6
Randomized control trials in ALS: lessons learned Hiroshi Mitsumoto, Paul Gordon, Petra Kaufmann, Clifton Gooch, Serge Przedborski and Lewis P Rowland The Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center Introduction As we stated in the Preface, a major impetus for organizing the ALS Clinical Trial Meeting held in Tarrytown, New York, was the overall great frustration we have had in treating ALS; that is, since 1996 when riluzole received FDA approval, we have not identified any new medications for ALS. Although we have been prescribing riluzole, its efficacy is modest at best. We desperately need more effective drugs. Although a cure may come only after the cause of ALS is elucidated, we do not have the luxury of waiting when we are confronted by the progressive paralysis of our patients. Vigorous basic research in ALS has offered new hypotheses and opportunities to test novel drugs. In the past two decades, we have had more than 25 randomized clinical trials (RCTs) in ALS (Table 1). Although we surely have made great progress in these trials, the collective experience is still less than that in many other diseases. An important lesson is always to learn from others and from the past. We have therefore decided to provide here not a review but rather an overview of the collective experience in past ALS RCTs, which hopefully will serve as a useful Introduction to this supplement. The issues that we raise are examined in detail by the other contributors to the supplement. Bench to clinic Drug effects in animal models have not been reliable predictors of human effects A critical limitation of RCTs is that our experience in ALS trials is not large enough. Because only riluzole has given positive RCT results, we cannot determine what type of results in preclinical studies in animal models will predict the results in human trials. At present, positive results in animals are rather misleading at best, and it is uncertain how much we actually have relied on the preclinical results. For example, the decision to test riluzole in humans was not based on studies in any animal models, only on its known anti- glutamate effects in in vitro studies. Riluzole was tested in SOD transgenic mice (termed SOD1 mice in this article) only after riluzole had already been approved (and also because transgenic mice carrying the human familial ALS mutation became generally available after the approval of riluzole). The effect size was small in both the mice and humans. 18,28 The SOD1 mouse is now considered to be the best animal model for preclinical testing in ALS. Earlier, neurotrophic factors were tested in other mouse models, such as the wobbler, pmn, and mnd mice, with some positive results that led to human trials of neurotrophic factors, including ciliary neurotrophic factors (CNTF), brain-derived neurotrophic factors (BDNF), and insulin-like growth factor-I (IGF-I). 29 When a number of neurotrophic factors were tested in SOD1 mice, negative findings were not published and trials were performed in spite of such findings. When most neurotrophic factors available for testing showed no clinical benefit in patients, trialists in retrospect felt that preclinical results in these less acceptable animal models had led to the negative results in human studies, and subsequently to the loss of interest in neurotrophic factor therapy. 29 Some major RCTs have been done without any preliminary studies in animal models, or were performed despite the fact that studies in animal models were negative, as seen in the examples of riluzole and xaliproden. Thus, it seems fair to state that animal studies have not been a critical factor in the decision- making for RCTs. Another issue is the timing of drug treatment in SOD1 mice. Presymptomatic treatment may not be ideal because it can be done only in those at risk for familial ALS, not in those with sporadic ALS. Because survival in SOD1 mice is usually less than 2 weeks after the onset of clinically detectable disease, particularly in the most commonly used mice (G93A SOD), almost all drug treatments have been initiated long before symptoms appear. At least two studies, which showed dramatic responses to therapeutic agents, commenced treatment after disease onset in SOD1 mice. Those studies include IGF-I gene transfer using adeno- associated virus 36 and a preliminary report with Mn- porphyrin treatment (J. Crow et al; reported at the 14th International ALS/MND Symposium, Milan 2003). Other complicating issues in SOD1 mice include the question of whether mutant SOD1 transgenic mouse results (which correspond to familial ALS) can be generalized in the majority of patients with sporadic ALS , the variations in progression and life span depending on the particular transgenic mouse model used, and the time and costs involved in careful animal testing. High hopes for clinical trials Demanding too much effect size? Modest or unequivocal benefit? In the past, some trialists thought that not many patients were needed to test drug efficacy in ALS. If a drug showed unequivocal benefit (something like wheelchair-bound patients starting walking again), we would know this immediately. In reality, it is possible that some effective drugs will have only a modest benefit that can be DOI: 10.1080/17434470410019942 ALS and other motor neuron disorders 2004 5(Suppl 1), 8–13 # 2004 ALS and other motor neuron disorders. All rights reserved. ISSN 1466-0822 8 Introduction Amyotroph Lateral Scler Downloaded from informahealthcare.com by CDL-UC Davis on 10/26/14 For personal use only.

Randomized control trials in ALS: lessons learned

  • Upload
    lewis-p

  • View
    212

  • Download
    0

Embed Size (px)

Citation preview

Randomized control trials in ALS: lessons learned

Hiroshi Mitsumoto, Paul Gordon, Petra Kaufmann, Clifton Gooch, Serge Przedborski and Lewis P RowlandThe Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center

Introduction

As we stated in the Preface, a major impetus for organizing

the ALS Clinical Trial Meeting held in Tarrytown, New York,

was the overall great frustration we have had in treating ALS;

that is, since 1996 when riluzole received FDA approval, we

have not identified any new medications for ALS. Although

we have been prescribing riluzole, its efficacy is modest at

best. We desperately need more effective drugs. Although a

cure may come only after the cause of ALS is elucidated, we

do not have the luxury of waiting when we are confronted by

the progressive paralysis of our patients. Vigorous basic

research in ALS has offered new hypotheses and opportunities

to test novel drugs. In the past two decades, we have had

more than 25 randomized clinical trials (RCTs) in ALS

(Table 1). Although we surely have made great progress in

these trials, the collective experience is still less than that in

many other diseases. An important lesson is always to learn

from others and from the past. We have therefore decided to

provide here not a review but rather an overview of the

collective experience in past ALS RCTs, which hopefully will

serve as a useful Introduction to this supplement. The issues

that we raise are examined in detail by the other contributors

to the supplement.

Bench to clinic

Drug effects in animal models have not been reliablepredictors of human effects

A critical limitation of RCTs is that our experience in ALS

trials is not large enough. Because only riluzole has given

positive RCT results, we cannot determine what type of results

in preclinical studies in animal models will predict the results

in human trials. At present, positive results in animals are

rather misleading at best, and it is uncertain how much we

actually have relied on the preclinical results. For example,

the decision to test riluzole in humans was not based on

studies in any animal models, only on its known anti-

glutamate effects in in vitro studies. Riluzole was tested in

SOD transgenic mice (termed SOD1 mice in this article) only

after riluzole had already been approved (and also because

transgenic mice carrying the human familial ALS mutation

became generally available after the approval of riluzole). The

effect size was small in both the mice and humans.18,28

The SOD1 mouse is now considered to be the best animal

model for preclinical testing in ALS. Earlier, neurotrophic

factors were tested in other mouse models, such as the

wobbler, pmn, and mnd mice, with some positive results that

led to human trials of neurotrophic factors, including ciliary

neurotrophic factors (CNTF), brain-derived neurotrophic

factors (BDNF), and insulin-like growth factor-I (IGF-I).29

When a number of neurotrophic factors were tested in SOD1

mice, negative findings were not published and trials were

performed in spite of such findings. When most neurotrophic

factors available for testing showed no clinical benefit in

patients, trialists in retrospect felt that preclinical results in

these less acceptable animal models had led to the negative

results in human studies, and subsequently to the loss of

interest in neurotrophic factor therapy.29 Some major RCTs

have been done without any preliminary studies in animal

models, or were performed despite the fact that studies in

animal models were negative, as seen in the examples of

riluzole and xaliproden. Thus, it seems fair to state that

animal studies have not been a critical factor in the decision-

making for RCTs.

Another issue is the timing of drug treatment in SOD1

mice. Presymptomatic treatment may not be ideal because it

can be done only in those at risk for familial ALS, not in

those with sporadic ALS. Because survival in SOD1 mice is

usually less than 2 weeks after the onset of clinically

detectable disease, particularly in the most commonly used

mice (G93A SOD), almost all drug treatments have been

initiated long before symptoms appear. At least two studies,

which showed dramatic responses to therapeutic agents,

commenced treatment after disease onset in SOD1 mice.

Those studies include IGF-I gene transfer using adeno-

associated virus36 and a preliminary report with Mn-

porphyrin treatment (J. Crow et al; reported at the 14th

International ALS/MND Symposium, Milan 2003). Other

complicating issues in SOD1 mice include the question of

whether mutant SOD1 transgenic mouse results (which

correspond to familial ALS) can be generalized in the

majority of patients with sporadic ALS , the variations in

progression and life span depending on the particular

transgenic mouse model used, and the time and costs

involved in careful animal testing.

High hopes for clinical trials

Demanding too much effect size? Modest orunequivocal benefit?

In the past, some trialists thought that not many patients

were needed to test drug efficacy in ALS. If a drug showed

unequivocal benefit (something like wheelchair-bound

patients starting walking again), we would know this

immediately. In reality, it is possible that some effective

drugs will have only a modest benefit that can be

DOI: 10.1080/17434470410019942

ALS and other motor neuron disorders 2004 5(Suppl 1), 8–13# 2004 ALS and other motor neuron disorders. All rights reserved. ISSN 1466-0822 8

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.

demonstrated only through statistical analysis of a large RCT.

Also, if research funds are limited, one tends to look for a

large effect size (large difference in the primary outcome

between placebo and drug) when testing a new medication.

Many RCTs in ALS have aimed for at least a 30% difference

from placebo. Such a large effect helps to reduce study costs,

partly by decreasing the number of patients required.

Furthermore, this approach effectively eliminates drugs that

have no major impact on the disease; on the other hand, it

would miss drugs that have modest benefits – less than 30%.

With the growing belief that the benefits of any new

treatment might be modest, we may have to lower the

effect size in future trials. As oncologists have done in RCTs,

we need to change our bias for large effects, and instead look

for effects of perhaps only 10% to 20%. A smaller effect size

requires a larger number of enrolled patients; however, there

is a recent trend towards enrolling more patients in ALS RCTs,

so this may not be a problem.

While a large effect size was an unrealistic approach to

identifying drugs with modest benefits, it may be useful when

the trial objective is different. A Phase II trial with a large

effect size would allow rapid screening of potential drugs

using a relatively small number of patients. Drugs found to

have large effects would then have top priority for testing in

subsequent Phase III trials. Alternatively, trialists could change

their statistical approach, setting the null hypothesis to be that

the new drug is effective, which would control for the Type I

error of eliminating a potentially beneficial medication. This will

allow us to decrease the sample size while setting the desired

effect size at a reasonable level. In essence, effect size must be

determined based on the goal of the trial.

Small clinical trials

There have been a number of small negative trials, which are

reviewed elsewhere.31,32 For this overview, we have defined

small negative studies as those with negative results that were

potentially underpowered. Those small studies guided us in the

direction ALS trials took because of their negative results, but in

retrospect many of these trials may have had potential problems

with sample size. By the same token, some RCTs listed in Table 1

may also have had the same problem. The relatively small

sample sizes in these studies may have left them underpowered

to test for efficacy, suggesting potential Type II errors rather than

truly negative results for the tested medications.

Searching for reliable primaryendpoints

Survival is the gold standard, but defining death iscomplex

FDA approval of riluzole influenced subsequent ALS RCTs.

The riluzole trial used survival as the primary endpoint; and

Table 1

Previous randomized controlled trials in ALS

Name Date Single or Multi-center Location Reference

Transfer factor 1979 Single New York 1

Bovine gangliosides 1984 Single Boston 2Thyrotropin releasing hormone 1984-1986 Multi USA 3, 4, 5, 6

Cyclosporine 1988 Single Houston 7

Branched-chain amino acids (BCAA) 1993-1996 Multi Europe, USA 8, 9, 10

Lamotrigine 1993 Single Vancouver 11Riluzole (1) 1994 Multi European 12

Acetylcysteine 1995 Single Amsterdam 13

Nimodipine 1996 Multi WALS 14

Verapamil 1996 Multi WALS 15CNTF 1996 Multi ACTS 16

CNTF 1996 Multi CASG/WALS 17

Riluzole (2) 1996 Multi Intercontinental 18Gabapentin (1) 1996 Multi WALS 19

IGF-I (1) 1997 Multi USA 20

IGF-I (2) 1998 Multi European 21

Selegiline 1998 Two centers USA 22Xaliproden 1998 Multi Intercontinental –

BDNF (Subcutaneous) 1999 Multi USA 23

BDNF (High-dose subcutaneous) 1999 Multi USA –

BDNF (Intrathecal) 1999 Multi Intercontinental –Gabapentin (2) 2001 Multi WALS 24

RiluzolezVitamin E 2001 Multi France 25

DMzQuinidine 2002 Multi USA –Topiramate 2003 Multi NEALS 26

Creatine (1) 2003 Multi Holland 27

Creatine (2) 2003 Multi NEALS –

Abbreviations: IGF-I=Insulin-like growth factor-I; BDNF=brain-derived neurotrophic factor; CNTF=ciliary neurotrophic factor; DM=dextromethorphan; WALS=Western

ALS study group; ACTS=ALS CNTF Treatment Study; CASG=CNTF ALS Study Group; NEALS=North Eastern ALS Study Group; (1) or (2) in the first column indicates

the first or second RCT with the same medication; – indicates no publication in refereed journals but a report exists in abstract or chapter form.

Randomized control trials in ALS: lessons learned 9

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.

survival was defined as time to death or tracheostomy.

Because riluzole trials were positive and riluzole was

approved, survival became the primary endpoint in trials

that followed. However, trials of BDNF and xaliproden gave

negative results, and the difficulties engendered by using

survival as the primary endpoint began to become apparent.

The main issue in using survival as the primary endpoint

concerns how death is defined for the purpose of RCTs in

ALS. How to define survival has become more difficult

because defining death has become less clear with the advent

of life-extending measures such as non-invasive positive-

pressure ventilation (NIPPV). For example, should any use of

NIPPV be considered equivalent to death in an ALS trial or

should more extended use, perhaps 24 h a day for 7 days, be

considered the defining marker? Clearly, defining death is

crucial if survival is to be used as the primary endpoint in

RCTs. The same problem applies to other life-extending

measures. For example, the majority of tracheostomized

patients require a permanent ventilator at the time of

tracheostomy. However, occasionally patients undergo tra-

cheostomy without ventilator support for management of

excessive secretions, and on rare occasions, proactive

tracheostomy is performed. How is death defined in these

situations? Making the matter more complicated, a few

reports have suggested that percutaneous endoscopic gastro-

stomy (PEG) prolongs survival in patients with ALS. Finally,

although riluzole marginally increases survival, retrospective

studies suggest that the survival effects may extend life by

about two months more than originally indicated.33 These

findings raise concern about confounding effects of pro-

longed survival in patients taking riluzole plus the test

therapy during an RCT. Therefore, we must conclude that

survival is strongly influenced by many factors, including

standard of care and management.

Outcome measures other than survival

One US study of insulin-like growth factor-I (IGF-I) demon-

strated a significant delay in functional deterioration as

measured by the Appel ALS scale.20 IGF-I, however, was not

approved by the FDA because a companion European RCT

showed no benefit.21 The beneficial changes in vital capacity

also were found significant in patients treated with xaliproden

(V. Meininger reported at the International ALS/MDA

Symposium, Arhus, 2001). Forced vital capacity (FVC) also

modestly but significantly differed between a subset of

patients receiving BDNF and those receiving placebo in a

Phase II RCT of BDNF. A Phase II study of gabapentin also

revealed a marginal difference (P=0.057) in the slope of

maximum voluntary muscle contraction (MVIC), favoring the

study drug;19 however, this difference proved to be due to the

more rapid decline in the placebo group.24 RCTs of CNTF

(ACTS group) or topiramate demonstrated significant wor-

sening of MVIC and FVC in those who received the study

drugs.16,26 Although these data are the painful reality, they

strongly support the idea that various outcome measures

other than survival or death can be useful for detecting

changes over time. A leading expert on clinical measures in

neuromuscular diseases once expressed the somewhat cynical

view that when facing repeated negative results in clinical

trials, investigators tend to shift from using one measure to

another. Without any doubt we are facing clear difficulty in

selecting primary outcomes. Simplicity, practicality, and

economy may be some of the important determining factors

when choosing the primary measure. Reflecting these goals,

the ALS functional rating scale (ALSFRS) and revised ALSFRS

seem to be having increasingly broad acceptance for use as

primary outcome measures in clinical trials.34 These topics

will be discussed in the supplement. We predict that the

second gold standard will be determined by the next primary

outcome measure that shows positive results in future RCTs.

Difficulties in using quantitative measures

The issue of intra- and inter-rater variability has been a

concern ever since quantitative measures were first applied in

multicenter ALS studies.17 Certification for the evaluators and

continued training during an RCT are imperative. Also

troublesome is that data are more likely to be missing

when changes are measured quantitatively in ALS trials.

Patients become progressively more disabled during the trial

and find it increasingly difficult to visit the study center for

quantitative studies. Furthermore, when the measures require

the patient’s cooperation to move, position, and give

maximum effort, as seen with MVIC, the test itself becomes

laborious and tiring, increasing the likelihood that data will

not be collected. Incomplete data obviously decrease the

power of the study. In particular, when the change in a

measure over time is used as the outcome, missing data

severely affect the determination of the rate of progression of

disease, i.e., determination of slope. Moreover, how to impute

missing data is a critical issue that has yet to be resolved.

Improving study design

Determining drug dose levels

ALS trialists tend to use higher doses or maximally tolerated

doses of the test medication to identify clinical benefits. This

practice is intended to ensure that the drug maximally

penetrates the central nervous system and to prevent trial

failure because the dose is deemed ‘‘insufficient’’. For

example, in a gabapentin trial, the dose was increased from

Phase II to Phase III,19,24 and in a topiramate trial, a

maximally tolerated dose was used.26 Other dose levels were

not included in either RCT. Multiple dose levels would be

prohibitively expensive, and investigator-initiated RCTs tend

to use a single dose level that would give the highest chance

of benefit. On the other hand, high doses may increase the

chance of intolerable side- effects or worsening of ALS, as

seen in trials of CNTF, topiramate, and gabapentin.16,24,26

Therefore, higher doses are not necessarily better than lower

doses, and multiple dose levels should be considered in RCTs.

Concomitant use of riluzole in RCTs

Although there is no evidence at this point, without doubt

almost all ALS experts and patient advocates suspect that ALS

10 H. Mitsumoto et al

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.

must be treated with a combination of multiple effective

drugs in the future, as seen in any other difficult diseases

which are usually treated with multiple medications. The next

logical assumption, although it might sound naıve, is that

adding riluzole is intuitively a ‘‘good thing’’. In fact, several

years ago, after a US IGF-I trial showed benefit, investigators

designed a combination trial of riluzole and IGF-I. This plan

was aborted when the FDA did not approve IGF-I, but the

combination was appropriate at that time. An ideal

combination trial with riluzole was conducted for the RCT

of xaliproden (Table 1). There were two protocols: one in

which patients received a high or low dose of xaliproden plus

riluzole, and one in which patients received xaliproden alone.

However, the trial results showed that administering riluzole

with another study drug might put patients at risk. A drop of

vital capacity to less than 50% was seen more often in those

who took both the higher xaliproden dose and riluzole than

in those who took xaliproden alone (data reported by V.

Meininger et al. at the International ALS/MND Symposium,

Arhus, 2001). In a wobbler mouse study, disease progressed

more rapidly in mice receiving both BDNF and riluzole than

in those receiving either drug alone (T. Ishiyama, unpub-

lished observation 2003). Preclinical studies of combined

treatment with riluzole and the study drug would seem

essential. Inclusion of riluzole without careful preclinical and

clinical consideration may be risky because drug interactions

in ALS are presently unknown.

Trial center and country effects may jeopardize studyresults

The use of NIPPV, PEG, and riluzole varies from center to

center.35-37 When the number of patients enrolled in a study

is not balanced among study centers in a multicenter RCT,

this imbalance may affect study results. In the Phase III

riluzole study, there was a large imbalance in patient

enrollment in different centers. Although the hypothesis is

totally speculative, such imbalance might explain an unusual

result of that trial:18 the Kaplan-Meier survival curve for

European patients receiving riluzole was identical to that of

the North American patients receiving placebo. What we

describe here may have nothing to do with center or country

effects, but it is suggestive of such a problem. In conducting

an ideal RCT, unbalanced enrollment between centers or

regions must be avoided lest it confound the results.

The ‘natural history’ of the placebo group isuncertain

Munsat 38,39 advocated using placebo patient groups from

earlier RCTs, and such historical controls would be sufficient

if an ALS treatment showed unequivocal benefit. Trials using

historical controls might be useful for rapid screening of

multiple new drugs, or to decrease study costs. However, this

design has not been accepted, because the ‘natural history’ in

the placebo arm may not be truly natural because it is

modified by placebo effects 2-4 and selection bias according to

inclusion and exclusion criteria. Furthermore, the natural

histories of control groups in past RCTs differ from one

another.19,22,24 As a result, there is now little support for trials

without placebo controls. As clinical trial experience

increases, however, the use of historical placebo controls

may again seem reasonable.

Another impediment to designing trials with historical

controls is the continuing paucity of historical control data.

Placebo data are not available for every ALS RCT. For

instance, in industry-sponsored trials some data were

considered proprietary and not released to the public. In

our experience, the leaders of the RCTs left the project or firm,

which then lost contact with them, and access to data is now

impossible. In the future, regardless of whether studies are

industry- or investigator-initiated, all placebo data must be

the property of the ALS community. The WALS study group is

providing its control data to other ALS trialists upon request,

and the NEALS group (Dr. Merit Cudkowicz, personal

communication, 2003) has initiated a program to store all

placebo data in an NINDS control databank. In other

neurological disorders, such as multiple sclerosis, virtual control

groups are created by entering all RTC data into a large,

multinational database [http://www.nationalmssociety.org].

In ALS, developing an organized system for storing and

accessing all placebo data is essential to improving ALS

research and RCT design.

Lessons about data and safetymonitoring

Patient safety and data management

An effective Data and Safety Management Program (DSMP) is

key to protecting the study patients from avoidable serious

adverse effects. To ensure rapid and effective triage and

decision-making systems, effective and accurate data manage-

ment and administrative systems are required. Investigator-

initiated studies in ALS are usually constrained by costs. Data

management may suffer most because sophisticated data

management is costly and is therefore a greater obstacle in

investigator-initiated studies than in industry-supported trials.

In the past decade, the NIH has established increasingly

stringent DSMP guidelines so that the DSMPs for NIH-funded

RCTs are now similar to those of industry-designed RCTs.

Well-planned DSMP and data management are imperative for

reliable RCTs.

Lessons for the investigators

Providing open-label medication after a RCT

Patients are often willing to participate in a RCT because they

are desperate and want to take the active drug. If a study does

not guarantee access to the test drug for all participants in an

‘open-label’ component after the formal trial, patients may be

discouraged from enrolling, and the dropout rate may

increase if patients suspect they have been given the placebo.

Access to the test drug is obviously one of the important

benefits and motivations for those who courageously

participate in prolonged and arduous trials. In fact, providing

Randomized control trials in ALS: lessons learned 11

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.

the active drug to all the patients in an RCT is one of the WFN

guidelines for RCTs.40

However, conducting the open-label phase after a RCT may

not be simple. The investigators must be certain that the agent

is safe. The open-label phase often is considered as merely an

extension of the blind phase, particularly when the agent is

an off-label drug that has been used in other disorders long

enough to guarantee safety. Consequently, safety might be

taken for granted because it already has been proven, and

participating investigators may have experience with the drug

in other diseases. Patients with ALS, however, may have a

different response to off-label drugs, because of 1. impending

alveolar hypoventilation caused by respiratory muscle weak-

ness, 2. dehydration due to dysphagia, and 3. weakened

immune responses because of malnutrition. Thus, off-label

drugs might act more adversely in patients with ALS than in

other diseases. For example, adverse events resulted in

discontinuation of the open-label phase of the topiramate

trial for ALS.26,41 ALS investigators will be studying more off-

label drugs and will need to pay strict attention to adverse

effects, particularly in patients with rapidly deteriorating

motor neuron function.

Publishing the results

The publication of RCT results, positive or negative, is the

investigators’ responsibility. The pharmaceutical industry has

been criticized for not publishing negative results, and, for

investigators, the experience can be frustrating if the sponsor

loses interest or declines to publish. Negative ALS study

results that have not yet been published include the second

BNDF high-dose subcutaneous study and the BNDF intra-

thecal study. We have been told that manuscripts for the

xaliproden studies are in preparation.

Some industry sponsors have published the results of

negative studies; for example, the IGF-I USA and European

studies, two CNTF studied by the CNTF Study Group and the

ALS CNTF Treatment Study group, and BDNF Study Group

were industry-initiated and had negative results. The inves-

tigators themselves may have been the driving force for

publication of these trials. Negative investigator-driven studies

may also go unreported or be reported in unrefereed journals,

such as a subcutaneous thyrotropin-releasing hormone

multicenter study and the Pan European BCAA study.

However, these studies were done years ago, and perhaps

the importance of publishing negative study results in ALS

was not realized. Journal editors are also likely to resist using

precious pages for the publication of negative results.

In planning a new RCT, the publication plan should be

clearly defined. Without an explicit plan to publish the

results, positive or negative, the RCT should not be

implemented. The development of RCTs for ALS is still in

the early stages, and we must learn as much as possible from

each RCT. Publication is imperative if we are to develop

effective and efficient RCTs for discovering useful ALS

treatments.

Conclusions

In this introduction we have described what we believe we

have learned from past RCTs for ALS. We hope that our

comments will generate discussion and even argument. Every

past RCT in ALS has provided opportunities to learn about

the issues involved in conducting trials. The experience and

the knowledge so generated provide the basis for developing

more effective, more efficient, and more innovative study

designs. We hope this introductory overview will serve as

background for the in-depth discussions, new ideas, opinions,

and critical suggestions that follow in this supplement. We

believe that ALS clinical trials have reached a decisive point

because of the use of diverse outcome measures, study

designs, available drugs, the number of ALS study groups, and

funding opportunities. Moreover, the ALS community is

beginning to work more cohesively. We have come a long

way – RCTs in ALS have matured to a level likely to assure the

proper evaluation of new drugs that can be approved by the

FDA for the treatment of ALS. Much, however, remains to be

accomplished, and it is up to us, particularly the young

investigators, to ensure that future trials are ideally designed,

implemented, analyzed, and published.

Acknowledgements

This publication is in part supported by the MDA Wings Over

Wall Street Fund. Cassandra Talerico-Kaplin provided editor-

ial assistance.

References

1. Olarte MR, Gersten JC, Zabriskie J, Rowland LP. Transfer factor

is ineffective in amyotrophic lateral sclerosis. Ann Neurol 1979;

5(4): 385–388.

2. Bradley WG, Hedlund W, Cooper C et al. A double-

blind controlled trial of bovine brain gangliosides in

amyotrophic lateral sclerosis. Neurology 1984; 34: 1079–1082.

3. Brooke MH, Florence JM, Heller SL et al. Controlled

trial of thyrotropin releasing hormone in amyotrophic lateral

sclerosis. Neurology 1986; 36: 146–151.

4. Mitsumoto H, Salgado ED, Negroski D et al. Amyotrophic

lateral sclerosis: effects of acute intravenous and chronic

subcutaneous administration of thyrotropin-releasing

hormone in controlled trials. Neurology 1986; 36: 152–159.

5. Brooks BR, Sufit RL, Montgomery GK, Beaulieu DA, Erickson LM.

Intravenous thyrotropin releasing hormone in patients with

amyotrophic lateral sclerosis: dose response and

randomized concurrent placebo controlled pilot studies.

Neurol Clin 1987; 5: 143–158.

12 H. Mitsumoto et al

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.

6. The National TRH Study Group. Multicenter controlled trial:

no effect of alternate-day 5mg/kg subcutaneous thyrotropin-

releasing hormone (TRH) on isometric strength decrease in

amyotrophic lateral sclerosis [abstract]. Neurology 1989; 39

(suppl): 322.

7. Appel SH, Stewart SS, Appel V et al. A double-blind study of the

effectiveness of cyclosporine in amyotrophic lateral sclerosis.

Arch Neurol 1988; 45(4): 381–386.

8. Steiner TJ. Motor Neurone Disease: Experience with Large

Multicenter Trials. In: Guiloff RJ, ed. Clinical Trials in Neurology.

London: Springer-Verlag, 2001: 451–463.

9. The Italian ALS Study Group. Branched-chain amino acids and

amyotrophic lateral sclerosis: a treatment failure? Neurology

1993; 43: 2466–2470.

10. Tandan R, Bromberg MB, Forshew D et al. A controlled trial of

amino acid therapy in amyotrophic lateral sclerosis: I. Clinical,

functional, and maximum isometric torque data. Neurology

1996; 47: 1220–1226.

11. Eisen A, Stewart H, Schulzer M, Cameron D. Anti-glutamate

therapy in amyotrophic lateral sclerosis: a trial using lamotrigine.

Can J Neurol Sci 1993; 20: 297–301.

12. Bensimon G, Lacomblez L, Meininger V. A controlled trial of

riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study

Group. N Engl J Med 1994; 330: 585–591.

13. Louwerse ES, Weverling GJ, Bossuyt P et al. Randomized,

double-blind, controlled trial of acetylcysteine in amyotrophic

lateral sclerosis. Arch Neurol 1995; 52: 559–564.

14. Miller RG, Shepherd R, Dao H et al. Controlled trial of

nimodipine in amyotrophic lateral sclerosis. Neuromuscul

Disord 1996 6; 20: 101–104.

15. Miller RG, Smith SA, Murphy JR et al. A clinical trial of verapamil

in amyotrophic lateral sclerosis.MuscleNerve 1996; 19: 511–551.

16. The ALS CNTF Treatment Study (ACTS) Phase III Study Group.

A double-blind placebo controlled trial of subcutaneous recom-

binant human ciliary neurotrophic factor (rHCNTF) in amyo-

trophic lateral sclerosis. Neurology 1996; 46: 1244–1249.

17. Miller RG, Petajan JH, Bryan WW et al. A placebo controlled trial

of recombinant human ciliary neurotrophic (rhCNTF) factor in

amyotrophic lateral sclerosis. Ann Neurol 1996; 39: 256–260.

18. Lacomblez L, Bensimon G, Leigh PN et al. Dose-ranging study of

riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral

Sclerosis/Riluzole Study Group II. Lancet 1996; 347: 1425–1431.

19. Miller RG, Moore D, Young LA et al. Placebo controlled trial of

gabapentin in patients with amyotrophic lateral sclerosis.

Neurology 1996; 47: 1383–1388.

20. Lai EC, Felice KJ, Festoff BW et al. Effect of recombinant human

insulin-like growth factor-I on progression of ALS: a placebo

controlled study. The North America ALS/IGF-I Study Group.

Neurology 1997; 49: 1621–1630.

21. Borasio GD, Robberecht W, Leigh PN et al. A placebo controlled

trial of insulin-like growth factor-I in amyotrophic lateral

sclerosis. European ALS/IGF-I Study Group. Neurology 1998;

51: 583–586.

22. Lange DJ, Murphy PL, Diamond B et al. Selegiline is ineffective

in a collaborative double-blind controlled trial for treatment of

amyotrophic lateral sclerosis. Arch Neurol 1998; 55: 93–96.

23. BDNF Study Group. A controlled trial of recombinant methionyl

human BDNF in ALS. The BDNF Study Group (Phase III).

Neurology 1999; 52: 1427–1433.

24. Miller RG, Moore DH Jr, Gelinas DF et al. Phase III randomized

trial of gabapentin in patients with amyotrophic lateral sclerosis.

Neurology 2001; 56: 843–848.

25. Desnuelle C, Dib M, Garrel C, Favier A. A double-blind, placebo

controlled randomized clinical trial of alpha-tocopherol (vitamin

E) in the treatment of amyotrophic lateral sclerosis. ALS riluzole-

tocopherol Study Group. Amyotroph Lateral Scler Other Motor

Neuron Disord 2001; 2: 9–18.

26. Cudkowicz ME, Shefner JM, Schoenfeld DA et al. A randomized,

placebo controlled trial of topiramate in amyotrophic lateral

sclerosis. Neurology 2003; 61: 456–464.

27. Groeneveld GJ, Veldink JH, van der Tweel I et al. A randomized

sequential trial of creatine in amyotrophic lateral sclerosis. Ann

Neurol 2003; 53: 437–445.

28. Gurney ME, Fleck TJ, Himes CS, Hall ED. Riluzole preserves

motor function in a transgenic model of familial amyotrophic

lateral sclerosis. Neurology 1998; 50: 62–66.

29. Mitsumoto H, Tsuzaka K. Neurotrophic factors and neuro-

muscular diseases. Part I and Part II. Muscle Nerve 1999; 22:

983–999 (I) and 1000–1021 (II).

30. Kaspar BK, Llado J, Sherkat N, Rothstein JD, Gage FH. Retrograde

viral delivery of IGF-1 prolongs survival in a mouse ALS model.

Science 2003; 301: 839–342.

31. Murray B, Mitsumoto H. Drug therapy in amyotrophic lateral

sclerosis. In: Pourmand R, Harati Y, eds. Neuromuscular

Disorders. New York: Lippincott, Williams and Wilkins.

Advances in Neurology 2002; 88 63–82.

32. Rowland LP, Schneider NA. Medical progress: amyotrophic

lateral sclerosis. N Engl J Med 2001; 344: 1688–1700.

33. Traynor BJ, Alexander M, Corr B et al. Riluzole and prognosis in

ALS: findings of the Irish ALS registry from 1995–2000. ALS and

other Motor Neuron Diseases 2001; 2 (Suppl 2): 43–44.

34. The ALS CNTF Treatment Study (ACTS) Phase I-II Study Group.

The Amyotrophic Lateral Sclerosis Functional Rating Scale.

Assessment of activities of daily living in patients with

amyotrophic lateral sclerosis. Arch Neurol 1996; 53: 141–147.

35. Bryan WW, McIntire D, Camperlengo L et al. Factors influencing

the use of riluzole by ALS patients. 8th International Symposium

on ALS/MND. November1997(abstract).Published in J Neurol Sc.

36. Cedarbaum JM, Stambler N. Disease status and use of ventilatory

support by ALS patients. BDNF Study Group. Amyotroph Lateral

Scler Other Motor Neuron Disord 2001; 2: 19–22.

37. Mitsumoto H, Davidson M, Moore DH et al. Percutaneous

endoscopic gastrostomy (PEG) in patients with ALS and bulbar

dysfunction. Amyotrophic Lateral Scler Other Motor Neuron

Disord 2003; 4: 177–185.

38. Munsat TL, Andres PL, Finison L, Conlon T, Thibodeaur L. The

natural history of motor neuron loss in amyotrophic lateral

sclerosis. Neurology 1988; 38: 409–413.

39. Munsat TL, Andres P, Skerry L. Therapeutic trials in amyotrophic

lateral sclerosis: measurement of clinical deficit. In: Rose FC, ed.

Amyotrophic Lateral Sclerosis. New York: Demos, 1990: 65–76.

40. The World Federation of Neurology Research Group on

Neuromuscular Diseases, Subcommittee on Motor Neuron

Disease: Airlie House Guidelines. Therapeutic trials in amyo-

trophic lateral sclerosis. J Neurol Sci 1995; 129 (suppl): 1–10.

41. Kaufmann P, Lomen-Hoerth C. ALS treatment strikes out

while trying for a homer:the topiramate trial. Neurology 2003;

61: 434–435.

Randomized control trials in ALS: lessons learned 13

Introduction

Am

yotr

oph

Lat

eral

Scl

er D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y C

DL

-UC

Dav

is o

n 10

/26/

14Fo

r pe

rson

al u

se o

nly.