Treatment of Parkinson's Disease after the Wearing Off Sets in

Yoshikuni Mizuno

Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan


In 2018, we wrote a paper on the drug treatment of Parkinson's disease. In this article, we obtained that the wearing off was observed in 77%, but the incidence of dyskinesia was 37.7% for the Parkinson?s disease patients from the onset of the disease 16-20 years. In this review article, we will discuss some of the newer treatments of Parkinson?s disease first, i.e., transplantation with induced pluripotent stem cell-derived cells, gene therapy, deep brain stimulation, levodopa/ carbidopa intrajejunal gel infusion, MRI-supported focused ultrasound, and IPX066. Then, we will discuss our opinion on the mechanism of wearing off and dyskinesia, and modifications of levodopa treatment after the wearing off sets in.


In 2018, we wrote a paper on the drug treatment of Parkinson’s disease (PD)1. We observed wearing off (shortening of the levodopa beneficial effects on parkinsonism) to be 77% and the incidence of dyskinesia (abnormal involuntary movements mainly due to the overdose of levodopa) 37.7% for the PD patients from the onset of 16 – 20 years (Table). In this review of the treatment of PD, we will touch on the newer treatments but would not intend to go in, if such treatments would be good or bad to the patients with PD. We believe that newer treatments as described below were introduced for the goodness of patients with PD. Then, we will discuss modifications of levodopa treatment after wearing off sets in.

Table 1. From reference 1, modified, Data indicate the number of patients who were positive in respective symptoms and the percentage.
From the onset No. Wearing off Dyskinesia Freezing
5 years or less 135 24 17.8% 3 2.2% 41 30.4%
6–10 years 158 89 56.3% 28 17.7% 88 55.7%
11–15 years 114 86 75.4% 36 31.6% 67 58.8%
16–20 years 53 39 73.6% 20 37.7% 35 66.0%
21 years or more 38 29 76.3% 21 55.3% 29 76.3%
Total 498 267 53.6% 108 21.7% 260 52.2%

Newer treatments have been introduced to the treatment of PD. Most of the newer treatments have developed as to treat the motor fluctuations of the PD.

Takahashi and Yamanaka2 were successful inducing pluripotent stem cells (iPS) from mouse embryonic or adult fibroblasts by introducing four factors. Since then PD has become a candidate, which may be benefitted by transplant by dopaminergic neurons derived from the iPS3. The first patient of PD was done in autumn of the year 2018. It will take 2 years or more to obtain the result. In addition, as they are using a MHC (major histocompatibility complex)-matching human iPS for this transplantation, they have to use an immuno-suppressor, tacrolimus4.

It is not known if the treatment using iPS is better than using fetal midbrains for transplantation. Olanow et al.5 reported on patients with PD by transplantation using the midbrain of the fetus. UPDRS (Unified PD rating scale) of the transplanted patients with PD were better until 15 months after the surgery. But after the 15 months, there was no significant difference with placebo-treated patients.

The progress of the gene therapy as of 2012 is well described in the review of Nakata et al.6. Among them, aromatic amino acid decarboxylase (AADC), glutamic acid decarboxylase (GAD), and neurturin appear to be of worth mentioning. Amino acid decarboxylase activity in the nigrostriatal system in the advanced PD is very low7.

Muramatsu et al.8 studied 6 patients with advanced PD for 6 months using adeno-associated virus vector 2 and AADC infusion in the striatum. They found that motor functions in the off-medication state improved an average of 46%. Mittemeyer et al.9 reported on ten patients the elevated PET signal to be persisted over 4 years. The UPDRS in all patients off medication improved in the first 12 months but displayed a slow deterioration in subsequent years.

Glutamate is an excitatory transmitter in the subthalamus10. When the glutamic acid decarboxylase is infused to the subthalamus, it may reduce the excitatory tone by changing glutamate to gamma amino butyric acid (GABA). LeWitt et al.11 did a bilateral subthalamic infusion of AAV2(adeno-associated virus 2)-GAD (N = 33) compared with sham surgery in patients (N = 22) with advanced PD. At the 6-month, the AAV2-GAD group showed significantly greater improvement from baseline compared with the sham group.

Glial cell-derived neurotrophic factor (GDNF) enhances the function of nigrostriatal dopaminergic neuron12. However, a double-blind trial did not show a bene?cial e?ect of GDNF13. Using neurturin, a naturally occurring structural analogue of GDNF14, Olanow et al.15 reported no significant difference after the infusion of neurturin from the sham surgery group.

Deep brain stimulation (DBS) became a standard procedure in the surgical treatment of PD since 199316-17. Short term effects up to five years are better in surgically treated patients than medically treated patients18-22. For the prognosis for more than 10 years, it is not clear. Merola et al.23 reported that the clinical course of STN-DBS (subthalamic DBS) progressively worsened during the 20 years period; 64% falls, 86% dysphagia, 57% urinary incontinence and 43% dementia. Janssen et al.24 reported that motor performance was mainly due to a deterioration in bradykinesia and axial symptoms in a study of 5 years on 26 of PD. Rizzone et al.25 reported that motor complications were well controlled at 11 years from the surgery, but that the UPDRS-II-on score worsened by 88.5%. Bang Henriksen et al.26 reported that 29 were demented, 19 were in nursing homes, and 24 have died among 79 patients who received STN-DBS 10 years before. Constantinescu et al.27 reported on 26 men and 7 women operated 15 years before and a sustained benefit was maintained in 19 of them (83%) but STN-DBS was inactivated in four (17%) due to inefficacy. All patients deteriorated slowly, and a majority developed severe non-motor and axial symptoms.

Regarding side effects of the DBS, voice and speech disorders are one of the most important issues28-29. Tanaka et al.29 reported that the STN-DBS group, especially females, showed widespread impairment of voice parameters (n=37) than the medical-therapy-alone group (n=40). Regarding the technical problems, infection, lead migration, skin erosion, and pain over the leads are well known30.

Thus, a long-term follow-up of more than 10 years is not always good for DBS. These symptoms may be a part of the natural course of PD, but it is important to compare with medically treated patients.

Intrajejunal gel infusion of levodopa/carbidopa (LCIG) started in 1988 for the treatment of severe wearing off31. This method can be applied to patients 70 years or over with or without mild to moderate dementia31-32. This method is also useful in non-motor symptoms of PD33. Olanow et al.34 reported that mean off-time decreased by 4.04 h on 35 patients with LCIG compared with a decrease of 2.14 h for 31 patients allocated to immediate-release oral levodopa-carbidopa at 12 weeks. Regarding side effects, Fernandez et al.35 reported that the most common were abdominal pain (30.7%), complication of device insertion (21.4%), procedural pain (17.7%), dyskinesia (10.9%), postoperative wound infection (10.4%), and excessive granulation tissue (13.5%) in 168 patients during 54-weeks treatment with LCIG. Wang et al.32 made meta-analysis on 8 trials with 384 patients of advanced PD with motor fluctuations. They concluded that compared with the control group, LCIG significantly decreased off-time, but more randomized double-blind controlled studies were needed to further confirm the efficacy and safety of LCIG. The device and procedural complications were common in LCIG, and it does not seem a safe treatment of PD.

MRI guided-focused ultrasound is a novel technique36. It is a treatment for essential tremor, PD, and neuropathic pain. It has been brought into clinical medicine since 201436-37. It seems good for unilateral intermedio-ventral thalamotomy in essential tremor38 and PD patients with dyskinesia39. Although it appears to be promising, long-term studies are needed.

IPX066 is a novel extended-release CD–LD (carbidopa-levodopa) formulation. It is designed to achieve rapid absorption and onset of clinical benefit and to provide longer duration of clinical benefit for 4 to 6 hours40. Hauser et al.41 reported that plasma levodopa concentrations increased at a rapid rate and was sustained above 50% of peak concentration for 4 hours in a single dose of IPX066, whereas immediate-release carbidopa-levodopa was sustained 1.4 hours. Hauser et al.42 reported that 201 patients treated double-blind with extended-release carbidopa-levodopa had greater reductions in off-time than did 192 patients treated double-blind with immediate-release carbidopa-levodopa. Stocchi et al.43 reported that 84 patients in an advanced stage with motor fluctuations demonstrated a lower percent off time, lower off time, and higher on time without troublesome dyskinesia with IPX066.

Hsu et al.44 reported that the single-dose pharmacokinetics of ER CD-LD (sustained release levodopa/carbidopa) in healthy subjects sustained for approximately 5 hours and not to decrease to 10% of peak until 10.1 hours, whereas immediate release LD reached peak concentrations at 1 hour; then decreased rapidly. With CD-LD-entacapone, LD Cmax occurred at 1.5 hours, and concentrations were less than 10% of peak by 6.3 hours. Concomitant use of dopamine agonists or monoamine oxidase inhibitors did not interfere with the beneficial effects of IPX006145.

For the first 5 to 6 years since the start of levodopa treatment, 100 -150 mg of levodopa with carbidopa three to four times a day before or after the meal is suffice to most of the PD patients1, 46-47. After 5 years or later since the levodopa was started, a patient with PD may notice worsening of the symptoms. Motor symptoms approximately follow the blood levels of levodopa. Patients of PD may have non-motor symptoms while off.

The blood half-life of levodopa is approximately 2 hours and a half44. Why no wearing off in the beginning of levodopa treatment. When the motor symptoms of PD occurs, approximately 50% of nigral neurons may be remaining48. Therefore, the ingested levodopa enters the dopaminergic terminals. Here, the dopamine transporters exist. Therefore, dopamine released to the synaptic cleft soon will be picked up into the original dopaminergic neurons after stimulating the dopamine receptors. But the time goes on, most of the dopaminergic neurons have degenerated. Then levodopa may be picked up by the serotonin neurons, which can produce dopamine, because they have aromatic amino acid decarboxylase. These serotonin neurons do not have dopamine transporters. Therefore, once released dopamine cannot be re-up-taken to those serotonergic neurons. Dopamine must be metabolized extracellularly and intracellularly in glial cells. This may be the underlying mechanism of wearing off. Dopamine floating in the synaptic cleft may induce dyskinesia. But the speed of neurodegeneration in serotonergic neurons may be slower than the dopaminergic neurons in PD49. Once the serotonergic neurons in the striatum undergo neurodegeneration, neuroglia may take place to produce dopamine. But the neuroglia-induced dopamine will not be sufficing to undertake the motor disturbance of PD. After a long run, patients with PD respond rather poorly to levodopa with less wearing off. This clinical course of PD seems to fit well to what may be going in the brain.

Giving Levodopa/Carbidopa 4 to 9 Times a Day: When the drug on period becomes below 3 hours, we have to give the patients with PD levodopa/carbidopa 5 to 7 times a day. Eventually one may need to give levodopa/carbidopa 8 or 9 times to the patients. Each dose of levodopa may be 75 to 200 mg.

Giving Levodopa/Carbidopa before meals: Giving levodopa/carbidopa before meals may give you somewhat higher blood levels than giving levodopa after meals.

Modify the drug interval: Patients with PD frequently claim that the duration of the drug interval varies a lot. Instruct the patient to take the levodopa at the beginning of the worsening of symptoms to reduce the off time. Most of the patients aware of the effect of levodopa/carbidopa is disappearing. It takes for approximately 30 minutes for levodopa/carbidopa to become effective.

Giving liquid levodopa/carbidopa. When the beneficial effect of levodopa to appear takes more than one hour, try to give the patient water soluble levodopa. Carbidopa/levodopa is easy to water soluble50. A levodopa/carbidopa tablet may stick to the esophageal or stomach wall. By the liquid levodopa, this can be avoided. Liquid preparation of levodopa is made to put a levodopa/carbidopa tablet into 50cc of water51. It is better to keep it in a refrigerator.

Modify each dose: Usually, patients become worse towards late afternoon. If an usual day dose is 100 mg every 3 hours, try to give 150 mg at 3 o’clock or so.

In case of early morning akinesia: Give levodopa while in a bed, then move one’s arms and legs in the bed for 15 to 20 minutes.

In case of dyskinesia: Try to discontinue selegiline or rasagiline if the drug is used. These drugs may worsen dyskinesia. Try to reduce or discontinue a dopamine agonist. These drugs may worsen dyskinesia when dopamine agonists are used together with levodopa. Try to discontinue istradefylline, which may aggravate dyskinesia. Try to decrease or discontinue COMT inhibitors. The incidence of dyskinesia is higher in those who were treated levodopa/carbidopa + entacapone that levodopa/carbidopa + placebo46. If all procedures fail to control dyskinesia, reduce the individual dose of levodopa and increase the number of dosages in a day.

In case of hallucination: Decrease individual dose of levodopa/carbidopa and increase the number of dosages in a day. Decrease or stop medication other than levodopa/carbidopa. One may add quetiapine 25 to 150 mg two or three times daily, donepezil 5 -10 mg a day, or both.

In case of freezing: Freeing tends to occur during the off period. Try to keep on as possible. Freezing tends to occur on the corner of the home and changing the position in the bathroom. Instruct the patients to walk on the heels with arms swing lightly, and to raise their heads. Freezing may also occur when she or he is engaged in drying the washing in the sun or to carrying a tray to bring food. In such occasions, nerve impulses going to the lower extremities become weak. Then, freezing or fall sets in. Patients with PD should be instructed that no drug will help freezing and freezing should be avoided by their own mind.

With the meticulous management of levodopa, majority of the patients with PD are managed medically for a long period. Newer treatments such as the gene therapy, DBS, MRI-guided focused ultrasound, and IPX066 are at times good for PD patients. But usually, they do not want surgery or any devices to be put on their bodies.

  1. Mizuno Y, Shimoda S, Origasa H. Long-term treatment of Parkinson's disease with levodopa and other adjunctive drugs. J Neural Transm Vienna. 2018 Jan; 125(1): 35-43. doi: 10.1007/s00702-016-1671-x. Epub 2017 Jan 16.
  2. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006 Aug 25; 126(4): 663-76. Epub 2006 Aug 10.
  3. Kikuchi T, Morizane A, Doi D, et al. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson's disease model. Nature. 2017 Aug 30; 548(7669): 592-596. doi: 10.1038/nature23664.
  4. Morizane A, Kikuchi T, Hayashi T, et al. MHC matching improves engraftment of iPSC-derived neurons in non-human primates. Nat Commun. 2017 Aug 30; 8(1): 385. doi: 10.1038/s41467-017-00926-5
  5. Olanow CW, Goetz CG, Kordower JH, et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease. Ann Neurol. 2003 Sep; 54(3): 403-14.
  6. Nakata Y, Yasuda T, Mochizuki H. Recent progress in gene therapy for Parkinson's disease. Curr Mol Med. 2012 Dec; 12(10): 1311-8.
  7. Ciesielska A, Samaranch L, San Sebastian W, et al. Depletion of AADC activity in caudate nucleus and putamen of Parkinson's disease patients; implications for ongoing AAV2-AADC gene therapy trial. PLoS One. 2017 Feb 6; 12(2): e0169965. doi: 10.1371/journal.pone.0169965. eCollection 2017.
  8. Muramatsu S, Fujimoto K, Kato S, et al. A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson's disease. Mol Ther. 2010 Sep; 18(9): 1731-5. doi: 10.1038/mt.2010.135. Epub 2010 Jul
  9. Mittermeyer G, Christine CW, Rosenbluth KH, et al. Long-term evaluation of a phase 1 study of AADC gene therapy for Parkinson's disease. Hum Gene Ther. 2012 Apr; 23(4): 377-81. doi: 10.1089/hum.2011.220. Epub 2012 Apr 10.
  10. Alexander GE, DeLong MR, Strick PL. Parallel organization of functionally segregated circuits linking basal ganglia and cortex. Annu Rev Neurosci. 1986; 9: 357-381.
  11. LeWitt PA, Rezai AR, Leehey MA, et al. AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol. 2011 Apr; 10(4): 309-19. doi: 10.1016/S1474-4422(11)70039-4.
  12. Kordower JH, Emborg ME, Bloch J, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson’s disease. Science. 2000; 290: 767–73.
  13. Nutt JG, Burchiel KJ, Comella CL, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology. 2003; 60: 69–73.
  14. Kotzbauer PT, Lampe PA, Heuckeroth RO, et al. Neurturin, a relative of glial-cell-line-derived neurotrophic factor. Nature. 1996; 384: 467–70.
  15. Olanow WC, Bartus RT, Baumann TL, et al. Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: A double-blind, randomized, controlled trial. Ann Neurol. 2015 Aug; 78(2): 248-57. doi: 10.1002/ana.24436. Epub 2015 Jun 10.
  16. Pollak P, Benabid AL, Gross C, et al. Effects of the stimulation of the subthalamic nucleus in Parkinson disease. Rev Neurol Paris. 1993; 149(3): 175-6.
  17. Benabid AL, Pollak P, Gross C, et al. Acute and long-term effects of subthalamic nucleus stimulation in Parkinson's disease. Stereotact Funct Neurosurg. 1994; 62(1-4): 76-84.
  18. Deuschl G, Schade-Brittinger C, Krack P, et al. A randomized trial of deep-brain stimulation for Parkinson's disease. N Engl J Med. 2006 Aug 31; 355(9): 896-908.
  19. Kleiner-Fisman G, Herzog J, Fisman DN, et al. Subthalamic nucleus deep brain stimulation: summary and meta-analysis of outcomes. Mov Disord. 2006 Jun; 21 Suppl 14: S290-304.
  20. Vizcarra JA, Situ-Kcomt M, Artusi CA, et al. Subthalamic deep brain stimulation and levodopa in Parkinson's disease: a meta-analysis of combined effects. J Neurol. 2018 Jun 16. doi: 10.1007/s00415-018-8936-2. [Epub ahead of print]
  21. Lee DJ, Dallapiazza RF, De Vloo P, et al. Current surgical treatments for Parkinson's disease and potential therapeutic targets. Neural Regen Res. 2018 Aug; 13(8): 1342-1345. doi: 10.4103/1673-5374.235220.
  22. Yamamoto T, Uchiyama T, Higuchi Y, et al. Long term follow-up on quality of life and its relationship to motor and cognitive functions in Parkinson's disease after deep brain stimulation. J Neurol Sci. 2017 Aug 15; 379: 18-21. doi: 10.1016/j.jns.2017.05.037. Epub 2017 May 19.
  23. Merola A, Zibetti M, Angrisano S, et al. Parkinson's disease progression at 30 years: a study of subthalamic deep brain-stimulated patients. Brain. 2011 Jul; 134(Pt 7): 2074-84. doi: 10.1093/brain/awr121. Epub 2011 Jun 11.
  24. Janssen ML, Duits AA, Turaihi AH, et al. Subthalamic nucleus high-frequency stimulation for advanced Parkinson's disease: motor and neuropsychological outcome after 10 years. Stereotact Funct Neurosurg. 2014; 92(6): 381-7. doi: 10.1159/000366066. Epub 2014 Oct 29.
  25. Rizzone MG, Fasano A, Daniele A, et al. Long-term outcome of subthalamic nucleus DBS in Parkinson's disease: from the advanced phase towards the late stage of the disease? Parkinsonism Relat Disord. 2014 Apr; 20(4): 376-81. doi: 10.1016/j.parkreldis.2014.01.012. Epub 2014 Jan 23.
  26. Bang Henriksen M, Johnsen EL, Sunde N, et al. Surviving 10 years with deep brain stimulation for Parkinson's disease--a follow-up of 79 patients. Eur J Neurol. 2016 Jan; 23(1): 53-61. doi: 10.1111/ene.12614. Epub 2014 Dec 9.
  27. Constantinescu R, Eriksson B, Jansson Y, et al. Key clinical milestones 15 years and onwards after DBS-STN surgery-A retrospective analysis of patients that underwent surgery between 1993 and 2001. Clin Neurol Neurosurg. 2017 Mar; 154: 43-48. doi: 10.1016/j.clineuro.2017.01.010. Epub 2017 Jan 18.
  28. Tripoliti E, Zrinzo L, Martinez-Torres I, et al. Effects of subthalamic stimulation on speech of consecutive patients with Parkinson disease. Neurology. 2011 Jan 4; 76(1): 80-6. Epub 2010 Nov 10.
  29. Tanaka Y, Tsuboi T, Watanabe H, et al. Voice features of Parkinson's disease patients with subthalamic nucleus deep brain stimulation. J Neurol. 2015 May; 262(5): 1173-81. doi: 10.1007/s00415-015-7681-z. Epub 2015 Feb 26.
  30. Jitkritsadakul O, Bhidayasiri R, Kalia SK, e al. Systematic review of hardware-related complications of Deep Brain Stimulation: Do new indications pose an increased risk. Brain Stimul. 2017 Sep - Oct; 10(5): 967-976. doi: 10.1016/j.brs.2017.07.003. Epub 2017 Jul 13.
  31. Kurlan R, Nutt JG, Woodward WR, et al. Duodenal and gastric delivery of levodopa in parkinsonism. Ann Neurol. 1988 Jun; 23(6): 589-95.
  32. Wang L, Li J, Chen J. Levodopa-Carbidopa Intestinal Gel in Parkinson's Disease: A Systematic Review and Meta-Analysis. Front Neurol. 2018 Jul 30; 9: 620. doi: 10.3389/fneur.2018.00620. eCollection 2018.
  33. Honig H, Antonini A, Martinez-Martin P, et al. Intrajejunal levodopa infusion in Parkinson's disease: a pilot multicenter study of effects on nonmotor symptoms and quality of life. Mov Disord. 2009 Jul 30; 24(10): 1468-74. doi: 10.1002/mds.22596.
  34. Olanow CW, Kieburtz K, Odin P, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson's disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol. 2014 Feb; 13(2): 141-9. doi: 10.1016/S1474-4422(13)70293-X. Epub 2013 Dec 20.
  35. Fernandez HH, Vanagunas A, Odin P, et al. Levodopa-carbidopa intestinal gel in advanced Parkinson's disease open-label study: interim results. Parkinsonism Relat Disord. 2013 Mar; 19(3): 339-45. doi: 10.1016/j.parkreldis.2012.11.020. Epub 2013 Jan 1.
  36. Dobrakowski PP, Machowska-Majchrzak AK, Labuz-Roszak B, et al. MR-guided focused ultrasound: a new generation treatment of Parkinson's disease, essential tremor and neuropathic pain. Interv Neuroradiol. 2014 May-Jun; 20(3): 275-82. doi: 10.15274/NRJ-2014-10033. Epub 2014 Jun 17.
  37. Bauer R, Martin E, Haegele-Link S, et al. Noninvasive functional neurosurgery using transcranial MR imaging-guided focused ultrasound. Parkinsonism Relat Disord. 2014 Jan; 20 Suppl 1: S197-9. doi: 10.1016/S1353-8020(13)70046-4.
  38. Zaaroor M, Sinai A, Goldsher D, et al. Magnetic resonance-guided focused ultrasound thalamotomy for tremor: a report of 30 Parkinson's disease and essential tremor cases. J Neurosurg. 2018 Jan; 128(1): 202-210. doi: 10.3171/2016.10.JNS16758. Epub 2017 Feb 24.
  39. Jung NY, Park CK, Kim M, et al. The efficacy and limits of magnetic resonance -guided focused ultrasound pallidotomy for Parkinson's disease: a Phase I clinical trial. J Neurosurg. 2018 Aug; 1: 1-9. doi: 10.3171/2018.2.JNS172514. [Epub ahead of print]
  40. Hauser RA. IPX066: a novel carbidopa-levodopa extended-release formulation. Expert Rev Neurother. 2012 Feb; 12(2): 133-40. doi: 10.1586/ern.11.195.
  41. Hauser RA, Ellenbogen AL, Metman LV, et al. Crossover comparison of IPX066 and a standard levodopa formulation in advanced Parkinson's disease. Mov Disord. 2011 Oct; 26(12): 2246-52. doi: 10.1002/mds.23861. Epub 2011 Jul 13.
  42. Hauser RA, Hsu A, Kell S, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson's disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol. 2013 Apr; 12(4): 346-56. doi: 10.1016/S1474-4422(13)70025-5. Epub 2013 Feb 26.
  43. Stocchi F, Hsu A, Khanna S, et al. Comparison of IPX066 with carbidopa-levodopa plus entacapone in advanced PD patients. Parkinsonism Relat Disord. 2014 Dec; 20(12): 1335-40. doi: 10.1016/j.parkreldis.2014.08.004. Epub 2014 Aug 15.
  44. Hsu A, Yao HM, Gupta S, et al. Comparison of the pharmacokinetics of an oral extended-release capsule formulation of carbidopa-levodopa (IPX066) with immediate-release carbidopa-levodopa (Sinemet(®)), sustained-release carbidopa-levodopa (Sinemet(®) CR), and carbidopa-levodopa-entacapone (Stalevo(®)). J Clin Pharmacol. 2015 Sep; 55(9): 995-1003. doi: 10.1002/jcph.514. Epub 2015 May 20.
  45. LeWitt PA, Verhagen Metman L, et al. Effect of Concomitant Medications on the Safety and Efficacy of Extended-Release Carbidopa-Levodopa (IPX066) in Patients With Advanced Parkinson Disease: A Post Hoc Analysis. Clin Neuropharmacol. 2018 Mar/Apr; 41(2): 47-55. doi: 10.1097/WNF.0000000000000269.
  46. Schrag A, Quinn N.?Dyskinesias and motor fluctuations in Parkinson's disease. A community-based study. Brain. 2000; 123: 2297-2305.
  47. Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE-PD study. Ann Neurol. 2010 Jul; 68(1): 18-27. doi: 10.1002/ana.22060.
  48. Brooks DJ. Neuroimaging in Parkinson's disease. NeuroRx. 2004 Apr; 1(2): 243-54.
  49. Kish SJ, Tong J, Hornykiewicz O, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain. 2008; 131(Pt 1): 120–31. doi: 10. 1093/brain/awm239 PMID: 17956909
  50. Metman LV, Hoff J, Mouradian MM, et al. Fluctuations in plasma levodopa and motor responses with liquid and tablet levodopa/carbidopa. Mov Disord. 1994 Jul; 9(4): 463-5.
  51. Pappert EJ, Goetz CG, Niederman F, et al. Liquid levodopa/carbidopa produces significant improvement in motor function without dyskinesia exacerbation. Neurology. 1996 Dec; 47(6): 1493-5.
 

Article Info

Article Notes

  • Published on: February 18, 2019

Keywords

  • Dyskinesia

  • Parkinsonism

*Correspondence:

Dr. Yoshikuni Mizuno, MD
Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Telephone No: 81-3-3824-3139; Fax No: 81-3-3824-3139
Email: y_mizuno@juntendo.ac.jp.