ArticlePDF Available

Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis

Authors:

Abstract and Figures

Alemtuzumab is a lymphocyte depleting monoclonal antibody that has demonstrated superior efficacy over interferon β-1a for relapsing-remitting multiple sclerosis (MS), and is currently under investigation in phase 3 trials. One unresolved issue is the duration and significance of the lymphopenia induced. The long term effects on lymphocyte reconstitution of a single course, and the consequences that this has on disability, morbidity, mortality and autoimmunity, were examined. The lymphocyte reconstitution (n=36; 384 person years) and crude safety data (n=37; 447 person years) are reported for the first patients with progressive MS to receive alemtuzumab (1991-1997). Reconstitution time was expressed as a geometric mean or, when a non-negligible number of individuals failed to recover, as a median using survival analysis. Geometric mean recovery time (GMRT) of total lymphocyte counts to the lower limit of the normal range (LLN; ≥1.0×10(9) cells/l) was 12.7 months (95% CI 8.8 to 18.2 months). For B cells, GMRT to LLN (≥0.1×10(9)/l) was 7.1 months (95% CI 5.3 to 9.5); median recovery times for CD8 (LLN ≥0.2×10(9) cells/l) and CD4 lymphocytes (LLN ≥0.4×10(9) cells/l) were 20 months and 35 months, respectively. However, CD8 and CD4 counts recovered to baseline levels in only 30% and 21% of patients, respectively. No infective safety concerns arose during 447 person years of follow-up. Lymphocyte counts recovered to LLN after a single course of alemtuzumab in approximately 8 months (B cells) and 3 years (T cell subsets), but usually did not recover to baseline values. However, this long lasting lymphopenia in patients with a previously normal immune system was not associated with an increased risk of serious opportunistic infection.
Content may be subject to copyright.
RESEARCH PAPER
Long term lymphocyte reconstitution after
alemtuzumab treatment of multiple sclerosis
Grant A Hill-Cawthorne,
1,2
Tom Button,
1
Orla Tuohy,
1
Joanne L Jones,
1
Karen May,
1
Jennifer Somerfield,
1,3
Alison Green,
4
Gavin Giovannoni,
5,6
D Alastair S Compston,
1
Michael T Fahey,
7,8
Alasdair J Coles
1
ABSTRACT
Background Alemtuzumab is a lymphocyte depleting
monoclonal antibody that has demonstrated superior
efficacy over interferon
b
-1a for relapsingeremitting
multiple sclerosis (MS), and is currently under
investigation in phase 3 trials. One unresolved issue is
the duration and significance of the lymphopenia
induced. The long term effects on lymphocyte
reconstitution of a single course, and the consequences
that this has on disability, morbidity, mortality and
autoimmunity, were examined.
Methods The lymphocyte reconstitution (n¼36; 384
person years) and crude safety data (n¼37; 447 person
years) are reported for the first patients with progressive
MS to receive alemtuzumab (1991e1997).
Reconstitution time was expressed as a geometric mean
or, when a non-negligible number of individuals failed to
recover, as a median using survival analysis.
Results Geometric mean recovery time (GMRT) of total
lymphocyte counts to the lower limit of the normal range
(LLN; $1.0310
9
cells/l) was 12.7 months (95% CI 8.8
to 18.2 months). For B cells, GMRT to LLN ($0.1310
9
/l)
was 7.1 months (95% CI 5.3 to 9.5); median recovery
times for CD8 (LLN $0.2310
9
cells/l) and CD4
lymphocytes (LLN $0.4310
9
cells/l) were 20 months
and 35 months, respectively. However, CD8 and CD4
counts recovered to baseline levels in only 30% and 21%
of patients, respectively. No infective safety concerns
arose during 447 person years of follow-up.
Conclusions Lymphocyte counts recovered to LLN after
a single course of alemtuzumab in approximately
8 months (B cells) and 3 years (T cell subsets), but
usually did not recover to baseline values. However, this
long lasting lymphopenia in patients with a previously
normal immune system was not associated with an
increased risk of serious opportunistic infection.
INTRODUCTION
Campath-1H is a humanised monoclonal antibody
that binds CD52 and depletes lymphocytes,
monocytes and NK cells.
1
Marketed as alemtu-
zumab, now Lemtrada, it was approved for the
treatment of chronic lymphocytic leukaemia in
2001.
2
Since 1991, we have investigated its use as
a treatment for multiple sclerosis (MS): a phase
2 trial has been published
3
and phase 3 trials are
ongoing.
45
However, despite the potential for its
widespread use in young systemically healthy
adults with MS, the extent and clinical signicance
of the lymphopenia that alemtuzumab induces is
not well known. Experience from alemtuzumab
treatment of other conditions is not representative,
as in lymphocytic malignancies there is abnormal
lymphocyte proliferation, and in treatment resis-
tant systemic autoimmune disease, patients are
older, unwell
6
and have been exposed to multiple
immunotherapies.
7
The aim of this study was to describe the long
term safety effects of a single course of alemtu-
zumab in treatment naïve people with MS. We
report data from the rst 37 patients, each with
progressive MS, treated between 1991 and 1997.
While alemtuzumab successfully reduced the
relapse, we have previously reported that patients
continued to experience progressive disability.
8
Previously, assuming linear kinetics of reconstitu-
tion after alemtuzumab, data from this cohort led
to estimates of median recovery time to baseline
levels for CD4 and CD8 T cells of 61 and
30 months, respectively, with B cells reaching
baseline levels and overshootingmore rapidly.
9
Linear reconstitution is a reasonable model for the
rst 12e18 months but we now show that after
18 months the rate decelerates to a point that
linear kinetics becomes an inappropriate model.
We now re-address the extent of lymphocyte
recovery in this cohort after a longer interval using
analysis techniques that do not assume linear
reconstitution.
We also report the long term safety prole of this
small cohort. We previously reported low rates of
infections in the rst few years after alemtuzumab,
strikingly lower than patients with HIV infection
with similar CD4 counts
10
; one potential explana-
tion being that, after alemtuzumab, lymph nodes
retain a substantial number of healthy lympho-
cytes that escape deletion.
11e13
We report the effect
of alemtuzumab on CSF oligoclonal bands (OCB)
in the context of progressive MS. Finally, we
explore the relationship between lymphocyte
reconstitution and the development of secondary
autoimmune diseases: up to 30% of alemtuzumab
treated MS patients develop autoimmune thyroid
disease,
13
and other autoimmune diseases such as
Goodpastures disease are also seen.
9
METHODS
This is a review of safety and lymphocyte recon-
stitution of the rst 37 patients to receive alem-
tuzumab as a treatment for MS, in Cambridge,
UK.
1
Department of Neurology,
University of Cambridge,
Cambridge, UK
2
Pathogen Genomics
Laboratory, King Abdullah
University of Science and
Technology (KAUST), Thuwal,
Mekkah, Saudi Arabia
3
Department of Medicine,
University of Auckland,
Auckland, New Zealand
4
The National CJD Surveillance
Unit, University of Edinburgh,
Edinburgh, UK
5
Institute of Neurology,
University College London,
London, UK
6
Centre for Neuroscience and
Trauma, Blizard Institute, Barts
and The London School of
Medicine and Dentistry, Queen
Mary University of London,
London, UK
7
Centre for Applied Medical
Statistics, Department of Public
Health and Primary Care,
University of Cambridge,
Cambridge, UK
8
Division of Mathematics,
Informatics and Statistics,
Commonwealth Scientific and
Industrial Research Organisation
(CSIRO), Clayton South, Victoria,
Australia
Correspondence to
Dr A J Coles, Department of
Neurology, Box 165,
Addenbrooke’s Hospital,
Cambridge CB2 2QQ, UK;
ajc1020@medschl.cam.ac.uk
Received 24 June 2011
Revised 22 September 2011
Accepted 23 September 2011
Published Online First
5 November 2011
298 J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
Patients and treatment
The rst cohort of patients, treated between 1991 and 1993,
consisted of seven patients, six with secondary progressive and
one with primary progressive disease.
14
Six patients were treated
with 12 mg of alemtuzumab daily for 10 days and one patient
received 60 mg in total. Early
14
and later
89
data on efcacy have
been reported previously. Five of the seven patients were re-
treated between 2 and 4 years after the rst dose. The second
cohort, treated between 1995 and 1997, consisted of 29 patients,
all with secondary progressive MS.
8
All patients received 100 mg
of alemtuzumab over 5 days. Fourteen of the 29 patients were
also treated with a novel humanised IgG4 anti-CD4 antibody
(200 mg over the subsequent 5 days), which was designed,
successfully, to be non-depleting. For the purposes of analysis,
both cohorts are considered together. No antimicrobial prophy-
laxis was administered to either cohort and no other immuno-
suppressant medications were taken during follow-up. One
additional patient with progressive MS, treated in 1997, is also
included in the analysis of reconstitution. This study was
approved by the local research ethics committees and all patients
gave written informed consent. GAHC, TB and MTF analysed
the data and all authors had access to the primary clinical data.
Assessments
All patients were offered continued follow-up at our site,
3 monthly for the rst 3 years after alemtuzumab, then bian-
nually for 2 years, and annually thereafter. At each of these
visits, blood was taken for total lymphocyte count, subsets
(CD4, CD8, CD19), autoantibody screen, liver function and
renal function. Blood tests were variably available between 1990
and 2009 depending on the patients treatment date and
availability for follow-up. Disability was assessed annually using
Kurtzkes Expanded Disability Status Score (EDSS).
15
Eleven
patients, who lived at a distance, declined these assessments.
All living patients were reviewed within the past 2 years or
received a telephone interview. As a minimum, data on auto-
immune disease, major illnesses or death were collected, as well
as a crude estimate of disability. Autoimmunity was dened as
the clinical development of secondary autoimmune disease or
persistently abnormal thyroid function tests in the presence of
autoantibodies indicating thyroid disease, as in our previous
studies.
16
Assessment of recovery
Total lymphocyte counts and subset analyses were carried out
whenever patients attended the clinic. Analysis was restricted to
measurements taken at least 1 day after completing the rst
cycle of alemtuzumab and not confounded by any subsequent
treatment. That is, all data recorded after subsequent treatments
were excluded in this analysis. The number of repeated
measurements per patient ranged from 5 to 46 (median 18, IQR
12e23). Four components of immune reconstitution were
examined: total lymphocyte counts, CD19, CD4 and CD8
lymphocyte subsets.
Total and T lymphocyte cell counts were analysed to two
endpoints. The rst endpoint was time taken for recovery to
a predened lower limit of normallevel (LLN). These LLNs
were derived from our laboratory ranges and were 1.0310
9
,
0.4310
9
and 0.2310
9
cells/l for total, CD4 and CD8 lympho-
cytes, respectively. Throughout this manuscript we shall refer to
this endpoint as reconstitution to normal. The second endpoint
was time to recovery to the patients relevant lymphocyte count
before alemtuzumab treatment. This second endpoint will be
referred to as reconstitution to baseline level.
B lymphocytes were only analysed to the rst
endpointdnamely, reconstitution to normaldue to a lack of
baseline measurements. For B lymphocytes, LLN was
0.1310
9
cells/l.
Fifteen patients from the second cohort consented to lumbar
puncture before and after alemtuzumab (table 1). Median
disease duration was 4 years (range 1e13). Unconcentrated CSF
and paired serum samples were assessed using isoelectric
focusing in agarose gel with immunoxation. The blots were
read independently by two experienced observers blinded with
respect to the identity of each patient and the time-point of the
sample.
Statistical analysis
Median and IQR were used to describe data. To make statistical
inference on recovery time for reconstitution, the geometric
mean or median was used. When the number of individuals who
failed to recover was negligible, recovery time was log trans-
formed to improve symmetry and the geometric mean
computed. When there was a non-negligible number of indi-
viduals failing to recover, their recovery times were treated as
censored and a median was estimated using survival analysis.
The relationship with age at baseline and autoimmune status
was examined using a scatterplot for the former and a box and
whisker plot for the latter. To examine the effect of autoim-
munity, a t test of zero mean difference in log recovery time was
calculated. This result was back transformed to the original time
scale and reported as the ratio of the geometric means for
autoimmunity absent divided by autoimmunity present along
with its 95% CI and p value. Median recovery times to baseline
level (as well as to normal level for CD4 and CD8) were esti-
mated using survival analysis to allow inclusion of patients who
did not recover to their baseline level in the analysis. These
results were presented in cumulative incidence (KaplaneMeier)
plots of recovery to baseline level against time since rst treat-
ment, and summarised by percentiles of the recovery time
distribution.
RESULTS
Patients
Thirty-seven patients (17 men and 20 women) were followed
altogether. All patients had primary or secondary progressive
MS, with a median age of 39 years (23e56) at the time of rst
alemtuzumab treatment and median duration since disease
onset of 9 years (range 1e23). Median disability at treatment
was EDSS 6 (4e8). Twenty-eight of 37 patients received a single
cycle of alemtuzumab; 9/37 patients received an additional
cycle, a median of 3.5 years after the rst exposure. Baseline
lymphocyte subset levels were not available in 3/37 patients;
follow-up lymphocyte counts were available for a median of
Table 1 Details of the 15 patients with secondary progressive
multiple sclerosis that consented to lumbar punctures before and
after alemtuzumab treatment
No of patients 15
No of females (%) 10 (67)
Age at time of treatment (years) (median (range)) 40 (23e49)
Disease duration (years) (median (range)) 4 (1e13)
EDSS pretreatment (median (range)) 6 (4e7)
Time of pretreatment LP (days) (median (range)) 1 (0e37)
Time of post-treatment LP (months) (median (range)) 11 (3e28)
EDSS, Expanded Disability Status Score; LP, lumbar puncture.
J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826 299
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
12 years (0.5e16) and clinical information (including by tele-
phone contact) for a median of 14 years (2e18). No patients
were lost to follow-up. The total duration of follow-up was 384
person years for lymphocyte counts and 447 person years for
clinical data. Twelve of 37 patients developed secondary auto-
immune disease.
Lymphocyte reconstitution
Figure 1 illustrates the total, CD19, CD4 and CD8 lymphocyte
counts from our cohort for a maximum of 16 years after a single
cycle of alemtuzumab. Total, CD4 and CD8 lymphocyte counts
all increased at a high rate of reconstitution for the rst 12e18
months before the rate decreased. The median baseline
lymphocyte counts (with IQR) and upper and lower normal
limits (ULN, LLN, all 310
9
cells/l) were: total lymphocyte count
1.8 (IQR 1.4e2.3) (LLN 1.0, ULN 3.5); CD4 0.8 (0.71e1.01) (0.4,
1.5); CD8 0.42 (0.35e0.45) (0.2, 0.9); and CD19 0.19 (0.17e0.25)
(0.1, 0.5).
Total lymphocyte counts reconstituted to a normal level
(dened as $1.0310
9
cells/l) in 34/36 patients over a median of
12 years; the two patients not recovering to normal are excluded
from this analysis. The distribution of time to recovery was
strongly skewed to the right, with most patientstotal
lymphocyte counts recovering within 2 years. Median recovery
time was 12.6 months (IQR 6.1e29.8 months). The log trans-
formed distribution of time taken to recovery was approxi-
mately symmetric (gure 2A) with the geometric mean closely
approximating the median. The geometric mean of recovery to
the normal level was 12.7 months (95% CI 8.8 to 18.2 months).
The total lymphocyte count recovery time to normal showed
little association with baseline age (r¼0.11) and a moderate
(negative) association with baseline total lymphocyte count
(r¼0.29). Total lymphocyte recovery tended to be 1.8 times
slower (95% CI 0.92 to 3.6, p¼0.08) for those patients who did
not develop autoimmunity (15.4 months) compared with those
who did (8.5 months, gure 2B).
Recovery of total lymphocyte counts to individual baseline
levels was achieved in only 14/36 patients (39%). To avoid
excluding these individuals, median recovery time was esti-
mated using survival analysis, yielding a gure of 151 months
(gure 2C; 95% CI 91 to 212). The 25th percentile of recovery
time was 38 months and more reliably estimated than the
median, as indicated by the condence bands around the curve
at this point.
Thirty-four of 36 patients recovered B cell lymphocyte
counts to a normal level ($0.1310
9
/l), mostly within 2 years
(gure 3A). Median recovery time was 8.4 months. After log
transformation, the distribution showed greater symmetry than
on the original scale, but with a visual indication of bimodality
(gure 3B). The geometric mean was 7.1 months (95% CI 5.3 to
9.5). There was no association between autoimmune status and
CD19 recovery to normallevels.
In 31/36 patients (86%), CD8 lymphocyte counts recovered to
a normal level ($0.2310
9
/l); 27 patients in the rst 4 years and
four in the next 8 years of follow-up (gure 4A). Median
recovery time was 20 months (95% CI 5 to 36). The 25th
percentile of recovery time was 6 months. No relationship was
found between mean CD8 recovery time to normal and auto-
immune status. In only 10/33 patients did CD8 lymphocyte
counts recover to baseline levels (gure 4B); median recovery
time was 155 months while the 25th percentile of recovery time
was 66 months.
Reconstitution of CD4 lymphocytes was slower; only 28/36
patients recovered to a normal CD4 level ($0.4310
9
/l) over
Figure 1 Raw data plots of total
lymphocyte counts (A), and CD19 (B),
CD4 (C) and CD8 subgroups (D) (all
310
9
cells/l) for years after
alemtuzumab. Broken lines indicate
upper and lower limits of normal range.
The solid line indicates the median (and
the grey shaded area the upper and
lower quartiles) of the baseline values.
300 J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
a median of 12 years (gure 4C). In the cumulative incidence
plot, the 25th percentile of recovery was more reliably estimated
than the median, being 27 months and 35 months, respectively.
Geometric mean recovery time to normal varied little by auto-
immunity and the differences were not signicant. CD4 counts
recovered to baseline in only 7/33 patients (three patients having
missing baseline measurements). The median cannot be esti-
mated but the 25th percentile of recovery time was 112 months
(gure 4D).
Disability
Disability in all but one of the 37 patients continued to worsen
progressively despite alemtuzumab treatment, as previously
reported.
8 9
At the last recorded follow-up, a median of 14 years
post-treatment, the median disability estimated in 35/37
patients was EDSS 7.5 (range 4.5e9). Relapses were uncommon
but were not systematically captured.
Morbidity and mortality
Information on infections was not systematically collected from
the patients and many minor infections, common in the normal
population, will neither have been reported by patients nor have
generated a hospital record. From the information available there
were 11 major infections among the 37 patients, with two
occurring in the same patient. Most of these were pneumonia at
advanced stages of disability, with all ve cases contributing to
death (one of which was noted to be secondary to aspiration).
Three cases of urinary sepsis were classied as severe because
they were notied as the cause of death. Deaths rates in the two
original cohorts of patients were 29% for the rst cohort and
33% for the second cohort. Three further episodes of infection
were caused by necrotising gingivitis (previously reported),
9
a cervical epidural abscess and septicaemia secondary to a breast
abscess. Five patients reported segmental varicella zoster
virus reactivations, the majority occurring within 2e3 years of
treatment, with one reactivation 8 years after treatment. Again,
major infection rates were similar in the rst and second cohorts
(29% and 30%, respectively). For patients undergoing surgical
procedures, including two cholecystectomies, a hernia repair and
a hemiarthroplasty, there was no excess post-surgical morbidity.
Fourteen patients developed Gravess disease after treatment
with alemtuzumab (37.8%). Autoimmune disease in these two
cohorts appeared to be associated with female sex; 12 out of 21
women (57%) versus two out of 15 men (13%). However, this
has not been a consistent nding in the larger clinical trial
cohorts.
3 9
Two malignant tumours were recorded in the 37 patients at
follow-up: a case of prostatic adenocarcinoma in a patient who
was 55 years old at the time of treatment and one example of
skin basal cell carcinoma. One benign tumour was recorded: an
incidental meningioma found during MRI scanning. As of 9
August 2010, there had been 12 deaths among the 37 patients,
giving a mortality rate of 2.68 per 100 person years. Leaving
aside the two patients who committed suicide, the median
disability of the patients who died was EDSS 8 (range 6e9),
measured on average 3 years before death. Subsequent clinic
visits to measure EDSS became impossible as the patients
became increasingly dependent and institutionalised. Causes of
death in this group were infections associated with advanced
disability (table 2).
Figure 2 Recovery of total
lymphocyte counts (LC) following
alemtuzumab treatment. Thirty-four of
the 36 patients recovered their total LC
to a normal level, defined as
$1.0310
9
cells/l. (A) Distribution of
time taken to recovery to a normal level
after log transformation. (B) Box and
whisker plot of log total LC recovery
time to a normal level by autoimmune
status. 0¼no autoimmune disease,
1¼autoimmune disease (see methods
for definition, p¼0.08). One outlier
(greater than 1.53IQR) can be seen in
the autoimmune status¼1 group. (C)
Cumulative incidence curve plotting the
recovery distribution where an event is
the occurrence of total LC recovery to
the patient’s baseline level (solid line).
Vertical lines on the curve indicate
censored observationsdthat is,
patients were not followed-up further
due to death, retreatment or lack of
lymphocyte data. Broken lines are
95% CIs.
J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826 301
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
OCB in cerebrospinal fluid
Paired CSF samples were taken before (range 0e37 days) and
after (range 3e28 months) the rst (12 patients) or second
(three patients) cycle of alemtuzumab treatment. In all 15 cases,
analysis of the CSF demonstrated the persistence of OCB
following treatment with alemtuzumab.
DISCUSSION
We have reported the longest follow-up to date of patients with
MS after a single course of alemtuzumab treatment: 384 person
years of lymphocyte counts and 447 person years of clinical data
from 37 patients treated between 1991 and 1997. We have
shown that lymphocyte counts recovered to the lower limit of
the normal range within 8 months (B cells) and 3 years (T cell
subsets), but rarely returned to baseline values. No long
term safety signal emerges from this small cohort, other than
conrmation of the increased risk of autoimmunity.
As previously reported,
3 9
we observed faster reconstitution of
B cells after alemtuzumab than T cells; we have previously
shown that the B cell subtypes return at varying rates.
17
There
was a suggestion that those patients with a high baseline total
lymphocyte count recover to a normal, albeit lower, lymphocyte
count more rapidly than others. Interestingly, T cell numbers
returned to the normal range in nearly all patients (78% of
patients for CD4, 86% for CD8), but rarely to baseline levels
(21% of patients for CD4 and 30% for CD8). We speculate that
in adults with little thymic function, reconstitution of the T cell
pool after alemtuzumab may be resetto a lower threshold.
Furthermore, there may be sustained alterations within the
lymphocyte subsetsdsuch as those shown at 12 months.
18
This
suggests that simple lymphocyte counts may not be a reliable
assessment of immunocompetence.
Our previous study of lymphocyte reconstitution in this
cohort, assuming linear kinetics, suggested more rapid recon-
stitution of Tcells.
9
However, it is now clear that reconstitution
is initially linear and rapid, followed either by slowing of the rate
of increase and/or a subsequent fall in lymphocyte count. A
similar pattern of immune reconstitution is seen after alemtu-
zumab treatment of other autoimmune diseases or for organ
transplantation,
13 19e21
and after alemtuzumab in an hCD52
transgenic mouse, albeit on a much contracted timescale.
12
Long
term follow-up was possible in patients treated with alemtu-
zumab for refractory rheumatoid arthritis between 1991 and
1994.
22 23
This cohort was relatively older (median age 54 years,
range 25.5e70) and had received a median of four disease
modifying antirheumatic drugs (range 1e8) before treatment. At
a median of 11.8 years (range 10.5e13.3), post-treatment CD4,
CD8 and CD19 counts were 0.5, 0.26 and 0.11310
9
cells/l,
respectively.
23
Most had CD4 and CD8 T cell counts within the
normal range but, in contrast with this study, B cell counts were
subnormal in 50%. No excess mortality or infections were seen
in patients treated with alemtuzumab compared with a hospital
based rheumatoid arthritis cohort. Likewise, this lymphocyte
reconstitution prole after alemtuzumab was similar to that
seen following lymphopenia in other contexts, suggesting that it
is driven by common homeostatic mechanisms, for instance
after haematopoietic stem cell transplantation.
6132425
The lag
in CD4 cell recovery correlates with age of the recipient and
probably reects impaired thymic function. Indeed, thymus
enlargement is evident radiographically in younger patients
following hematopoietic stem cell transplantation (HSCT)
26
and is seen in individuals treated with HSCT for both non-
Hodgkins lymphoma and MS.
27
Interestingly, CD4 reconstitu-
tion after HSCT for rheumatoid arthritis is considerably delayed,
an observation attributed to poor memory T cell expansion
associated with low levels of circulating interleukin (IL)-7.
28
However, in people with MS, serum IL-7 levels rise signicantly
after alemtuzumab.
18
As expected from previous reports,
3 13
one-third of MS
patients develop autoimmunity after alemtuzumab, particularly
those prone to excessive IL-21 secretion.
16
Here we demon-
strated a non-signicant trend that those patients who devel-
oped autoimmunity reconstituted their total lymphocytes
quicker (31.8) than those without autoimmunity. There are too
few data on lymphocyte subsets to judge whether their recon-
stitution differentiates between those with and without
autoimmunity.
We have also shown that alemtuzumab does not alter the
persistence of OCB in the CSF of patients with progressive MS
following treatment with alemtuzumab. Treatment with other
effective immunotherapies, such as rituximab
29
and autologous
HSCT, also do not eradicate intrathecal antibody
production.
30e36
These therapies, which so radically alter the
peripheral immune compartment, are clearly unable either to
Figure 3 Recovery of CD19 B cell counts following alemtuzumab
treatment. Thirty-four of the 36 patients recovered their B cells to
a normal level, defined as $0.1310
9
cells/l. (A) Distribution of time
taken for B cell recovery to a normal level. (B) Log transformation of time
taken for B cell recovery to a normal level.
302 J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
access or inuence the plasma cells producing antibodies
detected in CSF.
No particular safety signal emerged from this study. No
patient was lost to follow-up and our data on death and major
safety events are complete. Segmental varicella zoster virus
reactivation, a feature of alemtuzumab treatment in the
CAMMS223 trial,
3
was seen in 5/37 patients. However, minor
adverse events were not systematically collected and the cohort
described here is small. The phase 3 trials of alemtuzumab will
add further information on long term safety after alemtuzumab
in the context of MS.
The fact that 12/37 of this progressive MS cohort died is
consistent with our experience of managing people with
untreated MS at this level of disability. Two of these 12
committed suicide, which is more common in MS.
37
The mean
disability of the remaining 10 patients was EDSS 8.1 on average
3 years before their death. The cause of death in these patients,
overwhelmingly due to sepsis from a urinary or chest source, is
typical of that in untreated patients with advanced disability
from MS.
38
Mortality increases with disability in MS: in
a Canadian cohort, people with an EDSS of $7.5 had an
increased death rate of four times that of controls
39
whereas it
Figure 4 Recovery of CD8 and CD4 T
cell counts following alemtuzumab
treatment. (A) Cumulative incidence
curve plotting the recovery distribution
where an event is the occurrence of
CD8 lymphocyte recovery to a normal
level, defined as $0.2310
9
cells/l
(solid line). Vertical lines on the curve
indicate censored observationsdthat
is, patients were not followed-up
further due to death, retreatment or lack
of lymphocyte data. Broken lines are
95% CIs. (B) Similar cumulative
incidence curve where an event is
defined as CD8 lymphocyte recovery to
the patient’s baseline level. (C)
Cumulative incidence curve where an
event is the recovery of CD4 T cells to
a normal level, defined as
$0.4310
9
cells/l. (D) Similar curve for
recovery of CD4 T cells to patient’s
baseline level.
Table 2 Details of the 12 patients that died from the cohort of 37 treated with alemtuzumab for progressive multiple sclerosis
Patient No
Age at first
alemtuzumab
treatment (years)
Time from first
dose to death (years) Last EDSS recorded
Time from last EDSS
to death (years) Cause of death
2 33 9 8 5.5 Pneumonia/MS
3 33 6 7.5 0.5 Urinary sepsis
4 48 10 7 4.0 Urinary sepsis
9 30 11 7.5 4.5 Unknown
17 31 10 8 0.5 Pneumonia
22 55 11 7.5 1.5 Prostate carcinoma
24 36 11 9 4.0 Pneumonia
25 34 13 9 7.0 Pneumonia
29 40 13 9 1.5 Pneumonia
35 23 6 8 1.0 Suicide
36 40 12 8.5 1.5 Urinary sepsis
37 42 3 6 1.0 Suicide
EDSS, Expanded Disability Status Score; MS, multiple sclerosis.
J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826 303
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
was increased eightfold in a French cohort with similar levels of
disability.
40
There is no indication that alemtuzumab treatment
or its complications were directly implicated in the deaths of
any patient in our study.
We conclude, from this small cohort, that one cycle of alem-
tuzumab has long lasting effects on the immune system,
possibly by resetting the target for reconstitution of T
lymphocytes to below baseline values: CD4 and CD8 T counts
enter the normal range by 3 years. Throughout, our patients
appeared immunocompetent, and the main complication of
alemtuzumab treatment remained autoimmunity. A caveat is
that current trial protocols for the treatment of rela-
psingeremitting MS require two cycles of alemtuzumab,
12 months apart, with possible re-treatment with evidence of
the return of disease activity. The long term effects of such
multiple alemtuzumab treatments have yet to be studied.
Acknowledgements The authors are grateful to Geoff Hale, Jenny Phillips and
members of the Therapeutic Antibody Centre, Oxford, for producing the
alemtuzumab (then called Campath-1H) used in this study. The authors are also
grateful to Dr Graham Wood and the Immunology Department of Addenbrookes
Hospital for lymphocyte phenotyping, some of which was supported by a grant from
NHS R&D, held by Professor Geoff Hale of the University of Oxford.
Funding During the course of this work, AJC was supported by an MRC Clinical
Training Fellowship, Wellcome Intermediate Fellowship and now by the Biomedical
Research Centre, Cambridge, NIHR. JLJ is also supported by the Biomedical Research
Centre, Cambridge, NIHR. The original studies were supported by a grant from
MuSTER.
Competing interests AJC, DASC and JLJ have received personal travel costs,
occasional honoraria and departmental support from Genzyme Corporation. GG has
received honoraria and consulting fees from Genzyme Corporation and Sanofi-Aventis.
Ethics approval Ethics approval was provided by the local research ethics
committees.
Contributors GAHC designed and performed the research, collected and interpreted
the data, and wrote the manuscript; TB performed the research, and collected and
interpreted the data; OT, JLJ, KM, JS, AG and GG conducted and performed research;
DASC designed the research and participated in the writing of the manuscript; MTF
performed the statistical analysis; and AJC designed and performed the research,
interpreted the data and participated in the writing of the manuscript.
Provenance and peer review Not commissioned; externally peer reviewed.
REFERENCES
1. Hale G,Waldmann H. From laboratory to clinic: the story of CAMPATH-1. Methods
Mol Med 2000;40:243e66.
2. Hale G,Dyer MJ, Clark MR, et al. Remission inducation in non-Hodgkin lymphoma
with reshaped human monoclonal antibody CAMPATH-1H. Lancet
1988;332:1394e9.
3. Coles AJ,Compston DA, Selmaj KW, et al. Alemtuzumab vs. interferon beta-1a in
early multiple sclerosis. N Engl J Med 2008;359:1786e801.
4. Genzyme Oncology.Comparison of alemtuzumab and Rebif efficacy in multiple
sclerosis, study one. 2007. http://www.genzymeoncology.com/onc/clinical/
trialdetailresults.asp?nct¼NCT00530348 (accessed 17 Jun 2011).
5. Genzyme Oncology.Comparison of alemtuzumab and Rebif efficacy in multiple
sclerosis, study two. 2007. http://www.genzymeoncology.com/onc/clinical/
trialdetailresults.asp?nct¼NCT00548405 (accessed 17 Jun 2011).
6. Isaacs JD,Watts RA, Hazleman BL, et al. Humanised monoclonal antibody therapy
for rheumatoid arthritis. Lancet 1992;340:748e52.
7. Lockwood CM,Hale G, Waldman H, et al. Remission induction in Behcet’s disease
following lymphocyte depletion by the anti-CD52 antibody CAMPATH 1-H.
Rheumatology 2003;42:1539e44.
8. Coles AJ,Wing MG, Molyneux P, et al. Monoclonal antibody treatment exposes
three mechanisms underlying the clinical course of multiple sclerosis. Ann Neurol
1999;46:296e304.
9. Coles AJ,Cox A, Le Page E, et al. The window of therapeutic opportunity in multiple
sclerosis: evidence from monoclonal antibody therapy. J Neurol 2006;253:98e108.
10. Fahey JL,Taylor JMG, Detels R, et al. The prognostic value of cellular and serologic
markers in infection with human immunodeficiency virus type-1. N Engl J Med
1990;322:166e72.
11. Thierfelder S,Hoffmannfezer G, Rodt H, et al. Anti-lymphocytic antibodies and
marrow transplantation. VI. Absence of immunosuppression in vivo after injection of
monoclonal antibodies blocking graft-versus-host reactions and humoral antibody
formation in vitro. Transplantation 1983;35:249e54.
12. Hu YP,Turner MJ, Shields J, et al. Investigation of the mechanism of action of
alemtuzumab in a human CD52 transgenic mouse model. Immunology
2009;128:260e70.
13. Coles AJ,Wing M, Smith S, et al. Pulsed monoclonal antibody treatment and
autoimmune thyroid disease in multiple sclerosis. Lancet 1999;354:1691e5.
14. Moreau T,Thorpe J, Miller D, et al. Preliminary evidence from magnetic-resonance-
imaging for reduction in disease-activity after lymphocyte depletion in multiple-
sclerosis. Lancet 1994;344:298e301.
15. Kurtzke JF.Rating neurologic impairment in multiple sclerosis: an expanded
disability status scale (EDSS). Neurology 1983;33:1444e52.
16. Jones JL,Phuah CL, Cox AL, et al. IL-21 drives secondary autoimmunity in patients
with multiple sclerosis, following therapeutic lymphocyte depletion with
alemtuzumab (Campath-1H). J Clin Invest 2009;119:2052e61.
17. Thompson SA,Jones JL, Cox AL, et al. B-cell reconstitution and BAFF after
alemtuzumab (Campath-1H) treatment of multiple sclerosis. J Clin Immunol
2009;30:99e105.
18. Cox AL,Thompson SA, Jones JL, et al. Lymphocyte homeostasis following
therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol
2005;35:3332e42.
19. Dalakas MC,Rakocevic G, Schmidt J, et al. Effect of alemtuzumab (CAMPATH 1-H)
in patients with inclusion-body myositis. Brain 2009;132:1536e44.
20. Armstrong N,Buckley P, Oberley T, et al. Analysis of primate renal allografts after T-
cell depletion with anti-CD3-CRM9. Transplantation 1998;66:5e13.
21. Kirk AD,Hale DA, Mannon RB, et al. Results from a human renal allograft tolerance
trial evaluating the humanized CD52-specific monoclonal antibody alemtuzumab
(Campath-1H). Transplantation 2003;76:120e9.
22. Isaacs JD,Greer S, Sharma S, et al. Morbidity and mortality in rheumatoid arthritis
patients with prolonged and profound therapy-induced lymphopenia. Arthritis Rheum
2001;44:1998e2008.
23. Lorenzi AR,Clarke AM, Wooldridge T, et al. Morbidity and mortality in rheumatoid
arthritis patients with prolonged therapy-induced lymphopenia. Arthritis Rheum
2008;58:370e5.
24. Isaacs JD,Thiel A. Immune reconstitution. Best Pract Res Clin Haematol
2004;17:345e58.
25. Brett S,Baxter G, Cooper J, et al. Repopulation of blood lymphocyte sub-populations
in rheumatoid arthritis patients treated with the depleting humanized monoclonal
antibody, CAMPATH-1H. Immunology 1996;88:13e19.
26. Mackall CL,Fleisher TA, Brown MR, et al. Age, thymopoiesis, and CD4+ T-
lymphocyte regeneration after intensive chemotherapy. N Engl J Med
1995;332:143e9.
27. Fassas A,Anagnostopoulos A, Kazis A, et al. Autologous stem cell transplantation in
progressive multiple sclerosisdan interim analysis of efficacy. J Clin Immunol
2000;20:24e30.
28. Ponchel F,Verburg R, Bingham SJ, et al. Interleukin-7 deficiency in rheumatoid
arthritis: consequences for therapy-induced lymphopenia. Arthritis Res Ther
2005;7:R80e92.
29. Cross AH,Stark JL, Lauber J, et al. Rituximab reduces B cells and T cells in
cerebrospinal fluid of multiple sclerosis patients. J Neuroimmunol 2006;180:63e70.
30. Nash RA,Bowen JD, McSweeney PA, et al. High-dose immunosuppressive therapy
and autologous peripheral blood stem cell transplantation for severe multiple
sclerosis. Blood 2003;102:2364e72.
31. Burt RK,Traynor AE, Cohen B, et al. T cell-depleted autologous hematopoietic stem
cell transplantation for multiple sclerosis: report on the first three patients. Bone
Marrow Transplant 1998;21:537e41.
32. Carreras E,Saiz A, Marin P, et al. CD34(+) selected autologous peripheral
blood stem cell transplantation for multiple sclerosis: report of toxicity and
treatment results at one year of follow-up in 15 patients. Haematologica
2003;88:306e14.
33. Mondria T,Lamers CH, Boekhorst P, et al. Bone-marrow transplantation fails to halt
intrathecal lymphocyte activation in multiple sclerosis. J Neurol Neurosurg Psychiatry
2008;79:1013e15.
34. Openshaw H,Lund BT, Kashyap A, et al. Peripheral blood stem cell transplantation
in multiple sclerosis with busulfan and cyclophosphamide conditioning: report of
toxicity and immunological monitoring. Biol Blood Marrow Transplant
2000;6:563e75.
35. Saccardi R,Mancardi GL, Solari A, et al. Autologous HSCT for severe progressive
multiple sclerosis in a multicenter trial: impact on disease activity and quality of life.
Blood 2005;105:2601e7.
36. Saiz A,Carreras E, Berenguer J, et al. MRI and CSF oligoclonal bands after
autologous hematopoietic stem cell transplantation in MS. Neurology
2001;56:1084e9.
37. Bronnum-Hansen H,Stenager E, Stenager EN, et al. Suicide among Danes with
multiple sclerosis. J Neurol Neurosurg Psychiatry 2005;76:1457e9.
38. Ragonese P,Aridon P, Mazzola MA, et al. Multiple sclerosis survival: a population-
based study in Sicily. Eur J Neurol 2010;17:391e7.
39. Sadovnick AD,Ebers GC, Wilson RW, et al. Life expectancy in patients attending
multiple sclerosis clinics. Neurology 1992;42:991e4.
40. Leray E,Morrissey SP, Yaouanq J, et al. Long-term survival of patients with multiple
sclerosis in West France. Mult Scler 2007;13:865e74.
PAGE fraction trail=7
304 J Neurol Neurosurg Psychiatry 2012;83:298e304. doi:10.1136/jnnp-2011-300826
Multiple sclerosis
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
doi: 10.1136/jnnp-2011-300826
online November 5, 2011 2012 83: 298-304 originally publishedJ Neurol Neurosurg Psychiatry
Grant A Hill-Cawthorne, Tom Button, Orla Tuohy, et al.
sclerosisalemtuzumab treatment of multiple
Long term lymphocyte reconstitution after
http://jnnp.bmj.com/content/83/3/298.full.html
Updated information and services can be found at:
These include:
References http://jnnp.bmj.com/content/83/3/298.full.html#ref-list-1
This article cites 38 articles, 11 of which can be accessed free at:
service
Email alerting the box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in
Collections
Topic
(605 articles)Multiple sclerosis (1264 articles)Immunology (including allergy)
Articles on similar topics can be found in the following collections
Notes
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
group.bmj.com on April 1, 2012 - Published by jnnp.bmj.comDownloaded from
... The total B cell numbers quickly recover to baseline values, or even above, in 3-6 months 1 25-28 . Other studies observed a slower median recovery time of 8.4 months; only a few patients reached a final B cell count below the baseline values 29 . On the other hand, after ALTZ T lymphocytes repopulate much slower, driven by homeostatic proliferation of survived cells. ...
... In our patient the peripheral B cell counts remained as low as 6.6% (n.v. 7-21%) at 4 years after ALTZ, as also occasionally reported 29 , and after RTX became undetectable for the following 10 months. This is likely the result of a cumulative effect of ALTZ and RTX, further hindering the B cell repopulation 36 . ...
Article
Full-text available
BACKGROUND Secondary thyroid autoimmunity, especially Graves’ disease (GD), frequently develops in patients with multiple sclerosis (MS) following alemtuzumab treatment (ALTZ; anti-CD52). Thyroid Eye Disease (TED) can also develop, and rituximab (RTX; anti-CD20) is a suitable treatment. METHODS Immunophenotyping of blood and thyroid-derived lymphocytes in a patient treated with both ALTZ and RTX. RESULTS A 37-year-old woman with MS developed steroid-resistant active moderate-to-severe TED three years after ALTZ, that successfully responded to a single 500 mg dose of i.v. RTX. Before RTX peripheral B-cells were low, and were totally depleted immediately after therapy. Follow-up analysis four years post-ALTZ and one year post-RTX, showed persistent depletion of B cells, and relative reduction of T regulatory cells in both peripheral blood and thyroid tissue obtained at thyroidectomy. CONCLUSIONS RTX therapy successfully inactivated TED in a patient with low B-cell count derived from previous ALTZ treatment. B-cell depletion in both thyroid and peripheral blood was still present one year after RTX, as a likely cumulative effect of both treatments.
... The primary receptors involved in the activity of the therapeutic proteins include (parenthetical entry shows the number of such receptors): The primary receptors for approved protein therapeutics include: Glucagon-like peptide 1 receptor (3), Insulin receptors (3), Heat-stable enterotoxin receptors (2), Adrenocorticotropic hormone receptor, Angiotensin II type 2 (AT-2) receptor, Corticotropin-releasing factor receptor 1, Glucagonlike peptide 2 receptor, Gonadotropin-releasing hormone receptor, Notch signaling pathway, Oxytocin receptor, Parathyroid hormone receptor, Parathyroid hormone/parathyroid hormonerelated peptide receptor, Prothrombin, Receptor tyrosine-protein kinase erbB-2, Secretin receptor, Somatostatin receptor 2, Somatostatin receptor 5, Type-1 angiotensin II receptor, Vasopressin V1a receptor, Vasopressin V1b receptor, Vasopressin V1a receptor, Vasopressin V1b receptor, Vasopressin V2 receptor. [209] ( Table 1) [210] GPIIb/IIIa Adalimumab (Humira) [211] TNFα Alemtuzumab (Lemtrada) [212] CD52 Atezolizumab (Tecentriq) [213] PD-L1 Basiliximab (Simulect) [214] CD25 Belimumab (Benlysta) [215] BLyS Bevacizumab (Avastin) [216] VEGF Cetuximab (Erbitux) [217] EGFR Daclizumab (Zinbryta) [218] CD25 Daratumumab (Darzalex) [219] CD38 Denosumab (Prolia) [220] RANKL Dupilumab (Dupixent) [221] IL-4Rα Eculizumab (Soliris) [222] C5 Infliximab (Remicade) [223] TNFα Ipilimumab (Yervoy) [224] CTLA-4 Nivolumab (Opdivo) [225] PD-1 Obinutuzumab (Gazyva) [226] CD20 Ofatumumab (Arzerra) [227] CD20 Omalizumab (Xolair) [228] IgE Palivizumab (Synagis) [229] RSV F protein Pembrolizumab (Keytruda) [230] PD-1 Rituximab (Rituxan) [231] CD20 Sarilumab (Kevzara) [232] IL-6R Secukinumab (Cosentyx) [233] IL-17A Tocilizumab (Actemra) [234] IL-6R Trastuzumab (Herceptin) [235] HER2/neu Vedolizumab (Entyvio) [236] α4β7 integrin ...
Preprint
Full-text available
Demonstrating biosimilarity entails comprehensive analytical evaluations, clinical pharmacolo-gy profiling, and efficacy testing for at least one medical indication in patients. These require-ments are stipulated by the U.S. Biologics Price Competition and Innovation Act (BPCIA). The costliest element—efficacy testing—can be waived if other compliance benchmarks are satisfied, including comparing functional pharmacodynamic (PD) biomarkers, even when they do not di-rectly correlate with clinical outcomes. Most biological drugs, such as monoclonal antibodies (mAbs), lack identifiable PD biomarkers. The FDA has employed various 'omics' technologies to identify potential PD biomarkers, including proteomics, glycomics, transcriptomics, genomics, epigenomics, and metabolomics. Although these efforts provide a robust scientific basis for estab-lishing biosimilarity, they are neither practical nor necessarily superior to existing functional biomarkers, such as receptor binding and mode-of-action outcomes. As we report for the first time, these functional biomarkers can effectively serve as PD indicators for all FDA-licensed bio-logical drugs. We recommend that the FDA consider officially listing these functional biomarkers to expedite and reduce the cost of biosimilar development, thereby increasing the accessibility of biological drugs. PD surrogates, like the receptor binding and pharmacokinetic profiles, are more robust and offer a rational solution to finding PD markers to compare for establishing biosimi-larity.
Article
Full-text available
Background Different therapeutic strategies are available for the treatment of people with relapsing‐remitting multiple sclerosis (RRMS), including immunomodulators, immunosuppressants and biological agents. Although each one of these therapies reduces relapse frequency and slows disability accumulation compared to no treatment, their relative benefit remains unclear. This is an update of a Cochrane review published in 2015. Objectives To compare the efficacy and safety, through network meta‐analysis, of interferon beta‐1b, interferon beta‐1a, glatiramer acetate, natalizumab, mitoxantrone, fingolimod, teriflunomide, dimethyl fumarate, alemtuzumab, pegylated interferon beta‐1a, daclizumab, laquinimod, azathioprine, immunoglobulins, cladribine, cyclophosphamide, diroximel fumarate, fludarabine, interferon beta 1‐a and beta 1‐b, leflunomide, methotrexate, minocycline, mycophenolate mofetil, ofatumumab, ozanimod, ponesimod, rituximab, siponimod and steroids for the treatment of people with RRMS. Search methods CENTRAL, MEDLINE, Embase, and two trials registers were searched on 21 September 2021 together with reference checking, citation searching and contact with study authors to identify additional studies. A top‐up search was conducted on 8 August 2022. Selection criteria Randomised controlled trials (RCTs) that studied one or more of the available immunomodulators and immunosuppressants as monotherapy in comparison to placebo or to another active agent, in adults with RRMS. Data collection and analysis Two authors independently selected studies and extracted data. We considered both direct and indirect evidence and performed data synthesis by pairwise and network meta‐analysis. Certainty of the evidence was assessed by the GRADE approach. Main results We included 50 studies involving 36,541 participants (68.6% female and 31.4% male). Median treatment duration was 24 months, and 25 (50%) studies were placebo‐controlled. Considering the risk of bias, the most frequent concern was related to the role of the sponsor in the authorship of the study report or in data management and analysis, for which we judged 68% of the studies were at high risk of other bias. The other frequent concerns were performance bias (34% judged as having high risk) and attrition bias (32% judged as having high risk). Placebo was used as the common comparator for network analysis. Relapses over 12 months: data were provided in 18 studies (9310 participants). Natalizumab results in a large reduction of people with relapses at 12 months (RR 0.52, 95% CI 0.43 to 0.63; high‐certainty evidence). Fingolimod (RR 0.48, 95% CI 0.39 to 0.57; moderate‐certainty evidence), daclizumab (RR 0.55, 95% CI 0.42 to 0.73; moderate‐certainty evidence), and immunoglobulins (RR 0.60, 95% CI 0.47 to 0.79; moderate‐certainty evidence) probably result in a large reduction of people with relapses at 12 months. Relapses over 24 months: data were reported in 28 studies (19,869 participants). Cladribine (RR 0.53, 95% CI 0.44 to 0.64; high‐certainty evidence), alemtuzumab (RR 0.57, 95% CI 0.47 to 0.68; high‐certainty evidence) and natalizumab (RR 0.56, 95% CI 0.48 to 0.65; high‐certainty evidence) result in a large decrease of people with relapses at 24 months. Fingolimod (RR 0.54, 95% CI 0.48 to 0.60; moderate‐certainty evidence), dimethyl fumarate (RR 0.62, 95% CI 0.55 to 0.70; moderate‐certainty evidence), and ponesimod (RR 0.58, 95% CI 0.48 to 0.70; moderate‐certainty evidence) probably result in a large decrease of people with relapses at 24 months. Glatiramer acetate (RR 0.84, 95%, CI 0.76 to 0.93; moderate‐certainty evidence) and interferon beta‐1a (Avonex, Rebif) (RR 0.84, 95% CI 0.78 to 0.91; moderate‐certainty evidence) probably moderately decrease people with relapses at 24 months. Relapses over 36 months findings were available from five studies (3087 participants). None of the treatments assessed showed moderate‐ or high‐certainty evidence compared to placebo. Disability worsening over 24 months was assessed in 31 studies (24,303 participants). Natalizumab probably results in a large reduction of disability worsening (RR 0.59, 95% CI 0.46 to 0.75; moderate‐certainty evidence) at 24 months. Disability worsening over 36 months was assessed in three studies (2684 participants) but none of the studies used placebo as the comparator. Treatment discontinuation due to adverse events data were available from 43 studies (35,410 participants). Alemtuzumab probably results in a slight reduction of treatment discontinuation due to adverse events (OR 0.39, 95% CI 0.19 to 0.79; moderate‐certainty evidence). Daclizumab (OR 2.55, 95% CI 1.40 to 4.63; moderate‐certainty evidence), fingolimod (OR 1.84, 95% CI 1.31 to 2.57; moderate‐certainty evidence), teriflunomide (OR 1.82, 95% CI 1.19 to 2.79; moderate‐certainty evidence), interferon beta‐1a (OR 1.48, 95% CI 0.99 to 2.20; moderate‐certainty evidence), laquinimod (OR 1.49, 95 % CI 1.00 to 2.15; moderate‐certainty evidence), natalizumab (OR 1.57, 95% CI 0.81 to 3.05), and glatiramer acetate (OR 1.48, 95% CI 1.01 to 2.14; moderate‐certainty evidence) probably result in a slight increase in the number of people who discontinue treatment due to adverse events. Serious adverse events (SAEs) were reported in 35 studies (33,998 participants). There was probably a trivial reduction in SAEs amongst people with RRMS treated with interferon beta‐1b as compared to placebo (OR 0.92, 95% CI 0.55 to 1.54; moderate‐certainty evidence). Authors' conclusions We are highly confident that, compared to placebo, two‐year treatment with natalizumab, cladribine, or alemtuzumab decreases relapses more than with other DMTs. We are moderately confident that a two‐year treatment with natalizumab may slow disability progression. Compared to those on placebo, people with RRMS treated with most of the assessed DMTs showed a higher frequency of treatment discontinuation due to AEs: we are moderately confident that this could happen with fingolimod, teriflunomide, interferon beta‐1a, laquinimod, natalizumab and daclizumab, while our certainty with other DMTs is lower. We are also moderately certain that treatment with alemtuzumab is associated with fewer discontinuations due to adverse events than placebo, and moderately certain that interferon beta‐1b probably results in a slight reduction in people who experience serious adverse events, but our certainty with regard to other DMTs is lower. Insufficient evidence is available to evaluate the efficacy and safety of DMTs in a longer term than two years, and this is a relevant issue for a chronic condition like MS that develops over decades. More than half of the included studies were sponsored by pharmaceutical companies and this may have influenced their results. Further studies should focus on direct comparison between active agents, with follow‐up of at least three years, and assess other patient‐relevant outcomes, such as quality of life and cognitive status, with particular focus on the impact of sex/gender on treatment effects.
Article
Full-text available
Demonstrating biosimilarity entails comprehensive analytical assessment, clinical pharmacology profiling, and efficacy testing in patients for at least one medical indication, as required by the U.S. Biologics Price Competition and Innovation Act (BPCIA). The efficacy testing can be waived if the drug has known pharmacodynamic (PD) markers, leaving most therapeutic proteins out of this concession. To overcome this, the FDA suggests that biosimilar developers discover PD biomarkers using omics technologies such as proteomics, glycomics, transcriptomics, genomics, epigenomics, and metabolomics. This approach is redundant since the mode-action-action biomarkers of approved therapeutic proteins are already available, as compiled in this paper for the first time. Other potential biomarkers are receptor binding and pharmacokinetic profiling, which can be made more relevant to ensure biosimilarity without requiring biosimilar developers to conduct extensive research, for which they are rarely qualified.
Article
Thymic and bone marrow outputs were evaluated in 13 sequential samples of 68 multiple sclerosis patients who initiated alemtuzumab and were clinically followed for 48 months. Three months after alemtuzumab infusions, the levels of new T lymphocytes were significantly reduced, but progressively increased reaching the highest values at 36 months, indicating the remarkable capacity of thymic function recovery. Newly produced B cells exceeded baseline levels as early as 3 months after alemtuzumab initiation. Heterogeneous patterns of new T- and B-cell recovery were identified, but without associations with age, sex, previous therapies, development of secondary autoimmunity or infections, and disease re-emergence. Trial registration version 2.0-27/01/2016.
Article
Background: Multiple sclerosis (MS) is an autoimmune, T-cell-dependent, inflammatory, demyelinating disease of the central nervous system, with an unpredictable course. Current MS therapies focus on treating and preventing exacerbations, and avoiding the progression of disability. At present, there is no treatment that is capable of safely and effectively reaching these objectives. Clinical trials suggest that alemtuzumab, a humanized monoclonal antibody, could be a promising option for MS. Objectives: To evaluate the benefits and harms of alemtuzumab alone or associated with other treatments in people with any form of MS. Search methods: We used standard, extensive Cochrane search methods. The latest search date was 21 June 2022. Selection criteria: We included randomized controlled trials (RCTs) in adults with any subtype of MS comparing alemtuzumab alone or associated with other medications versus placebo; another active drug; or alemtuzumab in another dose, regimen, or duration. Data collection and analysis: We used standard Cochrane methods. Our co-primary outcomes were 1. relapse-free survival, 2. sustained disease progression, and 3. number of participants experiencing at least one adverse event. Our secondary outcomes were 4. participants free of clinical disability, 5. quality of life, 6. change in disability, 7. fatigue, 8. new or enlarging lesions on resonance imaging, and 9. dropouts. We used GRADE to assess certainty of evidence for each outcome. Main results: We included three RCTs (1713 participants) comparing intravenous alemtuzumab versus subcutaneous interferon beta-1a for relapsing-remitting MS. Participants were treatment-naive (two studies) or had experienced at least one relapse after interferon or glatiramer (one study). Alemtuzumab was given at doses of 12 mg/day or 24 mg/day for five days at months 0 and 12, or 24 mg/day for three days at months 12 and 24. Participants in the interferon beta-1a group received 44 μg three times weekly. Alemtuzumab 12 mg: 1. may improve relapse-free survival at 36 months (hazard ratio [HR] 0.31, 95% confidence interval [CI] 0.18 to 0.53; 1 study, 221 participants; low-certainty evidence); 2. may improve sustained disease progression-free survival at 36 months (HR 0.25, 95% CI 0.11 to 0.56; 1 study, 223 participants; low-certainty evidence); 3. may make little to no difference on the proportion of participants with at least one adverse event at 36 months (risk ratio [RR] 1.00, 95% CI 0.98 to 1.02; 1 study, 224 participants; low-certainty evidence), although the proportion of participants with at least one adverse event was high with both drugs; 4. may slightly reduce disability at 36 months (mean difference [MD] -0.70, 95% CI -1.04 to -0.36; 1 study, 223 participants; low-certainty evidence). The evidence is very uncertain regarding the risk of dropouts at 36 months (RR 0.81, 95% CI 0.57 to 1.14; 1 study, 224 participants; very low-certainty evidence). Alemtuzumab 24 mg: 1. may improve relapse-free survival at 36 months (HR 0.21, 95% CI 0.11 to 0.40; 1 study, 221 participants; low-certainty evidence); 2. may improve sustained disease progression-free survival at 36 months (HR 0.33, 95% CI 0.16 to 0.69; 1 study, 221 participants; low-certainty evidence); 3. may make little to no difference on the proportion of participants with at least one adverse event at 36 months (RR 0.99, 95% CI 0.97 to 1.02; 1 study, 215 participants; low-certainty evidence), although the proportion of participants with at least one adverse event was high with both drugs; 4. may slightly reduce disability at 36 months (MD -0.83, 95% CI -1.16 to -0.50; 1 study, 221 participants; low-certainty evidence); 5. may reduce the risk of dropouts at 36 months (RR 0.08, 95% CI 0.01 to 0.57; 1 study, 215 participants; low-certainty evidence). For quality of life, fatigue, and participants free of clinical disease activity, the studies either did not consider these outcomes or they used different measuring tools to those planned in this review. Authors' conclusions: Compared with interferon beta-1a, alemtuzumab may improve relapse-free survival and sustained disease progression-free survival, and make little to no difference on the proportion of participants with at least one adverse event for people with relapsing-remitting MS at 36 months. The certainty of the evidence for these results was very low to low.
Article
Full-text available
Multiple sclerosis (MS) is a disease of the central nervous system characterized by immune-mediated destruction of myelin. In patients with progressive deterioration, we have intensified immunosuppression to the point of myeloablation. Subsequently, a new hematopoietic and immune system is generated by infusion of CD34-positive hematopoietic stem cells (HSC). Three patients with clinical MS and a decline of their Kurtzke extended disability status scale (EDSS) by 1.5 points over the 12 months preceding enrollment and a Kurtzke EDSS of 8.0 at the time of enrollment were treated with hematopoietic stem cell (HSC) transplantation using a myeloablative conditioning regimen of cyclophosphamide (120 mg/kg), methylprednisolone (4 g) and total body irradiation (1200 cGy). Reconstitution of hematopoiesis was achieved with CD34-enriched stem cells. The average time of follow-up is 8 months (range 6-10 months). Despite withdrawal of all immunosuppressive medications, functional improvements have occurred in all three patients. We conclude that T cell-depleted hematopoietic stem cell transplantation can be performed safely in patients with severe and debilitating multiple sclerosis. Stem cell transplantation has resulted in modest neurologic improvements for the first time since onset of progressive disease although no significant changes in EDSS or NRS scales are evident at this time.
Article
Full-text available
Introduction Treatment with alemtuzumab is highly effective in relapsing–remitting multiple sclerosis; however, 30% of patients develop autoimmunity. Alemtuzumab (previously called Campath 1-H) induces a prolonged T-cell lymphopenia with memory cells dominating the reconstituting T-cell pool for at least 3 months. Results Here we show that B-cell recovery is rapid, returning to baseline by 3 months and rising to 165% of baseline by 12 months after treatment. Immature transitional 1 B cells are the predominant cell type 1 month after treatment. This coincides with a surge in serum B-cell activating factor (BAFF), which remains elevated by 33% for at least 12 months after alemtuzumab. BAFF is critical for transition to the mature naive B-cell phenotype, which dominates from 3 months after alemtuzumab. Differentiation to memory B cells is slow so there are radical and prolonged alterations to the B-cell pool after alemtuzumab.
Article
Full-text available
Phase II clinical trials revealed that the lymphocyte-depleting humanized monoclonal antibody alemtuzumab (Campath-1H) is highly effective in the treatment of early relapsing-remitting multiple sclerosis. However, 30% of patients develop autoimmunity months to years after pulsed exposure to alemtuzumab, usually targeting the thyroid gland and, more rarely, blood components. In this study, we show that autoimmunity arose in those patients with greater T cell apoptosis and cell cycling in response to alemtuzumab-induced lymphocyte depletion, a phenomenon that is driven by higher levels of IL-21. Before treatment, patients who went on to develop secondary autoimmunity had more than 2-fold greater levels of serum IL-21 than the nonautoimmune group. We suggest that serum IL-21 may, therefore, serve as a biomarker for the risk of developing autoimmunity months to years after alemtuzumab treatment. This has implications for counseling those patients with multiple sclerosis who are considering lymphocyte-depleting therapy with alemtuzumab. Finally, we demonstrate through genotyping that IL-21 expression is genetically predetermined. We propose that, by driving cycles of T cell expansion and apoptosis to excess, IL-21 increases the stochastic opportunities for T cells to encounter self antigen and, hence, for autoimmunity.
Article
Full-text available
Sporadic inclusion-body myositis (sIBM) is the most common disabling, adult-onset, inflammatory myopathy histologically characterized by intense inflammation and vacuolar degeneration. In spite of T cell-mediated cytotoxicity and persistent, clonally expanded and antigen-driven endomysial T cells, the disease is resistant to immunotherapies. Alemtuzumab is a humanized monoclonal antibody that causes an immediate depletion or severe reduction of peripheral blood lymphocytes, lasting at least 6 months. We designed a proof-of-principle study to examine if one series of Alemtuzumab infusions in sIBM patients depletes not only peripheral blood lymphocytes but also endomysial T cells and alters the natural course of the disease. Thirteen sIBM patients with established 12-month natural history data received 0.3 mg/kg/day Alemtuzumab for 4 days. The study was powered to capture > or =10% increase strength 6 months after treatment. The primary end-point was disease stabilization compared to natural history, assessed by bi-monthly Quantitative Muscle Strength Testing and Medical Research Council strength measurements. Lymphocytes and T cell subsets were monitored concurrently in the blood and the repeated muscle biopsies. Alterations in the mRNA expression of inflammatory, stressor and degeneration-associated molecules were examined in the repeated biopsies. During a 12-month observation period, the patients' total strength had declined by a mean of 14.9% based on Quantitative Muscle Strength Testing. Six months after therapy, the overall decline was only 1.9% (P < 0.002), corresponding to a 13% differential gain. Among those patients, four improved by a mean of 10% and six reported improved performance of daily activities. The benefit was more evident by the Medical Research Council scales, which demonstrated a decline in the total scores by 13.8% during the observation period but an improvement by 11.4% (P < 0.001) after 6 months, reaching the level of strength recorded 12 months earlier. Depletion of peripheral blood lymphocytes, including the naive and memory CD8+ cells, was noted 2 weeks after treatment and persisted up to 6 months. The effector CD45RA(+)CD62L(-) cells, however, started to increase 2 months after therapy and peaked by the 4th month. Repeated muscle biopsies showed reduction of CD3 lymphocytes by a mean of 50% (P < 0.008), most prominent in the improved patients, and reduced mRNA expression of stressor molecules Fas, Mip-1a and alphaB-crystallin; the mRNA of desmin, a regeneration-associated molecule, increased. This proof-of-principle study provides insights into the pathogenesis of inclusion-body myositis and concludes that in sIBM one series of Alemtuzumab infusions can slow down disease progression up to 6 months, improve the strength of some patients, and reduce endomysial inflammation and stressor molecules. These encouraging results, the first in sIBM, warrant a future study with repeated infusions
Article
Full-text available
Alemtuzumab, a humanized monoclonal antibody that targets CD52 on lymphocytes and monocytes, may be an effective treatment for early multiple sclerosis. In this phase 2, randomized, blinded trial involving previously untreated, early, relapsing-remitting multiple sclerosis, we assigned 334 patients with scores of 3.0 or less on the Expanded Disability Status Scale and a disease duration of 3 years or less to receive either subcutaneous interferon beta-1a (at a dose of 44 microg) three times per week or annual intravenous cycles of alemtuzumab (at a dose of either 12 mg or 24 mg per day) for 36 months. In September 2005, alemtuzumab therapy was suspended after immune thrombocytopenic purpura developed in three patients, one of whom died. Treatment with interferon beta-1a continued throughout the study. Alemtuzumab significantly reduced the rate of sustained accumulation of disability, as compared with interferon beta-1a (9.0% vs. 26.2%; hazard ratio, 0.29; 95% confidence interval [CI], 0.16 to 0.54; P<0.001) and the annualized rate of relapse (0.10 vs. 0.36; hazard ratio, 0.26; 95% CI, 0.16 to 0.41; P<0.001). The mean disability score on a 10-point scale improved by 0.39 point in the alemtuzumab group and worsened by 0.38 point in the interferon beta-1a group (P<0.001). In the alemtuzumab group, the lesion burden (as seen on T(2)-weighted magnetic resonance imaging) was reduced, as compared with that in the interferon beta-1a group (P=0.005). From month 12 to month 36, brain volume (as seen on T(1)-weighted magnetic resonance imaging) increased in the alemtuzumab group but decreased in the interferon beta-1a group (P=0.02). Adverse events in the alemtuzumab group, as compared with the interferon beta-1a group, included autoimmunity (thyroid disorders [23% vs. 3%] and immune thrombocytopenic purpura [3% vs. 1%]) and infections (66% vs. 47%). There were no significant differences in outcomes between the 12-mg dose and the 24-mg dose of alemtuzumab. In patients with early, relapsing-remitting multiple sclerosis, alemtuzumab was more effective than interferon beta-1a but was associated with autoimmunity, most seriously manifesting as immune thrombocytopenic purpura. The study was not powered to identify uncommon adverse events. (ClinicalTrials.gov number, NCT00050778.)
Article
We previously demonstrated prolonged, profound CD4+ T-lymphopenia in rheumatoid arthritis (RA) patients following lymphocyte-depleting therapy. Poor reconstitution could result either from reduced de novo T-cell production through the thymus or from poor peripheral expansion of residual T-cells. Interleukin-7 (IL-7) is known to stimulate the thymus to produce new T-cells and to allow circulating mature T-cells to expand, thereby playing a critical role in T-cell homeostasis. In the present study we demonstrated reduced levels of circulating IL-7 in a cross-section of RA patients. IL-7 production by bone marrow stromal cell cultures was also compromised in RA. To investigate whether such an IL-7 deficiency could account for the prolonged lymphopenia observed in RA following therapeutic lymphodepletion, we compared RA patients and patients with solid cancers treated with high-dose chemotherapy and autologous progenitor cell rescue. Chemotherapy rendered all patients similarly lymphopenic, but this was sustained in RA patients at 12 months, as compared with the reconstitution that occurred in cancer patients by 3–4 months. Both cohorts produced naïve T-cells containing T-cell receptor excision circles. The main distinguishing feature between the groups was a failure to expand peripheral T-cells in RA, particularly memory cells during the first 3 months after treatment. Most importantly, there was no increase in serum IL-7 levels in RA, as compared with a fourfold rise in non-RA control individuals at the time of lymphopenia. Our data therefore suggest that RA patients are relatively IL-7 deficient and that this deficiency is likely to be an important contributing factor to poor early T-cell reconstitution in RA following therapeutic lymphodepletion. Furthermore, in RA patients with stable, well controlled disease, IL-7 levels were positively correlated with the T-cell receptor excision circle content of CD4+ T-cells, demonstrating a direct effect of IL-7 on thymic activity in this cohort.
Article
Objective Therapies that deplete lymphocytes often improve symptoms in patients with otherwise refractory autoimmune disease but may result in long-term lymphopenia, the consequences of which are uncertain. To assess the impact of prolonged lymphopenia on morbidity and mortality, we studied patients who had previously received lymphocytotoxic monoclonal antibody (mAb) therapy for rheumatoid arthritis (RA).Methods Fifty-three patients who received the lymphocytotoxic mAb CAMPATH-1H between 1991 and 1994 in the United Kingdom were assessed for mortality and infectious and malignant morbidity, by interview and case-note review. In addition, patients were monitored via the National Health Service Central Registry, to verify notification of death. Peripheral blood lymphocyte subsets were analyzed by flow cytometry. A retrospective, matched-cohort study of mortality was also performed with 102 control subjects selected from the European League Against Rheumatism database, which comprises patients with rheumatic disorders who have received immunosuppressive drugs.ResultsThere was profound and persistent peripheral blood lymphopenia in the mAb-treated patients, affecting predominantly the CD4+ subset. Median CD4+, CD8+, and CD19+ peripheral blood lymphocyte counts at 73–84 months after therapy were 185 cells/μl, 95 cells/μl, and 115 cells/μl, respectively. At a median followup of 71 months (range 14–90), 13 patients had died (24.5%), compared with 18% of the matched controls, providing a mortality rate ratio of 1.45 (95% confidence interval 0.65–3.13). During 283 patient-years of followup, there were 36 infections classified as major (12.7 per 100 patient-years). The causes of death and the spectrum of infections documented were similar to those expected in a hospital-based RA cohort. Patients who received more than 1 course of therapy had more severe lymphopenia than did patients who received a single course, but this did not have an impact on mortality or morbidity.Conclusion Despite the occurrence of profound and long-lasting lymphopenia following treatment with antilymphocyte mAb therapy for RA, this therapy is not associated with a large excess of mortality nor with an unusual spectrum of infections, at least during a medium-term period of followup. These data are also relevant to patients receiving lymphocytotoxic mAb therapy for other indications, and to patients receiving other lymphodepleting therapies such as autologous stem cell transplantation.
Article
In 1890 it was shown that resistance to diphtheria toxin could be transferred from one animal to another by transfer of serum (1). From this discovery, passive antibody therapy was developed as an effective treatment for infectious diseases and for neutralization of toxins, and continues to be used to this day. Meanwhile, there have been continued efforts to use antibodies for cancer therapy, starting with the pioneering work of Hericourt and Richet in 1895 (2), which was the forerunner of the "magic bullet" concept. However, all of the early work on tumor therapy led ultimately to disappointment (3). The problems were readily acknowledged, i.e., lack of specificity and reproducibility, lack of purity, and the xenogeneic immune response. Developments over the past 20 years, as described throughout this book, have effectively overcome all of these technical problems, often in very ingenious ways. The difficulty we have now is different. There are just too many potential new antibody-based treatments for them all to be properly evaluated in the clinic. Many will still fail because of factors that are hard to predict from experiments: unexpected toxicity, biological heterogeneity of the target disease, or lack of access to the appropriate tissue.
Article
There are few population-based surveys on multiple sclerosis (MS) survival. To investigate MS survival in MS patients recruited during surveys conducted in Sicily. Multiple sclerosis patients identified during previous surveys were randomly matched to two referent subjects by residence, year of birth, and gender. Living status was obtained by municipality records (end of follow-up June, 30th 2007) and, for the deceased, date and causes of death were searched. Kaplan-Meier plots were used to calculate differences in mortality between MS patients and referent subjects. MS risks for mortality with 95% confidence intervals (CI) were also calculated. We included 194 MS patients and 388 matched persons. Thirty MS patients (15.5%) and 28 referents (7.2%) had died until the end of follow-up. Mean survival from onset of the disease to death was 20.6 years. Mean age at death was 55.5 for MS patients and 64.8 for the referents. Adjusted Hazard Ratios for mortality in MS was 1.81 (95% CI 1.36-2.40). Kaplan-Meier estimates showed a higher mortality amongst patients compared to referent subjects (P < 0.001). The present study confirms the higher mortality risk in MS patients with no significant gender difference. Causes of death are related to complications of high disability and to increasing age.
Article
Alemtuzumab is a humanized monoclonal antibody against CD52, an antigen found on the surface of normal and malignant lymphocytes. It is approved for the treatment of B-cell chronic lymphocytic leukaemia and is undergoing Phase III clinical trials for the treatment of multiple sclerosis. The exact mechanism by which alemtuzumab mediates its biological effects in vivo is not clearly defined and mechanism of action studies have been hampered by the lack of cross-reactivity between human and mouse CD52. To address this issue, a transgenic mouse expressing human CD52 (hCD52) was created. Transgenic mice did not display any phenotypic abnormalities and were able to mount normal immune responses. The tissue distribution of hCD52 and the level of expression by various immune cell populations were comparable to those seen in humans. Treatment with alemtuzumab replicated the transient increase in serum cytokines and depletion of peripheral blood lymphocytes observed in humans. Lymphocyte depletion was not as profound in lymphoid organs, providing a possible explanation for the relatively low incidence of infection in alemtuzumab-treated patients. Interestingly, both lymphocyte depletion and cytokine induction by alemtuzumab were largely independent of complement and appeared to be mediated by neutrophils and natural killer cells because removal of these populations with antibodies to Gr-1 or asialo-GM-1, respectively, strongly inhibited the activity of alemtuzumab whereas removal of complement by treatment with cobra venom factor had no impact. The hCD52 transgenic mouse appears to be a useful model and has provided evidence for the previously uncharacterized involvement of neutrophils in the activity of alemtuzumab.