Crimson Publishers Publish With Us Reprints e-Books Video articles

Full Text

Surgical Medicine Open Access Journal

Surgical Implantation of Autologous Dopamine Neuron Progenitor Cells (DANPCs) Into the Putamen of Patients with Parkinson’s Disease

Michael AS Guth*

Risk Management Consulting, USA

*Corresponding author:Michael AS Guth, Risk Management Consulting, Oak Ridge, Tennessee, USA

Submission: November 20, 2024;Published: December 16, 2024

DOI: 10.31031/SMOAJ.2024.06.000629

ISSN 2578-0379
Volume6 Issue1

Abstract

Objective: To describe a new clinical trial testing the safety and efficacy of implanting autologous Dopamine Neuron Progenitor Cells (DANPCs) into the brain of patients with Parkinson’s Disease (PD) as regenerative therapy for dopamine deficiency caused by PD.
Background: PD affects the putamen through multiple mechanisms. Dopamine depletion occurs as PD causes a gradual loss of dopaminergic neurons in the substantia nigra pars compacta, leading to reduced dopamine levels in the putamen. This dopamine depletion then contributes to motor dysfunction.
Additionally, the putamen can undergo atrophy, which is associated with both motor symptoms and cognitive impairment. PD can be managed with medications for a limited period, but PD has no cure. The standard treatments for PD have remained unchanged for several decades and do not slow the disease progression. Cell Replacement Therapy (CRT) presents an alternative approach that has the potential to restore striatal dopaminergic input and ameliorate debilitating motor symptoms in PD.
Methods: The ASPIRO clinical trial (Currently in Phase 1 and 2a) purports to be the first multi-center trial for an autologous neuron replacement therapy for Parkinson’s disease. The clinical trial tests an experimental biologic product derived from autologous skin cells converted to induced pluripotent stem cells. The stem cells are differentiated into Dopamine Neuron Progenitor Cells (DANPCs). Dose escalation will be achieved by bilateral injection of these DANPCs into the putamen. After drilling a burr hole in the skull, the neurosurgeon inserts a small catheter with the help of a smart frame through the parenchymal tissue to the putamen.
Data sources & results: The ASPIRO Trial is currently enrolling patients by invitation, but no results are yet available. The first trial patient has been successfully dosed with his autologous DANPCs.
Discussion: Case studies involving male patients with fetal nigral transplants in the striatum have demonstrated that the transplanted dopamine neurons can survive and reestablish connections in the striatum for a minimum of 10 years, even as the disease continues to progress and destroy the patients’ native dopamine neurons. Intraparenchymal injections are more surgically invasive than other administration routes. Still, they have some advantages: the dose of the vector can be lower when using intraparenchymal injections, and preexisting neutralizing antibodies have little effect on the transduction efficacy of biologics injected directly into the putamen. A prominent factor contributing to acute cell death during injection is the influence of mechanical forces on cells; specifically, the extensional force arising from differing velocities distorts cell length.

Keywords:Parkinson’s disease; Putamen; Caudate nucleus; Induced pluripotent stem cells; Sporadic Parkinson’s; Substantia nigra pars compacta; MRI-guided neurosurgery; Cell replacement therapy; Neurosurgical delivery

Introduction

Parkinson’s Disease (PD) ranks as the second most prevalent neurodegenerative disorder, following Alzheimer’s disease. It is a chronic condition affecting the brain, leading to a variety of symptoms. These include motor difficulties such as uncontrollable tremors, rigidity, slowness of movement, challenges with balance and coordination, and an overall inability to initiate movement. Additionally, individuals may experience mental and behavioral changes, including depression, anxiety, memory impairments, and visual hallucinations. Sleep disturbances, characterized by excessive periodic limb movements during sleep, are also common. Other associated symptoms may encompass loss of smell, constipation, excessive salivation, pain, and urinary issues. Symptoms typically emerge gradually and tend to escalate over time, with the initial sign often being a tremor in one hand, although it can also impact the arms, legs, feet, and facial muscles. While medications can help manage the condition, there is currently no known cure for Parkinson’s disease. As of 2024, the most recent advancements in the treatment of Parkinson’s Disease (PD) symptoms encompass a minimum of nine pharmacological and non-pharmacological approaches. (1) Produodopa has received approval from the United Kingdom’s National Institute for Health and Care Excellence (NICE) for individuals with PD who suffer from movement-related symptoms.

This treatment combines several medications, including foslevodopa, which is converted into dopamine and is administered continuously via a small pump. A syringe linked to the pump is connected to a cannula that is inserted beneath the skin, with the pump being worn in a specialized vest or pouch. (2) Vyalev, a solution of carbidopa and levodopa prodrugs (foscarbidopa and foslevodopa), has been approved for 24-hour continuous subcutaneous infusion to address motor fluctuations in PD. (3) Crexont is a newly formulated medication that combines immediateand extended-release levodopa with carbidopa. Levodopa serves as the natural precursor to dopamine, which is deficient in PD, while carbidopa facilitates the entry of levodopa into the brain and mitigates side effects. (4) Safinamide is an innovative experimental drug that demonstrates promise in treating both PD and epilepsy by blocking voltage-dependent sodium and N-type calcium channels, thereby inhibiting glutamate release. (5) Methylphenidate and atomoxetine are noradrenaline reuptake inhibitors currently under investigation for their potential effects on balance and gait. (6) Levodopa inhalation powder is a formulation that can be utilized as needed when the effects of other medications diminish. (7) Focused ultrasound represents a minimally invasive treatment that employs heat beams to generate acoustic energy, precisely targeting brain areas responsible for voluntary movement. (8) Deep brain stimulation is a surgical intervention that entails the implantation of electrodes in the brain to deliver electrical impulses, disrupting abnormal neural circuitry. (9) Bemdaneprocel is a cell therapy that has shown encouraging results in clinical trials, including enhancements in motor function and symptom management.

Clinical trials investigating the transplantation of cells into the brains of Parkinson’s disease patients include the following:
a. Fetal ventral mesencephalic tissue: This approach involved two patients who received grafts of tissue enriched with dopaminergic neuroblasts. Long-term evaluations conducted 15 and 18years post-transplantation assessed the efficacy of these grafts [1].
b. Dopamine neurons from fetal pigs: A phase I study reported promising outcomes; however, subsequent results from a double-blind phase II trial indicated that recipients of porcine neurons exhibited no significant clinical benefits compared to those undergoing sham surgery [2].
c. Patient-derived dopaminergic progenitor cells: Research findings hinted at potential benefits over a 24-month period, demonstrating stability in clinical and imaging assessments. Notably, patients did not report adverse events or a decline in function during this timeframe [3].
d. Allogeneic transplantation: Several clinical trials are currently underway, including [4].
- Kyoto University/CiRA/Sumitomo: Phase I trial initiated
- Sloan Kettering/BlueRock Therapeutics/Bayer: Phase I safety trial completed in August 2023, with a Phase II trial proposed for 2024
- Lund University/StemPD/Novo Nordisk: Phase I trial initiated
- Scripps Research/Aspen Neuroscience: Phase I trial initiated

Stem cell-based strategies have emerged as a central focus in the field of regenerative medicine for the treatment of Parkinson’s disease.

In light of the decreasing effectiveness of symptom treatments as the disease advances, a novel surgical clinical trial is currently being conducted to implant autologous Dopamine Neuron Progenitor Cells (DANPCs) into the putamen of patients diagnosed with Parkinson’s Disease (PD) [5]. The putamen is a rounded structure situated at the base of the forebrain, specifically within the telencephalon. Together with the caudate nucleus, the putamen constitutes the dorsal striatum. This subcortical structure is part of a collection of structures known as the basal ganglia or basal nuclei [6]. The term “basal nuclei” is more precise, as “ganglia” typically refers to clusters of nerve cell bodies located outside the central nervous system. The putamen is located adjacent to the globus pallidus, and the two are sometimes collectively referred to as the lentiform or lenticular nucleus. The putamen plays a significant role in reward-related learning and motor control, encompassing functions such as speech articulation, language processing, reward mechanisms, cognitive abilities, and addiction [7,8]. PD affects the putamen through multiple mechanisms. Dopamine depletion occurs as PD causes a gradual loss of dopaminergic neurons in the substantia nigra pars compacta, leading to reduced dopamine levels in the putamen. This dopamine depletion then contributes to motor dysfunction. Additionally, the putamen can undergo atrophy, which is associated with both motor symptoms and cognitive impairment.

Shape changes, particularly in the right putamen, are also observed. Furthermore, decreased activity in the putamen is another effect of PD [9-11]. PD affects various brain regions beyond the motor areas. The caudate and nucleus accumbens often experience dopamine depletion, which can have significant consequences. Early dopaminergic denervation in the caudate is linked to an increased risk of cognitive impairment, depression, and gait problems within the next 4 years [12]. Similarly, dopamine loss in the nucleus accumbens, sometimes referred to as “Mavridis’ atrophy,” is associated with cognitive deficits such as impaired reward processing, reduced motivation, apathy, and potential depression in PD patients [13,14]. While pharmacological dopamine replacement therapy can partially restore motor function, longterm use of these drugs may lead to motor complications. Overall, the widespread impact of dopamine depletion in PD underscores the need for comprehensive treatment approaches targeting both motor and non-motor symptoms.

Methods

“Sporadic Parkinson’s” refers to the most common form of PD, where the cause is unknown and there is no clear family history. PD is the broader term that encompasses both sporadic and familial (genetic) forms. Clinical trial NCT06344026, captioned “Phase 1/2a Study of ANPD001 in Parkinson Disease (ASPIRO),” will test the efficacy and safety of implanting the biologic, ANPD001, into the putamen of patients with sporadic PD. ANPD001, is an experimental product derived from autologous skin cells converted to induced pluripotent stem cells. The stem cells were differentiated into Dopamine Neuron Progenitor Cells (DANPCs). Dose escalation will be achieved by bilateral injection of these DANPCs into the putamen [5].

Preparing the DANPCs

To prepare autologous dopamine neuron progenitor cells, researchers initiate the process by obtaining a skin biopsy from the patient to isolate fibroblasts. These fibroblasts are subsequently reprogrammed into Induced Pluripotent Stem Cells (iPSCs) through the introduction of specific reprogramming factors. Following this, the iPSCs are differentiated in a controlled laboratory setting to yield dopamine neurons. This methodology effectively generates a population of progenitor cells capable of maturing into functional dopamine-producing neurons, all originating from the patient’s own somatic cells, thus ensuring their autologous nature [3]. Key steps in the process:
a. Skin biopsy: A small sample of skin is taken from the patient to obtain fibroblasts.
b. Reprogramming to iPSCs: The fibroblasts are genetically reprogrammed using specific factors (like Oct4, Sox2, Klf4, and c-Myc) to become pluripotent stem cells capable of developing into any cell type in the body [15].
c. Differentiation into dopamine neurons: The iPSCs are then cultured in a specific environment with specific chemical signals to guide their differentiation into dopamine neurons, specifically targeting the Midbrain Dopamine (mDA) neuron lineage [16].
d. Purification and selection: The resulting cell population is carefully screened to ensure a high proportion of dopamine neurons and to remove any remaining undifferentiated cells [3,16].
e. Benefits of autologous dopamine neuron progenitor cells:
i. Immune compatibility: Since the cells are derived from the patient’s own body, there is no risk of immune rejection, eliminating the need for immunosuppressive drugs.
ii. Personalized treatment: The cells can be tailored to the specific genetic profile of each patient.
f. Challenges of autologous dopamine neuron progenitor cell therapy:
i. Complex process: Generating a sufficient quantity of high-quality dopamine neurons from iPSCs can be technically challenging.
ii. Potential for mutations: There is a risk that the reprogramming process could introduce genetic mutations into the iPSCs.
iii. Clinical translation: Further research is needed to optimize the differentiation process and ensure the safety and efficacy of autologous cell therapies in clinical applications. Hence, the ASPIRO trial attempts to prove this technology can be used to treat patients with PD.

How DANPCs are cultured

The iPSCs are carefully differentiated into dopamine neuron progenitors through a controlled process involving specific growth factors and signaling pathways, essentially mimicking the early stages of brain development in the ventral midbrain region, where dopamine neurons originate, allowing for the generation of a patient-matched population of dopamine neuron progenitors ready for potential transplantation back into the patient [3,16]. Key steps in the process:
Cell collection: Reprogramming to iPSCs: Neural induction, differentiation into dopamine neurons: The iPSCs are cultured in a specific medium to initiate differentiation into neural progenitors, often by manipulating signaling pathways like the Wnt and Sonic Hedgehog (Shh) pathways [16,4].
Midbrain specification: Further differentiation steps are taken to guide the neural progenitors towards a midbrain identity, the region in the brain where dopamine neurons originate, by using specific growth factors like GDNF and BDNF [17-19].
Cell selection and purification: The differentiated cells are then purified using techniques like FACS (Fluorescence-Activated Cell Sorting) to select for cells expressing markers specific to dopamine neurons (e.g., Tyrosine Hydroxylase (TH)). The cells are exposed to additional factors to promote the development of mature dopamine neurons, identifiable by markers like Tyrosine Hydroxylase (TH) [16,19].
Expansion and maturation: The purified dopamine neuron progenitors can be further expanded in culture to produce a sufficient quantity of cells suitable for transplantation. Frequently, the population of dopamine neuron progenitors is enriched utilizing cell sorting techniques, which ensures a high yield of functional cells prior to the transplantation process [20,21].
Important considerations: Throughout the entire process, it is essential to implement stringent quality control measures to guarantee that the cells exhibit genetic stability, adequately express relevant markers, and remain free from contamination. For potential clinical applications, the cell culture process must adhere to strict Good Manufacturing Practice (GMP) guidelines to ensure the quality and safety of the cells. Regular testing is conducted to assess the purity, viability, and functionality of the differentiated dopamine neurons. The cultured dopamine neuron progenitors are typically transplanted into the targeted area of the brain utilizing stereotactic surgery. Additionally, the cell culture process must be optimized to align with GMP standards [16,21,22].

The ASPIRO trial will assess the effects on Parkinson’s Disease (PD) symptoms, safety, tolerability, and cell survival for 5 years post-transplant, using MRI and PET imaging scans of the brain. Safety and tolerability will be further evaluated annually for an additional 10 years via telephone follow-up, for a total of 15 years. The primary outcome measures are the incidence and severity of Treatment-Emergent Adverse Events (TEAEs) and serious adverse events. The secondary outcome measures will include “ON” time without troublesome dyskinesia, with a 1-year primary follow-up and 5-year long-term follow-up. Additionally, the trial will report the post-injection change in the Movement Disorder Society - Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) Part II (Activities of Daily Living-ADL) and Part III (motor score) in the “ON” state. The “ON” state refers to the period when Parkinson’s medication is effectively managing symptoms, allowing for normal movement with minimal side effects [5].

The majority of dopaminergic neurons are typically lost by the time Parkinson’s is diagnosed, leading to progressive deterioration of motor and neurological function. To replace these lost cells, neurosurgeons must target the putamen in the basal nuclei with a high degree of precision. The ClearPoint System’s® advanced intraoperative MRI-guided techniques allow surgeons to transplant the patient’s new Dopamine-Producing Cells (DANPCs), one microliter at a time, directly into the putamen where they are most needed. This transplantation procedure utilizes the SmartFlow® Cannula and a metered syringe. The minimally invasive SmartFlow® Cannula, less than 2 millimeters in diameter, enables precise delivery of therapeutic agents to the patient’s brain. The ClearPoint System provides real-time MRI-based navigation for the neurosurgeon, confirming the desired anatomical target is reached with sub millimetric accuracy.

No diagrams or pictures are provided in the National Library of Medicine’s clinicaltrials.gov file for the ASPIRO trial (NCT06344026). However, ClearPoint Neuro, the medical device company that manufactures both the Clear Point System and the SmartFlow Cannula provides the video shown below that demonstrates a general neurosurgical procedure with the two devices. In place of the pump shown at the end of the video to deliver the biologics to the brain, the ASPIRO trial neurosurgeons will connect Aspen Neurosciences’ syringe prefilled with autologous DANPCs to the canula and depress the plunger. Note the image shown below is a link to a video. If for any reason, the reader cannot get the video link to deploy correctly by clicking on the triangle in the center of the image, the video can be viewed at the following URL https://www. youtube.com/watch?v=cc3GZ5cvEEw and shows how the DANPCs are delivered to the putamen in the mid-brain region (Figure 1).

Figure 1:How the DANPCs are delivered to the putamen in the mid-brain region.


During the surgery, the patient’s head is securely held in place using a stable fixation frame that is anchored to the skull with bilateral bolts and plug endcaps. A smartgrid, which connects to the software used for navigating through the brain, is then attached to the shaved scalp. This smartgrid consists of a 6x6 array of squares, with each square containing four potential entry points for accessing the skull, located at each corner. The neurosurgeon can simultaneously view high-resolution MRI scans of the patient’s brain through the ClearPoint workstation software. These scans help the surgeon select the best trajectory to reach the putamen target. The software then determines the optimal entry point on the smartgrid for the operation. To begin, the surgeon makes an incision in the scalp to access the skull and drills a small burr hole at the preferred entry point on the smartgrid. A circular ring is placed around the burr hole to prevent the scalp from covering it during the procedure. Next, the surgeon affixes the ClearPoint smartframe securely over the burr hole by inserting screws into the skull at each of the four corners of the frame. The smartframe software detects any discrepancies between the current and desired surgical trajectory and recommends adjustments to ensure extreme accuracy. The surgeon makes these adjustments using color-coded hand controllers located on the smartframe. Outer knobs control pitch and roll, while inner knobs enable finer tuning of the trajectory to reach the desired location in the putamen. After confirming the alignment, a small cannula (<2mm diameter) is inserted into the brain through the central lumen of the smartframe. This cannula is visible on the MRI, allowing for verification of target accuracy. By using a single insertion, the procedure minimizes the number of intraparenchymal passes and creates a tissue seal to ensure that the infusion is delivered precisely as intended. The infusion into the putamen can be monitored in real-time through the workstation display. The design of the medical device setup is intended to provide submillimeter accuracy, resulting in a minimal incision, increased safety, rapid closure, and faster recovery.

The Autologous-derived Study of a Parkinson’s Investigational Regenerative therapy in an Open-label trial (ASPIRO) is a Phase 1/2a clinical trial that aims to assess the safety, tolerability, and potential efficacy of ANPD001 in patients with moderate to severe Parkinson’s disease. The secondary outcomes measures include [5]:
a. “ON” time without troublesome dyskinesia [Time Frame: 1 year (primary follow up) and 5 years (long term follow up)].
b. Post-injection change in Movement Disorder Society- Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) Part II (Activities of Daily Livin-ADL) and Part III (motor score) in the ON state (total score and scores for Parts I-IV) [Time Frame: 1 year (primary follow up) and 5 years (long term follow up)]. The Part II score range is 0 to 52, with 0 being normal and 30 and above being severe.
c. Post-injection change in the Movement Disorder Society- Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) Part III score in the practically defined OFF state [Time Frame: 1 year (primary follow up) and 5 years (long term follow up)]. The Part III score range is 0 to 132 with 0 being normal and 59 and above being severe.
d. Post-injection change in the 18F-DOPA uptake in the putamen [Time Frame: 1 year (primary follow up) and 5 years (long term follow up)]. The post-injection change in the 18-fluorodopa uptake in the putamen as measured from baseline via Positron Emission Tomography (PET).
e. Incidence and severity of treatment emergent adverse events during long term follow-up (Continued Safety and Tolerability) [Time Frame: 14 years of long term follow up (4 years in-person visits and 10 additional years follow up via telephone call)] [5].

ASPIRO trial inclusion criteria

Participants must be between 50-70 years of age at the time of consent for the trial-ready cohort study ANPD001-01.
a. Participants must meet all eligibility requirements for inclusion in the trial-ready cohort in the clinical study ANPD001-01.
b. Participants must have been diagnosed with Parkinson’s Disease at least 4 years prior.
c. Participants must have an unequivocal motor response to Levodopa treatment.

ASPIRO trial exclusion criteria

a. Prior brain surgery that, in the neurologist’s or neurosurgeon’s opinion, contraindicates administration of ANPD001.
b. History of intracranial therapy for Parkinson’s Disease (PD), including Deep Brain Stimulation (DBS), Focused Ultrasound (FUS), gene therapy, or other biological therapy.
c. History of cognitive impairment or dementia.
d. History of clinically significant Dopa Dysregulation syndrome.
e. History of epilepsy, stroke, multiple sclerosis, poorly controlled or progressive neurological disease (other than PD), or poorly controlled cardiovascular disease.
f. Inability to temporarily stop anticoagulation or antiplatelet therapy for at least 2 weeks.
g. History of malignancy (cerebral or systemic) within the prior 5 years, except for treated cutaneous squamous or basal cell carcinomas.
h. Contraindication to MRI and/or use of gadolinium.
i. Weight>300lbs or Body Mass Index (BMI) >35.
j. Uncontrolled diabetes (HbA1c>7.0%) or any other acute or chronic medical condition that would significantly increase the risks of a neurosurgical procedure.
k. Pregnancy or lactation.
l. Significant drug-induced dyskinesia (>2 for any body part on the Abnormal Involuntary Movement Scale [AIMS]).
m. Male or female with reproductive capacity who is unwilling to use barrier contraception for 3 months postadministration of the investigational product.
n. Unable to comply with the protocol procedures, including frequent and prolonged follow-up assessments.
o. Any significant issue raised by the neurologist or neurosurgeon.

Data and Results

The ASPIRO Trial is currently enrolling patients by invitation, but no results are yet available. The first patient has successfully been dosed with autologous DANPCs. To monitor the behavior of DANPCs following transplantation into the putamen, researchers predominantly utilize advanced brain imaging techniques, specifically Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET). The application of the tracer 18F-DOPA is particularly noteworthy, as it facilitates the visualization of dopamine uptake in the transplanted region, thereby providing insights into the survival and functionality of the grafted neurons. In addition, histological analysis, conducted through tissue sampling, enables a direct evaluation of cell morphology and marker expression within the graft site. PET imaging utilizing 18F-DOPA represents the gold standard for evaluating the activity of dopamine neurons by measuring the uptake of this radiolabeled precursor. This technique yields critical insights into the functional capacity of transplanted cells and their ability to synthesize dopamine. In addition, MRIs serve as valuable tools for assessing graft volume, thereby offering insights into cell survival and potential proliferation within the putamen and substantia nigra. Post-mortem analysis of brain tissue samples could enable the comprehensive examination of grafted cells through the application of antibodies targeting specific markers, such as Tyrosine Hydroxylase (TH), a pivotal enzyme in dopamine synthesis. However, post-mortem examination of the patients who undergo DANPC transplants may not be part of the ASPIRO trial. Behavioral assessments and motor function assessments following transplantation provide indirect evidence regarding the efficacy of the graft in alleviating symptoms associated with dopamine deficiency. To that end, employing standardized scoring systems during behavioral evaluations ensures consistency and rigor in data analysis. Serial imaging sessions are essential for monitoring temporal changes in graft function, encompassing both early post-transplant survival and long-term functionality. Accurate identification of the transplanted area within the putamen is critical for the precise interpretation of imaging data.

Discussion

In the late 1980s, researchers began transplanting human fetal dopamine neurons into the brains of individuals with Parkinson’s disease. This development occurred less than ten years after studies in rats demonstrated that embryonic dopamine neurons from a specific developmental stage were viable for transplantation [2]. This approach to cell replacement was based on numerous animal studies showing that restoring dopamine levels in the striatum through embryonic neural grafts could result in significant and enduring functional improvement [23,24]. Moreover, at least two case studies involving male patients with fetal nigral transplants in the striatum have demonstrated that the transplanted dopamine neurons can survive and reestablish connections in the striatum for a minimum of 10 years, even as the disease continues to progress and destroy the patients’ native dopamine neurons [24- 26]. In the late 1980s, open-label trials established a proof of concept demonstrating that the direct transplantation of Human Fetal Ventral Mesencephalic (hfVM) cells into the striatum could ameliorate certain motor deficits in individuals with Parkinson’s disease, enabling them to reduce their medication intake [27-29]. Following President Bill Clinton’s election, which lifted the ban on federally funded research utilizing human fetal tissue in the United States, two NIH-funded double-blind clinical trials examining hfVM grafts were published in the early 2000s [30,31]. Although these trials failed to achieve their primary endpoints and presented unforeseen graft-induced dyskinesias, subsequent postmortem evaluations and patient follow-ups suggested long-term clinical improvements and sustained graft survival [1,26,32-34]. The design and execution deficiencies observed in the NIH-funded clinical trials [28,35] prompt concerns regarding the potential to address these limitations in the forthcoming ASPIRO trial.

The ASPIRO trial will utilize direct intraparenchymal injection of the putamen and deep brain tissue, which emerged from the earlier studies as the only clinically effective approach for neural cell transplantation in PD patients. Intraparenchymal injections are more surgically invasive than other administration routes. Still, they have some advantages: the dose of the vector can be lower when using intraparenchymal injections, and preexisting neutralizing antibodies have little effect on the transduction efficacy of biologics injected directly into the putamen. Prior to the ASPIRO trial, cell suspensions were considered to be the ideal method in which to deliver cells to the central nervous system [28] but that conclusion may be more relevant to transplanting embryonic stem cells and hfVM grafts vs. the DANPCs injected in the ASPIRO trial. Unlike certain deep brain stimulation or ablative procedures, Cell Replacement Therapy (CRT) does not necessitate waking patients from anesthesia during the operation. However, CRT patients should still be anesthetized by experienced neuroanesthesia specialists, and the surgeries should be conducted in high-volume functional neurosurgery centers to adhere to the ‘Getting It Right First Time’ (GIRFT) principles [36-39].

The ASPIRO trial is utilizing the best practices that emerged from prior CRT clinical trials for PD patients: intra-operative MRI-assisted visualization, stereotactic targeting system, framebased and (distortion-corrected) MRI-guided surgery, choice of the putamen as the injection site, minimal needle or cannula diameter, and maximizing the number of cells injected on a single pass so that multiple transcortical penetrations are unnecessary. Neurosurgical delivery devices are generally composed of a straight cannula connected to an external syringe. The plunger of the syringe regulates the rate at which cells are delivered. Nonetheless, these systems pose several challenges that can adversely affect graft delivery. A prominent factor contributing to acute cell death during injection is the influence of mechanical forces on cells [40]; specifically, the extensional force arising from differing velocities causes distortion in cell length [41]. Cells exhibit heightened susceptibility to this mechanical force at the transition zone of the instrumentation, where they move from the larger diameter syringe to the smaller diameter cannula. Reducing the outer diameter of the delivery cannula while maintaining the syringe diameter constant will amplify the magnitude of the extensional force. Furthermore, increasing the length of the cannula prolongs the exposure of cells to this detrimental force [42]. A brief interval of just 20 minutes between the filling of the cannula and the subsequent injection has been associated with notable cell sedimentation and retention [28,43]. This observation holds particular significance in the context of neurosurgical delivery, where delays often occur before the syringe can be affixed to the stereotactic frame, hindering timely cell injection into the target area. To mitigate cell adhesion, it has been proposed to utilize specialized low-friction coatings such as silicone and to adjust factors that may affect chemical and electrostatic interactions. Additionally, the material composition of the delivery instrument has been shown to influence cell retention, with glass exhibiting higher retention rates compared to metal [28,43].

Patient Safety and Ethical Guidelines

When conducting the transplantation of autologous DANPCs into the putamen of patients with Parkinson’s disease, it is imperative to adhere to essential ethical guidelines ensuring patient safety. These include obtaining rigorous informed consent, implementing comprehensive pre-transplant screening protocols, maintaining stringent quality control of the transplanted cells, and conducting vigilant monitoring for any potential complications. Furthermore, outcomes must be transparently reported within the framework of a meticulously designed clinical trial, overseen by an ethics committee. Patients should be made fully aware of the experimental nature of the treatment, including potential risks and benefits, and they must retain the right to withdraw from the study at any stage. Specific considerations for ethical guidelines:

Patient selection

a. Only include patients with advanced Parkinson’s disease who have not responded adequately to standard treatments.
b. Carefully assess patients’ cognitive function and psychological state to ensure they can provide informed consent.
c. Consider potential risks associated with the patient’s overall health and medical history.

Cell preparation and quality control

a. Implement rigorous quality control protocols to ascertain the purity, viability, and differentiation capacity of autologous dopamine neuron progenitor cells.
b. Vigilantly assess for any potential genetic anomalies or tumorigenic risks associated with the cell lines.
c. Systematically document each phase of cell isolation, expansion, and differentiation to maintain comprehensive records.

Informed consent

a. It is essential to provide comprehensive information regarding the experimental aspects of the proposed treatment, including potential risks such as complications like bleeding, infection, and graft-induced dyskinesias, as well as possible benefits.
b. Patients must be informed about the long-term followup requirements and the potential necessity for additional interventions.

Surgical considerations

a. The procedure should be performed by neurosurgeons with significant experience in stereotactic surgery and cell transplantation.
b. To minimize the risk of complications, the use of advanced imaging techniques for precise targeting during the surgical procedure is recommended.

Post-transplant monitoring

a. Patients require close monitoring for any adverse events, which may encompass neurological changes, immune reactions, and indicators of infection.
b. Regular assessments of motor function and other clinical parameters should be conducted to ascertain the efficacy of the treatment.

Data collection and reporting

a. Research findings must be disseminated in peer-reviewed journals, inclusive of both positive and negative outcomes.
b. It is imperative to adhere to ethical guidelines governing data analysis and reporting to uphold scientific rigor and transparency.

Final ethical considerations

a. Potential for adverse effects: Despite the use of autologous cells, there remains a risk of complications or adverse reactions associated with the procedure.
b. Efficacy uncertainty: The long-term effectiveness of transplantation involving autologous dopamine neuron progenitor cells continues to be subject to ongoing research and evaluation.
c. Access to therapeutic interventions: The issue of equitable access to experimental therapies warrants attention, particularly in regions that have limited healthcare resources.

Conflict of Interest

The author was not affiliated with nor had any financial interest in any pharmaceutical or medical device firm while drafting this article.

References

  1. Kefalopoulou Z, Marios P, Paola P, Niccolo M, Kailash B, et al. (2014) Long-term clinical outcome of fetal cell transplantation for Parkinson disease: Two case reports. JAMA Neurol 71(1): 83-87.
  2. Freed CR, Zhou W, Breeze RE (2011) Dopamine cell transplantation for Parkinson's disease: The importance of controlled clinical trials. Neurotherapeutics 8(4): 549-561.
  3. Schweitzer JS, Bin S, Todd MH, Tae YP, Nayeon L, et al. (2020) Personalized iPSC-derived dopamine progenitor cells for Parkinson's disease. N Engl J Med 382(20): 1926-1932.
  4. Clark BJ, Lelos MJ, Loring JF (2024) Advancing Parkinson’s disease treatment: Cell replacement therapy with neurons derived from pluripotent stem cells. Stem Cells 42(9): 781-790.
  5. (2024) Phase 1/2a Dose escalation study of ANPD001 in sporadic Parkinson disease.
  6. Mehrnoosh G, Sunil M (2024) Neuroanatomy, Putamen. StatPearls Publishing LLC, USA.
  7. Haber SN (2016) Corticostriatal circuitry. Dialogues Clin Neurosci 18(1): 7-21.
  8. Viñas GN, Yan JW (2017) The role of the putamen in language: A meta-analytic connectivity modeling study. Brain Struct Funct 222(9): 3991-4004.
  9. Kinoshita K, Takehito K, Yoshie H, Kojiro M (2022) Putamen atrophy is a possible clinical evaluation index for Parkinson's disease using human brain magnetic resonance imaging. J Imaging 8(11): 299.
  10. Jue W, Jia RZ, Yu FZ, Tao W (2018) Consistent decreased activity in the putamen in Parkinson's disease: A meta-analysis and an independent validation of resting-state fMRI. GigaScience 7(6): giy071.
  11. Aiping L, Sue JL, Taomian M, Xun C, Piu C, et al. (2018) Decreased subregional specificity of the putamen in Parkinson's disease revealed by dynamic connectivity-derived parcellation. Neuroimage Clinical 20: 1163-1175.
  12. Pasquini J, Rory D, Louise W, Morten GS, Lynn R, et al. (2019) Clinical implications of early caudate dysfunction in Parkinson’s disease. J Neurol Neurosurg Psychiatry 90(10): 1098-1104.
  13. Mavridis IN, Pyrgelis ES (2022) Nucleus accumbens atrophy in Parkinson's disease (Mavridis' atrophy): 10 years later. Am J Neurodegener Dis 11(2): 17-21.
  14. Mavridis IN (2015) Is nucleus accumbens atrophy correlated with cognitive symptoms of Parkinson's disease? Brain 138(1): e319.
  15. Akram AA, Siew CN, Nadine N, Suleiman YA, Syahril A (2020) Induced pluripotent stem cells: Reprogramming platforms and applications in cell replacement therapy. Biores Open Access 9(1): 121-136.
  16. Bin S, Young C, Sanghyeok K, Jeha J, Nayeon L (2020) Human autologous iPSC-derived dopaminergic progenitors restore motor function in Parkinson's disease models. J Clin Invest 130(2): 904-920.
  17. Granholm AC, Reyland M, Albeck D, Sanders L, Gerhardt G, et al. (2000) Glial cell line-derived neurotrophic factor is essential for postnatal survival of midbrain dopamine neurons. J Neurosci 20(9): 3182-3190.
  18. Lindholm P, Saarma M (2022) Cerebral dopamine neurotrophic factor protects and repairs dopamine neurons by novel mechanism. Molecular Psychiatry 27(3): 1310-1321.
  19. Bathina S, Das UN (2015) Brain-derived neurotrophic factor and its clinical implications. Arch Med Sci 11(6): 1164-1178.
  20. Benjamin MH, David JM, Cayla AT, Nathaniel AE, Howard F, et al. (2022) Optimizing maturity and dose of iPSC-derived dopamine progenitor cell therapy for Parkinson’s disease. Regenerative Medicine 7(1): 24.
  21. Stoker TB (2018) Stem cell treatments for Parkinson’s Disease. Parkinson’s Disease: Pathogenesis and Clinical Aspects.
  22. Takahashi J (2020) iPS cell-based therapy for Parkinson's disease: A Kyoto trial. Regen Ther 13: 18-22.
  23. Björklund A, Lindvall O (2000) Cell replacement therapies for central nervous system disorders. Nat Neurosci 3(6): 537-544.
  24. Lindvall O, McKay R (2003) Brain repair by cell replacement and regeneration. Proc Natl Acad Sci USA 100(13): 7430-7431.
  25. Kordower JH, Freeman TB, Snow BJ, Vingerhoets FJ, Mufson EJ, et al. (1995) Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson's disease. N Engl J Med 332(17): 1118-1124.
  26. Piccini P, Brooks DJ, Björklund A, Gunn RN, Grasby PM, et al. (1999) Dopamine release from nigral transplants visualized in vivo in a Parkinson's patient. Nat Neurosci 2(12): 1137-1140.
  27. Parmar M, Torper O, Drouin OJ (2019) Cell-based therapy for Parkinson's disease: A journey through decades toward the light side of the force. Eur J Neurosci 49(4): 463-471.
  28. Saumya M, Harith A, Harry B, Suneil KK, Asuka M, et al. (2024) Dopaminergic cell replacement for Parkinson's disease: Addressing the intracranial delivery hurdle. J Parkinsons Dis 14(3): 415-435.
  29. Barker RA, Drouin OJ, Parmar M (2015) Cell-based therapies for Parkinson disease-past insights and future potential. Nat Rev Neurol 11(9): 492-503.
  30. Freed CR, Greene PE, Breeze RE, Tsai WY, Dillon S, et al. (2001) Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N Engl J Med 344(10): 710-719.
  31. Warren OC, Christopher GG, Jeffrey HK, Stoessl AJ, Vesna S, et al. (2003) A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease. Ann Neurol 54(3): 403-414.
  32. Yilong M, Chengke T, Thomas C, Paul G, Robert B, et al. (2010) Dopamine cell implantation in Parkinson's disease: Long-term clinical and (18)F-FDOPA PET outcomes. J Nucl Med 51(1): 7-15.
  33. Marios P, Wolfgang HO, Kit W, Niall PQ, Oliver P, et al. (2011) Graft-induced dyskinesias in Parkinson's disease: High striatal serotonin/dopamine transporter ratio. Mov Disord 26(11): 1997-2003.
  34. Jia YL, Wen L (2021) Postmortem studies of fetal grafts in Parkinson's disease: what lessons have we learned? Front Cell Dev Biol 9: 666675.
  35. Roger AB, Jessica B, Sarah LM, Anders B (2013) Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson's disease. Lancet Neurol 12(1): 84-91.
  36. Gatfield SA, Atkinson KV, Fountain D, Machin JT, Navaratnam AV, et al. (2023) Getting it right first time: National survey of surgical site infection 2019. Ann R Coll Surg Engl 105(6): 513-522.
  37. Mukerji N (2022) Variation, expectations, consent, litigation and GIRFT. Br J Neurosurg 36(1): 1-2.
  38. Oluwatobi OO, Nicola FF, Francis B, Oliver MS, Timothy WB, et al. (2020) Multiple injections for low back pain: What's the future? Eur Spine J 29(3): 564-578.
  39. Adam JW, David AC, Peter JH, Ryan KM, Nick P (2022) Mortality as an indicator of quality of neurosurgical care in England: A retrospective cohort study. BMJ Open 12(11): e067409.
  40. Mahetab HA, Felicity RAJR, Kevin MS, Michel M, Lisa JW (2017) Translational considerations in injectable cell-based therapeutics for neurological applications: Concepts, progress and challenges. NPJ Regen Med 2: 23.
  41. Brian AA, Widya M, James S, Kyle JL, Sarah CH (2012) Improving viability of stem cells during syringe needle flow through the design of hydrogel cell carriers. Tissue Eng Part A 18(7-8): 806-815.
  42. Potts MB, Silvestrini MT, Lim DA (2013) Devices for cell transplantation into the central nervous system: Design considerations and emerging technologies. Surg Neurol Int 4(Suppl 1): S22-S30.
  43. Eduardo MT, Matthieu T, Stephen BD (2015) Translation of cell therapies to the clinic: Characteristics of cell suspensions in large-diameter injection cannulae. Cell Transplant 24(4): 737-749.

© 2024 Michael AS Guth. This is an open access article distributed under the terms of the Creative Commons Attribution License , which permits unrestricted use, distribution, and build upon your work non-commercially.