participant*s cognitive and
functional abilities, agrees to provide information at clinic visits, which
require partner input for scale completion, and signs the necessary consent
form, per local guidelines.
12. Clinical diagnosis of probable AD dementia based on National Institute on
Aging Alzheimer*s Association criteria.
13. Screening MMSE score of 16-28 points, inclusive.
14. Screening Clinical Dementia Rating-Global Score (CDR-GS) of 0.5, 1.0, or
2.0.
15. Positive amyloid-PET scan by qualitative read, as defined in the PET
Imaging Charter.
16. If already taking cholinesterase inhibitor and/or memantine therapy for AD,
on a stable dose for at least 4 weeks prior to screening. There should be no
intent to initiate, discontinue, or alter the dose of any therapy for AD for
the duration of the study.
In addition, for MD Cohort M (i.e. participants with AD with a TREM2 mutation):
17. The participant must carry at least 1 of the TREM 2 mutations: R47H or
R62H.
Exclusion criteria
1. Pregnant, lactating, or intending to become pregnant within 16 weeks after
last dose of study drug.
2. Participation in a clinical trial within 30 days before randomization; use
of any experimental oral therapy within 30 days or 5 half-lives prior to Day 1,
whichever is greater; or use of any biologic therapy within 12 weeks or 5
half-lives prior to Day 1, whichever is greater. Participants who have received
an experimental therapy that has no half-life, like a vaccine, should have
completed that therapy at least 12 weeks prior to Day 1. Participants who have
received an experimental vaccine against a central nervous system target, such
as beta-amyloid or tau, are not eligible for this study.
3. Any non-experimental vaccine within 2 weeks of randomization, until 2 weeks
after the last dose. It is advised that prospective participants receive their
annual influenza vaccine as early as possible in advance of the flu season, and
then wait 2 weeks prior to randomization. It is permitted to receive the annual
influenza vaccine during the screening period.
4. Surgery or hospitalization during the 4 weeks prior to screening.
5. Planned procedure or surgery during the study.
6. Blood transfusion within 8 weeks prior to screening.
7. Donation or loss of blood (excluding the volume of blood that will be drawn
during screening procedures) as follows: 50-499 mL of blood within 30 days or >
499 mL of blood within 56 days prior to study drug administration.
8. Poor peripheral venous access.
9. History of major depression (within the past 5 years) unless effectively
treated at enrollment and for the duration of the study, at the discretion of
the Investigator. History of schizophrenia, schizoaffective disorder, or
bipolar disorder.
10. Alcohol and/or substance abuse or dependence (according to the Diagnostic
and Statistical Manual of Mental Disorders, Fifth Edition) within the past 2
years.
11. Within the last 2 years, unstable or clinically significant cardiovascular
disease (e.g. myocardial infarction, angina pectoris, New York Heart
Association Class II or more cardiac failure).
12. Uncontrolled abnormal blood pressure
a. For healthy volunteers, as indicated by sustained supine systolic blood
pressure (BP) > 140 or < 90 mm Hg or supine diastolic blood pressure > 90 or
<50 mm Hg at screening or admission. Duplicate assessments will be performed
and the average of the 2 assessments of BP will be used to exclude a
participant.
b. For MD participants with AD, sustained diastolic blood pressure >95 mm Hg
performed either sitting or supine. No repeated measurements for eligibility
are required for multidose participants.
13. Resting heart rate at screening of >100 or < 40 beats per minute.
14. Chronic kidney disease as indicated by a screening creatinine clearance <
30 mL/min as calculated by the central laboratory using the Cockcroft Gault
formula, which remains < 30 mL/min if retested.
15. Impaired hepatic function as indicated by screening aspartate
aminotransferase (AST) or alanine aminotransferase (ALT) * 2 or total bilirubin
* 1.5 x the upper limit of normal, which remains above these limits if retested
due to a slightly elevated initial result or abnormalities in synthetic
function tests that are judged by the Investigator to be clinically significant.
16. History of, or known to currently have, Human Immunodeficiency Virus (HIV)
infection, hepatitis B or hepatitis C infection that has not been adequately
treated in the opinion of the Investigator.
17. History or presence of infections of the central nervous system (e.g.
syphilis, Lyme, or borreliosis, viral or bacterial meningitis/encephalitis, HIV
encephalopathy).
18. History or presence of central nervous system or systemic autoimmune
disorders including but not limited to rheumatoid arthritis, multiple
sclerosis, lupus erythematosus, anti-phospholipid antibody syndrome, Behçet
disease.
19. Present or past history of uveitis requiring medical intervention, chronic
inflammatory or degenerative condition of the eye, current eye infection, any
ongoing eye disorder requiring injectable medical therapy (e.g. ranibizumab or
aflibercept for macular degeneration) or planned invasive eye procedure during
the study period.
20. Systemically, clinically significantly immunocompromised patients, owing to
continuing effects of immune suppressing medication.
21. Positive for Hepatitis C virus (HCV) antibody, Hepatitis B surface antigen
(HBsAg), or HIV antibody.
22. History of cancer except:
a. if considered likely to be cured,
b. is not being actively treated with anti-cancer therapy or radiotherapy and,
in the opinion of the Investigator, is not likely to require treatment in the
ensuing 3 years,
c. considered to have low probability of recurrence (with supporting
documentation from the treating oncologist if possible),
d. for prostate cancer or basal cell carcinoma, no significant progression over
the last 2 years,
e. Adequately resected squamous cell skin cancer.
23. Known history of severe allergic, anaphylactic, or other hypersensitivity
reactions to chimeric, human, or humanized antibodies or fusion proteins.
24. Past history of seizures, with the exception of childhood febrile seizures.
25. At risk of suicide in the opinion of the Investigator.
26. Any serious medical condition or abnormality in clinical laboratory tests
that, in the Investigator*s judgment, precludes the participant*s safe
participation in and completion of the study.
27. History or presence of an abnormal ECG that is clinically significant in
the Investigator*s opinion, including complete left bundle branch block,
second- or third-degree heart block, evidence of prior myocardial infarction
(except if due to a myocardial infarction that occurred more than 2 years
before screening).
28. History of ventricular dysrhythmias or risk factors for ventricular
dysrhythmias such as structural heart disease (e.g. severe left ventricular
systolic dysfunction, left ventricular hypertrophy), coronary heart disease
(symptomatic or with ischemia demonstrated by diagnostic testing), clinically
significant electrolyte abnormalities (e.g. hypokalemia, hypomagnesemia,
hypocalcemia), or family history of sudden unexplained death or long QT
syndrome.
29. Current treatment with medications that are well known to prolong the QT
interval, at the Investigator*s discretion.
30. Immunosuppression caused by disease (such as HIV) or medications,
immunosuppressive therapy (such as long-term systemic corticosteroid therapy)
within 12 months before screening through the entire study period.
31. Smoking more than 5 cigarettes, 1 cigar, or 1 pipe daily.
32. Contraindication to lumbar dural puncture, including coagulopathy,
concomitant anticoagulation (except for platelet inhibitor such as aspirin or
clopidogrel), thrombocytopenia, or other factor that precludes safe lumbar
puncture in the opinion of the Investigator.
In addition, for SAD cohorts (i.e. healthy adults):
33. QT interval corrected using Fridericia*s formula (QTcF) > 450 msec
demonstrated by at least 2 ECGs > 30 minutes apart.
34. The use of all prescribed medication is not allowed (unless discussed and
agreed upon by both the Sponsor and the PI) at least 30 days prior to admission
to the clinical research center until follow-up. The use of all over the
counter medication, vitamin preparations and other food supplements or herbal
medications (e.g. St. John*s Wort) is not allowed (unless discussed and agreed
upon by both the Sponsor and the PI) for at least 14 days prior to admission to
the clinical research center until follow-up. The use of
paracetamol/acetaminophen (up to 2000 mg/day) is allowed for the treatment of
headache or any other pain. Other medication to treat AEs may only be
prescribed if deemed necessary by the Investigator.
In addition, for the MD cohorts (i.e. participants with AD):
35. A lack of ability to consent, in accordance with the local regulations,
guidelines, and independent ethics committee (IEC) or institutional review
board (IRB).
36. Dementia due to a condition other than AD, including, but not limited to,
Frontotemporal Dementia, Parkinson*s disease, dementia with Lewy bodies,
Huntington disease, or vascular dementia.
37. History or presence of clinically evident vascular disease potentially
affecting the brain (e.g. clinically significant carotid, vertebral stenosis or
plaque; aortic aneurysm; intracranial aneurysm; cerebral hemorrhage;
arteriovenous malformation) that in the opinion of the Investigator has the
potential to affect cognitive function.
38. History or presence of stroke within the past 2 years or documented history
of transient ischemic attack within the last 12 months.
39. History of severe, clinically significant (persistent neurologic deficit or
structural brain damage) central nervous system trauma (e.g., cerebral
contusion).
40. Inability to tolerate MRI procedures or contraindication to MRI, including,
but not limited to, presence of pacemakers (with the exception of MRI-safe
pacemakers), aneurysm clips, artificial heart valves, ear implants, or foreign
metal objects in the eyes, skin, or body that would contraindicate an MRI scan;
or any other clinical history or examination finding that, in the judgment of
the Investigator, would pose a potential hazard in combination with MRI.
41. MRI evidence of:
a. more than 2 lacunar infarcts,
b. any territorial infarct > 1 cm3, or
c. significant FLAIR hyperintense lesions in the cerebral white matter that
may, in the Investigator*s opinion, contribute to cognitive dysfunction.
42. Any other severe or unstable medical condition that, in the opinion of the
Investigator or Sponsor, could be expected to progress, recur, or change to
such an extent that it could put the participant at special risk, bias the
assessment of the
clinical or mental status of the participant to a significant degree, interfere
with the participant*s ability to complete the study assessments, or would
require the equivalent of institutional or hospital care.
43. Residence in a skilled nursing facility such as a convalescent home or
long-term care facility. Participants who subsequently require residence in
these facilities during the study may continue in the study and be followed for
efficacy and safety, provided that they have a study partner who meets the
minimum requirement.
44. The following medications are prohibited as daily treatment from 1 month
prior to screening until the end of the study. They are, however, permitted on
an intermittent, as needed basis at any point during the study, provided that
no dose is taken within 2 days before any neurocognitive assessment:
a. typical anti-psychotic or neuroleptic medication,
b. narcotic analgesics,
c. sedative, hypnotic, or benzodiazepine medication,
d. tricyclic antidepressant medications,
e. any sedating antihistamine medication (diphenhydramine or other similar over
the counter antihistamine therapy).
45. QT interval corrected using Fridericia*s formula (QTcF) > 470 msec
demonstrated by at least 2 ECGs > 30 minutes apart for male participants and QT
interval corrected using Fridericia*s formula (QTcF) > 480 msec demonstrated by
at least 2 ECGs > 30 minutes apart for female participants.
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 | EUCTR201900020630-NL |
CCMO | NL71749.056.19 |