Sei sulla pagina 1di 10

Key Symposium | doi: 10.1111/j.1365-2796.2010.02315.

Immunotherapy for Alzheimer’s disease


D. Morgan
From the Alzheimer’s Institute, University of South Florida, Tampa, FL, USA

Abstract. Morgan D. (Alzheimer’s Institute, University of deposition. Additional clinical trials with passive
South Florida, Tampa, FL, USA) Immunotherapy for immunotherapy confirmed occasional appearance of
Alzheimer’s Disease (Key Symposium). J Intern Med microhaemorrhage and occurrence of vasogenic
2011; 269: 54–63. oedema in some patients, particularly those with the
apolipoprotein E4 genotype. Recent data with posi-
In the year1999, a vaccine approach was found to tron emission tomography demonstrates trial partici-
reduce amyloid deposits in transgenic mice overpro- pants passively immunized with anti-Aß antibodies
ducing the amyloid precursor protein. This was have reduced signals with amyloid binding ligands
followed closely by demonstrations that vaccines or after 18 months of therapy. Several anti-Aß immu-
passive immunotherapy could rescue memory defi- notherapies have reached phase 3 testing, and
cits in these mice. Initial human clinical trials re- immunotherapy is likely to be the first test of the
vealed apparent autoimmune reactions in a subset of amyloid hypothesis of Alzheimer’s disease. Identify-
patients, but also some cases of cognitive benefit and ing antibody variants that retain amyloid clearance
amyloid clearance. Further work with passive immu- with fewer adverse reactions remains a major focus of
notherapy in mouse models confirmed exceptional translational research in this area.
clearing abilities of anti-amyloid antibodies even in
older mice. However, in parallel with parenchymal Keywords: Alzheimer’s disease, antibody, microhaem-
amyloid clearance was the appearance of micro- orrhage, transgenic mice.
haemorrhages and increased vascular amyloid

Introduction Although debated intensely for the past 25 years,


there is now a reasonable consensus emerging
Alzheimer’s disease is the most common form of or-
regarding the pathogenesis of Alzheimer’s disease.
ganic dementia. It affects roughly10% of the popula-
The initiating factor, which appears necessary but
tion over 65 and 40% of those over 85 years of age. In
not sufficient for Alzheimer’s disease, is the accumu-
the US, 7–8% of all medical costs are related to
lation of amyloid aggregates consisting of the Aß pep-
dementia. Unlike other leading causes of death, mor-
tide. The genetics of familial forms of Alzheimer’s dis-
tality from Alzheimer’s disease is increasing. The
ease and Down’s syndrome cases (which result in
number of cases is predicted to escalate dramatically
precocious Alzheimer pathology) have as a common
over the next 2–3 decades as success in treating heart
element overproduction of a longer C-terminal form
disease and cancer permit more individuals to reach
of the Aß peptide (42 amino acids in length). This form
the age of risk for dementias.
is more prone to form beta sheet structures and
aggregate into oligomers and fibrils [3]. Evidence is
The definition of Alzheimer’s dementia is based on a
now emerging that these amyloid deposits may be
case report by Alois Alzheimer (1907) based on a pla-
present up to a decade prior to the initiation of cogni-
que and tangle pathology within the brain of a women
tive symptoms of the disorder [4, 5].
succumbing to the disorder in her fifties. Until re-
cently, the definitive diagnosis of Alzheimer’s re-
A second step in the pathogenesis of the disease is the
quired either biopsy or postmortem histopathology
formation of intraneuronal neurofibrillary tangles
[1]. The introduction of ligands labelling amyloid pla-
formed from hyperphosphorylated forms of the
ques with positron emitting isotopes and spinal fluid
microtubule binding protein tau. Other neurodegen-
measurements of specific molecules combined with
erative disorders can be formed by the tau pathology
cognitive testing now permits more definitive diagno-
in the absence of amyloid deposits, but these differ
ses to be ascertained without requiring histopathol-
from Alzheimer’s both in clinical presentation and in
ogy [2].

54 ª 2010 The Association for the Publication of the Journal of Internal Medicine
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

the location of the pathology regionally in the brain. ‘‘Alzheimer’s mice’’ and that treatments effective in
In tau transgenic mouse models, the tau deposits can the mice should be effective in Alzheimer patients.
be precipitated by intracranial injection of amyloid [6] Unfortunately, the APP mice lack a number of critical
or breeding with mice producing Aß deposits [7]. features of Alzheimer’s, including hyperphosphory-
Moreover, interrupting the amyloid deposition with lated tau pathology and significant neuron loss. At
anti-Aß immunotherapy can diminish the progres- best they resemble an early preclinical phase of the
sion of tau pathology in mice expressing multiple disease, nonetheless one which may be the optimal
transgenes [8]. Moreover, the tau pathology appears phase in which to initiate therapy that prevents the
to be more closely correlated with cognitive status disorder.
than the amyloid pathology, consistent with it being
more proximal cause of the mental dysfunction [9]. The seminal paper in the field of immunotherapy for
By uncertain mechanisms, these pathologies result Alzheimer’s disease was published in 1999. Schenk
in the loss of synaptic function, synapses and ulti- and his collaborators at Elan Pharmaceuticals (nee
mately a loss of neurons leading to considerable brain Athena Neuroscience) demonstrated that treating
atrophy. There are multiple hypotheses regarding the mice with a vaccine against the Aß peptide in an APP
mechanistic steps involved, some of which argue the mouse model prior to the formation of deposits was
intermediate sized aggregates of amyloid and ⁄ or tau, effective in blocking amyloid accumulation as the
referred to as oligomers, may be more directly causing mice aged. Within weeks of this publication, our re-
the toxicity, but demonstrations in vivo are still lack- search group in Tampa, Florida began treating APP
ing. Even in the mildest stages of the disorder, the so- mice at a later age, when amyloid deposits had al-
called ‘‘mild cognitive impairment’’ phase, there ap- ready become manifest. One pathogenic component
pears to be considerable accumulation of plaque and of Alzheimer’s disease pathology was argued to be the
tangle pathology, and neuron loss [10]. Increasingly inflammation associated with amyloid deposits, and
these observations suggest that treating the disorder we had concerns that the immune activation associ-
at the earliest possible stages, much as is performed ated with the vaccine might aide in clearing the amy-
with cardiovascular disease, will be essential to con- loid plaques, but may also cause destructive bystan-
trolling Alzheimer’s disease. der effects to the surrounding tissue. We also had
recently detailed the time course of memory deficien-
cies developing in our APP + PS1 mouse model [17]
Studies of active immunotherapy in animal models
as the amyloid accumulated [18]. After several
The gene responsible for the protein containing the months of repeated immunization, we tested the mice
Aß peptide is the amyloid precursor protein (APP). for behavioural deficiencies, but found no deficits in
APP was cloned in the late 1980s following the either the treated or untreated mice. However, when
sequencing of the vascular Aß deposits by Glenner we continued the treatment to an age when we ex-
et al. [11]. Several years later, it was identified that pected memory deficits to be present, we found the
some families inheriting Alzheimer’s disease in an vaccinated mice were protected from these deficits,
autosomal dominant fashion had mutations in APP whilst mice given a control vaccination were unable
near the cleavage sites releasing the Aß peptide [12]. to learn the task [19]. A second research group work-
This led to a very substantial number of largely failed ing independently found qualitatively similar effects
attempts to develop transgenic mice overexpressing on memory in a different mouse model [20]. These
mutant forms of human APP [13]. Finally, two models observations that vaccines or antibodies against the
emerged [14, 15] which demonstrated amyloid depo- Aß peptide can rescue the memory deficits in these
sition in locations and with features common to that models have been replicated extensively, in some
found in Alzheimer’s disease. Although some claim cases demonstrating that rescue can be seen within
that certain of these APP transgenic models are supe- days of treatment (before clearance of plaque deposits
rior and other APP models lacking, the remarkable can be detected; [21, 22].
feature is that the patterns of Aß deposition are lar-
gely the same in all models in spite of different muta- Importantly, the efficacy of the vaccine in reversing
tions and promoters (cortical and hippocampal dif- amyloid deposits is not only found in transgenic mice.
fuse Aß deposits, compacted neuritic plaques and Nonhuman primates also develop amyloid deposits
vascular amyloid are found in most APP models and like Alzheimer patients and associated memory defi-
Alzheimer’s tissue [16]). These transgenic mice are cits (although like the mice, they do not develop tau
superb representations of amyloid deposition. pathology) [23]. These can be reversed within months
Unfortunately, many authors argue that they are using vaccination against the Aß peptide [24]. Beagle

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63 55
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

dogs also develop diffuse amyloid deposits and mem-


ory dysfunction with age. Two years of active immuni-
zation were found to clear these diffuse (not com-
pacted) deposits, but not have impact on the age-
associated memory impairments in these animals
[25]. It is important to recognize that most mammals
develop age-associated memory impairments and
the majority lack amyloid deposition of any type.
Hence, memory deficiencies with age are not always
associated with amyloid deposits.

The development of novel vaccine strategies and ad-


juvants against the Aß peptide has been an area of in-
tense creativity. In most instances, the goal has been
to develop B-cell activation and antibody production, Fig. 1 Opsonization and Phagocytosis. Anti-Ab antibodies
with minimal T-cell involvement (at least against Aß), bind to Ab aggregates within the CNS. The immune complex
because of the adverse events found in human trials then stimulates phagocytosis by Fc-gamma receptor bearing
with vaccines against Aß (see below). These impres- macrophages. The ingested Ab is then digested and ⁄ or
exported from the CNS. Reprinted from Ugen, K. and Morgan,
sive approaches have been carefully described in a
D. DNA and Cell Biology 20:677–678.
recent review [26] and are outside the scope of
the present review. However, long-term, assuming
immunotherapy is successful against Alzheimer’s
pathology, these may become standard approaches one form of Aß can clear other forms, assuming that
to preventing the disease. heteromeric fibrils containing these different forms
are present [30, 31].
Mechanisms of immunotherapy action in reducing Aß deposits
A second proposed mechanism assumes that the
The success of the immunotherapy approach to brain penetration of the antibodies into CNS is not a critical
disorder surprised a number of investigators given step and that the presence of circulating antibodies
the dogma that the brain is an immune-privileged or- creates a ‘‘peripheral sink’’ which alters the equilib-
gan with minimal immune surveillance. Nonetheless, rium across the blood brain barrier for Aß to favour ef-
the normal range of CSF IgG (5–50 mg L)1) is roughly flux owing to the reduced free Aß concentration in
0.1% of the plasma IgG concentration, with an aver- blood [32] (Fig. 2). Certainly, many antibodies with
age concentration of 0.1 lmol L)1. Studies in rodent high affinity for Aß do dramatically elevate circulating
with iodinated anti-Aß antibodies confirm that concentrations. There also appear to be mechanisms
0.11% of the circulating antibody enters the CNS by which circulating Aß does cross into the CNS,
[27]. This has led to several proposed mechanisms which would be expected to decline as free Aß
regarding the reduction in Aß deposition associated dropped [33]. However, it has also been argued that
with anti-Aß immunotherapy. the major cause of increased Aß in circulation with
antibody administration is owing to the reduced rates
The first proposed mechanism relied upon the tradi- of clearance from the blood [34]. A variant of this idea
tional role of antibody to opsonize antigens leading to is that the antibodies in the brain ventricular fluid
macrophage phagocytosis and complement activa- might bind Aß and act as a sink within the central
tion [28] (Fig. 1). This approach assumes that suffi- nervous system.
cient antibody enters the brain and binds to the amy-
loid to trigger this phagocyte action of either resident A third option is that antibodies act catalytically to
microglia, or infiltrating monocytes ⁄ macrophages. modify the secondary structure of the Aß monomers
Certainly, our own work and that of others have dem- into a conformation that is less likely to form the
onstrated decoration of amyloid deposits after sys- aggregates associated with oligomeric or fibrillar
temic administration of anti-amyloid antibodies [29]. forms (Fig. 3). This was first proposed by Beka Solo-
One advantage of this mechanism is that the stoichi- mon et al. before the capacity of antibodies to affect
ometry to the antibody to Aß can be considerably Aß in vivo was demonstrated [35, 36]. She has dem-
<1 : 1 and still have a meaningful impact on deposi- onstrated that stoichiometries of antibodies as low as
tion. It also may explain how antibodies selective to 1 : 10 effectively blocked Aß fibril formation in vitro.

56 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

saturable and inhibited by Fc fragments. One possi-


ble consequence is that excess antibody may lead to
competition within this efflux mechanism by increas-
ing transport of antibody without bound antigen. Our
own work and that of others has found that excess
anti-Aß monoclonal antibody administered systemi-
cally can inhibit clearance of amyloid plaques [39,
40].

Critically, none of these mechanisms are mutually


exclusive. Moreover, different antibodies may utilize
different mechanisms to different degrees. For exam-
ple, antibodies not binding fibrillar amyloid may be
less able to induce phagocytosis, yet function excep-
tionally as a peripheral sink. Some antibodies upon
binding Aß may not modify the secondary structure,
Fig. 2 Peripheral Sink. Circulating anti-Ab antibodies bind
but still activate macrophages and FcRn-mediated
free Ab in the blood and increase the efflux of Ab down its con-
centration gradient and ⁄ or block the influx of Ab from the cir- transport. The extent to which different antibodies
culation and back into the brain. Reprinted from Ugen, K. and utilize different mechanisms may also confer differ-
Morgan, D. DNA and Cell Biology 20:677–678. ent adverse event profiles for each of the antibodies.

Active immunization (vaccine) clinical trials


Given the remarkable success of vaccination in ani-
mal models and the absence of any alternative dis-
ease modifying therapy for Alzheimer’s disease, Elan
partnered with Wyeth to initiate an active immuniza-
tion trial against the Aß peptide in Alzheimer pa-
tients. The vaccine consisted of the full length pep-
tide, which contains both B- and T-cell epitopes,
aggregated into fibrils. Initially, about 60 patients
were treated with one or more doses of the vaccine
Fig. 3 Catalytic Modification of Conformation. Antibody and the cases monitored in a phase 1 safety trial. One
binds Ab and modifies the secondary structure to one which of the initial observations was a variable antibody re-
minimizes the formation of aggregates. Reprinted from Ugen, sponse, with many patients failing to develop detect-
K. and Morgan, D. DNA and Cell Biology 20:677–678. able titres against the antigen. Approximately half
way through the trial, there was a change in the adju-
vant to QS-21 in an attempt to enhance this response.
We have confirmed this and extended the results to There were no adverse responses detected during the
indicate that stoichiometries of antibody to Aß as low trial portion of the phase I study [41].
as 1 : 1000 can specifically modify the secondary
structure of Aß and block fibril formation using ATR- This led Elan and Wyeth to pursue a phase 2a trial
FTIR and atomic force microscopy [37]. Obviously, with the AN1792 formulation including the QS-21
this catalytic modification hypothesis benefits from adjuvant. The goal was to immunize patients to a pre-
being active at low stoichiometries and may be active set antibody titre using multiple inoculations. Within
in the absence of macrophage activation, a circum- a short time of initiating the trial, however, the immu-
stance that might still lead to adverse bystander ef- nizations were halted as a result of a fraction of the
fects. patients developing aseptic meningoencephalopa-
thy, an inflammatory reaction in the CNS, which in
A fourth mechanism has been suggested to be active most instances responded well to steroid therapy.
at least for intracranially administered antibody. Subsequently, one case from the phase 1 trial also
That is neonatal Fc receptor (FcRn) medicated efflux developed these symptoms of CNS inflammation and
of antibody antigen complexes across the blood brain died from pulmonary embolism a short time later
barrier [38]. This mechanism was shown to be [42]. A second case from the phase 2a study was also

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63 57
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

collected [43]. In both cases, a T-cell infiltrate of the applied antibodies and detailed a rapid time course of
CNS was apparent with signs of meningeal inflamma- changes in amyloid deposits and microglial activa-
tion. The T cells did not appear directed to the amyloid tion. In a series of papers, we found that diffuse amy-
deposits, and there was no relationship between anti- loid was cleared within 1 day, whilst congophilic
body titre and incidence of the adverse event, but it deposits, associated with microglial activation, were
was concluded the response was an autoimmune cleared within 3 days. By 1 week, the regions infused
reaction caused he vaccine [44]. with antibody were largely cleared of deposits and free
of antibody [46, 47]. We further found that systemic
In spite of the interruption of the trial, some cases did administration of antibodies in aged amyloid deposit-
develop significant antibody titres. In the cohort eval- ing APP mice (18–22 months; analogous to 65- to 75-
uated in Zurich, these titres were measured using a years-old humans) caused a time-dependent clear-
brain amyloid histology assays (TAPIR) without ance of pre-existing deposits that after 3 months re-
breaking the blind [45]. Cognitive function tests in duced the congophilic compacted plaques by 90%
this cohort (35 overall) indicated that those patients compared to mice given a control antibody[29]. How-
with the highest titre declined less over the ensuing ever, a prior brief report had indicated that passive
year than those without detectable antibody titres. immunotherapy caused appearance of microhaem-
Some of these cases remained stable for years after orrhage in aged amyloid depositing mice [48]. When
the original treatments. Whilst admittedly anecdotal, we stained sections for haemosiderin, we found a
these observations implied that the immunotherapy time-dependent increase in the number of micro-
approach may have benefits in spite of the adverse haemorrhages associated with the antibody-medi-
events observed. ated removal of the parenchymal amyloid deposits
[49]. Moreover, we were the first to report an increase
A more complete follow-up of the patient cohort stud- in the number of vascular amyloid deposits and asso-
ied in the truncated phase 2a vaccination trial has ciation of the haemorrhages with these deposits.
been conducted at 4–5 years postimmunization. This However, remarkably, these aged mice that had held
study examined cognitive test performance of 25 ori- amyloid in their brain from over half their lifetimes
ginal cases that responded to the immunization and endured microhaemorrhages, still obtained the
(reaching a titre of 1 : 2200) and had 129 cases over- benefit of restored memory performance with the
all. A significant difference in Disability Assessment antibody treatment. Subsequently, others confirmed
for Dementia (an activities of daily living measure) that treatment with anti-Aß antibodies in older mice
and the Dependence Scale was found in the initial that already possess deposits when treatment is initi-
responders indicating a benefit of the treatment. ated (a therapeutic rather than prophylactic model)
Although no differences in the rate of brain shrinkage leads to microhaemorrhage development [50, 51].
by MRI or cognitive performance were observed, these
results are encouraging that some benefit could be After confirming our observation that Rinat’s anti-
obtained from a trial that was suspended before body caused microhaemorrhage, we cautiously ad-
reaching the intended duration of therapy. vised them of this significant concern. This led them
to modify the antibody to minimize interaction with
effector proteins such as Fc-gamma receptors and
Studies of passive immunotherapy in animals
complement proteins (by enzymatic deglycosylation).
The combination of low responsiveness to vaccines When we tested this antibody using intracranial
and the appearance of T-cell dependent adverse administration, it was capable of removing amyloid
events with active immunization quickly led multiple deposits without causing activation of microglia [52].
groups to evaluate monoclonal antibody treatments Further tests using systemic administration showed
to clear brain amyloid deposits. The first publication dramatically reduced formation of microhaemor-
of the success of this approach [28] was followed rhages and a smaller increase in vascular amyloid
shortly by others [32]. Our own work in this area was deposition with the modified antibody. Although
facilitated by a collaboration with a small biotechnol- there was a slight reduction in amyloid clearance,
ogy start-up in the San Francisco Bay area, Rinat both the modified and native antibodies rescued the
Neurosciences. Rinat shared with us antibodies they memory deficits in the aged amyloid depositing trans-
had developed against specific Aß epitopes which we genic mice [53].
then tested in our murine models of amyloid deposi-
tion (using research support from the NIA ⁄ NIH). Our One potential explanation for the increase in micro-
first studies examined the effects of intracranially haemorrhage is that antibodies capable of binding

58 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

amyloid fibrils in the vasculature can cause a local using monoclonal antibodies against the Aß peptide.
inflammatory reaction. Ultimately this may weaken By far the most advanced of these is an N terminal-
the endothelial cell barrier and result in vascular specific antibody from Elan-Wyeth referred to as
leakage. A second possibility is that the opsonization bapineuzimab (now in development by Wyeth-Pfizer
of parenchymal Aß deposits leads to macrophage and Janssen). The phase 1 trial of this antibody used
phagocytosis which then redistributes the phagocy- single administrations of 0.5, 1.5 or 5 mg kg)1 of the
tosed material to the vasculature. Fiala and col- antibody[55]. Patients were tested cognitively both
leagues used an in vitro blood brain barrier to demon- before administration and 16 weeks later. With re-
strate that macrophages with engulfed amyloid die spect to safety, 3 of 10 patients at the highest dose
when attempting to pass through the endothelial cell developed vasogenic oedema by MRI. One patient
barrier [54]. This leads to local deposition of the amy- developed a punctuate MRI hyperintensity that was
loid material they have phagocytosed. By reducing not present prior to the treatment and remained
the activation of effector proteins, the deglycosylated 12 months later. This was interpreted as a micro-
antibody used in our mouse work may diminish the haemorrhage. Although not designed to measure effi-
formation of vascular deposits and microhaemor- cacy, exploratory analyses found that the MMSE val-
rhages by reducing the extent of microglial ⁄ macro- ues improved in the 0.5 and 1.5 mg kg)1 doses, with
phage activation and phagocytosis. However, effects no benefit found in the highest dose. This is similar to
mediated by the peripheral sink and the catalytic our dose–response data mentioned above, with bene-
modification of Aß would be retained. Our results fits to APP mice at 3 and 10 mg kg)1 of antibody and
with Rinat’s modified antibody led to their acquisition no benefit at 30 mg kg)1 [39]. The causes for these
by Pfizer. A humanized version of this antibody is atypical dose responses are not clear, although satu-
presently in phase II clinical testing (ponezumab; ration of FcRn transport is one option. A similar inac-
Table 1). tivity at high doses was also reported previously in ab-
stract form [40].
Passive immunization in Alzheimer patients
A phase II clinical trial with bapineuzimab has also
Given the issues associated with active immuniza- been completed. The study included 230 patients
tion, several companies have started clinical trials and four doses of the drug (0.15, 0.5, 1 or 2 mg kg)1)

Table 1 Immunotherapeutic approaches in clinical development

Company Approach Abeta Epitope Biological Stage


Janssen ⁄ Wyeth (Elan) Passive N-terminus Bapineuzumab Phase III
Eli Lilly Passive Central domain Solanezumab Phase III
Baxter Passive IVIg – mix Gammaguard Phase III
Janssen ⁄ Wyeth (Elan) Active N-terminus ACC-001 Phase II
Novartis Active N-terminus CAD106 Phase II
Pfizer Passive C-terminus Ponezumab Phase II
GSK ⁄ Affiris Active Abeta mimetic Affitope AD1 and AD2 Phase II
Roche Passive N-terminus + central domain Gantunerumab R1450 Phase I
Merck Active Conformational V950 Phase I
GlaxoSmithKline Passive GSK933776A Phase I
Janssen ⁄ Wyeth (Elan) Passive N-terminus Bapineuzumab s.c. Phase I
Eisai ⁄ BioArctic Passive Protofibrils BAN2401 Phase I
Abbott Passive Conformational Preclinical
Elan ⁄ Wyeth Passive Conformational AAB-002 Preclinical
Genentech ⁄ ACImmune Passive Conformational Preclinical
BiogenIdec ⁄ Neurimmune Passive Preclinical
Boehringer ⁄ Ablynx Nanobodies Abeta Preclinical

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63 59
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

receiving six infusions 13 weeks apart [56]. Vasogen- phase, prior to overt clinical dementia, considerable
ic oedema was found in roughly 10% of the cases trea- neuron and synapse loss is evident [10].
ted with bapineuzimab, with half of these being
symptomatic. There was propensity for the vasogenic
The way forward
oedema to be more common in the higher dose groups
and in apolipoprotein E4 carriers. Intention to treat One of the more exciting developments of the last
analysis failed to identify a significant cognitive bene- 2 years in Alzheimer’s disease research is the identifi-
fit of bapineuzimab (all dose groups combined), but cation of methods to positively identify Alzheimer de-
there were significant benefits in patients that com- ments from other disorders without requiring au-
pleted all six antibody infusions and in patients that topsy. One method to achieve this is positron
were nonapolipoprotein E4. Thus, in spite of the ab- emission tomography (PET) using amyloid binding li-
sence of a dose–response effect, a phase III trial is gands. The first of these was a [11C]-labelled com-
presently underway, with a separate analysis of E4 pound referred to as Pittsburgh compound B [63].
carriers and noncarriers. It should be noted that prior Additional ligands are in various stages of develop-
studies also identified less efficacy of treatments in ment using [18F] labelling, a longer-lived isotope per-
apolipoprotein E4 carriers for tacrine [57] or rosigli- mitting a broader application of the methods (i.e. not
tizone [58]. requiring an adjacent cyclotron and radiochemistry
facility). An increasing number of publications dem-
onstrate a reasonable correlation of the PET ligand
Pathological analyses of vaccinated patients
signals and amyloid deposition in brain. Perhaps
Roughly 8 years have passed since the first patients most important, there now appear to be individuals
received anti-Aß vaccines. A number of these cases who are cognitively normal, but carry positive PET
have now come to autopsy. The Southampton group amyloid ligand signals. A recent report monitoring
led by James Nicoll have collected tissue from a num- these individuals has found a high predictability of
ber of patients (up to 12 at present) who were in the the PET signals in normals and subsequent conver-
phase I trial and have issued multiple reports on anal- sion to dementia [4]. In addition, there are now cere-
yses from these specimens [59–62]. Based upon the brospinal fluid analyses that appear to specify Alzhei-
time postvaccination, they describe the following se- mer cases, and these also appear in some individuals
quence of events in the small patient population they that do not yet have dementia. Together these raise
have. At relatively short intervals (<1 year), they ob- the possibility of defining, with some precision, a set
serve the presence of ragged ‘‘moth-eaten’’ amyloid of individuals with dramatically increased risk of
deposits and the presence of microglia with ingested dementia. At the recent ICAD meeting in Honolulu, it
immunopositive amyloid. Between 1 and 3 years po- was suggested these signals may precede disease on-
stimmunization, they report a partial clearance of set by up to a decade [5].
amyloid deposits (compared to age-matched Alzhei-
mer samples that were not part of the study), and the This raises the possibility of attempting to prevent the
appearance of increased numbers of vascular amy- development of Alzheimer’s dementia. Screening
loid deposits and increased microhaemorrhage. By high-risk populations for these amyloid signatures
4–6 years of postimmunization, they identified two using PET scans and spinal fluid analyses may iden-
cases that have a virtually complete removal of amy- tify those with amyloid deposits before substantial
loid deposits, including vascular deposits, and no evi- neural damage has occurred. Treatments could then
dence of residual microhaemorrhage. Nonetheless, be attempted which reduce the amyloid in anticipa-
there was no resolution of tau pathology. Perhaps tion of slowing the onset of the disorder or preventing
most disturbing was that no cognitive benefits of the the disease altogether. In this context, a subset of the
amyloid clearance could be detected in these end patients in the phase 3 bapineuzimab trial were
stage cases [59]. This implies that (i) amyloid has no screened for amyloid deposits using PET scans before
direct impact on cognitive status in humans, (ii) these and after the 18 months treatment with the
patients had some other form of dementia to begin agent[64]. The antibody treatment significantly re-
with or (iii) that the damage is so extensive by the time duced the intensity of the amyloid signals over the
that cognitive symptoms are apparent and that res- 18 months of the trials compared to patients receiv-
cue is not feasible. In any event, if these two cases are ing placebo therapy. Even if the issue of vasogenic oe-
representative, this does not bode well for any anti- dema complicates the use of bapineuzimab, there are
amyloid approach in patients that already have the a large number of other immunotherapy approaches
disease. It should be noted that even in the MCI also in clinical or preclinical development (see

60 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

Table 1). We remain hopeful that one or more of these plaques and neurofibrillary tangles ‘‘do count’’ when staging dis-
will succeed in reducing amyloid presymptomatically ease severity. J Neuropathol Exp Neurol 2007; 66: 1136–46.
10 Morris JC, Price AL. Pathologic correlates of nondemented aging,
and that this will be sufficient to avert the onset of the
mild cognitive impairment, and early-stage Alzheimer’s disease.
cognitive problems associated with Alzheimer’s J Mol Neurosci 2001; 17: 101–18.
dementia, permitting us to prevent Alzheimer’s dis- 11 Glenner GG, Wong CW. Alzheimer’s disease: initial report of the
ease from occurring. purification and characterization of a novel cerebrovascular
amyloid protein. Biochem Biophys Res Commun 1984; 120: 885–
90.
Acknowledgements 12 Goate A, Chartier-Harlin MC, Mullan M et al. Segregation of a
missense mutation in the amyloid precursor protein gene with fa-
This work was supported by NIH grants AG-04418, milial Alzheimer’s disease. Nature 1991; 349: 704–6.
AG 15490 and AG18478. We thank Karen Ashe and 13 Greenberg BD, Savage MJ, Howland DS et al. APP transgenesis:
Karen Duff for early access to their transgenic mouse approaches toward the development of animal models for Alzhei-
models of amyloid deposition. mer disease neuropathology. Neurobiol Aging 1996; 17: 153–71.
14 Games D, Adams D, Alessandrini R et al. Alzheimer-type neuro-
pathology in transgenic mice overexpressing V717F beta-amy-
Conflict of interest statement loid precursor protein. Nature 1995; 373: 523–7.
15 Hsiao K, Chapman P, Nilsen S et al. Correlative memory deficits,
In the past decade, Dr Morgan has consulted with the Abeta elevation, and amyloid plaques in transgenic mice. Science
following pharmaceutical companies on issues re- 1996; 274: 99–102.
lated to Alzheimer’s Immunotherapy. Pfizer, Wyeth, 16 Gordon MN, Holcomb LA, Jantzen PT et al. Time course of the de-
Rinat Neuroscience, Merck, Astra-Zeneca, Bristol- velopment of Alzheimer-like pathology in the doubly transgenic
Myers-Squibb, Eisai, Forest, Lundbeck, Neurim- PS1+APP mouse. Exp Neurol 2002; 173: 183–95.
17 Holcomb L, Gordon MN, McGowan E et al. Accelerated Alzhei-
mune, Elan and Baxter. Dr Morgan does not have
mer-type phenotype in transgenic mice carrying both mutant
current research support from any of the above-men- amyloid precursor protein and presenilin 1 transgenes. Nat Med
tioned companies. 1998; 4: 97–100.
18 Gordon MN, King DL, Diamond DM et al. Correlation between
cognitive deficits and Aá deposits in transgenic APP+PS1 mice.
References Neurobiol Aging 2001; 22: 377–85.
1 DeKosky ST, Harbaugh RE, Schmitt FA et al. Cortical biopsy in 19 Morgan D, Diamond DM, Gottschall PE et al. A beta peptide vac-
Alzheimer’s disease: diagnostic accuracy and neurochemical, cination prevents memory loss in an animal model of Alzheimer’s
neuropathological, and cognitive correlations. Intraventricular disease. Nature 2000; 408: 982–5.
Bethanecol Study Group. Ann Neurol 1992; 32: 625–32. 20 Janus C, Pearson J, McLaurin J et al. A beta peptide immuniza-
2 Tarawneh R, Holtzman DM. Critical issues for successful immu- tion reduces behavioural impairment and plaques in a model of
notherapy in Alzheimer’s disease: development of biomarkers Alzheimer’s disease. Nature 2000; 408: 979–82.
and methods for early detection and intervention. CNS Neurol 21 Kotilinek LA, Bacskai B, Westerman M et al. Reversible memory
Disord Drug Targets 2009; 8: 144–59. loss in a mouse transgenic model of Alzheimer’s disease. J Neu-
3 Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s dis- rosci 2002; 22: 6331–5.
ease: progress and problems on the road to therapeutics. Science 22 Dodart JC, Bales KR, Gannon KS et al. Immunization reverses
2002; 297: 353–6. memory deficits without reducing brain Abeta burden in Alzhei-
4 Morris JC, Roe CM, Grant EA et al. Pittsburgh compound B mer’s disease model. Nat Neurosci 2002; 5: 452–7.
imaging and prediction of progression from cognitive normality 23 Lemere CA, Oh J, Stanish HA et al. Cerebral amyloid-beta protein
to symptomatic Alzheimer disease. Arch Neurol 2009; 66: 1469– accumulation with aging in cotton-top tamarins: a model of early
75. Alzheimer’s disease? Rejuvenation Res 2008; 11: 321–32.
5 Mintun MA, Vlassenko A, Sheline YI, Morris JC. Longitudinal PIB 24 Lemere CA, Beierschmitt A, Iglesias M et al. Alzheimer’s disease
PET imaging of the appearance and accumulation of beta-amy- abeta vaccine reduces central nervous system abeta levels in a
loid in cognitively normal middle and late life adults. Alzheimers non-human primate, the Caribbean vervet. Am J Pathol 2004;
Dement 2010; 6: S2. 165: 283–97.
6 Gotz J, Chen F, Van Dorpe J, Nitsch RM. Formation of neurofi- 25 Head E, Pop V, Vasilevko V et al. A two-year study with fibrillar
brillary tangles in P301l tau transgenic mice induced by Abeta 42 beta-amyloid (Abeta) immunization in aged canines: effects on
fibrils. Science 2001; 293: 1491–5. cognitive function and brain Abeta. J Neurosci 2008; 28: 3555–
7 Lewis J, Dickson DW, Lin WL et al. Enhanced neurofibrillary 66.
degeneration in transgenic mice expressing mutant tau and APP. 26 Tabira T. Immunization therapy for Alzheimer disease: a compre-
Science 2001; 293: 1487–91. hensive review of active immunization strategies. Tohoku J Exp
8 Oddo S, Billings L, Kesslak JP, Cribbs DH, LaFerla FM. Abeta Med 2010; 220: 95–106.
immunotherapy leads to clearance of early, but not late, hyper- 27 Pan W, Solomon B, Maness LM, Kastin AJ. Antibodies to beta-
phosphorylated tau aggregates via the proteasome. Neuron Amyloid Decrease the Blood-to-Brain Transfer of beta- Amyloid
2004; 43: 321–32. Peptide. Exp Biol Med (Maywood) 2002; 227: 609–15.
9 Nelson PT, Jicha GA, Schmitt FA et al. Clinicopathologic correla- 28 Bard F, Cannon C, Barbour R et al. Peripherally administered
tions in a large Alzheimer disease center autopsy cohort: neuritic antibodies against amyloid beta-peptide enter the central

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63 61
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

nervous system and reduce pathology in a mouse model of Alz- 46 Wilcock DM, Munireddy SK, Rosenthal A, Ugen KE, Gordon MN,
heimer disease. Nat Med 2000; 6: 916–9. Morgan D. Microglial activation facilitates Abeta plaque removal
29 Wilcock DM, Rojiani A, Rosenthal A et al. Passive amyloid immu- following intracranial anti-Abeta antibody administration. Neuro
notherapy clears amyloid and transiently activates microglia in a biol Dis 2004; 15: 11–20.
transgenic mouse model of amyloid deposition. J Neurosci 2004; 47 Wilcock DM, DiCarlo G, Henderson D et al. Intracranially admi-
24: 6144–51. nistered anti-Abeta antibodies reduce beta-amyloid deposition
30 Levites Y, Das P, Price RW et al. Anti-Abeta42- and anti-Abeta40- by mechanisms both independent of and associated with micro-
specific mAbs attenuate amyloid deposition in an Alzheimer dis- glial activation. J Neurosci 2003; 23: 3745–51.
ease mouse model. J Clin Invest 2006; 116: 193–201. 48 Pfeifer M, Boncristiano S, Bondolfi L et al. Cerebral hemorrhage
31 Karlnoski RA, Rosenthal A, Kobayashi D et al. Suppression of after passive anti-Abeta immunotherapy. Science 2002; 298:
amyloid deposition leads to long-term reductions in Alzheimer’s 1379.
pathologies in Tg2576 mice. J Neurosci 2009; 29: 4964–71. 49 Wilcock DM, Rojiani A, Rosenthal A et al. Passive immunother-
32 DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, apy against Abeta in aged APP-transgenic mice reverses cognitive
Holtzman DM. Peripheral anti-A beta antibody alters CNS and deficits and depletes parenchymal amyloid deposits in spite of in-
plasma A beta clearance and decreases brain A beta burden in a creased vascular amyloid and microhemorrhage. J Neuro inflam-
mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A mation 2004; 1: 24.
2001; 98: 8850–5. 50 Racke MM, Boone LI, Hepburn DL et al. Exacerbation of cerebral
33 Deane R, Du YS, Submamaryan RK et al. RAGE mediates amy- amyloid angiopathy-associated microhemorrhage in amyloid
loid-beta peptide transport across the blood-brain barrier and precursor protein transgenic mice by immunotherapy is depen-
accumulation in brain. Nat Med 2003; 9: 907–13. dent on antibody recognition of deposited forms of amyloid beta.
34 Levites Y, Smithson LA, Price RW et al. Insights into the mechan- J Neurosci 2005; 25: 629–36.
isms of action of anti-Abeta antibodies in Alzheimer’s disease 51 Schroeter S, Khan K, Barbour R et al. Immunotherapy reduces
mouse models. FASEB J 2006; 20: 2576–8. vascular amyloid-beta in PDAPP mice. J Neurosci 2008; 28:
35 Solomon B, Koppel R, Frankel D, Hanan-Aharon E. Disaggrega- 6787–93.
tion of Alzheimer beta-amyloid by site-directed mAb. Proc Natl 52 Carty NC, Wilcock DM, Rosenthal A et al. Intracranial adminis-
Acad Sci U S A 1997; 94: 4109–12. tration of deglycosylated C-terminal-specific anti-Abeta antibody
36 Solomon B, Koppel R, Hanan E, Katzav T. Monoclonal antibodies efficiently clears amyloid plaques without activating microglia in
inhibit in vitro fibrillar aggregation of the Alzheimer beta-amyloid amyloid-depositing transgenic mice. J Neuroinflammation 2006;
peptide. Proc Natl Acad Sci U S A 1996; 93: 452–5. 3: 11.
37 Alcantar NA, Jimenez J, Morgan D. Direct observation of the ki- 53 Wilcock DM, Alamed J, Gottschall PE et al. Deglycosylated anti-
netic mechanisms for Aß peptide aggregation: Towards elucidat- amyloid-beta antibodies eliminate cognitive deficits and reduce
ing Alzheimer plaque dissolution. Alzheimers Dement 2010; 6: parenchymal amyloid with minimal vascular consequences in
S247. aged amyloid precursor protein transgenic mice. J Neurosci
38 Deane R, Sagare A, Hamm K et al. IgG-assisted age-dependent 2006; 26: 5340–6.
clearance of Alzheimer’s amyloid beta peptide by the blood-brain 54 Zaghi J, Goldenson B, Inayathullah M et al. Alzheimer disease
barrier neonatal Fc receptor. J Neurosci 2005; 25: 11495–503. macrophages shuttle amyloid-beta from neurons to vessels, con-
39 Karlnoski RA, Rosenthal A, Alamed J et al. Deglycosylated anti- tributing to amyloid angiopathy. Acta Neuropathol 2009; 117:
Abeta antibody dose-response effects on pathology and memory 111–24.
in APP transgenic mice. J Neuroimmune Pharmacol 2008; 3: 187– 55 Black RS, Sperling RA, Safirstein B et al. A single asce-
97. nding dose study of bapineuzumab in patients with
40 Gitter BD, Gannon KS, Cummins DJ et al. Reduction in brain Alzheimer disease. Alzheimer Dis Assoc Disord 2010; 24: 198–
amyloid b burden and reversal of memory impairment in APP 203.
V717F transgenic mice following chronic administration of the 56 Salloway S, Sperling R, Gilman S et al. A phase 2 multiple ascend-
anti-amyloid b antibody m266.2. Neurobiol Aging 2002; 23: ing dose trial of bapineuzumab in mild to moderate Alzheimer
s105. disease. Neurology 2009; 73: 2061–70.
41 Schenk D. Amyloid-beta immunotherapy for Alzheimer’s 57 Farlow MR, Lahiri DK, Poirier J, Davignon J, Schneider L, Hui SL.
disease: the end of the beginning. Nat Rev Neurosci 2002; 3: Treatment outcome of tacrine therapy depends on apolipoprotein
824–8. genotype and gender of the subjects with Alzheimer’s disease.
42 Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller Neurology 1998; 50: 669–77.
RO. Neuropathology of human Alzheimer disease after immuni- 58 Risner ME, Saunders AM, Altman JF et al. Efficacy of
zation with amyloid-beta peptide: a case report. Nat Med 2003; 9: rosiglitazone in a genetically defined population with mild-to-
448–52. moderate Alzheimer’s disease. Pharmacogenomics J 2006; 6:
43 Ferrer I, Boada RM, Sanchez Guerra ML, Rey MJ, Costa-Jussa F. 246–54.
Neuropathology and pathogenesis of encephalitis following amy- 59 Holmes C, Boche D, Wilkinson D et al. Long-term effects of
loid-beta immunization in Alzheimer’s disease. Brain Pathol Abeta42 immunisation in Alzheimer’s disease: follow-up of a ran-
2004; 14: 11–20. domised, placebo-controlled phase I trial. Lancet 2008; 372:
44 Orgogozo JM, Gilman S, Dartigues JF et al. Subacute menin- 216–23.
goencephalitis in a subset of patients with AD after Abeta42 im- 60 Boche D, Nicoll JA. The role of the immune system in clearance of
munization. Neurology 2003; 61: 46–54. Abeta from the brain. Brain Pathol 2008; 18: 267–78.
45 Hock C, Konietzko U, Papassotiropoulos A et al. Generation of 61 Boche D, Zotova E, Weller RO et al. Consequence of A{beta}
antibodies specific for beta-amyloid by vaccination of patients immunization on the vasculature of human Alzheimer’s disease
with Alzheimer disease. Nat Med 2002; 8: 1270–5. brain. Brain 2008; 131: 3299–310.

62 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63
D. Morgan
| Key Symposium: Immunotherapy for Alzheimer’s disease

62 Nicoll JA, Barton E, Boche D et al. Abeta species removal after blind, placebo-controlled, ascending-dose study. Lancet Neurol
abeta42 immunization. J Neuropathol Exp Neurol 2006; 65: 2010; 9: 363–72.
1040–8.
63 Klunk WE, Engler H, Nordberg A et al. Imaging brain amyloid in Correspondence: Dave Morgan, Byrd Alzheimer Institute, 4001 E
Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol Fletcher Ave, MDC 36, University of South Florida, Tampa, FL
2004; 55: 306–19. 33613, USA.
64 Rinne JO, Brooks DJ, Rossor MN et al. 11C-PiB PET assessment (fax: 1 813 396 1601; e-mail: scientist.dave@gmail.com).
of change in fibrillar amyloid-beta load in patients with Alzhei-
mer’s disease treated with bapineuzumab: a phase 2, double-

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 269; 54–63 63

Potrebbero piacerti anche