Monday, October 14, 2013

Present and future therapies for Parkinson's disease: A hopeful future



Those of us in the Biomedical Science Master’s program at Regis University where this post originates have spent the first quarter of the semester looking at lysosomal storage disorders including Parkinson’s disease.  Shachar et al. (2011) describes a causal relationship between these two diseases where mutations in the glucocerebrosidase gene caused by Parkinson’s disease results in an enzyme deficiency consistent with Gaucher disease, the most prevalent lysosomal storage disorder.  The main focus of our investigation of lysosomal storage disorders including Parkinson’s disease has been on gene therapy trials using murine models.  The current success in treating these diseases in the murine model and non-human primates is high and promising, however there is a large contrast in the success of treating these diseases in human subjects.  

Parkinson’s disease is the second most prevalent age related neurodegenerative disease with an average onset occurring close to 60 years of age, although early onset can occur.  The symptoms of the disease most commonly include tremor, rigidity, bradykinesia, and several non-motor symptoms more commonly associated with cognitive functioning (Douglas 2013).  Rush University Medical Center (2013) describes two methods currently being used for treating Parkinson’s disease in humans which include symptomatic therapies and protective therapies.  Symptomatic therapies focus on supplementing dopamine because the natural release of dopamine by cells is affected by the disease, many drugs fall into this category and have been available for many years to treat individuals with Parkinson’s disease.  Protective therapies work to delay the degenerative processes occurring in the brain and are largely focused on gene therapies, these treatments are currently only available to human participants in a handful of clinical trials. 

The success of treating Parkinson’s disease has recently hit the media with the full time return of Michael J. Fox to our television sets who postponed his acting career in 2000 because of his worsening symptoms of Parkinson’s disease.  His doctors have recently been able to find the right balance of medications and dosage levels that work for him.  While the exact treatment regimen used by Michael J. Fox is between him and his doctors, it is most likely through the use of current symptomatic therapies.  It is important to note that symptomatic therapies are not a cure for Parkinson’s disease and they are only able to mask the symptoms of the disease for some time. 

Symptoms of Parkinson’s disease are typically measured by the Unified Parkinson’s Disease Rating Scale, UPDRS (Kordower and Bjorklund 2013), which rates symptoms from zero to four with a score of zero representing normal functioning and a score of 4 representing severe impairment.  This rating scale can be used to measure both “on” and “off” scores.  “On” time is considered the period where therapies are successful in treating the symptoms and improving motor control with the right balance of dopamine supplemented to the system.  “Off” time is considered the period where the amount of dopamine being supplemented is out of balance and symptoms worsen, this can occur during the initial assessment of the appropriate dosage of dopamine which is highly variable per individual and as the effects of the drug where off which will typically occur after some time of the drug being effective.  Current symptomatic therapies are continually being improved to increase the amount of “on” time an individual experiences from the drugs and dosages are continually monitored by the prescribing doctors.

The most common symptomatic therapy is focused on increasing dopamine production within the nigral neurons and is accomplished through drugs like Levodopa and more often Levodopa in combination with Calbidopa (Douglas 2013).  Levodopa targets the remaining nigral neurons that are still exhibiting proper functioning to increase dopamine production. The first two years of Levodopa therapy on treating Parkinson’s disease shows large success in controlling the symptoms of the disease, however after this time the drug can show a wearing off effect and symptoms once masked by the therapy tend to return (Rush University 2013).  Following the wearing off effect of Levodopa dopamine agonist, Pramipexole and Ropinirole, along with COMT inhibitors, Entacapone and Tolcapone, can be used to increase delivery of Levodopa to the brain and decrease the breakdown of Levodopa (Rush University 2013).  Other symptomatic medications are available that work through similar mechanisms.

A second symptomatic treatment option for individuals with Parkinson’s disease is deep brain stimulation, referred to as DBS.  While the exact target within the brain is still widely debated, DBS is a surgical treatment that places an electrical wire into the brain tissue and high frequency stimulation is delivered through a small battery pack that is placed under the skin near the thoracic cavity (Rush University 2013).  DBS works by inhibiting glutamate release within the cells of the subthalamic nucleus (Douglas 2013).  This treatment is most often reserved for those individuals who show little improvement with symptomatic drug therapies.  While both DBS and combination drug therapies can work well to reduce symptoms of the disease for some time the future for treating Parkinson’s disease lies with protective therapies that aim to actually stop and reverse the effects of the disease on neurons within the CNS.

Current research being conducted on protective therapies is focused on gene therapy and stem cells.  The association of many mutated genes with Parkinson’s disease has been researched and includes, among many others, the PARK1 locus which encodes for alpha-synuclein, several autosomal recessive genes and proteins including Parkin, PINK1, DJ-1, and an autosomal dominant gene, LRRK2, which is the most correlated gene mutation found within individuals with Parkinson’s disease (Douglas 2013).  Protective therapies aim to stop and reverse the damage to the brain caused by Parkinson’s disease through introducing new stem cells to the affected areas of the brain or by introducing viral vectors that can deliver new functioning genes.  Although these therapies show positive clinical significance in the lab with both mice, rat, and non-human primate models, trials on humans have proven to be less effective.  The discrepancy of the effectiveness of treatment is related to both the blood brain barrier and general difficulties accessing the affected areas of the brain while delivering the therapy.

Several studies investigating the effects of glial cell line derived neurotrophic factor, GDNF, have found that protection and restoration of neurons in animal models was possible.  Clinical trials in humans were conducted by Nutt et al. (Kordower and Bjorklund 2013) and used infusions of recombinant GDNF proteins that were directly administered into the lateral ventricles through mechanical pumps. However the double blind study did not produce any significant results and a number of side effects of the treatment were reported.  A postmortem autopsy revealed that the GDNF was unable to diffuse out of the lateral ventricles and therefore unable to have an effect.  A second open label study used a similar method but injected the GDNF directly into the postcommissural putamen which showed promising results in phase I clinical trials and improvement of motor symptoms by about 33% were seen.   PET scans revealed increased fluoradopa uptake after about a half year of treatment.  However when taken to phase II clinical trials no significant benefit was seen and antibodies towards GDNF were found along with an identified risk for cerebellar damage that caused early termination of the trial (Kordower and Bjorklund 2013).

Other methods of delivery of GDNF proteins have been developed but have been yet to see human trials.  These include genetically engineered cells that are encapsulated in a semi-permeable membrane that can be delivered to the brain and avoid detection by the immune system while delivering therapeutic agents to the cell (Kordower and Bjorklund 2013).  Brendan Harmon from Northeastern University (Science Daily 2013) has also developed an intranasal approach for delivery of GDNF to the brain that is able to pass the blood brain barrier that may create a noninvasive method for delivery of therapeutic agents to the brain.  This method has proven to be successful in rat models but has not yet seen human trials (Science Daily 2013).

Other human trials investigated the use of adeno-associated virus vectors to deliver NTN to the brain, NTN is similar to and works in the same way as GDNF (Douglas 2013). This AAV2-NTN technique showed an 80% improvement level that lasted for up to 12 months following administration in Rhesus monkeys.  However when taken to clinical trials in humans complications in the delivery to the brain during surgery resulted in one death and other individuals who underwent successful surgeries showed several major side effects.  Despite these complications results were promising and this method was taken to a phase II clinical trial but again no significant results were found using human subjects after one year (Douglas 2013).  A follow up study did find significant results after 18 months which may suggest that the procedure was a success but may need more time to work and show corrections within human subjects, further trials with longer follow up times is under way. 

Within the dopaminergic biosynthetic pathway dopamine is converted to another molecule known as AADC and AADC has been used therapeutically to reduce Parkinsonism symptoms.  Adeno associated vectors using AADC have reached Phase I clinical trials and motor improvements up to 75% have been seen (Douglas 2013).  Outweighing the clinical success of the AAV-AADC many patients within the sample developed brain hemorrhages which were related to the surgery delivering the AAV.  A follow up to this particular study was performed and it was found that the positive effected waned after a one year period (Douglas 2013).

AAV vectors using CERE-120, which encodes for Neurturin, also known as NTRN, have also reached Phase II trials in humans based upon highly positive results in monkeys (Kordower and Bjorklund 2013).  Phase I trials using this vector again showed positive results although no significant difference was found to exist between low and high dosage groups but very few side effects were reported.  Given the low amount of side effects the AAV-CERE-120 vector reached Phase II clinical trials but again showed no significant results with only limited clinical benefit (Kordower and Bjorklund 2013).  Interestingly a follow up study did show more significant results after 15 and 18 month periods again suggesting that longer treatment follow ups may be necessary.

The first successful Phase II clinical trials seen in humans was with a gene therapy known as NLX-P101 which significantly reduced motor symptoms (USA Today Magazine 2012).  This treatment used glutamic acid decarboxylase, known as GAD, was injected into the brain using a virus vector.  GAD is able to produce GABA which is an inhibitory neurotransmitter that is able to decrease the firing of neurons.  So while dopamine loss within the brain is most often associated with the major mechanism of the symptoms of Parkinson’s disease a decrease in GABA is also seen.  By reintroducing GABA into the brain the dysfunction of the circuitry responsible for motor coordination can also be reduced (USA Today Magazine 2013).

In summary new methods for treating Parkinson’s disease are being developed and tested and the results are promising.  Both mouse and rat studies along with studies on non-human primates have produced significant data that at the very least gives hope in reducing and eliminating the effects of Parkinson’s disease on the brain.  However transferring the success of these trials to human trials have proved to be difficult for several reasons including the inability of therapeutic agents to cross the blood brain barrier and difficulties of surgical methods used to directly deliver the therapeutic agents to the correct regions of the brain.  The future for an individual diagnosed with Parkinson’s disease is getting better every day, with increased efficacy of symptomatic drugs that able to mask the symptoms for several years and the advancement and success of therapeutic drugs in correcting the effects on the brain gives hope that someday a cure for Parkinson’s disease may be found.  

References

Douglas MR.  2013.  Gene therapy for Parkinson’s disease.  Expert Rev Neurother.  13(6): 695-705

Federation of American Societies for Experimental Biology.  2013.  Science Daily.  A noninvasive avenue for Parkinson’s disease gene therapy.

Kordower JH, Bjorklund A. 2013. Trophic factor gene therapy for Parkinson’s disease.  Movement Disorder Society. 28(1): 96-109

Rush University Medical Center.  2013.   Clinical Services Parkinson’s disease.  Chicago

USA Today Magazine.  2013.  Gene therapy reverses symptoms.  The Society for Advancement in Medicine.  McLean, VA.



    


No comments:

Post a Comment