Parkinson’s Disease Awareness & Education Conference

Parkinson’s Disease Awareness & Education Conference
Jeff’s Catering & Conference Center 15 Littlefield Way, Brewer, ME 04412
Thursday, October 4, 2018

The following message is pasted from an announcement by The Maine Parkinson Society:

The Maine Parkinson Society will present its Parkinson’s Disease Awareness Conference event for Maine people with Parkinson’s disease, caregivers, health care professionals and the general public on Thursday, October 4, 2018, at Jeff’s Catering & Conference Center in Brewer, Maine.
Won’t you please help us support this conference of awareness and education by becoming a sponsor or an exhibitor? Please see the attached form; sponsorship at any level is greatly appreciated and welcomed! There are over 7,000 Maine families living with Parkinson’s disease and they welcome and deserve your consideration.
Featured speakers include Maine neurologists specializing in the treatment of Parkinson’s disease
Dr. Roople Unia of Eastern Maine Medical Center Neurology Specialists and Dr. William Stamey of Mid-Coast Neurology in addition to Michael Westphal, a well-known Maine-native and runner with Parkinson’s.

There will be an assortment of exhibitors and demonstrations throughout the day to help gain a better understanding of techniques and resources available to patients and caregivers.

Maine Parkinson Society is dedicated to improving the quality of life for Maine People with Parkinson’s disease through education, increasing public awareness and creating specialized programs that directly benefit patients and their families.
As the only non-profit organization in the State of Maine dedicated solely to Parkinson’s disease, we ask for your help in supporting this very vital event by becoming an exhibitor or sponsor.

Sincerely,
Gary Cole, Past President
Karen L Marsters
Karen Marsters, President-Elect

Pipeline drugs, part 4: apomorphine

Apomorphine (Apokyn) is a dopamine delivered by multi-dose glass cartridges, 30 mg/3 mL with a multiple dose pen injector (APOKYN Pen).  Apokyn is FDA approved in the U.S. for the acute, intermittent treatment of hypomobility, OFF episodes, and unpredictable ON/OFF episodes associated with advanced Parkinson disease (PD).  It is used by a relatively small number of patients in the U.S.  However, different formulations of apomorphine are available outside the U.S., and different formulations are under investigation within our borders.

One study evaluated APL-130277, a sublingual (beneath the tongue) apomorphine oral strip, as an acute, effective, noninvasive treatment for OFF episodes (1).  This was a phase II, open-label, proof-of-concept study.

In open-label studies, patients know they are taking the study drug and there is no placebo, thus no blind.  Proof-of-concept studies are designed to verify that some concept has practical potential, such as using a sublingual version of the drug for a certain indication.  

In this study patients presented to clinic in the morning hours “in the practically defined OFF state” (before  taking their morning medications) and were given APL-130277 10 mg.  PD motor scores using MDS-UPDRS III were recorded before dosing and at 15, 30, 45, 60, and 90 minutes.  If a full ON state was not achieved within 3 hours, the dose was increased in 5 mg increments until the patient was either ON, or had taken 30 mg.  Of the 19 people with PD treated, 15 (78.9%) reached a full ON response within 30 minutes, and 6 of the 15 patients reached ON within 15 minutes.  Average duration of ON was 50 minutes, and nine patients remained fully ON for 90 or more minutes.  No patients discontinued the drug due to adverse event, though dizziness occurred in 36.8%, sleepiness in 31.6%, and nausea in 21.1%.

NCT0254269 is an open-label phase III study to examine long-term safety, tolerability, and efficacy of APL-1320277 in doses ranging from 10-35 mg for the treatment of OFF episodes in patients with PD (2).  Estimated enrollment will be 226 patients and recruitment is ongoing at many centers in the U.S. and Canada.

Apomorphine has also been developed as a drug which may be administered by pump.  Already available in some countries, it is currently in trials in the U.S.  The apomorphine pump is being tested as a method to avoid fluctuations in the motor symptoms of PD.  In a study published in 2017, authors noted that people with advanced PD and contraindications for deep brain stimulation (DBS) might benefit from apomorphine as an add-on to existing medications, and evaluated the motor and nonmotor symptoms in advanced PD (3).

A small cohort of 12 patients with advanced PD were assessed before and after 6 months of apomorphine with a type of brain imaging called 18F-fluorodeoxyglucose positron emission tomography (PET scan) and “exhaustive clinical assessments.”  Authors noted that after 6 months of therapy they were able to significantly reduce oral PD meds (and thus reduce risk of side effects from those meds), and that motor and nonmotor scores improved “with a beneficial effect on executive functions, quality of life and apathy.”  Brain PET scan of these patients reportedly revealed significant metabolic changes consistent with the improvements in clinical scores.  The authors noted, “these preliminary results have to be confirmed by further studies.”

NCT02339064 is a phase III, 52-week safety study being called INFUS-ON, which is an actively recruiting, multicenter, open-label trial to assess the long-term safety and tolerability of continuous subcutaneous infusion of apomorphine in advanced PD patients with unsatisfactory motor fluctuations while using levodopa and at least one other class of drugs or mode of therapy for PD (4).  The study will also evaluate reductions in OFF time and improvements in ON time without troublesome dyskinesias.

 

REFERENCES

  1. Hauser, et al. Sublingual apomorphine (APL-130277) for the acute conversion of OFF to ON in Parkinson’s disease. Mov Disord. 2016;31(9):1366-72.
  2. https://clinicaltrials.gov/ct2/show/NCT02542696?term=sublingual+apomorphine&cond=Parkinson+Disease&rank=36
  3. Auffret, et al, Apomorphine pump in advanced Parkinson’s disease: Effects on motor and nonmotor symptoms with brain metabolism correlations. J Neurol Sci. 2017;372:279-287.
  4. https://clinicaltrials.gov/ct2/show/NCT02339064?term=apomorphine+pump&cond=Parkinson+Disease&rank=6

FDA approval of inhaled levodopa delayed

In the spring, 2018 issue of MPDN, the drug INBRIJA was discussed (1).  INBRIJA is a self-administered, orally inhaled form of levodopa (similar to an asthma inhaler) which if approved by the Food and Drug Administration (FDA), will be indicated for symptoms of OFF periods in people who take carbidopa/levodopa for Parkinson’s disease.  In other words, it will serve as a rescue drug for OFF periods.  As noted in that article, in February Acorda Therapeutics, Inc. announced the FDA had accepted the New Drug Application (NDA) for INBRIJA.  Under the Prescription Drug User Fee Act (PDUFA), the FDA set a target date of October 5, 2018.   However, the FDA announced on September 13 that PDUFA date for the drug been extended to January 5, 2019.   This delay comes after Acorda provided more data in response to FDA requests for additional information on chemistry, manufacturing, and controls (CMC).   Per the Acorda website press release, “FDA determined that these submissions constitute a major amendment and will take additional time to review” (2).

  1. Pipeline drugs, part 1: levodopa  https://mainepdnews.org/2018/03/24/pipeline-drugs-part-1-levodopa/
  2. http://ir.acorda.com/investors/investor-news/investor-news-details/2018/Acorda-Announces-FDA-Extends-INBRIJA-NDA-Review-Period/default.aspx

Jennifer Bryce to offer speech and language care at Aroostook Medical Center

Jennifer H. Bryce, MA, CCC-SLP, (speech & language pathologist), certified in LSVT-LOUD, has moved from the Boothbay and Damariscotta area, where in addition to her professional role she started and led the Parkinson disease support group for several years.   A loss to one region is a gain for others in Maine however.  She has returned home to Aroostook County, where she is available to provide treatment to people with PD.   If interested, please let your health care provider know that referrals can be sent to:   The Aroostook Medical Center,  Fax: 207-768-4734,  Phone: 207-768-4735.

Pipeline drugs, part 3: young blood

Aging is associated with many changes in the human brain.  We may notice, for example, that short-term memory is not as robust as we get older.  One reason for this is that the hippocampus, the area of the brain that deals with the formation of short-term memory and learning, is affected by normal aging.  The reason is complicated, but the more-or-less short version of the story has to do in part with genes.  Genes are the individual instructions for function in your genetic library, the DNA.  Your DNA contains over 20,000 genes and these may be turned on (upregulated), or turned off (downregulated).  Those on/off gene switches are often influenced by chemical messages from other cells.  Selection of which genes work is one way humans can start out as a single cell (a fertilized egg), and wind up trillions of cells in the collective that is an adult.

Every living cell in your body contains the full DNA library that makes you who and what you are.  The key is that each type of cell needs different instructions from the DNA to form and carry out basic functions that make them unique cell types of the brain, liver, skin, and so on.  Even within the brain, there are many different types of cells that make up different parts and have very different functions.  The cells of the hippocampus don’t appear or function in a way similar to the cells of the basal ganglia, for example.  And, as we age, the functions of cells change for better or worse in response to needs at the time and the dictates of our genes.

Brain cells communicate with each other at points called synapses.  Imagine a cell having many branches that connect with the branches of other cells. You have over 80 billion brain cells (neurons), and each forms up to 10,000 synapses with other neurons – though the average is probably closer to 7,000.   When you learn something, it is because you are able to form new connections between neurons through a process called synaptic plasticity.  In medicine, if something is plastic, it is able to change.  Synaptic plasticity allows a set of new connections that makes a new synaptic pathway, and through that pattern of connected neurons, you have learned something.  This new learning is stored as a memory that can be called upon again.

In the older person, synapses of the hippocampus may have trouble with one of the functions of memory formation called long-term potentiation (1).  The process is influenced by genes that control synaptic plasticity.  Those genes may be downregulated, and one culprit is thought to be a tiny chemical messenger in the tissue around the cell (2).

It turns out that in younger people there are proteins in the blood plasma that upregulate the same genes.  This might be why younger people have better memory.   

Multiple times over the last several years researchers have shown that exposing older cells to young blood can improve function (3), and even reverse age-related changes to brain cells (4) and cognitive function (5), though these studies were in mice, not humans.

Researchers at Stanford University have been on the cutting edge of this investigative work, and a paper published in April 2017 in the journal Nature (6) hypothesized that plasma from human umbilical cord blood would contain “a reservoir of such plasticity-promoting proteins.”  They showed that human cord plasma treatment revitalized the hippocampus and improved cognitive function in older mice, who were better able to learn mazes and avoid unpleasant situations.  Researchers did not just test wholesale injection of plasma however.  Over 60 compounds found in the umbilical plasma were tested, and a molecule was identified that by itself made positive changes.  TIMP2, known to be present in human cord plasma, young mouse plasma, and young mouse hippocampus, was found to increase synaptic plasticity at the level of the hippocampus in older mice, and it was shown that blocking TIMP2 prevented long-term potentiation (memory formation).

In the PLasma for Alzheimer SymptoM Amelioration Study (PLASMA) (NCT02256306), Stanford researchers next trialed intravenously-administered plasma from young donors to 18 patients with mild-moderate Alzheimer disease (AD) (7).   Participants were given one unit of plasma weekly over four weeks.  The primary endpoint was a measure of safety and tolerability.  Secondary outcome measures included changes over a nine-week period in cognitive tests, functional activities, activities of daily living, and depression.  Other lab and imaging studies were also included.  No results are posted on ClinicalTrials.gov.  However, Stanford Medicine News Center quoted investigator Dr. Sharon Sha, who noted the trial showed good tolerability, but “no significant changes in participants’ mood or their performance on tests of cognition … these kinds of changes are typically observed only in clinical trials whose durations exceed one year” (8).  Thus, larger, longer studies are needed to see if these improvements would occur.  Stage of disease may be another key factor.  Still, it is encouraging that the therapy was tolerated, and the door is open to new trials.

An ongoing study at Stanford is the The Stanford Parkinson’s Disease Plasma Study (SPDP) (NCT02968433) (9), which proposes to show that “young plasma infusions can be performed safely in patients with Parkinson’s Disease (PD).”  In the study, secondary outcomes include “behavioral and laboratory data that will support the next study that will inquire whether young plasma infusions improve or slow the progression of cognitive, mood and/or motor impairment and rate markers of the disease.”  All 15 participants will undergo neuropsychological, neuropsychiatric, and motor assessment prior to receiving infusions of one unit of young plasma, twice a week over a four week duration.  Following that period, participants will undergo reassessments.  There will be no placebo arm, or as investigators put it, “no deception will be used.”  The primary outcome measure will be an assessment of safety and adverse events.  Secondary outcome measurements will include change in motor function up to eight weeks out.  Finally, there will be a measure of potential change in cognitive ability.

If compounds in young blood are helpful in PD, the goal might be to isolate them and form new therapies.  This work in PD is in the early stages of development, though it is very helpful that so much of the groundwork has been done in mice and in Alzheimer disease.

 

REFERENCES

  1. Burke, S. N. & Barnes, C. A. Neural plasticity in the ageing brain. Nature Rev. Neurosci. 7. 2006;30–40
  2. Lee, et al. Gene-expression profile of the ageing brain in mice. Nature Genet.2000;25:294–297
  3. Conboy, et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature.2005;433:760–764
  4. Katsimpardi, et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science. 2014;344:630–634
  5. Villeda, S. A. et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nature Med. 2014;20:659–663
  6. Castellano, et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 2017; 544:488–492
  7. https://clinicaltrials.gov/ct2/show/NCT02256306?term=NCT02256306&rank=1
  8. https://med.stanford.edu/news/all-news/2017/11/clinical-trial-finds-blood-plasma-infusions-for-alzheimers-safe.html 
  9. https://clinicaltrials.gov/ct2/show/NCT02968433?term=nct02968433&rank=1

Focused ultrasound part II, what is the data now?

In the fall, 2017 issue of MPDN we discussed focused ultrasound (FUS) (1), a specialized technique that was FDA approved to treat essential tremor (ET) in 2016.  Parkinson disease (PD) tremor is not yet an FDA approved indication, but it seems likely that it will be in the future.  Also mentioned in the MPDN article was a study of patients with PD who were given FUS.  As that study was ongoing, no data was then available. That study is now complete and has been published in JAMA Neurology as discussed below, along with two other new scientific reports.  Here we review the topic and briefly discuss the new findings and considerations.

FUS

Patients undergoing FUS experience a noninvasive MRI-guided procedure in which beams of acoustic energy (sound waves) are directed toward the same target in the brain from 1,024 tiny transducers worn around the head in a stereotactic frame.  This energy combines on the target to generate friction and heat, which destroy the target.  One could imagine an unwanted electrical signal being stopped in this way.   There are a variety of potential targets in the brain.

VIM

For ET and most reported cases of PD, the target is the ventral intermediate nucleus (VIM) of the thalamus (a deep gray matter structure present on both sides of the brain).   VIM in some ways functions as a relay for the signal which causes tremor and has been a target for deep brain stimulation (DBS), cautery, and gamma knife approaches to tremor.   As with most parts of the brain, each side governs the opposite side of the body.   FUS of the VIM is meant to cause a lesion which will stop or reduce tremor.  As this heating up is taking place it is monitored by magnetic resonance thermometry.  In other words, researchers are able to watch the lesion occur because the patient is inside of an MRI at that time. This provides confirmation that the correct target is being destroyed.  There are multiple other parts of the thalamus nearby involving among other functions such as bodily sensation, hearing, and vision.  And, there are neighboring structures involved in muscular strength, muscle tone, and balance.  As of yet, with ET only one side of the brain is treated by FUS (under FDA approval).  And, studies with PD have used FUS on only one side, which limits the risk of adverse events (AE) with the procedure.   The most common AEs with VIM FUS are numbness on the side of the body tremor is being controlled and ataxia (the loss of coordination or control of bodily movements not due to weakness).  With VIM FUS the goal is to control severe, medication-refractory tremor.

We previously referenced a small study which included 9 patients with Parkinson disease (2) with VIM FUS.   In that study, the motor score (part III of the Unified Parkinson’s Disease Rating Scale, UPDRSIII) improved from 24.9 ± 8.0 to 16.4 ± 11 at one month, and 13.4 ± 9.2 at six months after treatment, a positive result (footnote).

A recently published study took place at two different academic centers (University of Virginia Health Sciences Center in Charlottesville, Virginia, and Swedish Neuroscience Institute, Seattle Washington), and sought to answer the questions as to how safe and efficacious FUS thalamotomy (VIM FUS) is for managing medically refractory, tremor-dominant PD (TDPD), and to ask the magnitude of the placebo response (3).   Researchers identified TDPD patients from the database, and included patients whose disease was medication-refractory, severe, and disabling.  Twenty-seven patients were randomized: 20 to FUS thalamotomy, and 7 to sham procedure.   Twenty-six of the patients were male (96%), average age 67.8 years.   The group receiving the sham procedure was offered open-label treatment after unblinding.  Patients undergoing the study protocol were prepared by having their heads shaved and a stereotactic frame placed on their heads for MRI stereotactic planning.  This meant that patients would each be placed in the MRI to localize the exact target before beginning the ultrasound treatment.   Before beginning therapy, the treatment team was informed of whether each patient was to receive the sham or FUS. Ultrasound was administered with increasing energy and patients were clinically monitored after each sonication.  Tremor was assessed at rest, posture, finger to nose, and drawing.  Researchers noted that tremor did not tend to decrease until temperatures exceeded 50°C (122°F).  The stated goal during the treatment was to reach tremor suppression “at an adequate thermal dose to the target.” During the treatment headache occurred in 65% and dizziness/vertigo and 42%.  All of this resolved with completion of the procedure.  Two patients had mild persistent hemiparesis (weakness on one side of the body) with gradual improvement.  One patient had persistent mild ataxia.  Patients returned for an MRI the next day and were then discharged from the hospital.  Assessments of tremor were performed at one and three months.   Neither the patient nor the evaluators were un-blinded until the 12 month assessment.  Hand tremor was measured using a clinical rating scale for tremor (CRST) and revealed improvements of 62%, and was still present at the 12 month assessment.  The more familiar UPDRSIII (motor subsection) revealed an overall 8 point benefit (from a baseline average of 23 points) , with 1.5 point improvement of resting tremor (from a baseline average of 4 points) and 1 point for postural tremor (from baseline average of 4 points). Three patients had inadequate improvement or even mild worsening of tremor scores.  Sham treatment did not result in benefit.  Six sham patients did go on to have the full treatment after unblinding.  The most common thalamotomy related adverse events were finger tingling (39%), ataxia (35%), facial tingling (27%).  At one year tingling persisted in 19% of patients and ataxia in 4%.

VOP

A single case report from a 73 year-old Japanese patient with refractory right-sided tremor (4).  The patient could not tolerate higher doses of levodopa or dopamine agonists due to hallucinations and excessive daytime sleepiness.  He was given FUS to ablate both the VIM and the ventro oralis posterior (VOP) nuclei of the thalamus (4).  By including the VOP, investigators hoped to improve muscular rigidity (increased tone).  Right-side resting tremor and rigidity were “abolished immediately following 10 sonications with an average maximum sonication time of 13.0 ± 3.4 seconds (range 10-17 seconds).”  Temperatures inside of the thalamus reached a maximum of 60°C (140°F).  No adverse events occurred during sonications.  However, bradykinesia (slowness of movement) was exacerbated after thalamotomy and was reportedly due to local swelling of brain tissue.  The patient was given steroids and the bradykinesia resolved after two weeks.

STN

Another target to consider is the subthalamic nucleus (STN). This is the more common site used for DBS in PD because of several factors, including the benefit that not only tremor, but bradykinesia (slowness of movement) and rigidity might be improved.

An open-label pilot study (not blinded, patients knew they were getting the treatment) was done at CINAC (Centro Integral de Neurociencias), UniversityHospital HM Puerta del Sur in Madrid, Spain (5).   Ten PD patients with markedly asymmetric parkinsonism poorly controlled with medications were included.   FUS subthalamotomy was targeted with several sonications above 55°C and adjusted according to clinical response.  Total number of sonications given ranged from 17-31, though the average number of sonications above 55°C was 9.  Time spent receiving sonication ranged from 144-337 seconds.   There were pauses between sonications, and thus time spent from first sonication to completion ranged from 83-174 minutes, thus the longest time under therapy was just under 3 hours.  The goal was to safely change the motor status of the treated side of the body in both off-medication and on-medication states at 6 months.

By the 6 month follow-up, 38 non-serious AEs had been recorded, though only 7 were still present.  Three AEs were directly related to subthalamotomy: off-medication dyskinesia, on-medication dyskinesia, and subjective speech disturbance.  Four of the adverse events present at 6 months were related to medical management, and included anxiety, fatigue, and weight gain.  The most frequent AEs were transient gait ataxia (6 patients), transient head pain related to the head frame (6 patients), and transient high blood pressure during the procedure (5 patients).   During the procedure back pain was reported, likely due to position in the MRI (2 patients), “warm cranial sensations (2 patients), and nausea (4 patients).  Transient facial asymmetry (1 patient) and moderate impulsivity (2 patients) were also noted.  At six months the average motor score in the treated side of the body improved by 53% in the off-medication state and 47% in the on-medication state.

Gpi

Globus pallidus interna is a target for DBS leads.  Surgical lesions of the GPi have been shown to improve tremor, bradykinesia, rigidity, and dyskinesias.  As yet there do not appear to be any published cases regarding Gpi FUS.  However, a trial is recruiting (NCT02003248) in Seoul, Korea to study safety and effectiveness of Gpi FUS in the treatment of dyskinesia (6).  Another trial (NCT03319485) with locations in Maryland, New York, and Ohio is also recruiting to evaluate the safety and efficacy of unilateral focused ultrasound pallidotomy in the management of dyskinesia or motor fluctuations for medication refractory, advanced PD (7).   A third trial (NCT02263885) is active, not recruiting, in multiple U.S. sites including California, Maryland, Massachusetts, Ohio, and Virgina (8).   This study is designed to evaluate the safety, and initial efficacy of unilateral lesion in the globus pallidus as an adjunct to PD medications in medication-refractory PD.

PPN

Pedunculopontine nucleus (PPN) is an experimental target for DBS in the treatment of freezing and other gait dysfunction.  As yet no studies or articles are available approaching this target.

Footnote:  Recall that UPDRSIII is a 108 point scale which measures motor functions such as tremor, stiffness, slowness, gait, etc.

 

REFERENCES

  1. https://mainepdnews.org/2017/09/28/how-close-are-we-to-focused-ultrasound-for-pd-in-maine/
  2. Zaaroor, et al.  Magnetic resonance-guided focused ultrasound thalamotomy for tremor: a report of 30 Parkinson’s disease and essential tremor cases. J Neurosurg. 2017 Feb 24:1-9.
  3. Bond, et al.   Safety and efficacy of focused ultrasound thalamotomy  for patients with medication-refractory, tremor-dominant Parkinson disease.  A randomized clinical trial.  JAMA Neurol.   2017;74 (12): 1412-1418.
  4. Ito, et al. Magnetic Resonance Imaging-guided Focused Ultrasound Thalamotomy for Parkinson’s Disease: A Case Report.Intern Med. 2017 Dec 21. .9586-17.
  5. Martínez-Fernández, et al.  Focused ultrasound subthalamotomy in patients with asymmetric Parkinson’s disease: a pilot study. Lancet Neurol 2018; 17: 54–63
  6. https://clinicaltrials.gov/ct2/show/NCT02003248?term=focused+ultrasound&cond=Parkinson+Disease&rank=2
  7. https://clinicaltrials.gov/ct2/show/NCT03319485?term=focused+ultrasound&cond=Parkinson+Disease&rank=9
  8. https://clinicaltrials.gov/ct2/show/NCT02263885?term=focused+ultrasound&cond=Parkinson+Disease&rank=10

Pipeline drugs, part 2: gastric emptying agents

Delayed gastric emptying (GE) affects over 70% of PD patients (1, 2).  In other words, food or medicines may take longer than normal to leave the stomach.  This may be a problem on many fronts.  First, there is the discomfort of a stomach that does not empty.  There is the issue of early satiety, or loss of appetite, too soon in the meal.  This may mean a person is not getting enough calories.  Additionally, there is the effect on absorption of nutrients and medications, which may be impaired for several reasons.  The stomach and the intestines have very specialized jobs to do, and these may be sidelined by slow emptying.  In the case of levodopa, absorption takes place just after leaving the stomach and entering the first part of the small intestine.  Delay of GE is associated with poor absorption of levodopa and subsequent delayed ON time (the time medications are working and symptoms are controlled), and worsening motor fluctuations (for example, unexpected medication failures such as doses that do not work at all, or suddenly stop working before they should) (3, 4).

For years, researchers have attempted to deal with the problem of GE.  In the 1990s, investigators showed that cisapride (Propulsid), a prescription drug FDA approved for severe nighttime heartburn in patients with gastroesophageal reflux disease (GERD), was associated with fewer motor fluctuations in PD (5-7).  The implication was that earlier emptying of stomach contents would improve drug absorption, and therefore function, in PD patients.  However, Janssen Pharmaceuticals Inc., made a voluntary action to withdraw the drug from the U.S. market after noting that as of December 31, 1999, use of cisapride had been associated with 341 reports of heart rhythm abnormalities, including 80 reports of death.  Per the press release, most of these adverse events occurred in patients who were taking other medications, or suffering from underlying conditions known to increase risk of cardiac arrhythmia associated with cisapride (8).

The problem is that there are not many drugs available to help GE.  In the U.S., the drug metoclopramide (Reglan) improves GE, but is also a very potent dopamine receptor blocker which can exacerbate symptoms of PD, and is generally not used in this population for that reason.  To be clear, metoclopramide is possibly the most common cause of drug-induced parkinsonism in the U.S., even among those who do not have underlying PD.

Outside of the U.S., domperidone is used, and generally does not alter PD motor symptoms because it affects serotonin, rather than dopamine, receptors.  Movement disorder and gastroenterology specialists in the U.S. still frequently prescribe the drug.  However, because there is no FDA approval, it is not covered by Medicare and patients must pay out of pocket.  Most of the time the drug is obtained by patients from Canada, though compounding pharmacists may also fill capsules in the U.S., and that is likely a more expensive option.   Interestingly, use of domperidone is associated with increases in plasma levodopa concentration among PD patients (9).

What is approved in the U.S.?  Erythromycin, an antibiotic FDA approved to treat infection, has long been known to assist in gastric motility and GE.  However, there are side effect and adverse event concerns, such as the development of bacterial resistance to antibiotics, and kidney and ototoxicity.  Erythromycin has been reported in some patients to cause severe and permanent damage to the inner ear, resulting in hearing loss, deafness, and severe vertigo.  The drug was trialed NCT02005029 (10) at Virginia Commonwealth University Parkinson’s Center, Richmond, Virginia, and results were posted online at ClinicalTrials.gov in February 2017, though it does not appear the data was published in a medical journal.  The study included 4 participants who received a single dose of erythromycin and 4 who received placebo.   There was a modest improvement of GE in the erythromycin group (105 minutes) versus placebo (180 minutes).  Also reported was a small increase in plasma levodopa concentration.  The drug is used off-label by some physicians and obviously requires monitoring of side effects.

There may be a better mechanism on the horizon.  In February 2018, international researchers in the United Kingdom, Europe, and Australia published a study in the journal Movement Disorders which tested another agent, camicinal (GSK962040), a “gastroprokinetic” drug (11).

Camicinal improves GE by binding to motilin receptors in the gut.  The primary outcome of the study was to evaluate the effect of camicinal on levodopa pharmacokinetics (the movement of drugs within the body) in PD patients.  The study was multicenter (11 sites).  Participants had PD with suboptimal control of motor function while taking levodopa.  Motor fluctuations were defined in the study as wearing off, peak-dose dyskinesia, and delayed or absent ON periods that impacted functional status or quality of life.  Patients had to have a stable levodopa dosing regimen for at least four weeks prior to screening, and therapy could not be altered during the study.  Participants further had to have a delayed GE of 70 or more minutes.

Nineteen patients who had been randomized to placebo, and 37 patients to caminical 50 mg once daily, completed the study.  This was a double-blinded investigation (patients and raters were blinded to whom was receiving placebo or study drug).  Patients were treated from seven to nine days, and there was a 14-day follow up period.  GE improved by an average of 5.3 minutes in the treatment group.  Though this seems modest, there were other measures which indicated improvement, and the trend toward improvement at the end of the study would seem to indicate that a longer duration trial would be in order.  For example, by the eighth day of the study the time for maximum level of levodopa in the plasma was 23 minutes sooner with camicinal than placebo.  And, MDS-UPDRS III scores (measures of motor function in PD) improved an average of 9 points by the eighth day.  With camincinal, there was a nearly two hour improvement in ON time.

This study was the first to describe the motilin receptor agonist camicinal motor response to levodopa in PD patients with motor fluctuations.

 

REFERENCES

  1. Cloud, et al.  Gastrointestinal features of Parkinson’s disease.  Curr Neurol Neursci Rep. 2011;11:379-384.
  2. Heetun, et al.Gastroparesis and Parkinson’s disease: a systematic review. Parkinsonism Relat Disord 2012;18:433-440.
  3. Doi, et al.  Plasma levodopa peak delay and impaired gastric emptying in Parkinson’s disease.  J Neurol Sci 2012;319:86-88.
  4. Muller, et al.  Impact of gastric emptying on levodopa pharmacokinetics in Parkinson disease patients.  Clin Neuropharmacol 2006;29:61-67.
  5. Djadldetti, et al.  Effect of cisapride on response fluctuations in Parkinson’s disease.  Mov Disord 1995;10:81-84.
  6. Cisapride was shown to improve plasma levodopa levels and therefore the motor function in patients with PD.
  7. Neira, et al.  the effects of cisapride on plasma L-DOPA levels and clinical response in Parkinson’s disease.  Mov Disord. 1995;10-66-70.
  8. https://www.fda.gov/ohrms/dockets/ac/00/backgrd/3634b1a_tab4a.htm
  9. Nishikawa, et al. Coadministration of domperidone increases plasma levodopa concentration in patients with Parkinson’s disease.  Clin Neuropharmacol 2012;35:182-184.
  10. https://clinicaltrials.gov/ct2/show/NCT02005029?term=erythromycin&cond=Parkinson+Disease&rank=1
  11. Marrinan, et al.  A randomized, double-blind, placebo-controlled trial of carmicinal in Parkinson’s disease.  Movement Disord. 2018;33(2):329-332.

 

New deep brain stimulation guideline by Dr. Rughani

Dr. Anand Rughani of Maine Medical Partners Neurosurgery is the first author of a new guideline for deep brain stimulation (DBS) in the treatment of Parkinson disease (PD). The paper was accepted for publication by the journal Neurosurgery in February, 2018 (1).    Neurosurgery is the official journal of the Congress of Neurological Surgeons, and publishes research on clinical and experimental neurosurgery.

The guideline by Rughani, et al. is for physicians helps establish whether the DBS target should be the subthalamic nucleus (STN), or globus pallidus internus (GPi), and is a useful tool which forms recommendations on the basis of seven key questions in order to tailor the choice for each patient.  The questions regard efficacy, reduction of medications, treatment of dyskinesias, improvement in quality of life, impact on cognitive function, impact on mood, and risk of adverse events.  The article can be read online (2).

Congratulations to Dr. Rughani, and thank you for this contribution to medicine.

REFERENCES

  1. Advani, et al. Congress of neurological surgeons systematic review and evidence-based guideline on subthalamic nucleus and globus pallidus internus deep brain stimulation for the treatment of patients with Parkinson’s disease: executive summary.  Neurosurgery.   2018 Mar 12. doi: 10.1093/neuros/nyy037. [Epub ahead of print]
  2. https://academic.oup.com/neurosurgery/advance-article/doi/10.1093/neuros/nyy037/4925265

Pipeline drugs, part 1: levodopa

A frequent question in the clinic is “What’s new in Parkinson’s?”  It is a deceptively simple question, because the truth is that there are thousands of researchers around the world working on Parkinson disease (PD), and there is no way to answer the question in anything close to the time of a follow-up appointment in the office. The volume of information is enormous.  A quick search for the term “Parkinson disease” in PubMed indicates that in 2017 there were 2,450 articles published in peer-reviewed medical journals. A search going back to 1947 yields 65,022 articles, with the majority in the last decade – a huge and growing database. Navigating that data requires a specialized skill set such as an advanced degree and training in medicine.  People with PD who don’t have that degree of training can arm themselves with knowledge by reading articles from MPDN, the Michael J. Fox Foundation, the National Parkinson Foundation, or other reputable sources. Mainers can attend meetings such as the April Parkinson Conference.

The purpose of this article, and the series on pipeline drugs that will follow, is to focus on some of the medications and therapies in development or under trial for the treatment of PD.  Search for the information occurred in two main ways. First was review of peer-reviewed medical journal articles describing completed studies. As described in the spring, 2018 MPDN article “the drug development process as outlined by the FDA,” studies proceed through phases prior to FDA approval.  Briefly, phase I trials provide information about how drugs work in the body and may be used to find appropriate drug doses and drug tolerance. Phase II trials are designed to give more safety data, refine research questions, develop research methods, and design new phase III protocols.  Phase III trials evaluate how effective a treatment is, and monitor for side effects or adverse reactions. After completion of a successful phase III trial a drug may receive FDA approval for a specific disease indication, such as PD, dyskinesias in PD, etc.  

If the drug is new, doctors may then begin to write prescriptions after FDA approval, though prescribing physicians are not limited to FDA approved indication.  

The second method in the search for information was via ClinicalTrials.gov, a freely accessible database provided by the U.S. National Library of Medicine that, as this article was being written, listed over 268,000 privately and publicly funded studies in the United States and 203 other countries.  Every study on the website is assigned a unique number, the ClinicalTrials.gov Identifier, a.k.a. the NCT number, which may be used to search for a particular trial.  With the ongoing trials described below NCT numbers are included for the reader because the contents are updated periodically and may give information regarding a more detailed description of the study itself, requirements of the study, participant inclusion versus exclusion criteria, study locations, contact information, and status of the study (recruiting, completed, etc.).  It is a very valuable resource for those who want to participate in, or keep up with certain areas of research, and it covers a wide array of worldwide PD investigation. On March 18, 2018 a search for the term “Parkinson disease” for example, produced 1,982 results. Fortunately, searches can be narrowed to contain only certain terms such as the NCT number, study drug, location, age of patient, etc.

As this article begins a series, I will divide into topics, which will include new developments with levodopa (this article), dopamine agonists, drugs already approved for another indication that have value in PD, some interventions that may seem unusual, such as blood transfusions for PD, and other topics.  

The accordion pill, a “novel gastroretentive delivery system,” is being developed by Intec Pharma.  The drug levodopa is only absorbed in the first part of the small bowel and competes with proteins for absorption, and thus diet may interfere (see the MPDN article “PD and Diet,” winter, 2016/17).  Likewise, many people with PD have problems with gastric motility (for eg., the stomach does not empty properly, to be covered later in the this series), constipation, or infection with Helicobacter pylori, all of which may change the rate of absorption of levodopa and negatively affect the window of opportunity for drug absorption. In other words, the drug may not wind up in the right place at the right time for absorption.  With immediate release levodopa, the half-life of time that a drug is at its therapeutic concentration in the blood, is only about 90 minutes; whereas the controlled release forms may have a half-life of several hours, though it is variable, and sometimes not well tolerated.  This is why some patients reach for liquid levodopa, Rytary, or the DUOPA pump (see article in this issue). A new attempt to address the problem is called the accordion pill because it expands in the GI tract to keep it in a region where dopamine will be absorbed while it slowly releases medication.  This approach to treating PD completed a phase II trial of 60 PD patients with doses of 250-500 mg levodopa, given once or twice daily over a 7-21 day period. Per the company’s website (unpublished data) there was a reduction in OFF time and dyskinesias. The baseline characteristics of the patients, including disease severity, were not apparent on the website.  The drug will need to complete a phase III trial before FDA approval is sought.

ClinicalTrials.gov lists two current phase III trials testing the accordion pill, though one must have completed the first 27 week trial to enter the second, a 12 month trial:

NCT02605434 is a  randomized, double-blind study in adult subjects with fluctuating PD (1).  Patients will be given a six week period to establish proper dosing of either carbidopa/levodopa (Sinemet) or the accordion pill (AP-CD/LD) before starting the double-blind maintenance period lasting 13 weeks.  In a double-blind study neither the patient nor the evaluator know which treatment is being given in order to eliminate bias.  Investigators plan to enroll 328 patients across 96 study sites (including Dartmouth and Boston University), and estimated primary completion is June, 2018.  Primarily, they want to measures a change in the percentage of daily OFF time (time medications have worn off and motor symptoms are not controlled) during waking hours.  Secondarily, they will measure a change from baseline through study completion in ON time (time medications are working and motor symptoms are improved or controlled) without troublesome dyskinesia during waking hours, a change in the number of total daily levodopa doses, and change in total Unified Parkinson Disease Rating Scale (UPDRS).  UPDRS is a standardized tool used by doctors to measure the effects of PD.   

NCT02615873  is an open extension trial taking place at 80 study sites that will follow patients who have completed NCT02605434 and evaluate the clinical effect and safety of the accordion pill over 12 months (2).

Subcutaneous (SC) levodopa, ND0612, is a patch/pump system being developed by Neuroderm, and is a delivery system of up to 360 mg levodopa over a 24 hour continuous infusion of a patch/pump (a patch with a small needle placed under the skin, thus subcutaneous).  It is designed for moderate to severe PD as an alternative to the Duopa pump or deep brain stimulation. Two phase II studies have shown the drug is safe and tolerable. Reportedly, the phase III studies will begin soon.  At this point, Boston will probably be the closest option.

NCT02726386 is an active trial, no longer recruiting, involving 100 advanced PD patients spread over 57 international study sites over a 12 month period (3). The primary outcome of this open-label safety study is to assess the long term safety and tolerability of continuous infusion of ND0612 by recording adverse events, vital signs, and local tolerability of the device and the drug.  Estimated study completion date is May, 2018.

NCT02782481, is a phase III, randomized, double-blind, parallel group study of the efficacy, tolerability, and safety of continuous ND0612 in addition to oral levodopa compared to placebo infusion over a 16 week study period (4).  The study is multicenter, and intended to include 150 people with PD who experience “motor complications despite optimized anti-PD therapy.” The study is past the estimated completion date, but is listed as still recruiting, though the single recruiting site listed on ClinicalTrials.gov is in Israel.  

NCT03462043  was announced in March, 2018 – not yet recruiting at announcement, and is an open-label, randomized, crossover study to assess the relative bioavailability (the proportion of the drug that will enter the circulation, and is thus able to have an effect) of ND0612 (the patch) versus jejunal pump levodopa (an external pump similar to DUOPA) in patients with advanced PD (5).  As yet, the only location listed is in Roma, Italy.

NCT02577523 is a multicenter, parallel-group, rater-blinded, randomized phase II clinical study in subjects with advanced PD investigating the efficacy, safety and tolerability of continuous SC infusion of 2 dosing regimens of ND0612H, a solution of  carbidopa/levodopa delivered continuous ND0612 infusion, compared to standard oral carbidopa/levodopa (6). Participants will be given the drug for a total of about 2.5 months. Investigators are recruited 38 participants at five international locations, and the study is ongoing, not recruiting.  

Inhaled levodopa, CVT-301, (INBRIJA) is a self-administered, inhaled form of levodopa by Acorda Pharmaceuticals.  The concept is to avoid the problems with GI tract absorption of levodopa by administering a powder with an inhaler which is absorbed by the lungs, enters the bloodstream, and is carried to the brain.  Phase I and II trials were in part funded by the Michael J. Fox Foundation for Parkinson’s Research.

A phase II study evaluated CVT-301, which PD patients self-administered to relieve OFF episodes (times when medications are not working and symptoms are not controlled) (7).  Study participants had to have two or more hours per day of OFF time despite oral levodopa four or more times per day. Patients were randomized: 43 to inhaled CVT-301, and 43 to inhaled placebo for 4 weeks.  Both groups used inhalers up to 3 times per day for OFF episodes. After two weeks the study-drug dose was increased from 35 to 50 mg. At the end of the fourth week investigators measured the change in UPDRS III score (a measure of the motor signs of PD) from OFF state to the post-dosing scores at 10, 20, 30, and 60 minutes.  The treatment effect was evident at 10 minutes and average OFF time changed by close to an hour daily. This might mean that as one was wearing off at the end of dose they used the inhaler to feel ON until the next dose.  Typically, this was the case for an average of two 25 minute periods daily. The most frequently reported adverse events among those receiving the drug were dizziness, cough, and nausea, each seen in 3 patients). Investigators concluded that CVT-301 was generally safe and well-tolerated.

The same group next entered a second phase II study which evaluated lung function in patients given a single dose of CVT-301 versus placebo, and in patients who received multiple doses/day over 4 weeks (8).  Assessment of pulmonary function (before and three hours after a dose) by spirometry (a portable way to measure lung function) was within normal ranges. The most common side effect was mild-to-moderate cough (21% in the single dose group, 7% in the four week treatment group).     

February, 2017 Acorda  announced positive phase III clinical trial results for CVT-301 with the SPAN-PD trial (9).   SPAN-PD was a phase III, randomized, double-blind, placebo-controlled clinical trial that evaluated the efficacy and safety of CVT-301 compared with placebo in people with PD who experienced motor fluctuations (OFF periods). All took a stable regimen of oral carbidopa/levodopa, and were maintained on other existing PD medications.  The study included 339 study participants who were randomized to CVT-301 84 mg, CVT-301 60 mg, or placebo. Participants self-administered treatment up to five times daily for 12 weeks.

At week 12 the change in UPDRS III score compared with placebo was measured at 30 minutes after using the inhaler.  SPAN-PD reportedly showed the 84 mg dose resulted in a 9.83 point improvement in the UPDRS III compared with a 5.91 drop for the placebo group.  In the 84 mg dose group nausea was reported in 5.3% (2.7% with placebo), cough in 15% (1.8% with placebo), upper respiratory tract infection in 6% (2.7% with placebo).   When cough was reported, it was “typically mild and reported once per participant during the course of treatment.” Three of 227 participants receiving CVT-301 discontinued the study due to cough.

Data from the SPAN-PD trial were presented at the International Congress of Parkinson’s Disease and Movement Disorders (MDS) in June 2017.

February 20, 2018 Acorda Therapeutics, Inc. announced that the FDA had accepted for filing its New Drug Application (NDA) for INBRIJA, “an investigational inhaled levodopa treatment for symptoms of OFF periods in people with Parkinson’s disease taking a carbidopa/levodopa regimen” (10).  Under the Prescription Drug User Fee Act (PDUFA), the FDA has set a target date of October 5, 2018.  

REFERENCES

  1. https://clinicaltrials.gov/ct2/show/NCT02605434?term=accordion+pill&cond=Parkinson+Disease&rank=1
  2. https://clinicaltrials.gov/ct2/show/NCT02615873?term=accordion+pill&cond=Parkinson+Disease&rank=2
  3. NCT02726386 https://clinicaltrials.gov/ct2/show/NCT02726386?term=subcutaneous&cond=Parkinson+Disease&rank=1
  4. https://clinicaltrials.gov/ct2/show/NCT02782481?term=subcutaneous+levodopa%2C+neuroderm&cond=Parkinson+Disease&rank=2
  5. https://clinicaltrials.gov/ct2/show/NCT03462043?term=subcutaneous+levodopa%2C+neuroderm&cond=Parkinson+Disease&rank=3
  6. https://clinicaltrials.gov/ct2/show/NCT02577523?term=subcutaneous+levodopa%2C+neuroderm&cond=Parkinson+Disease&rank=5
  7. LeWitt, et al.  A randomized trial of inhaled levodopa (CVT-301) for motor fluctuations in Parkinson’s disease. Mov Disord. 2016;31(9):1356-65.
  8. LeWitt, et al. Pulmonary Safety and Tolerability of Inhaled Levodopa (CVT-301) Administered to Patients with Parkinson’s Disease.J Aerosol Med Pulm Drug Deliv. 2017 Nov 21. doi: 10.1089/jamp.2016.1354. [Epub ahead of print] full artice: https://www.liebertpub.com/doi/10.1089/jamp.2016.1354
  9. http://ir.acorda.com/investors/investor-news/investor-news-details/2017/Acorda-Announces-Positive-Phase-3-Clinical-Trial-Results-for-CVT-301/default.aspx
  10. http://ir.acorda.com/investors/investor-news/investor-news-details/2018/Acorda-Announces-FDA-Acceptance-of-New-Drug-Application-for-INBRIJA-levodopa-inhalation-powder/default.aspx