Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
The clinical symptoms and signs of Parkinson disease (PD) can be conveniently divided into motor and nonmotor features, necessitating individualization of therapy tailored to each patient’s needs ( ). Although the nonmotor aspects of PD can be equally incapacitating for people with PD, it is the motor component that defines the disease, for without the motor component, the diagnosis cannot be established. This chapter focuses on the treatment of the motor features of PD and motor complications of dopaminergic treatment. The description and treatment of the nonmotor features of the disease are discussed in Chapter 8 . The surgical treatment of PD is discussed in Chapter 7 (with the exception of intrajejunal infusion of levodopa, which is covered in this chapter).
The most efficacious drug available for the medical treatment of PD is levodopa, which was introduced in its high-dose therapeutic form over 50 years ago ( , ). An entire issue of the journal Movement Disorders was devoted to this drug in celebration of 50 years of its use ( ), and the reader is referred to that issue for details regarding the discovery and development of levodopa. Levodopa remains the gold standard for reducing many of the motor features of PD. If levodopa could be used without the development of motor complications, the symptomatic treatment of this disease would be straightforward. However, 30% to 50% of PD patients develop motor complications after 5 years of treatment with levodopa. Motor complications include motor fluctuations with “wearing off,” dyskinesia, and dose failure. The occurrences of motor complications are primarily related to the progression of the underlying disease rather than the duration of levodopa therapy. Hence, there is no rationale for “levodopa phobia,” withholding symptomatic treatment with dopaminergic agents until disability or loss of employment ( ). But there is a need for addressing these complications. These include new delivery systems for levodopa, additional medications that can prolong the effect of levodopa, using other longer acting dopaminergic agents, and treatments for dyskinesia. Although deep brain stimulation (DBS) does address motor complications, it will be discussed in a separate chapter.
The typical natural history of PD is someone who develops the first symptoms around age 60 and lives approximately 20 years. A typical pattern of progression is shown in Fig. 6.1 . In early PD, treatment may be straightforward. As the disease progresses, motor and nonmotor complications make treatment more challenging. Topics in this chapter will include potential disease-modifying therapies, initiating pharmacologic treatment, and addressing motor complications.
Treating PD is complicated. There is heterogeneity of PD, with some patients having tremor predominant form, some with gait difficulty and postural instability, and some with mixed phenotype and additional motor and nonmotor features ( ). Variation in the clinical response to dopaminergic drugs occurs, with some patients responding dramatically and others with more modest improvement. There can be varying treatment goals, with some patients desiring to continue employment and others who want to minimize medications. Finally, numerous adverse effects can intervene and limit the usefulness of some of the treatments. The clinician treating PD must be familiar with the large variety of drugs to treat the motor and nonmotor symptoms, including the indications for the drug, side effect profile, and limitations.
Disease-modification is a term used to indicate slowing of clinical worsening and is not equivalent to neuroprotection (slowing of disease pathologic progression). All clinical trials discussed later were designed to test disease modification. In this chapter, we will use the term “neuroprotection” instead of “disease modification” in an effort to use just one label. Neuroprotection can be divided into at least three different classes of action: slowing the pathogenetic cascade that leads to cell death so that the natural history of the disease is less progressive (neuroprotection), restoring injured dysfunctional neurons to good health (neurorescue, neurorestoration), and replacing dead neurons ( Fig. 6.2 ).
Currently, despite many clinical studies, no agent has demonstrated restorative properties or efficacy in slowing progression of PD ( Table 6.1 ). In most of the studies in Table 6.1 , worsening of severity of PD was measured quantitatively by the Unified Parkinson’s Disease Rating Scale (UPDRS) [ ]. Drugs that have been specifically evaluated in controlled clinical trials for slowing disease progression include selegiline and tocopherol ( ), selegiline alone ( ; ; , ); TCH346 (an antiapoptotic propargyline that inhibits glyceraldehyde 3-phosphate dehydrogenase) ( ; ; ), riluzole ( ), neuroimmunophilin ( ), coenzyme Q 10 ( ; ), glial cell line–derived neurotrophic factor (GDNF) ( ; ), rasagiline ( ; ), minocycline and the energy enhancer creatine ( ; ; ), CEP-1347 (an inhibitor of the mixed lineage kinase-3 family) ( ), neurturin ( ; ), GM1 ganglioside ( ), pramipexole ( ), coenzyme Q10 ( ), intranasal reduced glutathione ( ), pioglitazone ( ), exenatide ( , ; ), isradipine ( , ; ), inosine ( ; Press Release, 2019A), isradipine ( ), and levodopa ( ; ; and most recently nilotinib ( (see Table 6.1 ).
Year/reference | Trial | Agent | N | Primary outcome variable |
---|---|---|---|---|
1993; Parkinson Study Group | DATATOP | Tocopherol | 800 | Need for levodopa |
2003; Rascol et al. | Riluzole | Riluzole | 1084 | Need for levodopa |
2003; Nutt et al. | Intraventricular infusion of GDNF | GDNF | 50 | Change in UPDRS |
2006; Lang et al. | Intraputaminal infusion of GDNF | GDNF | 34 | UPDRS and F-DOPA PET |
2006; Olanow et al. | Antiapoptotic propargylamine | TCH346 | 301 | Need for levodopa |
2006 2008; NINDS NET-PD Investigators | Anti-inflammatory; FS-1 study | Minocycline | 200 | UPDRS |
2007; NINDS NET-PD Investigators | Neuroimmunophilin-ligand compound; FS-Too study | (GPI-1485) | 213 | UPDRS and need for levodopa |
2007; Parkinson Study Group PRECEPT Investigators | Antiapoptotic drug; mixed lineage kinase inhibitor; PRECEPT study | CEP-1347 | 800 | Need for levodopa |
2007; Storch et al. | Mitochondrial agent | Coenzyme Q10 | 131 | UPDRS |
2010; Marks et al. | Trophic factor; intraputaminal infusion | AAV2 Neurturin | 58 | UPDRS practically defined off score |
2013; Schapira et al. | Dopamine agonist; PROUD study | Pramipexole | 535 | Delayed-start design; UPDRS |
2014; Parkinson Study Group QE3 Investigators | Mitochondrial agent; QE-3 trial | Coenzyme Q10 | 600 | UPDRS |
2015; Olanow et al | Trophic factor; intraputaminal and intranigral infusion | AAV2 Neurturin | 51 | UPDRS practically defined off score |
2015; Writing Group for the NINDS Exploratory Trials in Parkinson Disease (NET-PD) Investigators | LS-1 trial | Creatine | 1741 | Combination of multiple outcomes |
2015; NINDS Exploratory Trials in Parkinson Disease (NET-PD) FS-ZONE Investigators | Peroxisome proliferator activated receptor γ (PPAR-γ) agonist; FS-ZONE study | Pioglitazone | 210 | Change in UPDRS |
2019 Verschuur et al. | Levodopa delayed start design; LEAP study | Levodopa | 445 | Change in UPDRS |
2019 https://www.michaeljfox.org/news/parkinsons-inosine-trial-ending-early | Urate prodrug; SURE-PD3 study | Inosine | 298 | Change in MDS-UPDRS |
2020; Parkinson Study Group STEADY-PD III Investigators. | Calcium channel blocker; STEADY-PD III | Isradipine | 336 | Change in MDS-UPDRS |
Some controlled studies of disease-modifying treatments have demonstrated success in achieving the primary outcome, but each of these was confounded by the symptomatic effect of the agent being tested, and none are approved for neuroprotection ( Table 6.2 ).
Year/reference | Trial | Agent | N | Primary outcome variable |
---|---|---|---|---|
1993; Parkinson Study Group | MAO-B inhibitor; DATATOP | Selegiline | 800 | Need for levodopa |
1994; Parkinson Study Group | MAO-B inhibitor; ROADS | Lazabemide | 321 | Need for levodopa |
1995; Olanow et al. | MAO-B inhibitor; SINDEPAR | Selegiline | 101 | Change in UPDRS |
2002; Shoulson et al. | MAO-B inhibitor; BLIND-DATE | Selegiline | 368 | UPDRS and freezing of gait |
2004a; Parkinson Study Group | MAO-B inhibitor; TEMPO | Rasagiline | 404 | Delayed-start UPDRS |
2004b; Parkinson Study Group | ELLDOPA | Levodopa | 361 | UPDRS after washout |
2006; Palhagen | MAO-B inhibitor; Swedish study | Selegiline | 157 | Need for levodopa and UPDRS |
2009; Olanow et al. | MAO-B inhibitor; ADAGIO | Rasagiline | 1176 | Delayed-start UPDRS |
2017; Athauda et al. | Glucagon-like peptide-1 agonist | Exenatide | 62 | MDS-UPDRS motor scores in practically defined “off” state |
Despite the fact that many of the drugs listed previously showed promise in animal models and in small early studies, each failed in larger controlled trials or were confounded by the effect of the drug in improving motor symptoms. In part, some of the reasons for these failures may be the lack of appropriate animal models for progressive PD, the heterogeneity of PD that suggests there may be different mechanisms of disease, and lack of a biomarker for progression of disease.
Several studies are currently in progress looking at newer agents to determine whether they have such a disease-modifying effect. Alpha-synuclein antibodies and other antisynuclein strategies are currently being investigated in clinical trials ( ; ).
Although exercise is important to everyone to maintain health, it may be particularly helpful in PD. However, despite the increasing literature citing the benefits of exercise, many do not participate in a regular exercise regimen. A cross-sectional study noted that the obstacles to exercise in PD include lack of certainty of the benefit, lack of time, and fear of falling during exercise ( ). Many types of exercise have been reported to improve the symptoms of PD and reduce falls ( , , ; ; ; ; ; ; ; ; ). In fact, tai chi, progressive resistance, treadmill, forced bicycle riding, tango, and many other forms of exercise have improved symptoms. However, most of these require regular exercise of approximately 3 to 4 times per week for at least 30 minutes. The benefits from exercise tend to wane if the patient fails to adhere to the program. Freezing of gait, which is a difficult parkinsonian motor feature to treat, was found to be reduced in a controlled clinical trial evaluating exercises with motor complexity – so-called adapted resistance training with instability ( ).
Physical therapy is one technique that can motivate patients to exercise. However, many become noncompliant after completion of the therapy. Physical therapy strategies can lead to improved activities of daily living (ADLs), improved gait, and treatment of musculoskeletal issues that are secondary to PD, including frozen shoulder and back pain.
PD leads to decreased motivation and increased passivity. An active exercise program, even early in the disease, can help overcome this. Furthermore, such a program involves patients in their own care, allows muscle stretching and full range of joint mobility, and enhances a better mental attitude toward fighting the disease. By being encouraged to take responsibility in fighting the devastations of the disease, the patient becomes an active participant. Physical therapy, which can be implemented in the form of a well-constructed exercise program, is useful in all stages of disease. In early stages, a physical therapy program can instruct the patient in the proper exercises and the regimen forces the patient to exercise if he or she lacks the motivation to exercise on his or her own. In advanced stages of PD, physical therapy may be even more valuable by keeping joints from becoming frozen and providing guidance how best to remain independent in mobility. Therefore, exercise is beneficial in both the early and later stages. It has been shown that PD patients who exercise intensively and regularly have better motor performance [ ; ; ; ; ] and quality of life [ ; ].
Three relatively new exercise programs have become popular with patients: Lee Silverman Voice Treatment (LSVT) BIG ( ), Rock Steady Boxing, and dancing. When patients are in an exercise community, it appears that they are more likely to continue to exercise and view it as beneficial. The motivation provided by a group session that is ongoing is likely one of the reasons that these are successful. At this time, there are few studies of these interventions
A number of reports suggest that exercise may be more than a symptomatic treatment. Investigators have determined that exercise may have positive effects on the underlying pathophysiologic processes of PD. In animal models, there is an increase in neurotrophic factors, reduced neuroinflammation, and a reversal of dendritic spine loss in the striatum ( ; ; ; ; ). Basic scientific studies have discovered that exercise, particularly enriched exercise, can reduce the loss of dopaminergic neurons after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) exposure. When such exercise is initiated shortly after rodents were given experimental lesions of the nigrostriatal dopamine pathway, the result was a significantly lesser amount of damage to the dopamine pathway ( , , ; ; ; ; ). The mechanism appears to be to the induction of increased trophic factors, such as GDNF ( ; ) and brain-derived neurotrophic factor (BDNF) ( ). Exercise also appears to reverse dendritic spine loss in direct and indirect striatal medium spiny neurons ( ) and to enhance neuroplasticity in motor and cognitive pathways ( ). An imaging study found that habitual exercisers had greater dopamine release in the caudate nuclei than do sedentary subjects when measured before and after stationary cycling ( ).
Treatment of patients with PD can be divided into three major categories: physical (and mental health) therapy, medications, and surgery. Physical exercise and physiotherapy were discussed previously. Speech therapy plays a similar role in those with problems of communication. Dysarthria, palilalia, and tachyphemia are difficult to treat, but hypophonia can be overcome by training the patient to shout, known as the LSVT ( ). In a controlled clinical trial, LSVT LOUD was found to be statistically superior to increasing speech amplitude and clarity than another form of speech therapy or no therapy ( ). Psychiatric assistance may be required to handle depression and the social and familial problems that can develop with this chronic, disabling illness. Electroconvulsive therapy (ECT) may have a role in patients with severe, intractable depression; some psychiatrists have been promoting it to help overcome the motor symptoms of PD, but at best ECT provides only short-term motor benefit, and it may not be replicated on repeat treatments.
The treatment of PD needs to be individualized; that is, each patient presents with a unique set of symptoms, signs, response to medications, and a host of social, occupational, and emotional problems that need to be addressed. Although early in the disease the treatment may be straightforward, with progression of PD the occurrence of motor complications, drug side effects, and nonmotor issues such as dementia, depression, and apathy, increases the complexity of decision making when selecting pharmacologic interventions. The major goal is to keep the patient functioning independently as long as possible. Determine what the issues are for the individual patient by asking the patient to list the specific symptoms that trouble him or her the most ( ). Attempt to treat the most troublesome symptoms; this is the way to maximize quality of life.
As will be emphasized below, the most effective pharmacotherapeutic agent for the motor features of PD is dopamine replacement with its precursor, levodopa. A major problem is that 75% of patients have serious complications after 6 years of levodopa therapy ( Table 6.3 ) ( ), and younger patients (less than 60 years of age) are particularly prone to develop the motor complications of fluctuations and dyskinesias ( ; ; ; ). As the result of these motor complications, a number of other pharmaceuticals (and surgeries) have been developed to either substitute for levodopa or treat the motor complications of levodopa.
N ∗ | % | |
---|---|---|
|
83 | 25 |
|
142 | 43 |
|
67 | 20 |
|
14 | 4 |
|
27 | 8 |
The International Parkinson and Movement Disorder Society (MDS) published a review of evidence-based efficacy on the various treatments for the motor features of PD ( ). Selection of the most suitable drugs for the individual patient and deciding when to use them in the course of the disease are challenges to the treating clinician. Because PD is a chronic progressive disease, patients require lifelong treatment. Medications and their doses will change over time as adverse effects and new symptoms are encountered.
It has been more than 50 years since the introduction of levodopa for the treatment of PD. This followed the discovery that the motoric symptoms of PD (bradykinesia, rigidity, tremor) are related to striatal dopamine deficiency ( ). , reviewed the discovery of levodopa as a treatment for PD. Dopamine replacement therapy remains the major medical approach to treating the motor features of the disease. Table 6.4 lists these dopaminergic drugs. The most powerful drug is levodopa, the immediate precursor of dopamine. Dopamine, being a charged molecule, is unable to enter the brain via the blood-brain barrier. Levodopa crosses the gut and the blood-brain barrier via specific transporters. These large amino acid transporters are also used by the aromatic amino acids that are present in many proteins, which can interfere with L-dopa transfer. Levodopa is absorbed only in the proximal small intestine ( ). Hence, slowing of gastric emptying and reduction in transit time will affect the absorption. The absorption of levodopa may be increased by eradicating gastric Helicobacter pylori with omeprazole, amoxicillin, and clarithromycin in PD patients documented to be infected with this bacterium ( ; ). About 50% of the general population is infected with the bacterium.
Dopamine precursor (usually in combination with decarboxylase inhibitor carbidopa or benserazide) Levodopa (Sinemet, Madopar) Extended-release levodopa (Sinemet CR, Rytary), inhalational levodopa (Inbrija), orally dissolving carbidopa/levodopa (Parcopa), intestinal gel (Duopa) Dopamine agonists ∗ Pramipexole (immediate and extended release) (Mirapex), ropinirole (immediate and extended release) (Requip), rotigotine dermal patch (Neupro), piribedil † , apomorphine (injectable as rescue [Apokyn], sublingual (Kynmobi) or continuous infusion † ) NMDA receptor antagonist/indirect dopaminergic effects Amantadine, amantadine extended release Levodopa-enhancing agents Catechol-O-methyltransferase inhibitors: entacapone, tolcapone, opicapone Type B MAO inhibitor: selegiline (Eldepryl), Zydis selegiline (Zelapar), rasagiline (Azilect), safinamide (Xadago) |
∗ Ergoline-derived dopamine agonists bromocriptine, pergolide, cabergoline, and lisuride are not used in the treatment of Parkinson disease resulting from cardiac valvular fibrosis.
Currently marketed | Common trade name |
---|---|
Standard formulation, also known as immediate-release | Sinemet |
Orally disintegrating tablets | Parcopa |
Extended-release formulation | Sinemet CR |
Extended-release microspheres | Rytary |
Intestinal gel | Duopa in United States; DuoDopa elsewhere |
Levodopa dry powder aerosol | Inbrija |
Under Development | Research Label |
Deuterated levodopa | D3-l-DOPA |
Gastroretentive carbidopa/levodopa swelling tablets | DM-1992 |
Gastroretentive Accordion Pill carbidopa/levodopa | AP CD/LD |
Soluble carbidopa/levodopa solution | ND-0612 |
Levodopa prodrug | XP21279 |
Once in the central nervous system (CNS), levodopa is metabolized to dopamine by aromatic amino acid decarboxylase (dopa decarboxylase) and is pharmacologically active at striatal dopamine receptors, in essence providing the dopamine that is depleted through the degeneration of dopamine-producing cells in the substantia nigra (SN). Levodopa in the periphery is frequently associated with nausea and vomiting as a result of its peripheral conversion by dopa decarboxylase to dopamine, which stimulates the area postrema (vomiting center), an area of the brainstem where the blood-brain barrier does not exist. Therefore, L-dopa is usually combined with a peripheral decarboxylase inhibitor ( ), which prevents the peripheral conversion of levodopa to dopamine and alleviates the peripheral side effects, including nausea and vomiting. Further, by reducing the metabolism of levodopa in the periphery, more levodopa is available to enter the CNS. The peripheral decarboxylase inhibitors benserazide and carbidopa potentiate levodopa, allowing about a four-fold reduction of levodopa dosage to obtain the same benefit. Typically, if starting carbidopa/levodopa, the 25/100 mg tablet is used, providing the highest ratio of carbidopa to levodopa. If additional carbidopa is needed for patients in whom nausea persists, it can be prescribed (Lodosyn) and patients can obtain it from their pharmacy. Carbidopa and benserazide are peripheral dopa decarboxylase inhibitors. In the United States the only dopa decarboxylase inhibitor available is carbidopa. The combination of the two (carbidopa/levodopa) was initially branded as Sinemet (Latin for “without emesis”) but is now available primarily as a generic preparation. In many other countries, benserazide is also available and in combination with levodopa. Because both drugs are azides, they are unable to penetrate the blood-brain barrier and do not interfere with the central effects of L-dopa. Tablets of carbidopa/levodopa are available as 10/100, 25/100 and 25/250 mg. The generic preparations differ in their formulations, and some patients find that one generic product may be superior to another. In non-U.S. countries, benserazide is combined with levodopa, with the brand name Madopar. Because L-dopa is a short-acting drug with a half-life of approximately 90 minutes, it has been reformulated to increase its half-life. Extended-release carbidopa/levodopa, with the brand name Sinemet CR, is a polymer release tablet formulation available as a 25/100-mg or 50/200-mg product with a more prolonged half-life. However, the CR preparation has time to peak level approximating 2 hours, compared with 30 minutes for L-dopa, making its onset of action delayed. In a clinical setting, the onset of action and clinical benefit profile may be problematic and unpredictable in a patient ( ). However, the extended-release formulation may be more helpful, particularly at night when wearing off of levodopa may result in awakening from sleep. If the tablets are cut in half, the duration is not as prolonged.
The second extended-release formulation of carbidopa/levodopa is Rytary, formally IPX066, which was approved by the U.S. Food and Drug Administration (FDA) in 2015. Rytary is provided in capsules filled with microbeads of carbidopa and levodopa, dissolving at different rates. Rytary capsules contain a combination of both extended- and immediate-release forms, with 32% being immediate release ( ). An important element in the capsule is the presence of a weak acid, tartaric acid, within its own extended-release microbead. An acidic environment keeps levodopa stable ( ). The goal was to make the onset of action more predictable and yet have the effect be longer lasting. This formulation is intended for PD patients with end-of-dose wearing off of benefit. In the pivotal clinical trials there was an increase of hours of “on time,” and reduction in “off time” ( ). However, the dose of levodopa was approximately 30% higher in the Rytary group. Rytary comes in multiple dosing formulations (carbidopa/levodopa 23.75 mg/95 mg, 36.25 mg/145 mg, 48.75 mg/195 mg, 61.25 mg/245 mg). As clinical studies have demonstrated, patients are able to reduce the number of times per day that the medication is taken, although often they must take several capsules at a single dose. Altering the dose (increasing or decreasing) may require multiple prescriptions. The side effect profile is similar to that of levodopa. Switching a patient from regular carbidopa/levodopa to Rytary can be challenging in the more fragile patient. In Europe, the brand name is Numient ( ). In the United States, Rytary is available only through specialty pharmacies.
A formulation of levodopa that was approved by the FDA in 2018 is the inhalational powder, CVT-301 (Inbrija) ( ; ). Inhalational levodopa was approved as a rescue therapy for patients who have wearing off of their oral levodopa. Inhalational levodopa has the advantage of not being dependent on the gastrointestinal system and is absorbed directly through the pulmonary system. It uses a breath-actuated delivery system and is administered through a specially designed inhaler. Patients using this treatment need training in its administration, including the method of inhalation and timing of dosing. Using CVT-301, plasma levodopa concentrations increased more rapidly and predictably compared with oral levodopa. ( ) A randomized study in 86 PD patients taking oral levodopa with more than 4 “off” periods per day and more than 2 hours of “off” time demonstrated that CVT-301 in addition to their usual levodopa improved motor scores, reduced “off” time, and demonstrated a time to “on” that was superior to placebo ( ). The main side effects were cough, nausea, and dizziness, affecting approximately 7% of those enrolled. A subsequent study demonstrated that there were no changes in pulmonary function with continued use of CVT-301 ( ). Currently, CVT 301 is approved by the FDA as Inbrija for intermittent treatment of “off” episodes in PD patients currently taking oral levodopa. It is available in 42-mg capsules, with 2 capsules per dose. It is recommended for use up to five times per day and is available only through a specialty pharmacy.
There is also a dispersible formulation of carbidopa/levodopa that dissolves in the mouth and is swallowed with saliva (Parcopa) and is available as generic equivalents. Dispersible carbidopa/levodopa can be taken without water, which may be an advantage for some patients, such as those who have trouble swallowing or who need to be without food or water before and after surgery.
Some undergoing clinical trials with formulations of levodopa are deuterium-labeled levodopa ( ), gastric-retention levodopa ( ; Verhagen ; ), subcutaneous infusion levodopa ( ; ), and a levodopa prodrug ( ).
Although levodopa is an effective symptomatic treatment for PD, it does not treat some of the motor features that develop in advanced PD, such as flexed and twisted dystonic postures, loss of postural reflexes (a major cause of falling), and freezing of gait. Levodopa does not prevent continued worsening of the disease itself, it has limited effectiveness against most of the nonmotor features of PD (e.g., cognitive decline, mood alterations, loss of motivation, autonomic dysfunction) (covered in Chapter 8 ), and it is associated with motor fluctuations (wearing off, unpredictable “off”) and dyskinesias (discussed later in a separate section). Because of the latter problems, other dopaminergic drugs have been developed (see Table 6.4 ).
The association of levodopa with malignant melanoma has been controversial. Levodopa is an intermediary metabolite in the synthesis of skin melanin (via the enzyme tyrosinase). Whether this action of levodopa gives rise to melanoma remains a question. A review of the literature does not provide evidence of a definite relationship between treatment with levodopa and the development or reemergence of malignant melanoma ( ; ; ). Epidemiologic studies have shown that people with PD have an increased prevalence of malignant melanoma ( ). A clinical trial in which the development of melanoma was a secondary outcome measure showed that patients with PD on the placebo arm of the trial had a much higher rate of developing malignant melanoma than would have been predicted; no association between levodopa therapy and the incidence of melanoma was found ( ). Yet it would seem prudent not to treat with levodopa in patients with a history of a malignant melanoma if other antiparkinson agents remain effective. Once it becomes necessary to use levodopa to improve quality of life, these patients need to be informed of this potential risk and should be examined regularly by their dermatologist or oncologist.
Besides being metabolized by aromatic amino acid decarboxylase (often called dopa decarboxylase), levodopa is also metabolized by catechol-O-methyltransferase (COMT) to form 3-O-methyldopa. Three COMT inhibitors are currently available: tolcapone, entacapone, and opicapone. These agents extend the plasma half-life of levodopa with only a small increase in its peak plasma concentration, and can thereby prolong the duration of action of each dose of levodopa. These drugs are used in conjunction with levodopa to reduce the wearing-off effect ( ), a common motor fluctuation adverse effect of levodopa therapy. The net effect with multiple dosings per day, though, is to elevate the average plasma concentration while smoothing out the variations in the concentration. Tolcapone has two potential adverse effects that need to be explained to the patient. The most serious is that a small percentage of patients will develop elevated liver transaminases, and patients need to have baseline and follow-up liver function tests. Death from hepatic necrosis has occurred in three patients who had no liver function surveillance ( ). Entacapone has not been shown to induce these hepatic changes. In addition with tolcapone, a small percentage of patients will develop diarrhea that does not appear for about 6 weeks after starting the drug. The diarrhea can be explosive, so the patient might not have any warning. Entacapone appears not to have these adverse effects. Many clinicians think that tolcapone is more effective than entacapone in reducing motor fluctuations, but one should not prescribe the former unless the latter has not been effective in relieving wearing off. Patients on entacapone can be easily switched to tolcapone if the former had less than the desired effect, and a double-blind comparison showed tolcapone to be slightly more effective in reducing the amount of “off” time ( ). If tolcapone is to be used, we advise starting at a low dose of 100 mg/day and increase gradually to 100 mg three times daily. Entacapone has a short half-life, so a dose of 200 mg is taken with each dose of levodopa ( ). One formulation (Stalevo) combines carbidopa, levodopa, and 200 mg entacapone into a single tablet; the dose of a tablet of Stalevo represents the amount of levodopa in the tablet.
Adding entacapone to patients on levodopa who are not experiencing motor fluctuations did not add any improvement to motor performance in one study ( ) but improved the ADLs score in another ( ). In the FIRST-STEP study, levodopa/carbidopa/entacapone (LCE) 100/25/200 mg 3 times daily, was compared with levodopa/carbidopa (LC) 100/25 mg 3 times daily in patients with early PD for 39 weeks ( ). LCE treatment resulted in slightly better Unified Parkinson’s Disease Rating Scale (UPDRS) Part II ADLs scores ( P = 0.025) but not Part III motor scores.
The concept that intermittent brain levels of levodopa and dopamine contribute to the development of motor complications (see later in discussion of advanced PD) has led to the concept that continuous dopaminergic stimulation may avoid these complications from levodopa. So far, one study (STRIDE-PD) testing this hypothesis has yielded the opposite effect, that is, an earlier onset of dyskinesias ( ). A total of 747 patients with early PD were randomized to LCE 100/25/200 mg or LC 100/25 mg with flexible dosing to reach 400 mg/day, with a dose 3.5 hours apart. The results showed that time to dyskinesia was statistically significantly shorter in LCE-treated patients compared with LC-treated patients. The incidence of dyskinesia during the study period was higher in LCE-treated patients in comparison to the LC group.
The next most powerful drugs, after levodopa for the treatment of PD motor symptoms are the dopamine agonists. Available dopamine agonists for the treatment of PD are listed in Table 6.4 . The ergoline direct dopamine agonists are no longer considered a treatment for PD because of their association with cardiac valve fibrosis. Cabergoline is the longest acting and could be taken just once per day ( ; ); it theoretically could be most important in terms of preventing or reducing the wearing-off effect because of its long duration of action, but it has not been tested for this use. Piribedil is relatively weak but has been touted as having an antitremor effect.
Apomorphine and rotigotine cannot be absorbed with oral administration. Rotigotine can be absorbed transdermally and is marketed as an applied skin patch ( ; ; ; ; ), which is useful for patients unable to swallow medications, such as after gastrointestinal surgery. Despite its continuous absorption by transdermal patch, rotigotine did not prove effective in clinical trials in combating the wearing-off problem that is commonly seen with levodopa therapy. It takes higher doses of rotigotine to reduce “off” time ( ). Rotigotine is a relatively weak dopamine agonist. Apomorphine needs to be injected subcutaneously or absorbed sublingually. It is the strongest agonist and can mimic the effectiveness of levodopa. In Europe it is often used as a subcutaneous infusion to overcome the wearing-off problem seen with levodopa therapy. In the United States it is marketed as single intramuscular injections and as a sublingual film to rescue patients who are in a deep “off” state; clinical trials on the efficacy and safety of subcutaneous infusions are under way in the United States. The FDA failed to approve sublingual administration of apomorphine in 2019 and requested further information, particularly related to oral irritation. In the pivotal trial involving 109 patients who were randomly assigned to receive apomorphine sublingual film (n = 54) or placebo (n = 55), there was a significant and clinically meaningful reduction in the MDS-UPDRS Part III score between predose and 30-minute postdose at week 12 with sublingual apomorphine compared with placebo ( ). Besides oropharyngeal irritation, other treatment-emergent adverse events included somnolence and dizziness. The FDA ultimately approved sublingual apomorphine on May 21, 2020 (Press Release, 2020).
The other dopamine agonists in Table 6.4 are effective orally. Bromocriptine is the weakest clinically compared to the others. Pergolide, pramipexole, and ropinirole appear to be comparable in clinical practice, but some patients will respond better to one than the others. There are some differences of these agonists in their affinity for the dopamine receptor subtypes, as depicted in Tables 6.6 and 6.7 . Only apomorphine (strong) and pergolide (modest) have agonist activity at the dopamine D1 receptor. The activation of the D2 receptor is known to be important in obtaining an anti-PD response, whereas it is unknown how important D3 receptor activation is for improving the anti-PD response. Bromocriptine, pergolide, pramipexole, and ropinirole activate the dopamine D3 and D2 receptors, but their ratios of affinities for these two receptors are different (see Table 6.7 ) ( ). All dopamine agonists are less likely to induce dyskinesias compared with levodopa ( ). The agonists can be used as adjuncts to levodopa therapy (e.g., ; ) or as monotherapy (e.g., ; ; ; ; ). Adverse effects that are more common with dopamine agonists than with levodopa are impulse control disorders ( ), drowsiness, sleep attacks, confusion, orthostatic hypotension, nausea, and ankle/leg edema associated commonly with erythema ( ; ). Edema can spread to involve other areas of the body, including the arms and face.
Agonist | D1 | D2 | D3 | D4 | D5 |
---|---|---|---|---|---|
Bromocriptine | — | ++ | ++ | + | + |
Lisuride | + | ++ | ? | ? | ? |
Pergolide | + | ++ | +++ | ? | + |
Cabergoline | — | +++ | ? | ? | ? |
Ropinirole | — | ++ | ++++ | + | — |
Pramipexole | — | ++ | ++++ | ++ | ? |
Agonist | D1 | D2 | D3 | D2/D3 ratio |
---|---|---|---|---|
Bromocriptine | 0 | +++ | ++ | 10:1 |
Pergolide | + | +++ | +++ | 1:1 |
Ropinirole | 0 | ++ | +++ | 1:10 |
Pramipexole | 0 | ++ | +++ | 1:10 |
Apomorphine, being water soluble and injectable, can be employed as a rapidly acting dopaminergic agent to overcome “off” states, that is, provide a rescue. It is either injected, infused subcutaneously or absorbed sublingually. Because of its emesis-producing propensity, the patient must be pretreated with an antinauseant, such as domperidone or trimethobenzamide. In the United States, apomorphine is FDA-approved, so far, for subcutaneous injections to overcome an “off” episode. In Europe it is also being used by continuous subcutaneous infusion to provide a smooth response for patients who fluctuate between dyskinetic and “off” states. Apomorphine is the most powerful of the dopamine agonists and activates both the dopamine D1 and D2 receptors.
Having several dopamine agonists to choose from allows the opportunity to find one that is better tolerated and that might have a better effect. Adverse effects may be the deciding factor as to which drug will work best for a patient. Unfortunately, all of these drugs can induce confusion and hallucinations in elderly patients. Leg edema occurs in some patients, usually after a few years. Pramipexole, ropinirole, and other dopaminergics, though with probably less frequency, can cause daytime sleepiness and sleep attacks (sudden falling asleep without warning of drowsiness). This could be dangerous for the patient who drives an automobile, and motor vehicle accidents have occurred when patients fell asleep at the wheel ( ; ; ; ). So when deciding to place a patient on pramipexole or ropinirole, the physician should determine the extent of driving to be done by the patient, and warn the patient about this potential hazard. Short trips (e.g., 10 minutes or so) should be without risk. Should sudden falling asleep occur in any nondriving activity, this event can serve as a warning against driving or else it would be best to taper and even discontinue these medications if driving is necessary. Dopamine agonists also are more likely to induce impulse control problems, such as gambling, hypersexuality, shopping, and binge eating ( ) (see Chapter 8 ). The dopamine agonist withdrawal syndrome, in which symptoms of withdrawal are sustained, is another risk with these drugs ( ). The risk for impulse control problems with dopamine agonists has led many neurologists to shy away from using these drugs in favor of levodopa.
Amantadine has several actions, including antimuscarinic, that block dopamine reuptake from dopaminergic synapses. It is also a noncompetitive antagonist of the N-methyl-D-aspartic acid (NMDA) glutamate. Its dopaminergic action make it a useful drug to modestly reduce symptoms of PD, although this symptomatic effect tends to be transient, lasting a few months. The amantadine side effect profile includes livedo reticularis, ankle edema, visual hallucinations, and confusion, the latter two mainly in older individuals or those with a cognitive deficit. Amantadine is more commonly used to help overcome levodopa-induced dyskinesias. So far, it is the only drug that has shown this ability without worsening parkinsonism. Amantadine is also available in an extended-release formulation with the brand name Gocovri. This preparation of long-acting amantadine has been FDA-approved for reducing levodopa-induced dyskinesia. It is taken one time a day in the evening. During the night, the levels of amantadine increase and reach a stable plasma level during the day. The pivotal clinical trial demonstrated that there was an increase in “on” time of up to 2 hours compared with placebo and a reduction in dyskinesia. The side effect profile is similar to that of amantadine, as noted earlier ( ; ).
Domperidone is a peripherally active dopamine receptor blocker and is useful in preventing gastrointestinal upset from levodopa and the dopamine agonists. Although it does not enter the CNS, it can still block the dopamine receptors in the area postrema, thereby preventing nausea and vomiting. By not penetrating the CNS, it does not block the dopamine receptors in the striatum, thus not interfering with the action of dopamine or dopamine agonists. Domperidone is available in most countries world-wide, but is not marketed in the United States.
Monoamine oxidase (MAO) inhibitors (MAOIs) offer mildly effective symptomatic benefit. MAO is composed of two isoenzymes, MAO-A and MAO-B, which are located in the mitochondria; each has selective substrates that it deaminates and oxidizes. In brain, MAO-A acts on serotonin and norepinephrine; both MAO-A and MAO-B act on dopamine. MAO-A is located in neurons and MAO-B in glia. In peripheral organs, tyramine is a substrate for MAO-A, but not MAO-B ( ). The original MAOIs developed for their antidepressant effects (e.g., phenelzine and tranylcypromine) were associated with the “cheese effect” (hypertensive crisis). These agents are nonselective and inhibit both MAO-A and MAO-B activity ( ). The cheese effect is due to the inhibition of MAO-A in the intestinal tract; thus, the gut is unable to deaminate ingested tyramine, allowing tyramine to be absorbed into the body. Tyramine is present in certain food substances, particularly fermented meats and cheeses, soy sauce, and red wine. A high-tyramine meal results in absorption of tyramine from the gut. Tyramine is taken up into noradrenergic nerve terminals and acts as a false neurotransmitter, displacing norepinephrine from nerve terminals ( ). This excess release of norepinephrine from the nerve terminals produces the hypertensive crisis. The relatively selective MAO-B inhibitors selegiline, rasagiline, and safinamide, in the recommended therapeutic dosages, do not cause a hypertensive crisis, but they offer little antidepressant effect either. However, excessively high doses of these drugs will also inhibit MAO-A, so higher than the recommended doses could result in serious side effects.
Selegiline and rasagiline are both irreversible, selective MAO-B inhibitors with a similar chemical structure; both are propargylamine compounds. Safinamide is a reversible MAO-B inhibitor. None of these compounds have been compared head-to-head with each other to determine their relative effectiveness. The standard dose is 1 mg/day for rasagiline, 10 mg/day for selegiline, and 50 to 100 mg/day for safinamide. At these doses, the drugs are highly selective for MAO-B and do not require the patient to be on a low-tyramine diet. A controlled tyramine challenge showed that rasagiline up to 2 mg/day did not induce a significant blood pressure or pulse change when tyramine was added ( ). All three drugs are approved as adjunct therapy with levodopa. Their action is to enhance the action and prolong the duration of levodopa effect and thus reduce the wearing off.
Selegiline and rasagiline have mild symptomatic effects when used alone ( , , , , ; ; ). These two drugs are commonly used as initial pharmacotherapy for PD. Safinamide was reported not to be efficacious in reducing PD symptoms in the absence of levodopa ( ). When used in the presence of levodopa therapy, the MAO-B inhibitors potentiate levodopa’s effect ( ). The selective MAO-B inhibitors can be used safely with selective serotonin uptake inhibitors without producing the serotonin syndrome, provided the dosages are in the approved range of MAO-B selectivity and to avoid inhibiting MAO-A ( ; ).
Zydis selegiline is a form of selegiline that dissolves in the mouth and is absorbed through the oral mucosa, avoiding first pass metabolism in the liver ( ). Zydis selegiline is formulated as freeze-dried tablets and is marketed as Zelapar. High doses of zydis selegiline can inhibit MAO-A in the brain ( ) and by bypassing the intestinal tract avoid the cheese effect.
Selegiline is also available in a dermal patch (marketed as Emsam), in a dosage high enough to also inhibit MAO-A in the brain. Emsam is marketed as an antidepressive agent. Emsam does not cause the cheese effect because selegiline was absorbed transdermally, bypassing the gut and thereby avoiding the inhibition of intestinal MAO-A ( ). However, this is not approved for use in PD.
Rasagiline is metabolized to aminoindan, selegiline to L-amphetamine and L-methamphetamine, and safinamide to its acid form. Their effects in treating clinical fluctuations and as possible disease-modifying agents are discussed later in the appropriate sections (Treatment of motor fluctuations associated with levodopa therapy and Selegiline, rasagiline, and antioxidants).
Inhibitors of both MAO-A and MAO-B would offer greater inhibition of dopamine oxidation in the brain, and thus the combination would theoretically be more capable of reducing oxidative stress and providing more symptomatic effect ( ). But tranylcypromine and phenelzine (both nonselective inhibitors of MAO-A and MAO-B) cannot be taken in the presence of levodopa therapy because of the cheese effect, and even in the absence of levodopa, patients on these drugs need to adhere to a reduced-tyramine diet ( ).
We will return to discuss MAOIs and antioxidants later in their possible role in treating early-stage PD. Next, we will review the nondopaminergic drugs that are useful in treating the motoric problems of PD ( Table 6.8 ).
Antimuscarinics: trihexyphenidyl, benztropine, ethopropazine, etc. Antihistaminics: diphenhydramine, orphenadrine Antiglutamatergics: amantadine, dextromethorphan, riluzole Benzodiazepines: alprazolam, lorazepam, diazepam Muscle relaxants: cyclobenzaprine, diazepam, baclofen Adenosine A 2A receptor antagonist: istradefylline Neurotrophins: neuroimmunophilins, GDNF, and neurturin all failed in clinical trials Antioxidants: tocopherol and coenzyme Q10 failed in phase III clinical trials |
COMT inhibitors (COMTIs) are useful only in the presence of levodopa therapy. COMT catalyzes the methylation of catechols (e.g., levodopa and dopamine). By inhibiting this enzyme peripherally, the half-life of levodopa is lengthened somewhat. Drugs in this class are entacapone, tolcapone, and opicapone. Opicapone is a long-acting drug, taken at bedtime; it was approved by the FDA in 2019. Opicapone was approved by the FDA in 2019. All three are discussed in more detail in the section related to the treatment of the wearing-off effect, for which these drugs can be useful.
Some nondopaminergic agents (see Table 6.8 ) are also useful to treat motoric PD symptoms, particularly antimuscarinic drugs (commonly referred to as anticholinergics), which have been widely used since the 1950s, but these are much less effective than the dopaminergic agents, including amantadine. The original use of antimuscarinic alkaloids date back to the time of Charcot, and synthetic agents became available and popular in the 1950s (see review by ). Antimuscarinic drugs are somewhat helpful in reducing all symptoms of PD, but they have found special favor in reducing the severity of tremor. However, because of sensitivity to memory impairment in all groups and hallucinations in the elderly population, antimuscarinic drugs usually should be avoided in patients over the age of 70 years. The antihistaminics (diphenhydramine, orphenadrine, and others), tricyclics (amitriptyline and others), and cyclobenzaprine (Flexeril) have milder anticholinergic properties that can make them useful in PD, particularly in older patients who should not take the stronger anticholinergics because of their propensity to impair short-term memory at that age level.
Amantadine, listed in Table 6.4 as a dopaminergic agent, is listed also in Table 6.8 because it has antiglutamatergic effects; this property appears to account for its usefulness in reducing choreic dyskinesias induced by levodopa ( ; ; ; ). An extended-release form of amantadine was found to be effective to reduce levodopa-induced dyskinesias in a controlled clinical trial ( , ) and has been approved by the FDA for this indication. Dextromethorphan is another antiglutamatergic agent, and it has been found effective in reducing the severity of dyskinesias by 50% ( ).
Another useful class of drugs is the benzodiazepines to decrease parkinsonian tremor that is exacerbated by stress. These drugs reduce anxiety and by calming a patient provide a lessening of tremor. Diazepam is usually well tolerated and does not exacerbate parkinsonian symptoms, whereas chlordiazepoxide can ( ). Lorazepam and alprazolam are other useful benzodiazepine agents; the latter has the added benefit of being short-acting, so it can be used when needed. It also has some antidepressant effects. The benzodiazepines can cause drowsiness, so it is best to start with a small dose and increase it if tolerated. The muscle relaxants listed in Table 6.8 might help in treating “off” and peak-dose dystonias. Because oxidative stress appears to play a role in the pathogenesis of PD, high dosages of antioxidant vitamins, tocopherol, and coenzyme Q10 have been tried for patients with PD and failed in clinical trials ( ; ).
Stress, excitement, and anxiety make parkinsonian symptoms worse, especially tremor. In fact, tremor that is otherwise well controlled with medication can reemerge under stress, excitement, and anxiety. The benzodiazepines, by reducing anxiety, can partially offset this worsening of tremor. Apathy and fatigue are common in PD, and no medication as yet has been found satisfactory.
Adenosine A 2A receptors are located on gamma-aminobutyric acid (GABA) neurons in the striatum and antagonize the effect of dopamine on these neurons ( ). Antagonizing adenosine A2A receptors has a behavioral effect similar to enhancing dopaminergic transmission. Several of these receptor antagonists ( Table 6.8 ) have undergone clinical trials for patients with motor fluctuations ( ; ; ; ; ), but the results were mixed, with insufficient relief of fluctuations and enhanced dyskinesias ( ). However, after further clinical trials, istradefylline was found effective in reducing clinical fluctuations ( ) and was approved for this indication by the FDA in 2019 under the label of Nourianz. Because of its favorable side effect profile, istradefylline may be particularly useful in elderly patients with troublesome motor fluctuations who may have comorbid depression or cognitive impairment ( ).
Various sleep problems are encountered in PD. Excessive drowsiness can occur after a dose of levodopa or dopamine agonist. Modafinil and methylphenidate can sometimes help overcome this problem. Insomnia needs to be treated, otherwise quality of life suffers and daytime sleepiness is enhanced. For insomnia, mild hypnotic drugs such as melatonin, antihistamines, trazodone, and mirtazapine can be used with satisfying results. Stronger hypnotics, such as zolpidem and benzodiazepines, can be safely used if the milder drugs are not effective.
The earliest stage of PD begins when the first motor symptoms are noticed and the diagnosis is made. At this stage, symptoms are mild and there is no threat to the patient’s activities. The designation of “early stage” lasts until the symptoms begin to become troublesome to the patient or quality of life is affected, and intervention with symptomatic medications is needed. All symptomatic drugs can induce side effects, and if a patient is not troubled by mild symptoms socially or occupationally, the introduction of these drugs can be delayed until symptoms become more pronounced. The clinician needs to discuss this choice with the patient and the patient’s family. Levodopa, the most effective symptomatic drug, failed to show evidence of a neuroprotective effect in a delayed-start design clinical trial ( ) ( Fig. 6.3 ). Most neurologists do not use levodopa or other potent antiparkinson agents when the diagnosis is first established, and the disease manifests with no threat to physical, social, or occupational activities ( , ; ).
Because symptomatically beneficial medications are not needed, and because there is no proven neuroprotective treatment, patients in the early, recently diagnosed stage of PD are excellent candidates for participating in a controlled clinical trial in which a placebo is one of the treatment arms. In the past decades, several literature reviews of clinical trials related to neuroprotection in PD have been published ( ; ; and Suchowersky et al., 2006). The MAO-B inhibitors have figured largely in clinical trials for neuroprotection (disease-modification).
As can be seen from Table 6.2 , the MAO-B inhibitors that have been tested in neuroprotection trials were all able to slow the rate of worsening of the motor features of PD. The irreversible inhibitors, selegiline and rasagiline are available commercially, but even lazabemide, a reversible MAO-B inhibitor, never marketed, also was effective. Not a single study with an MAO-B inhibitor failed a clinical trial in being superior to a placebo. The major question is whether this effect is all from its symptomatic effect or some of its benefit is from a neuroprotection (disease-modification) effect.
The first controlled clinical trial for the purpose of evaluating medications as neuroprotective agents for PD was the test for selegiline in the Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study ( , , ). Selegiline was called deprenyl at the time the trial was begun. Selegiline is a propargyline drug and has a long duration of action (MAO-B inhibition half-life of 40 days) ( ). The DATATOP study evaluated the antioxidant, alpha-tocopherol (vitamin E), along with selegiline in a 2 × 2 design. Patients were enrolled in the study early in the course of their illness and did not require symptomatic therapy. They were placed on selegiline (5 mg twice daily), alpha-tocopherol (1000 IU twice daily), the combination, or double placebo, with approximately 200 subjects in each of the four treatment arms. The primary endpoint was the need for dopaminergic therapy, that is, levodopa. The study showed that tocopherol had no effect in delaying parkinsonian disability, but selegiline delayed symptomatic treatment by 9 months ( Fig. 6.4 ) ( ). It also reduced the rate of worsening of the UPDRS by half ( Table 6.9 ). Other investigators subsequently conducted other studies testing selegiline, showing similar beneficial results ( ; ; , ).
Treatment | Total UPDRS |
---|---|
Placebo | 14.02 ± 12.32 |
Tocopherol | 15.16 ± 16.12 |
Selegiline | 7.00 ± 10.76 |
Tocopherol and selegiline | 7.28 ± 11.11 |
P Value | <0.001 |
Because selegiline has a mild symptomatic effect that is long lasting ( ), one might explain its ability to delay progression of disability entirely on this symptomatic effect. In favor of some neuroprotective effect is that after 2 months of washout of the drug, patients had slightly milder PD than did those on placebo ( ). But because of selegiline’s very long duration of action as an inhibitor of MAO-B ( ), this observation could represent an insufficient washout period. Furthermore, selegiline’s benefit in delaying the introduction of levodopa gradually diminishes over time ( ), with the best results occurring in the first year of treatment. The odds ratio increased from 0.35 for the first 6 months, to 0.38 in the second 6 months, to 0.77 in the third 6 months, and to 0.86 after 18 months. Follow-up of DATATOP subjects showed that placebo-treated subjects fared better than selegiline-treated subjects when the drug was reintroduced after a 2-month washout period and that the two groups were identical in developing levodopa complications ( ). The net understanding by the year 2000 was that there is no convincing evidence that selegiline delayed the need for levodopa because of any protective effect; all results could be those of a drug with a continuing mild symptomatic benefit.
On the other hand, basic scientific research was finding that in animal models tiny doses of selegiline have a neuronal rescue effect ( ). This effect is separate from its MAO inhibitor mechanism of action. Ultimately this finding led to investigations of other agents for their rescue effect, resulting in the discovery of a propargyline drug without MAO inhibition (TCH346) that was tested in a clinical trial ( , ). This drug failed to be better than placebo ( ).
When the results of the DATATOP study were analyzed to better understand the development of freezing of gait, it was discovered that the group who were treated with selegiline had a statistically significantly decreased risk for developing freezing of gait ( Fig. 6.5 ) ( ). It could not be discerned whether this benefit was because of selegiline’s mild symptomatic benefit or of some unknown neuroprotection effect. Whichever it was, the authors concluded that one should consider using selegiline in patients who are likely to develop freezing of gait those with (absence of tremor, gait involvement as the initial symptom).
Subsequently, based on the BLIND-DATE study, there is evidence that the decreased risk for freezing of gait with selegiline is not simply from its symptomatic effect as an enhancer of dopamine. The investigators of the DATATOP study, while continuing to follow their subjects, carried out a rerandomization of the subjects into a new controlled trial (called the BLIND-DATE study). A total of 368 subjects who were now taking both selegiline and levodopa therapy agreed to be randomized to either selegiline or placebo while remaining on open-label levodopa. The results were dramatic. Over an almost 2-year period, the subjects on selegiline required a lower dosage of levodopa, had a slower rate of worsening of symptoms and signs of PD ( Table 6.10 ), and had less freezing of gait ( Fig. 6.6 ) ( ). These results support the view that selegiline does provide some neuroprotective effect or else it has a symptomatic effect separate from a dopaminergic effect. The possibility that this benefit is derived from an antiapoptotic effect rather than its antioxidative effect is discussed later.
Duration after randomization | Placebo | Selegiline | Difference |
---|---|---|---|
1 month | 0.50 ± 7.73 | –1.52 ± 7.54 | 2.02 |
3 months | 1.57 ± 9.41 | –0.85 ± 9.42 | 2.42 |
9 months | 4.18 ± 10.12 | 1.63 ± 10.61 | 2.55 |
15 months | 5.63 ± 10.73 | 0.46 ± 10.88 | 5.17 |
21 months | 7.06 ± 12.70 | 1.51 ± 10.36 | 5.55 |
↑ L-dopa mg/day | 181 ± 246 | 106 ± 205 | P = 0.003 |
A similar study was carried out in Sweden by Palhagen and colleagues (2006), who followed patients for at least 7 years after they entered a controlled clinical trial evaluating selegiline versus placebo in those with early, untreated PD. Then, when any subject required symptomatic therapy, open-label levodopa was added while the blind was maintained on selegiline versus placebo. During the 7 years of follow-up from the start of the study, the selegiline-treated group had a statistically significantly slower rate of worsening of clinical signs and symptoms as measured by UPDRS scores. Like the study by Shoulson and colleagues (2002) mentioned earlier, this also shows the added benefit that selegiline provides in slowing clinical symptoms. Whether this can be attributed to a neuroprotective effect or to a symptomatic effect that does not appear to be through dopamine is undetermined by the two studies.
The safety of selegiline was raised, though, in an open-label clinical trial in the United Kingdom ( ). The use of selegiline when combined with levodopa was reported to be associated with a higher mortality rate than was seen in the patients assigned to levodopa treatment alone. Analysis of this result by others found a number of flaws in the study to refute this conclusion ( ). The U.K. investigators followed up their report with a more detailed analysis of the cause of death (Ben- ). The excess mortality in the selegiline plus levodopa group was greatest in the third and fourth years of treatment. The cause of the increase in deaths showed the excess to be from PD only and to occur particularly in patients with dementia and a history of falls. No significant differences in mortality were found for revised diagnosis, disability rating scores, autonomic or cardiovascular events, other clinical features, or drug interactions. Other studies with selegiline have failed to find any excess mortality from the combination treatment with levodopa ( ; ; ). After being followed by the Parkinson Study Group an average of 8.2 years, the subjects in the DATATOP study showed no difference in mortality between the groups assigned to treatment with selegiline, tocopherol, or placebo; the death rate averaged 2.1% per year ( ), much lower than in the U.K. study.
A meta-analysis of 17 controlled clinical trials involving MAO-B inhibitors found that no significant difference in mortality existed between patients on the inhibitors compared with control subjects ( ). The analysis also found that subjects randomized to MAO-B inhibitors had significantly better total scores, motor scores, and ADL scores on the UPDRS at 3 months compared with patients taking placebo; they were also less likely to need additional levodopa or to develop motor fluctuations. No difference existed between the two groups in the incidence of side effects or withdrawal of patients.
High-dosage tocopherol also has been suggested to increase mortality, but analysis of the DATATOP cohort followed for up to 13 years failed to find any difference in mortality between the groups on vitamin E and the group on placebo ( ).
In an analysis of retrospective observational data from Scotland ( ) comparing PD patients with a comparable control population, the patients with PD had a higher rate of mortality than those without PD (rate ratio [RR], 1.76; 95% confidence interval [CI], 1.11–2.81). There was significantly greater mortality in monotherapy (RR, 2.45, 95% CI, 1.42–4.23) relative to the comparators, adjusting for previous cardiovascular drug use and diabetes. However, there was no significant difference in mortality in patients with PD who received combination therapy of selegiline with levodopa and other drugs in relation to the comparators (RR, 0.92, 95% CI, 0.37–2.31). Thus, from this study, selegiline did not increase the mortality rate, whether used as monotherapy or in combination with levodopa. In fact, levodopa monotherapy had the highest mortality rate.
Rasagiline, based on the delayed-start studies, TEMPO ( ) and ADAGIO ( ), also can reduce the rate of clinical worsening in patients with early PD. But there were inconsistent outcomes between these two studies. In TEMPO, the 2-mg dose of rasagiline had a superior result compared with the 1-mg dose. In ADAGIO, only the 1-mg dose was superior to placebo; the 2-mg dose was no better than placebo ( Fig. 6.7 ). Starting a symptomatically effective drug early does not automatically lead to a reduced rate of clinical worsening as tested by the delayed-start design. For example, pramipexole, an effective dopaminergic agent, does not give a superior clinical result if started early compared with starting it later ( ). Thus, there would appear to be a special property of the MAOIs to be able to delay clinical worsening in a delayed-start study.
As mentioned earlier, the dose of selegiline and rasagiline should not exceed their specificity as selective type B inhibitors of MAO. Selegiline greater than 10 mg/day and rasagiline greater than 2 mg/day will also inhibit MAO-A. In one example, a woman given rasagiline at 4 mg/day in the presence of levodopa therapy developed the serotonin syndrome of hyperpyrexia, confusion, agitation, and episodic periods of unconsciousness ( ). Because selegiline is metabolized to L-amphetamine and L-methamphetamine, insomnia could develop and one should avoid taking it late in the day. It may be necessary to limit selegiline to 5 or 10 mg in the morning only if insomnia is a problem. Male impotence is not common with MAO inhibitors. In the presence of levodopa, MAOIs potentiate levodopa’s effect, and lower doses of levodopa can usually be achieved ( ; ). Selegiline does not prevent the development of levodopa-induced complications of fluctuations and dyskinesias ( ). Selegiline decreases the risk for patients developing freezing of gait ( ; ). It is not clear if rasagiline has this ability. Interestingly, MAO-A inhibitors, but not MAO-B inhibitors, have been shown to reduce stress-induced freezing behavior in rats ( ).
The DATATOP study showed that selegiline inhibits MAO activity by about 20% in the CNS ( ). Because the original premise for the DATATOP study was that selegiline might be neuroprotective by inhibiting MAO (reducing formation of hydrogen peroxide and thereby decreasing oxidant stress), the CSF analysis of homovanillic acid indicates that selegiline is a poor inhibitor of CNS MAO. This finding could explain the lack of success of selegiline as a powerful neuroprotective agent. Whether a more potent MAOI could be more successful remains to be determined. In the meantime, it is reasonable for patients to consider an inhibitor of both types A and B, as possibly augmenting inhibition of MAO in brain. However, such MAOIs can induce the cheese effect, so a low-tyramine diet needs to be followed. Such MAOIs can be used only in the absence of levodopa because the presence of levodopa will create marked blood pressure fluctuations.
Another MAO-B inhibitor, safinamide, offers modest symptomatic benefit of PD ( ; ; ), but it has not been tested for a potential neuroprotective effect. Safinamide is a reversible MAO-B inhibitor, in contrast to selegiline and rasagiline, which are irreversible inhibitors. Safinamide also blocks sodium voltage-sensitive channels and modulates stimulated release of glutamate ( ). Thus, there may other clinical benefits or adverse effects from this drug. It was approved by the FDA in 2017 as an add-on drug for treating the wearing-off effect.
Dopamine agonists are the next most powerful drugs after levodopa in reducing the motor symptoms of PD. Controlled clinical trials compared four different dopamine agonists against levodopa in patients with PD who were in need of symptomatic therapy: cabergoline and levodopa ( ), ropinirole and levodopa ( ), pramipexole and levodopa (the CALM-PD trial) ( ), and pergolide and levodopa ( ). The clinical outcomes of these studies are discussed later.
In this section, the results of the neuroimaging component of these trials are discussed. In the CALM-PD (pramipexole versus levodopa) trial, the 4-year imaging results show a statistically significant lesser rate of decay of dopamine transporter (DAT) binding (β-CIT SPECT) (a marker of integrity of nerve terminals of the dopaminergic nigrostriatal fibers) in the striatum in the group originally assigned to pramipexole treatment ( Fig. 6.8 ) ( ). A separate study evaluating FDOPA positron emission tomography (PET) scans, a marker of dopa uptake and dopa decarboxylase activity, showed a similar statistically significant lesser rate of decay of labeling in the striatum in a controlled trial in the group assigned to ropinirole compared with the group assigned to levodopa therapy ( ).
Because there was no placebo comparator in either study, interpretation is difficult. Whether dopamine agonists slow the rate of progression of PD, whether levodopa hastens it, and whether both explanations are playing a role are possibilities. Another possibility would be a pharmacodynamic effect on the DAT and dopa decarboxylase by either the agonists or levodopa. For example, if levodopa downregulated the DAT, β-CIT SPECT binding would be reduced. If levodopa downregulated dopa decarboxylase, FDOPA PET binding would be reduced. Short trials of levodopa showed no change in these imaging markers, so there is no evidence that levodopa affects either type of imaging study in such a pharmacologic manner. But a consensus conference concluded that there is insufficient information about the effect of medications on dopaminergic imaging to recommend neuroimaging as a biomarker for disease progression in the presence of medication ( ). Without knowing whether the agonists actually slow the rate of progression, it is not possible to recommend the starting treatment on the basis of these results alone. Pramipexole was later tested in a delayed-start clinical trial and failed to show any slowing of PD ( ).
Substantia nigra pars compacta (SNc) dopamine neurons are autonomously active; that is, they generate action potentials at a clock-like 2 to 4 Hz in the absence of synaptic input ( )]. In this respect, they are much like cardiac pacemakers. Juvenile dopamine-containing neurons in the SNc use sodium influx as the pacemaking mechanisms common to neurons not affected in PD, but the sodium mechanism remains latent in adulthood ( ). Instead, the autonomous activity is generated by Ca ++ influx ( ; ; ). The SNc dopamine neurons rely on L-type Ca(v)1.3 Ca ++ channels. With increased intracellular calcium, mitochondria function can be affected with increased demand on oxidative phosphorylation, leading to increased production of reactive oxygen species and eventually cellular damage. As the cells undergo more stress over time, they thus “age faster.” This would be a link with the risk factor of age ( ). Blocking Ca(v)1.3 Ca ++ channels in adult neurons induces a reversion to the juvenile form of pacemaking. Such blocking (“rejuvenation”) protects these neurons in both in vitro and in vivo models of PD, pointing to a new strategy that could slow or stop the progression of the disease ( ; ).
As it turns out, use of calcium channel blockers for treating hypertension has been shown to be associated with less risk for developing PD in two database-mining studies ( ; ), but not in a third ( ). A clinical trial to evaluate the dihydropyridine isradipine, a calcium channel blocker, was shown to be well tolerated ( ; ), but the results of a phase III clinical trial failed to show any difference between isradipine and placebo in the progressive worsening of PD ( ; Parkinson Study Group STEADY-PD III Investigators, 2020) (see Table 6.1 ).
Because elevation in blood uric acid has been thought to be potentially neuroprotective, the prourate drug inosine has been tested in patients with early-stage PD, but the phase III trial (SURE-PD) ended early because the investigators thought they would not be able to show that inosine slows Parkinson’s progression ( https://www.michaeljfox.org/news/parkinsons-inosine-trial-ending-early ) (see Table 6.1 ).
In this earliest stage of PD, we follow a recommendation that Fahn has advised, namely patients need to be informed about the disease and the advantages of a daily exercise program. Furthermore, although there is no medication proven to slow the worsening of the disease, it is reasonable to initiate therapy with a MAO-B inhibitor listed in Table 6.1 (selegiline or rasagiline) that has consistently been shown to reduce motor severity compared to a placebo, and may have some disease-modifying effect. Because selegiline has been the only drug shown in controlled clinical trials (even in the presence of levodopa therapy) to delay development of freezing of gait ( ; Fig. 6.6 ), Fahn prefers this medication.
The mild stage of PD occurs when the signs and symptoms of the illness are beginning to interfere with daily activities or with quality of life. The judgment to initiate symptomatic drug therapy is made in discussions between the patient and the treating physician. According to a survey ( ) the most common problems that clinicians consider important for the decision to initiate symptomatic agents are (1) threat to employability; (2) threat to ability to handle domestic, financial, or social affairs; (3) threat to ability to handle ADLs; and (4) appreciable worsening of gait or balance. According to a Norwegian quality of life study ( ), the factors that produce the highest distress for PD patients compared with healthy elderly people are depressive symptoms, self-reported insomnia, and a low degree of independence, as measured by the Schwab and England scale. Severity of parkinsonian motor symptoms contributed, but to a lesser extent. A sense of lack of energy was seen in half of the PD patients compared with a fifth of controls, and this could be only partially accounted for by depressive symptoms and the UPDRS motor scores.
The choice of drugs to improve motor symptoms is wide (see Tables 6.4 and 6.8 ), but the degree of disability and the age (or mental acuity) of the patient are two critical factors in making the selection. If the delay in initiating symptomatic treatment was so prolonged that the symptoms threaten employment or endanger falling, one needs to begin levodopa to get a quick, effective response. The advantages of using levodopa when the symptoms are this pronounced, in preference to a dopamine agonist or other medications, are that a therapeutic response is both rapid and virtually guaranteed, because nearly all patients with PD will respond to levodopa and relatively quickly. In contrast, only a minority of patients with severe symptoms will benefit sufficiently from a dopamine agonist given alone, and it takes more time (often months) to build up the dose to adequate levels to discover this. If levodopa is to be used, inhibitors of type A MAO must be discontinued. If selegiline (or another selective type B MAOI) was the MAOI that was used, this drug can be continued.
If the symptoms are not severe enough to require levodopa and the patient is younger than 60 (younger than 70 if the patient is mentally young), we prefer to employ a dopa-sparing strategy to avoid, as long as possible, the development of levodopa-induced dyskinesias and motor fluctuations (mainly the wearing-off effect). These motor complications are more likely to occur in younger patients ( ; ; ; ). Besides MAO-B inhibitors, which the patient may already be taking, the choices are dopamine agonists, amantadine, and anticholinergics. Dopamine agonists are the most potent antiparkinsonian agents among this group of drugs. Four-year results of the pramipexole versus levodopa trial reveal that levodopa is clinically more potent but is also much more likely to induce dyskinesias and clinical fluctuations ( ). In general, for patients older than 70 years or those with any cognitive impairment, employ levodopa as the initial therapy. Not only is there less need for a dopa-sparing strategy in these elderly patients but they are also more susceptible to confusion, psychosis, or drowsiness from other antiparkinson drugs, including dopamine agonists. Levodopa provides the greatest benefit for the lowest risk of these adverse effects compared with the other drugs. Another consideration is for those patients who need to drive an automobile extensively for their work. The risk for falling asleep at the wheel is greatest for those taking dopamine agonists ( ; ).
As was mentioned earlier, patients younger than 60 years of age are particularly prone to develop the motor complications of fluctuations and dyskinesias ( ; ; ; ). Some physicians therefore recommend using a dopa-sparing strategy, rather than levodopa, in younger patients when beginning therapy, in an attempt to delay the onset of these problems ( ; ; ). But others prefer starting with levodopa ( ). A conference on this topic failed to produce a consensus ( ). There is evidence that duration and severity of disease is a more critical risk factor than duration of levodopa therapy in causing dyskinesias ( ).
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here