You are on page 1of 12

CME

Movement Disorders
Vol. 23, Suppl. 3, 2008, pp. S497–S508
Ó 2008 Movement Disorder Society

The History of Dopamine and Levodopa in the


Treatment of Parkinson’s Disease

Stanley Fahn, MD

Columbia University, New York, USA

Abstract: The discoveries of dopamine as a neurotransmitter went beyond a single clinical entity of parkinsonism, for it
in the brain, its depletion in patients with Parkinson disease, opened up the beginning of a much better understanding of
and its replacement with levodopa therapy were major revo- the role of dopamine in other neurologic movement disorders
lutionary events in the rise to effective therapy for patients and also in many psychiatric diseases. Ó 2008 Movement
with this disorder. This review describes these events and the Disorder Society
persons who carried out these accomplishments. Their impact Key words: levodopa; history

The introduction of high dosage levodopa by George written extensively on the early history of dopamine
Cotzias1,2 revolutionized the treatment of Parkinson’s and L-dopa, and some of the material in this review
disease (PD), and L-dopa remains until this day the draws upon his excellent articles.6,7
most efficacious treatment for patients with PD. In
addition, L-dopa therapy had a major impact on
THE BIOCHEMISTRY OF DOPAMINE
neurology, in general. (1) With the ability to improve
functional disability in a neurodegenerative disorder, L-dopa is an abbreviation of L-3,4-dihydroxyphenylal-

neurology became an attractive field for young medical anine. The decarboxylated amine 3,4-dihydroxyphenyle-
students. (2) Besides adding knowledge to the patho- thylamine (also known as 3-hydroxytyramine) has been
physiology of parkinsonism, L-dopa provided clues abbreviated to dopamine (see Fig. 1). Today, we know
about the nature of chorea and dystonia. (3) The that the synthesis of dopamine proceeds from L-tyrosine
response and adverse effects associated with L-dopa to L-dopa to dopamine (see Fig. 1).
treatment contributed to a better understanding to the According to Hornykiewicz,6 dopamine was first
organization of the basal ganglia.3–5 (4) The dramatic synthesized by George Barger and James Ewens in
benefit of L-dopa in PD attracted neurologists to study 1910 at the Wellcome labs in London, England. Subse-
this disorder, which in turned spawned the subspecialty quently, also at the Wellcome labs, Henry Dale (who
of movement disorders and the formation of The later became Sir Henry) found it to be a weak sympa-
Movement Disorder Society. thomimetic, epinephrine-like compound,8 and years
The impact of L-dopa in the treatment of PD and in later, in 1952, it was Dale who suggested its current
translational neuroscience cannot be overestimated, and name of dopamine. D,L-dopa was synthesized chemi-
therefore, the history of how L-dopa came about is cally by Casimir Funk in 1911.9 Two years later, Mar-
revealing and instructive as we move forward looking cus Guggenheim at the Roche laboratories in Switzer-
for even better treatments. Oleh Hornykiewicz has land isolated L-dopa from fava beans (Vicia faba).10
He ingested 2.5 g of L-dopa and discovered its emetic
Correspondence to: Stanley Fahn, MD, Neurological Institute, 710 effect; otherwise he found it essentially without phar-
West 168th Street, New York 10032.
E-mail: fahn@neuro.columbia.edu macologic activity (see Hornykiewicz7).
Potential conflict of interest: Nothing to report. Little work was done on dopamine for the next 30
Received 31 December 2007; Revised 28 January 2008; Accepted years, until Peter Holtz et al.11 in Germany discovered
18 February 2008
Published online in Wiley InterScience (www.interscience.wiley. the enzyme aromatic-L-amino-acid decarboxylase (also
com). DOI: 10.1002/mds.22028 called dopa decarboxylase), which, in mammalian tis-

S497
S498 S. FAHN

FIG. 1. Synthesis of levodopa (from its precursor L-tyrosine) and dopamine (from its precursor levodopa). In parentheses are the names of the
cofactors involved in the syntheses next to the enzymes each interacts with.

sues, converts L-dopa to dopamine (see Fig. 1). This sine to L-dopa is present in skin and other pigmented
discovery provided a mechanism for the formation of tissues, but not in sympathetic tissue or in brain. In the
dopamine in the brain from an exogenous source, since early 1960s, Toshiharu Nagatsu from Japan was a
L-dopa, unlike dopamine itself, can cross the blood postdoctoral fellow in Sidney Udenfriend’s laboratory
brain barrier. Shortly after the discovery of dopa decar- at the National Heart Institute, a component of the
boxylase, both Holtz12 and, independently, Hermann National Institutes of Health (NIH). Udenfriend’s lab
Blaschko13 at the physiological laboratories in Cam- was engaged in catecholamine biochemistry, especially
bridge proposed that L-dopa and dopamine were inter- as it relates to sympathetic tissue. Nagatsu et al.16 dis-
mediate metabolites in the biosynthetic pathway of the covered the enzyme tyrosine hydroxylase that converts
catecholamines, epinephrine, and norepinephrine (see L-tyrosine to L-dopa. This turned out to be a very
Fig. 2). This hypothesis was later shown to be correct, important discovery, since tyrosine hydroxylase is the
although it still failed to explain completely the physi- rate-limiting enzyme in forming L-dopa, dopamine, and
ologic significance of dopamine. Over the next decade indeed, all of the catecholamines. Nagatsu subse-
or two, Peter Holtz and others continued to study do- quently returned to Japan and continued his studies of
pamine and found that it could lower blood pressure in this pathway. In 1994, his lab identified the gene defect
animals and that it could be detected in many periph- responsible for dopa-responsive dystonia (DRD), also
eral tissues, including the adrenal medulla, heart, and known as Segawa’s disease or hereditary progressive
kidney.14 dystonia with diurnal fluctuations, a rare type of dysto-
In 1956, Oleh Hornykiewicz began working in nia that is responsive to L-dopa therapy. The gene re-
Blaschko’s laboratory, now located in Oxford, and was sponsible for this disease encodes for GTP cyclohydro-
assigned the task of determining whether blood pres- lase 1 (GCH1), which is the rate-limiting enzyme for
sure was directly affected by dopamine, or if some the biosynthesis of tetrahydrobiopterin, a cofactor for
other product of dopamine was responsible. Hornykie- the formation of tyrosine hydroxylase and other aro-
wicz showed that, indeed, dopamine did lower blood matic amino-acid hydroxylases.17 Molecular genetics
pressure and that L-dopa had a similar effect.15 research by Nygaard et al.18 had mapped the DRD
It was not known until later how L-dopa was gene to an area on chromosome 14q a few years ear-
formed; the enzyme tyrosinase which can convert tyro- lier, but the Nagatsu lab which had been studying

Movement Disorders, Vol. 23, Suppl. 3, 2008


DOPAMINE AND LEVODOPA IN THE TREATMENT OF PD S499

FIG. 2. Biochemical pharmacology of dopamine. The diagram shows the synthesis and catabolism of dopamine, along with the enzymes
involved. L-dopa and dopamine are precursors to norepinephrine, which in turn could be further metabolized to epinephrine (not shown). COMT
catalyzes the 3-O-methylation of levodopa (not shown) as well as dopamine (and other catechols). MAO deaminates and oxidizes the mono-
amines, dopamine, norepinephrine, epinephrine, and serotonin (only the dopamine pathway is shown). MAO, monoamine oxidase; COMT, cate-
chol-O-methyltransferase.

GCH1, was able to make the gene identification for cholamine, that was responsible for lowering blood
this autosomal dominant disorder. pressure and for the akinetic effect of reserpine. In 1957
he observed that norepinephrine and epinephrine dis-
appeared more or less completely from tissues, includ-
DOPAMINE IN THE BRAIN ing brain, after reserpine treatment,20 the same effect
As progress was being made throughout the 1950s that Brodie and coworkers had shown for serotonin.
and 1960s on defining the mechanisms involved in do- After returning to Sweden and establishing his own
pamine biosynthesis, physiologists and pharmacologists laboratory at the University of Lund, Carlsson showed
were making headway in understanding the physiologi- that D,L-dopa, not 5-hydroxytryptophan (the precursor
cal actions of various intermediates of the catechol- of serotonin), reversed the akinetic effect of reserpine
amine pathway. Arvid Carlsson, then a postdoctoral in rabbits.21 In dramatic fashion, reserpinized rabbits,
fellow in Bernard Brodie’s cardiac pharmacology lab after receiving D,L-dopa, went from being totally un-
at NIH, was investigating the mechanism whereby re- responsive to alert, with their drooping ears popping
serpine lowers blood pressure and slows heart rate. up almost immediately (see Fig. 3). The action of
Brodie’s lab had shown that reserpine lowered sero- DOPA was markedly potentiated by monoamine oxi-
tonin levels19 and had proposed that loss of serotonin dase (MAO) inhibitors, emphasizing that the observed
was responsible for reserpine’s cardiovascular effect effect was not caused by DOPA itself but rather by
and also for its neurologic effect of ‘‘tranquilisation’’ amines formed from it. This discovery really put dopa
(now recognized as akinesia). Carlsson, however, won- and catecholamines on the map, at least in regard to
dered if perhaps it was not serotonin, but rather a cate- reserpine’s akinetic effect, but which catecholamine

Movement Disorders, Vol. 23, Suppl. 3, 2008


S500 S. FAHN

FIG. 3. Response of reserpinized akinetic rabbits to D,L-dopa. Top: Reserpine-treated rabbits become akinetic and immobile (note the floppy
ears). After treatment with D,L-dopa, the rabbits perked up and became mobile, recovering from akinesia (note the erect ears). From Carlsson A,
Lindqvist M, Magnusson T. The 3,4-dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 1957;180:1200.

was responsible for the effect was not known at that of dopamine in dog brain, demonstrating that it was
time. Carlsson et al. were unable to detect any increase concentrated in the basal ganglia,27 quite distinct from
of norepinephrine or epinephrine in brain when DOPA the regional distribution of norepinephrine and epi-
was given to reserpinized animals, and wondered about nephrine; and this was simultaneously shown in human
dopamine, which had been assumed to be only an in- brain by another one of Carlsson’s collaborators, Isamu
termediate in the formation of norepinephrine and epi- Sano.28 All of these findings led Carlsson to speculate,
nephrine. The reversal of the akinetic effect of reser- at the 1959 international pharmacology meeting, that
pine in humans by L-dopa was reported in 1960 by dopamine was somehow responsible for PD.29 Carls-
Degkwitz and coworkers.22 son, of course, went on to win the 2000 Nobel Prize in
At about this time, Kathleen Montagu23 in Hans Physiology or Medicine for his work on dopamine,
Weil-Malherbe’s laboratory near London, along with along with Paul Greengard and Eric Kandel, for their
Weil-Malherbe and Bone,24 showed, for the first time, work on signal transduction in the nervous system and
that dopamine was present in the brain. Actually, 6 the molecular basis of memory, respectively.
years earlier, Raab and Gigee25 had discovered a cate-
cholamine-like compound in the brain with a regional
distribution in the striatum and that increased after DOPAMINE AND PARKINSON’S DISEASE
dopa administration; today their unknown brain sub- Hornykiewicz, back in Vienna from being a post-
stance is considered to have been dopamine.6,7 doc in Blaschco’s lab in Oxford, began to measure
Carlsson et al., meanwhile, developed a new chemi- postmortem brain dopamine in people with PD, posten-
cal fluorescent assay technique to measure dopamine in cephalitic parkinsonism, Huntington’s disease and in
tissue, for there was previously no method to measure other extrapyramidal disorders, as well as in control
the microgram quantities suspected. He then showed brains, and in 1960, published a landmark paper show-
that not only was it present in the brain, but that it was ing for the first time a marked depletion of dopamine
depleted with reserpine and restored with L-dopa.26 in the caudate and putamen of patients only in the PD
Working in Carlsson’s lab, two medical students, Åke and postencephalitic parkinsonian brains.30 Six years
Bertler and Evald Rosengren, mapped the distribution later, following several other of his seminal papers on

Movement Disorders, Vol. 23, Suppl. 3, 2008


DOPAMINE AND LEVODOPA IN THE TREATMENT OF PD S501

postmortem biochemistry of PD, Hornykiewicz pub- offering him about 2 g of L-dopa he had kept from
lished a major review article in Pharmacological Blaschko’s lab to test in PD patients. Intravenous
Reviews positing that striatal dopamine deficiency is injections of the L-dopa produced dramatic, but tran-
correlated with most of the motor symptoms of PD.31 sient, benefit in these patients, heralding the beginning
Over the next 5 years or so, using Nils-Ake Hillarp’s of the era of L-dopa therapy.35 Over the next several
technique of histofluorescence, Nils-Eric Andén, Ann- years, some investigators throughout the world showed
ica Dahlström, and Kjell Fuxe in Sweden mapped similar benefits using higher doses of L-dopa,36–41
dopamine pathways in the brain of rodents and dis- while other investigators reported no benefits with sim-
covered the nigrostriatal pathway that goes from cell ilar doses.42–45 The first person to use really high doses
bodies of the A9 neurons in the midbrain to their nerve of DOPA was Patrick McGeer in Vancouver, British
terminals in the striatum.32 At about the same time, Columbia. Even with 5 g of D,L-dopa per day, McGeer
Louis Poirier and Ted Sourkes in Montreal were meas- failed to show a benefit.46
uring dopamine in the nigra of animals and showed Then in 1967, the major breakthrough occurred. The
that striatal dopamine levels fall when the nigra is dose-limiting side effect of anorexia, nausea, and vom-
injured, thus tying together the nigrostriatal pathway iting had previously impeded using higher dosages of
anatomically and pathologically.33 Thus, there was ex- D,L-dopa. George Cotzias developed a new approach,
perimental evidence of dopamine in the nigrostriatal which in principle is still used today. He started
neurons and their depletion in the striatal target follow- patients on a very low dose of D,L-dopa to avoid the
ing a lesion in the cell bodies in the substantia nigra, gastrointestinal adverse effects, and then raised the
the known pathologic feature of human PD. dose very gradually, eventually giving up to 16 g per
In the brain, the enzymes involved in catecholamine day.1 Patients with PD tolerated this very slow, gradual
synthesis are transported from the cell body in the build-up of dosage and then began to achieve dramatic,
nigra into nerve terminals, where dopamine is formed. revolutionary benefit as therapeutic doses were
Dopamine is synthesized in the cytosol and transported reached. They reported marked improvement in 8
into synaptic vesicles via the vesicular monoamine patients and less improvement in two others. Of the 8
transporter 2 (VMAT2). From the synaptic vesicles, who received 12 g or more per day, 7 showed marked
dopamine is released in a quantal manner when a benefit. Granulocytopenia was seen in 4 patients, and
nerve impulse traveling down the axon reaches the bone marrow examination revealed vacuoles in the my-
nerve terminal. Once in the synaptic cleft, dopamine eloid cells in 4 of the 12 patients with bone marrow
can act on both postsynaptic and presynaptic dopamine examinations. Because of the hematologic problems
receptors. If there is excessive dopamine accumulation and because D-dopa is not metabolized to form
in nerve terminal cytoplasm or if dopamine transport dopamine, Cotzias et al. subsequently used L-dopa,2
into the synaptic vesicles is blocked by inhibition of and these hematologic problems were no longer
VMAT2 (a target of both reserpine and tetrabenazine), encountered.
elevated cytosolic dopamine levels results. This cyto- Interestingly, Cotzias initially was not trying to use
solic dopamine can be auto-oxidized into toxic pro- D,L-dopa to restore brain dopamine levels, but was try-
ducts that could lead to neurodegeneration; the neuron ing to repigment the brain, i.e. restore loss of neurome-
defends itself by condensing cytosolic dopamine into lanin.1 He apparently was not impressed with the con-
neuromelanin34 or metabolizing the dopamine by cept that parkinsonism could be due to low striatal do-
MAO and catechol-O-methyltransferase (COMT) to pamine. Before trying D,L-dopa, he tried phenylalanine,
form homovanillic acid. The latter mechanism accounts the precursor of tyrosine, in an attempt to increase neu-
for the depletion of brain catecholamines following re- romelanin, but parkinsonism worsened on phenylala-
serpine or tetrabenazine treatment. nine,1 probably reflecting competition of tyrosine’s
entry into brain due to oversaturation of the aromatic
amino-acid transport mechanism. With the success of
THE ERA OF L-DOPA THERAPY IS BORN high dosage D,L-dopa, Cotzias accepted the concept
Even before the nigrostriatal dopamine pathway had that the drug was restoring brain dopamine. It turns out
been mapped by histofluorescence, Oleh Hornykiewicz with subsequent investigations that the brain’s neuro-
in 1960, after discovering striatal dopamine deficiency melanin is not restored with L-dopa therapy.47 While
in the brains of patients with PD, began to consider L- Cotzias was testing high dosage L-dopa following his
dopa as a possible treatment for PD. He approached breakthrough results with D,L-dopa, Yahr et al. was
Walther Birkmayer, a clinical neurologist in Vienna, conducting a double-blind trial with L-dopa and

Movement Disorders, Vol. 23, Suppl. 3, 2008


S502 S. FAHN

showed similar dramatic benefit,48 as that observed by Dyskinesias were not seen during the first 3 weeks of
Cotzias et al.2 treatment, but occurred later on.
Many subsequent reports showed significant im- The next paper reporting on the use of L-dopa was
provements in 75% of patients with parkinsonism. by Yahr et al.48 In their 60 patients, gastrointestinal
Although a complete remission was rarely obtained, adverse effects were encountered in 51, dyskinesias in
akinesia and rigidity were generally most benefited, 37, hypotension in 14, cardiac abnormalities in 13, and
and many who had been unable to turn in bed or arise psychiatric symptoms in 10. By 1970 McDowell
from a chair became able to do so. Tremor had a et al.49 and Schwarz and Fahn50 were reporting that
more variable response; sometimes it was eliminated dyskinesias were as common as gastrointestinal side
by L-dopa, and in other patients, the tremor was resist- effects. They noted that the gastrointestinal effects could
ant. A number of other symptoms, including postural often be avoided by building up the dose of L-dopa
instability and speech disturbance, were typically unaf- very slowly, and that often patients build up tolerance,
fected by L-dopa therapy, suggesting these resistant with the result that few patients would have persistent
symptoms are not due solely to dopamine deficiency. gastrointestinal difficulties. On the other hand, dyskine-
As seen in the early days of L-dopa therapy, this drug sias, although occurring later, would persist, and would
does not have an immediate antiparkinsonian effect, increase and become more prominent with continuing
and it may take several days to weeks of high dosage treatment. By 1971, dyskinesias were noted to be the
therapy to achieve the desired degree of benefit. Once most common dose-limiting adverse effect.51 The
a patient has been primed, though, then restarting L- abnormal movements were seen in all parts of the
dopa after a withdrawal period brings on the benefit body, and most often were choreic in nature.
almost immediately. The first review article on L-dopa-induced dyskine-
We can interpret the introduction of high dosage sias was presented by Duvoisin,52 based on an analysis
L-dopa therapy by Cotzias as a revolutionary treatment of L-dopa therapy in 116 patients with PD. He found
for PD, not just an evolutionary one, for it brought a that by 6 months of treatment, 53% of patients had
new way to treat the disease, which still dominates the developed dyskinesias; by 12 months, 81% had.
current therapy of the disorder. Subsequent to the early Although described earlier by Cotzias et al.2 myoclonic
investigator-initiated clinical trials, Hoffman-La Roche, jerks in patients with PD, especially as a toxic reaction
where L-dopa was first isolated, was able to synthesize to L-dopa was further elaborated by Klawans et al.53
the compound and initiated company-sponsored clinical Besides dyskinesias, treating physicians began to
trials, leading to regulatory approval of L-dopa for become more aware of motor fluctuations, especially
treatment of PD. as the return of parkinsonian symptoms during these
episodes were more prominent due to the underlying
worsening of the disease. Various terms were coined to
COMPLICATIONS SEEN IN THE EARLY label these fluctuations. ‘‘On–off’’ was coined in 1974
DAYS OF L-DOPA THERAPY to describe a sudden loss of L-dopa’s benefit and
The initial paper by Cotzias et al.1 describing the replacing it with the parkinsonian state (the ‘‘off’’
successful use of high dosage D,L-dopa in patients with state).52,54–56 The speed of this change was likened to
PD did not mention motor complications. Adverse that of a light switch turning on and off. The ‘‘on’’
effects that were mentioned were predominantly ano- state was equated with the time when the patient was
rexia, nausea, vomiting, faintness, and hematologic having a good response from L-dopa; the ‘‘off’’ state,
changes. Cotzias et al.2 then substituted L-dopa for D,L- when L-dopa was not working. The reemergence of the
dopa, which eliminated the hematologic adverse ‘‘on’’ state was sometimes sudden, without even the
effects. That article also presented the first report of benefit of another dose of L-dopa. But often the ‘‘on’’
L-dopa-induced dyskinesias, as well as mental symp- state would not appear until another dose of
toms of irritability, anger, hostility, paranoia, insomnia, L-dopa was ingested. The more common gradual
and an awakening effect. The dyskinesias described development of the ‘‘off’’ state, taking many minutes to
were chorea, myoclonus, hemiballism (ipsilateral to the develop and appearing as the plasma levels of L-dopa
side of a prior thalamotomy), and dystonia. These had fallen, was labeled in 1976 as the ‘‘wearing-off’’
investigators noted that the adverse effects would sub- phenomenon by Fahn57 and also the ‘‘end-of-dose’’
side with a lowering of the dosage of L-dopa. They deterioration by Marsden and Parkes.58 Both of these
also reported that the appearance of dyskinesias is not terms refer to the identical clinical situation and have
an early occurrence after initiating L-dopa therapy. been used interchangeably since.

Movement Disorders, Vol. 23, Suppl. 3, 2008


DOPAMINE AND LEVODOPA IN THE TREATMENT OF PD S503

A new dyskinetic state related to the timing of given dose because peripheral metabolism to dopamine
L-dopa dosing was described in 1977. Up to this point, is blocked. More importantly, these agents block the
all dyskinesias were considered to occur at the peak gastrointestinal side effects due to peripheral dopamine
effect of the L-dopa dose. Muenter et al.59 described acting upon the vomiting center of the area postrema,
dyskinesias appearing at the beginning and at the end which is not protected by the blood-brain barrier. The
of the dose, which they called ‘‘D-I-D’’ for dystonia combination of L-dopa with carbidopa into a single tab-
(dyskinesia)-improvement-dystonia (dyskinesia). These let was commercially marketed under the trade name
workers contrasted this to the much more common of Sinemet, to indicate without (sine) emesis. Sinemet
peak-dose dyskinesia, labeled by Muenter as ‘‘I-D-I.’’ (carbidopa/L-dopa) was initially sold in 1:10 ratios as
Subsequently, the D-I-D phenomena have been labeled 10/100 and 25/250 mg strength tablets. But because
diphasic dyskinesias.60,61 Such dyskinesias most typi- many patients require at least 50–75 mg of carbidopa a
cally appear in the legs.60 day to have adequate inhibition of peripheral dopa de-
Not all dyskinesias appear at the peak, the begin- carboxylase, a 25/100 mg tablet was later marketed, and
ning, or the ending of the dose. Lees et al.62 described is probably the most common strength utilized today. In
that painful ‘‘off’’ period dystonic cramps can appear, some patients even 75 mg per day of carbidopa is inad-
especially in the foot in the early morning on awaken- equate, and nausea, anorexia, or vomiting still occurs.
ing. Melamed63 elaborated on this phenomenon and Because such patients could benefit from higher doses
labeled it early-morning dystonia, pointing out that this of carbidopa without needing additional L-dopa, carbi-
occurs when the effect of the previous night’s dose of dopa became available as a separate tablet, which is
L-dopa had completely worn off. This is a dyskinesia, available under the trade name of Lodosyn. The combi-
appearing as a dystonia, rather than as chorea, that nation of benserazide and L-dopa is marketed under the
occurs during the ‘‘off’’ state, a time when bradykine- brand name of Madopar. A number of studies showed
sia and other signs of the parkinsonian state would the advantage of L-dopa combined with a peripheral de-
have been manifested. Instead, the ‘‘off’’ state dystonia carboxylase inhibitor compared to L-dopa alone.65–72
is seen in place of parkinsonism. Early morning dysto- The next enzyme inhibitor to be studied as an
nia is the most common type of ‘‘off’’ dystonia, but adjunct to L-dopa in the treatment for PD was selegi-
these tight, cramped muscles can appear at other times line (formerly called deprenyl), an irreversible inhibitor
of the day when the medication wears off. In addition of MAO type B. The action of MAO is to oxidatively
to ‘‘off’’ dystonia, dystonic postures can be seen as a deaminate monoamines, including dopamine (see Fig.
peak-dose dyskinesia, but is less common than peak- 2). MAO comes in two genetically distinct isoforms,
dose chorea. Melamed later described the mechanism known as MAO-A and MAO-B. Each has relatively
of delayed ‘‘ons’’ and dose-failures following a dose of selective substrates. Dopamine is metabolized by both
64
L-dopa as due to poor gastric absorption. types, whereas serotonin and tyramine (present in some
foods) are preferential substrates of MAO-A. MAO-A
is more enriched in nerve terminal mitochondria,
STEPS TAKEN TO ENHANCE THE thereby metabolizing cytosolic dopamine that isn’t
EFFECTIVENESS AND REDUCE THE taken up and sequestered into the synaptic vesicles.
ADVERSE EFFECTS OF L-DOPA THERAPY MAO-B is located predominantly in glial mitochondria
L-aromatic amino acid decarboxylase inhibitors, such and is available to metabolize dopamine released into
as a-methyldopa, had been developed and were used the synaptic cleft and not taken back up into the nerve
for the treatment of hypertension. By attaching an terminal by the action of the dopamine transporter.
azide moiety to these compounds, they are no longer MAO-A inhibition makes patients susceptible to risks
able to penetrate the blood-brain barrier, and thus associated with high dietary levels of tyramine. If tyra-
could inhibit the conversion of L-dopa to dopamine in mine is not degraded by MAO-A present in the gut, ty-
the peripheral system, and still allow this enzymatic ramine enters the body and acts as a false transmitter,
step to occur centrally. The development of such pe- entering noradrenergic nerve terminals and synaptic
ripheral dopa decarboxylase inhibitors was the next vesicles displacing norepinephrine, which then can trig-
major step making L-dopa more potent while reducing ger sudden hypertension, leading to cerebral hemorrhage
gastrointestinal adverse effects. Carbidopa and bensera- or other complications. This is commonly known as the
zide are such peripheral decarboxylase inhibitors and ‘‘cheese-effect’’ because of the presence of high levels
are used commercially. When given with L-dopa, they of tyramine in some fermenting cheeses (as well as in
allow for up to a 4-fold increase in the availability of a other fermented foods such as aged meat and some red

Movement Disorders, Vol. 23, Suppl. 3, 2008


S504 S. FAHN

wines). Selective MAO-B inhibitors are not associated reported were spectacular, although transient, with
with this risk. Drugs that inhibit both MAO-A and patients getting out of their wheelchairs and walking.
MAO-B (nonselective inhibitors) have been commer- Similar dramatic results, but longer lasting, were seen
cially available since the 1950s for the treatment of by Cotzias with high dosage oral L-dopa.2 In the early
depression. Although excellent antidepressants, the risk 1970s, Fahn speculated that patients diagnosed with
of the cheese-effect (unless patients strictly adhere to a PD should be treated with L-dopa as soon as the diag-
low tyramine diet) has made such nonselective inhibi- nosis was made to allow the exogenous supply of do-
tors extremely unpopular, especially since many other pamine to maintain activation of dopamine receptors,
types of antidepressants have now been developed. thereby easing the overactive remaining dopamine
Use of nonselective MAO inhibitors with L-dopa neurons not to work so hard. Such overactivity of re-
(especially in the absence of a peripheral dopa decar- sidual dopamine neurons is one of the compensatory
boxylase inhibitor) can result in hyper- and hypoten- mechanisms for insufficient dopamine release in the
sive episodes. However, inhibitors of MAO-B can be striatum due to the marked loss of dopamine nerve
taken safely with L-dopa; these potentiate the sympto- terminals there. This compensatory mechanism was
matic benefit of L-dopa by about one-third lower dos- proposed by Hornykiewicz,110 who showed that the
age of L-dopa,73,74 probably by inhibiting brain metab- ratio of homovanillic acid to dopamine was increased
olism of dopamine formed from L-dopa therapy. The in the striatum of PD subjects compared to healthy
first MAO-B inhibitor studied was deprenyl, now subjects, indicating an increased activity of remaining
called selegiline.75–77 Two such MAO-B inhibitors are presynaptic dopamine neurons to keep dopamine
now commercially available, selegiline and rasagiline. receptors activated. By providing an exogenous route
Both drugs have been demonstrated to have anti-par- to activate dopamine receptors, treatment with L-dopa
kinsonian effects particularly in treating the wearing- (or dopamine agonists) early in PD might permit
off effect78–80 and both have been studied for their remaining dopamine neurons to be less active and
potential to slow the clinical progression of PD,74,81–84 hopefully not die so quickly. Although Fahn did not
but these topics are beyond the scope of this review. use the word then, this concept is in keeping with the
Another catabolic route for both dopamine and L- idea that L-dopa would be neuroprotective or at least
dopa is 3-O-methylation, catalyzed by the enzyme provide a disease modifying effect. Early treatment
COMT (see Fig. 2). Two inhibitors of this enzyme, tol- with L-dopa, however, brought with it unintended con-
capone and entacapone, are commercially available sequences: patients developed dyskinesias, wearing
and have been demonstrated to reduce the wearing-off off, and sudden on and off periods. These observa-
effect when used as adjuncts to L-dopa.85–92 These tions prompted Fahn and others to think that late
COMT inhibitors slow the peripheral metabolism of L- treatment would be more beneficial, and in 1978 he
dopa, thereby delaying the elimination of plasma L- wrote, with Donald Calne, a proposal suggesting that
dopa. Recent interest has focused on the potential use L-dopa treatment should be delayed until symptoms
of L-dopa plus a COMT inhibitor in early disease to had become severe to avoid these motor complica-
prevent the development of motor complications.93 tions.111
Another approach to provide a longer plasma half- Debates have continued ever since, with some, such
life of L-dopa is the development of delayed release as Markham and Diamond112 advocating earlier rather
formulations of L-dopa, and some clinical studies than later treatment based on retrospective analysis
showed effectiveness with such compounds.94–109 One that early treatment delays severity. When Cohen113
product is called carbidopa/L-dopa extended release, suggested that the neuron’s own dopamine can cause
with the brand name of Sinemet CR (for continuous oxidative stress by being oxidized to oxyradicals, and
release); another is Madopar hydrodynamically bal- that this could lead to degeneration of dopamine neu-
anced system. Dopamine agonists that act directly on rons if insufficient antioxidative mechanisms are avail-
the dopamine receptor have also been developed, but able; this concept raised the suspicion that perhaps
are not covered in this review. L-dopa by increasing cytosolic dopamine could be
toxic and hasten further loss of dopamine neurons. The
potential for L-dopa to promote dopaminergic neuronal
THE DEBATE OVER WHEN TO loss in patients with PD was debated without
START TREATMENT resolution. To hopefully resolve the uncertainty, the
When Birkmayer and Hornykiewicz35 first gave in- Parkinson Study Group developed the clinical trial,
travenous L-dopa to patients with PD, the results they Earlier versus Later L-dopa in PD (ELLDOPA trial),

Movement Disorders, Vol. 23, Suppl. 3, 2008


DOPAMINE AND LEVODOPA IN THE TREATMENT OF PD S505

FIG. 4. Photographs of the 3 giants who pioneered the development of levodopa therapy for Parkinson’s disease. Pictured are Arvid Carlsson,
Oleh Hornykiewicz, and George Cotzias, obtained from the World Wide Web. [Color figure can be viewed in the online issue, which is available
at www.interscience.wiley.com.]

which was designed to assess the effect of low, me- (see Fig. 4) stand out among all others as giants in
dium, and high doses of carbidopa/L-dopa on disease their contributions. Carlsson’s work led to the realiza-
progression, measured by first treating patients with tion that dopamine is a neurotransmitter in brain, that
early PD for 40 weeks and then withdrawing the symp- there is high dopamine concentration in the striatum,
tomatic drug for 2 weeks and assessing severity of the that dopamine depletion from reserpine is responsible
disease. Although the hypothesis was that L-dopa for the akinetic effect, and that PD is related to dopa-
would be toxic at higher doses and thereby hasten dis- mine in brain. Hornykiewicz showed that striatal dopa-
ease progression, surprisingly the clinical results sug- mine is deficient in PD and that the greater the deple-
gested that higher doses may be protective and should tion, the more severe the disease; he also was the first
be started early.114 However, neuroimaging data col- to suggest using L-dopa to treat the disease. Cotzias
lected in the same study contradicted the clinical showed how one could safely use high enough doses
results, suggesting instead that L-dopa may accelerate of L-dopa to be effective, and this led to the successful
the loss of nigrostriatal dopamine nerve terminals or introduction of L-dopa into the treatment of PD.
modify the dopamine transporter. Thus, the debate
remains unresolved.
At the Movement Disorder Congress in Kyoto, REFERENCES
Japan, in October 2006, Fahn raised the question to
1. Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino
the audience of over 1,000 attendees as to what dose acids and modification of parkinsonism. N Engl J Med 1967;
of L-dopa was the plateau dose they usually attempt to 276:374–379.
reach before proceeding to higher dosages if clinical 2. Cotzias GC, Papavasiliou PS, Gellene R. Modification of par-
kinsonism—chronic treatment with L-DOPA. N Engl J Med
benefit is insufficient; almost unanimously, the plateau 1969; 280:337–345.
dose was 25/100 mg of carbidopa/L-dopa three times 3. DeLong MR, Alexander GE, Mitchell SJ, Richardson RT. The
daily (Fahn, personal observations). Only one attendee contribution of basal ganglia to limb control. Prog Brain Res
1986;64:161–174.
went higher, to 37.5/150 mg three times daily. The 4. Penney JB, Jr, Young AB. Striatal inhomogeneities and basal
result of this ‘‘survey’’ suggests that without proven ganglia function. Mov Disord 1986;1:3–14.
benefit of a neuroprotective effect of L-dopa at higher 5. Crossman AR. Primate models of dyskinesia: the experimental
approach to the study of basal ganglia-related involuntary
dosages, clinicians cannot warrant using a higher dose movement disorders. Neuroscience 1987;21:1–40.
because of the potential for motor complications. 6. Hornykiewicz O. Dopamine miracle: from brain homogenate to
dopamine replacement. Mov Disord 2002;17:501–508.
7. Hornykiewicz O. L-DOPA: from a biologically inactive amino
CONCLUSION acid to a successful therapeutic agent. Amino Acids 2002;23:
65–70.
In looking back at the history of developing L-dopa 8. Barger G, Dale HH. Chemical structure and sympathomimetic
as an effective therapy for PD, three early pioneers action of amines. J Physiol 1910;41:19–59.

Movement Disorders, Vol. 23, Suppl. 3, 2008


S506 S. FAHN

9. Funk C. Synthesis of dl-3:4-dihydroxyphenylalanine. J Chem 34. Sulzer D, Bogulavsky J, Larsen KE, et al. Neuromelanin bio-
Soc 1911;99:554–557. synthesis is driven by excess cytosolic catecholamines not
10. Guggenheim M. Dioxyphenylalanin, eine neue Aminosäure aus accumulated by synaptic vesicles. Proc Natl Acad Sci USA
Vicia faba. Hoppe-Seyler’s Zeitschr Physiol Chem 1913;88: 2000;97:11869–11874.
276–284. 35. Birkmayer W, Hornykiewicz O. Der L-3,4-Dioxyphenylalanin
11. Holtz P, Heise R, Lüdtke K. Fermentativer Abbau von Ldioxy- (DOPA)—Effekt bei der Parkinson-Akinese. Wien Klin Wochenschr
phenylalanin (Dopa) durch Niere. Naunyn-Schmiedeberg’s Arch 1961;73:787–788.
Exp Path Pharmak 1938;191:87–118. 36. Barbeau A, Sourkes TL, Murphy GF. Les catecholamines dans
12. Holtz P. Dopadecarboxylase. Naturwissenschaften 1939;27: la maladie de Parkinson. In: de Ajuriaguerra J, editor. Sympo-
724–725. sium sur les monoamines et systeme nerveux central. Geneve:
13. Blaschko H. The specific action of L-DOPA decarboxylase. J Georg & Cie; 1962. p 247–262.
Physiol 1939;96:50. 37. Friedhoff AJ, Hekiman L, Alpert M, Tobach E. Dihydroxyphe-
14. Holtz P, Stock K, Westermann E. [On the effect of dopamine nylalanine in extrapyramidal disease. JAMA 1964;184:285.
on the blood pressure.] Naunyn Schmiedebergs Arch Exp 38. Umbach W, Bauman D. Die Wirksamkeit von L-Dopa bei Par-
Pathol Pharmakol 1963;246:133–146. kinson-Patienten mit und ohne sterotaktischen Herneingriff.
15. Hornykiewicz O. The action of dopamine on the arterial pres- Arch Psychiat Nervenkr 1964;205:281.
sure of the guinea pig. Brit J Pharmacol 1958;13:91–94. 39. Hirschmann J, Mayer K. Zur Beeinflussung von L-Dopa (L-
16. Nagatsu T, Levitt M, Udenfriend S. Tyrosine hydroxylase: the Dihydroxyphenylananin). Deutsch Med Wschr 1964;89:1877.
initial step in norepinephrine biosynthesis. J Biol Chem 1964; 40. Fazzagli A, Amaducci L. La Sperimentazione clinica del Dopa
239:2910–1917. nelle sindromi parkinsoniane. Riv Neurobiol 1966;12:138.
17. Ichinose H, Suzuki T, Inagaki H, Ohye T, Nagatsu T. Molecu- 41. Bruno A, Bruno SC. Effetti dell L-2,4-dihydossifenilalanina (L-
lar genetics of dopa-responsive dystonia. Biol Chem 1999;380: Dopa) nei pazienti parkinsoniani. Riv Sper Freniatr 1966;90:39.
1355–1364. 42. Greer M, Williams CM. Dopamine metabolism in Parkinson’s
18. Nygaard TG, Wilhelmsen KC, Risch NJ, et al. Linkage map- disease. Neurology 1963;13:73.
ping of dopa-responsive dystonia (DRD) to chromosome 14q. 43. Aebert K. Was leistet L-DOPA bei der Behandlung der Parkin-
Nat Genet 1993;5:386–391. son-Akinese? Deutsch Med Wschr 1967;92:483.
19. Pletscher A, Shore PA, Brodie BB. Serotonin release as a possi- 44. Fehling C. Treatment of Parkinson’s syndrome with L-DOPA, a
ble mechanism of reserpine action. Science 1955;122:374–375. double-blind study. Acta Neurol Scand 1966;42:367–372.
20. Carlsson A. Morphologic and dynamic aspects of dopamine in 45. Rinne UK, Bracco F, Chouza C, et al. Early treatment of Par-
the central nervous system. In: Costa E, Côté LJ, Yahr MD, kinson’s disease with cabergoline delays the onset of motor com-
editors. Biochemistry and pharmacology of the basal ganglia. plications. Results of a double-blind levodopa controlled trial.
Hewlett, NY: Raven Press; 1966. p 107–113. The PKDS009 Study Group. Drugs 1998;55 (Suppl 1):23–30.
21. Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenyl- 46. McGeer PL, Zeldowicz LR. Administration of dihyroxypheny-
alanine and 5-hydroxytroptophan as reserpine antagonists. Na- lalanine to parkinsonian patients. Canad Med Assoc J 1964;
ture 1957;180:1200. 90:463–466.
22. Degkwitz R, Frowein R, Kulenkampff C, Mohs U. Über die 47. Yahr MD, Wolf A, Antunes JL, Miyoshi K, Duffy P. Autopsy
Wirkungen des L-dopa beim Menschen und deren Beeinflus- findings in parkinsonism following treatment with levodopa.
sung durch Reserpin, Chlorpromazin, Iproniazid und Vitamin Neurology 1970;6 (Suppl 1):55–66.
B6. Klin Wochenschr 1960;38:120–123. 48. Yahr MD, Duvoisin RC, Schear MJ, Barrett RE, Hoehn MM.
23. Montagu KA. Catechol compounds in rat tissues and in brains Treatment of parkinsonism with levodopa. Arch Neurol 1969;
of different animals. Nature 1957;180:244–245. 21:343–354.
24. Weil-Malherbe H, Bone AD. Intracellular distribution of cate- 49. McDowell F, Lee JE, Swift T, Sweet RD, Ogsbury JS, Kessler
cholamines in the brain. Nat Lond 1957;180:1050–1051. JT. Treatment of Parkinson’s syndrome with dihydroxyphenyla-
25. Raab W, Gigee W. Concentration and distribution of ‘‘ence- lanine (levodopa). Ann Int Med 1970;72:29–35.
phalin’’ in the brain of humans and animals. Proc Soc Exp Biol 50. Schwarz GA, Fahn S. Newer medical treatments in parkinson-
Med 1951;76:97–100. ism. Med Clin North Am 1970;54:773–785.
26. Carlsson A, Lindqvist M, Magnusson T, Waldeck B. On the 51. Calne DB, Reid JL, Vakil SD, Pallis C. Problems with L-
presence of 3-hydroxytyramine in the brain. Science 1958;127: DOPA therapy. Clin Med 1971;78:21–23.
471. 52. Duvoisin RC. Hyperkinetic reactions with L-DOPA. In: Yahr
27. Bertler A, Rosengren E. Occurrence and distribution of MD, editor. Current concepts in the treatment of parkinsonism.
catechol amines in brain. Acta Physiol Scand 1959;47:350– New York: Raven Press; 1974. p 203–210.
361. 53. Klawans HL, Goetz C, Bergen D. Levodopa-induced myoclo-
28. Sano I, Gamo T, Kakimoto Y, Taniguchi K, Takesada M, Nish- nus. Arch Neurol 1975;32:331–334.
inuma K. Distribution of catechol compounds in human brain. 54. Sweet RD, McDowell FH. The ‘‘on-off’’ response to chronic L-
Biochim Biophys Acta 1959;32:586–587. DOPA treatment of parkinsonism. Adv Neurol 1974;5:331–338.
29. Carlsson A. The occurrence, distribution and physiological role 55. Yahr MD. Variations in the ‘‘on-off’’ effect. Adv Neurol 1974;
of catecholamines in the nervous system. Pharmacol Rev 5:397–399.
1959;11(2, Part 2):490–493. 56. Fahn S. ‘‘On-off’’ phenomenon with levodopa therapy in parkin-
30. Ehringer H, Hornykiewicz O. Verteilung von Noradrenalin und sonism: clinical and pharmacologic correlations and the effect of
Dopamin (3-Hydroxytyramin) im Gehirn des Mensachen und intramuscular pyridoxine. Neurology 1974;24:431–441.
ihr Verhalten bei Erkrankungen des extrapyramidalen Systems. 57. Fahn S. Medical treatment of movement disorders. In: Davis
Klin Wochenschr 1960;38:1238–1239. FA, editor. Neurological reviews 1976. Minneapolis: American
31. Hornykiewicz O. Dopamine (3-hydroxytyramine) and brain Academy of Neurology; 1976. p 72–106.
function. Pharmacol Rev 1966;18:925–964. 58. Marsden CD, Parkes JD. ‘‘On–off’’ effects in patients with Par-
32. Anden NE, Carlssson A, Dahlstrom A, Fuxe K, Hillarp NA, kinson’s disease on chronic levodopa therapy. Lancet 1976;1:
Larsson K. Demonstration and mapping out of nigro-neostriatal 292–295.
dopamine neurons. Life Sci 1964;3:523–530. 59. Muenter MD, Sharpless NS, Tyce GM, Darley FL. Patterns of
33. Poirier L, Sourkes TL. Influence of the substantia nigra on the dystonia (‘‘I-D-I’’ and ‘‘D-I-D’’) in response to L-DOPA therapy
catecholamine content of the striatum. Brain 1965;88:181–192. of Parkinson’s disease. Mayo Clin Proc 1977;52:163–174.

Movement Disorders, Vol. 23, Suppl. 3, 2008


DOPAMINE AND LEVODOPA IN THE TREATMENT OF PD S507

60. Marsden CD, Parkes JD, Quinn N. Fluctuations of disability in randomized, double-blind, parallel-group trial. Lancet 2005;
Parkinson’s disease—clinical aspects. In: Marsden CD, Fahn 365:947–954.
S, editors. Movement disorders. London: Butterworth Scientific; 81. Birkmayer W, Knoll J, Riederer P, Youdim MBH, Hars V,
1982. p 96–122. Marton J. Increased life expectancy resulting from addition of
61. Fahn S. Fluctuations of disability in Parkinson’s disease: patho- L-deprenyl to Madopar treatment in Parkinson’s disease: a
physiological aspects. In: Marsden CD, Fahn S, editors. Movement long-term study. J Neural Transm 1985;64:113–127.
disorders. London: Butterworth Scientific; 1982. p 123–145. 82. Parkinson Study Group. Effects of tocopherol and deprenyl on
62. Lees AJ, Shaw KM, Stern GM. ‘‘Off period’’ dystonia and ‘‘on the progression of disability in early parkinson’s disease. N
period’’ choreoathetosis in levodopa-treated patients with Par- Engl J Med 1993;328:176–183.
kinson’s disease. Lancet 1977;2:1034. 83. Palhagen S, Heinonen E, Hagglund J, Kaugesaar T, Maki-Ikola
63. Melamed E. Early-morning dystonia: a late side effect of long- O, Palm R;Swedish Parkinson Study Group. Selegiline slows
term levodopa therapy in Parkinson’s disease. Arch Neurol the progression of the symptoms of Parkinson disease. Neurol-
1979;36:308–310. ogy 2006;66:1200–1206.
64. Melamed E, Bitton V, Zelig O. Episodic unresponsiveness to 84. Parkinson Study Group. A controlled, randomized, delayed-start
single doses of L-Dopa in parkinsonian fluctuators. Neurology study of rasagiline in early Parkinson disease. Arch Neurol 2004;
1986;36:100–103. 61:561–566.
65. Yahr MD, Duvoisin RC, Mendoza MR, Schear MJ, Barrett RE. 85. Roberts JW, Cora-Locatelli G, Bravi D, Amantea MA, Moura-
Modification of L-DOPA therapy of Parkinsonism by alpha- dian MM, Chase TN. Catechol-O-methyltransferase inhibitor
methyldopa hydrazine (MK-486). Trans Am Neurol Assoc tolcapone prolongs levodopa/carbidopa action in parkinsonian
1971;96: 55–58. patients. Neurology 1993;43:2685–2688.
66. Papavasiliou PS, Cotzias GC, Duby SE, Steck AJ, Fehling C, 86. Limousin P, Pollak P, Gervasin-Tournier CL, Hommel M, Per-
Bell MA. Levodopa in Parkinsonism: potentiation of central ret JE. Ro 40-7592, a COMT inhibitor, plus levodopa in Par-
effects with a peripheral inhibitor. N Engl J Med 1972;286:8–14. kinson’s disease. Lancet 1993;341:1605.
67. Barbeau A, Gillo-Joffroy L, Mars H, Arsenault A. Treatment 87. Davis TL, Roznoski M, Burns RS. Effects of tolcapone in Par-
of Parkinson’s disease with L-DOPA alone or combined with kinson’s patients taking L-dihydroxyphenylalanine/carbidopa
Ro 4-4602. Rev Can Biol 1972;31 (Suppl):169–174. and selegiline. Mov Disord 1995;10:349–351.
68. Lieberman AN, Derby BM, Feigenson J, et al. MK 486 and 88. Kurth MC, Adler CH, Hilaire MS, et al. Tolcapone improves
levodopa in treatment of Parkinsonism. Dis Nerv Syst 1973;34: motor function and reduces levodopa requirement in patients with
167–171. Parkinson’s disease experiencing motor fluctuations: a multicen-
69. Fermaglich J, Chase TN. Methyldopa or methyldopahydrazine ter, double-blind, randomized, placebo-controlled trial. Tolca-
as levodopa synergists. Lancet 1973;1:1261–1262. pone Fluctuator Study Group I. Neurology 1997;48:81–87.
70. Birkmayer W, Linauer W, Mentasti M, Riederer P. 2-year 89. Kaakkola S, Teräväinen H, Ahtila S, Rita H, Gordin A. Effect
experiences with a combination treatment of Parkinsonism with of entacapone, a COMT inhibitor, on clinical disability and lev-
L-DOPA and a decarboxylase inhibitor (Benserazid, Ro 4- odopa metabolism in parkinsonian patients. Neurology 1994;
4602). Wien Med Wochenschr 1974;124:340–344. 44: 77–80.
71. Markham CH, Diamond SG, Treciokas LJ. Carbidopa in Par- 90. Merello M, Lees AJ, Webster R, Bovingdon M, Gordin A.
kinson disease and in nausea and vomiting of levodopa. Arch Effect of entacapone, a peripherally acting catechol-o-methyl-
Neurol 1974;31:128–133. transferase inhibitor, on the motor response to acute treatment
72. Sweet RD, McDowell FH. Wasterlain CO, Stern PH. Treatment with levodopa in patients with Parkinson’s disease. J Neurol
of ‘‘on-off effect’’ with a dopa decarboxylase inhibitor. Arch Neurosurg Psychiatry 1994;57:186–189.
Neurol 1975;32:560–563. 91. Ruottinen HM, Rinne UK. Entacapone prolongs levodopa
73. Lees AJ. Comparison of therapeutic effects and mortality data response in a one month double blind study in parkinsonian
of levodopa and levodopa combined with selegiline in patients with patients with levodopa related fluctuations. J Neurol Neurosurg
early, mild Parkinson’s disease. Br Med J 1995;311:1602–1607. Psychiatry 1996;60:36–40.
74. Shoulson I, Oakes D, Fahn S, et al; Parkinson Study Group. 92. Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and
Impact of sustained deprenyl (selegiline) in levodopa-treated clinical dose-response study of entacapone as an adjuvant to
Parkinson’s disease: a randomized placebo-controlled extension levodopa therapy in advanced Parkinson’s disease. Clin Neuro-
of the deprenyl and tocopherol antioxidative therapy of parkin- pharmacol 1996;19:283–296.
sonism trial. Ann Neurol 2002;51:604–612. 93. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine re-
75. Birkmayer W, Knoll J, Riederer P, Youdim MBH. (2)-Dep- ceptor stimulation in the treatment of parkinson’s disease: sci-
renyl leads to prolongation of L-DOPA efficacy in Parkinson’s entific rationale and clinical implications. Lancet Neurol
disease. Mod Probl Pharmacopsychiatry 1983;19:170–176. 2006;5:677–687.
76. Stern GM, Lees A, Hardie RJ, Sandler M. Clinical and pharma- 94. Hutton JT, Dippel Rl, Bianchine JR, Strahlendorf HK, Meyer
cological problems of deprenyl (selegiline) treatment in Parkin- PG. Controlled-release carbidopa/levodopa in the treatment of
son’s disease. Acta Neurol Scand Suppl 1983;95:113–116. parkinsonism. Clin Neuropharmacol 1984;7:135–139.
77. Lieberman AN, Gopinathan G, Neophytides A, et al. Deprenyl 95. Nutt JG, Woodward WR, Carter JH. Clinical and biochemical
in the treatment of Parkinson’s disease. A specific type B mon- studies with controlled-release levodopa/carbidopa. Neurology
oamine oxidase inhibitor. N Y State J Med 1984;84:13–16. 1986;36:1206–1211.
78. Golbe LI, Lieberman AN, Muenter MD, et al. Deprenyl in the 96. Juncos JL, Fabbrini G, Mouradian MM, Serrati C, Kask AM,
treatment of symptom fluctuations in advanced Parkinson’s dis- Chase TN. Controlled release levodopa treatment of motor fluc-
ease. Clin Neuropharmacol 1988;11:45–55. tuations in Parkinson’s disease. J Neurol Neurosurg Psychiatry
79. Parkinson Study Group. A randomized placebo-controlled trial 1987;50:194–198.
of rasagiline in levodopa-treated patients with Parkinson dis- 97. Cedarbaum JM, Breck L, Kutt H, McDowell FH. Controlled-
ease and motor fluctuations—The PRESTO study. Arch Neurol release levodopa/carbidopa. I. Sinemet CR3 treatment of res-
2005;62: 241–248. ponse fluctuations in Parkinson’s disease Neurology 1987;37:
80. Rascol O, Brooks DJ, Melamed E, et al; LARGO study group. 233–241.
Rasagiline as an adjunct to levodopa in patients with Parkin- 98. Goetz CG, Tanner CM, Klawans HL, Shannon KM, Carroll VS.
son’s disease and motor fluctuations (LARGO, Lasting effect in Parkinson’s disease and motor fluctuations: long-acting carbi-
Adjunct therapy with Rasagiline Given Once daily, study): a dopa/levodopa (CR-4-Sinemet). Neurology 1987;37:875–878.

Movement Disorders, Vol. 23, Suppl. 3, 2008


S508 S. FAHN

99. Juncos JL, Fabbrini G, Mouradian MM, Chase TN. Controlled 106. Quinn N, Marion MH, Marsden CD. Open study of Madopar
release levodopa-carbidopa (CR-5) in the management of HBS, a new formulation of levodopa with benserazide, in 13
parkinsonian motor fluctuations. Arch Neurol 1987;44:1010– patients with Parkinson’s disease and ‘‘on-off’’ fluctuations. Eur
1012. Neurol 1987;27 (Suppl 1):105–113.
100. Cedarbaum JM, Hoey M, Kutt H, McDowell FH. Controlled- 107. Rondot P, Ziegler M, Aymard N, Holzer J. Clinical trial of
release levodopa/carbidopa. III: sinemet CR5 treatment of Madopar HBS in parkinsonian patients with fluctuating drug
response fluctuations in Parkinson’s disease. Clin Neurophar- response after long-term levodopa therapy. Eur Neurol 1987;27
macol 1988;11:168–173. (Suppl 1):114–119.
101. Sage JJ, Mark MH. Comparison of controlled-release Sinemet 108. Rinne UK. Madopar HBS in the long-term treatment of parkin-
(CR4) and standard Sinemet (25 mg/100 mg) in advanced Par- sonian patients with fluctuations in disability. Eur Neurol 1987;
kinson’s disease: a double-blind, crossover study. Clin Neuro- 27 (Suppl 1):120–125.
pharmacol 1988;11:174–179. 109. Lees AJ. A sustained-release formulation of L-DOPA (Madopar
102. Goetz CG, Tanner CM, Shannon KM, et al. Controlled-release HBS) in the treatment of nocturnal and early-morning disabilities
carbidopa/levodopa (CR4-Sinemet) in Parkinson’s disease pa- in Parkinson’s disease. Eur Neurol 1987;27 (Suppl 1):126–134.
tients with and without motor fluctuations. Neurology 1988;38: 110. Hornykiewicz O. Metabolism of brain dopamine in human par-
1143–1146. kinsonism: neurochemical and clinical aspects. In: Costa E, Côté
103. Hutton JT, Morris JL, Bush DF, Smith ME, Liss CL, Reines S. LJ, Yahr MD, editors. Biochemistry and pharmacology of the
Multicenter controlled study of Sinemet CR vs Sinemet (25/ basal ganglia. Hewlett, NY: Raven Press; 1966. p 171–185.
100) in advanced Parkinson’s disease. Neurology 1989;39 111. Fahn S, Calne DB. Considerations in the management of par-
(Suppl 2):67–72. kinsonism. Neurology 1978;28:5–7.
104. Hutton JT, Morris JL. Long-acting carbidopa-levodopa in the 112. Markham CH, Diamond SG. Evidence to support early levodopa
management of moderate and advanced Parkinson’s disease. therapy in Parkinson disease. Neurology 1981;31:125–131.
Neurology 1992;42 (Suppl 1):51–56. 113. Cohen G. Monoamine oxidase, hydrogen peroxide, and Parkin-
105. Poewe WH, Lees AJ, Stern GM. Treatment of motor fluctua- son’s disease. Adv Neurol 1986;45:119–125.
tions in Parkinson’s disease with an oral sustained-release prep- 114. Fahn S, Oakes D, Shoulson I, et al; Parkinson Study Group.
aration of L-DOPA: clinical and pharmacokinetic observations. Levodopa and the progression of PD. New Engl J Med 2004;
Clin Neuropharmacol 1986;9:430–439. 351:2498–2508.

Movement Disorders, Vol. 23, Suppl. 3, 2008

You might also like