What drugs are in development for Parkinson Disease?

12 March 2025
Overview of Parkinson's Disease

Definition and Symptoms
Parkinson’s disease (PD) is a progressive, neurodegenerative disorder primarily affecting the dopaminergic neurons located in the substantia nigra, a brain region crucial for regulating movement. Clinically, PD is characterized by quintessential motor symptoms—bradykinesia (slowness of movement), resting tremor, rigidity, and postural instability—that often appear only after a substantial loss of dopamine-producing neurons has occurred. In addition to these motor disturbances, patients experience a range of nonmotor symptoms such as depression, cognitive impairment, sleep disturbances, autonomic dysfunction, pain, and, in some cases, psychosis. In fact, impaired cognitive and psychiatric function has been increasingly recognized as a critical element of the disease burden. These symptoms result from a multifaceted pathogenic process that includes abnormal protein aggregation (for instance, α‑synuclein accumulation leading to Lewy bodies), mitochondrial dysfunction, oxidative stress, inflammation, and alterations in various neurotransmitter systems.
Overall, Parkinson’s disease is much more than simply a movement disorder; it affects multiple physiological systems. The heterogeneity of symptoms—both motor and nonmotor—indicates that PD has a complex pathophysiology, including genetic and environmental influences. Advances in molecular biology have shed light on the mechanisms underlying neuronal death, with recent research focusing on factors such as misfolded proteins, neuroinflammation, and impaired autophagy. A thorough understanding of PD’s diverse clinical features is essential to shape the identification of new drug targets and treatments.

Current Treatment Options
At present, the mainstay of treatment for PD is symptomatic and is based on dopamine replacement strategies. Levodopa remains the gold standard—it is a precursor that can cross the blood–brain barrier and is decarboxylated to dopamine within the brain. However, due to its rapid peripheral metabolism, levodopa is typically administered in combination with decarboxylase inhibitors such as carbidopa (as seen in the commonly prescribed formulation Sinemet) to enhance its bioavailability and reduce side effects. Dopamine agonists such as ropinirole, pramipexole, and rotigotine provide an alternative approach by directly stimulating dopamine receptor subtypes, thereby partially compensating for the dopaminergic deficit. Moreover, monoamine oxidase-B (MAO-B) inhibitors like selegiline and rasagiline slow the enzymatic breakdown of dopamine, increasing its availability. Catechol-O-methyltransferase inhibitors (e.g., tolcapone and entacapone) are further used to prolong the duration of levodopa’s effect and minimize “wearing off” phenomena.
Despite these options, none of the available medications have been able to halt disease progression fully. Their efficacy may wane over time as symptomatic treatments do not target the underlying mechanisms responsible for neurodegeneration. This limitation in current treatments has spurred an active search for novel drugs that offer true disease modification alongside improved symptomatic control.

Drug Development Pipeline

Phases of Drug Development
The drug development process for Parkinson’s disease follows the general stages of clinical research – from preclinical investigations through multiple phases of clinical trials. Preclinical studies typically involve in vitro models and animal models (e.g., MPTP toxin‐induced models in primates and rodents) to assess safety, efficacy, and the mechanistic basis of candidate drugs. Promising agents then progress into early human trials (Phase 1), which focus on establishing safety and tolerability in small cohorts of healthy volunteers or patients. Phase 2 studies are aimed at getting preliminary efficacy data, often using surrogate endpoints such as improvements in motor scores or patient biomarker changes. These trials help fine-tune dosing regimens and gather further safety information. Later stage Phase 3 studies move to larger patient populations and are designed to definitively demonstrate clinical efficacy, though they are particularly challenging for PD because of the slow progression of the disease and the need for sensitive endpoints. In some instances, especially with advanced symptomatic treatments or novel delivery approaches, Phase 4 post-marketing surveillance studies also contribute vital data regarding long-term safety and effectiveness.
Notably, the complex heterogeneity in PD means that clinical trial designs must account for various disease stages and symptom clusters. The identification of early, intermediate, and late-stage patient cohorts has become critical. Moreover, the measurement of treatment effect in PD – particularly for disease-modifying treatments – often requires long trial durations, large sample sizes, and adoption of novel digital or imaging biomarkers to capture subtle changes over time.

Key Players and Companies
The global drug development pipeline for Parkinson’s disease is sustained by a diverse group of key players, including major pharmaceutical companies, biotechnology firms, academic institutions, and research foundations. Companies like Merck & Co., Eisai, ACADIA Pharmaceuticals, Bristol Myers Squibb, and Novartis have played central roles in both advancing symptomatic treatments and exploring disease-modifying agents. In addition, smaller biotech firms (such as Dragonfly Therapeutics and Exelixis) are increasingly contributing to the pipeline by focusing on niche targets, innovative formulations, and repurposing strategies.
Many strategic collaborations and licensing deals underscore the collaborative nature of PD drug development. For instance, partnerships between companies like Merck and Dragonfly Therapeutics, and collaborations between bio‐pharmaceutical companies and academic research groups, are common. These agreements help to leverage specialized expertise, share resources, and integrate new technologies (e.g., digital biomarkers, gene therapy advancements) into clinical trial design. Funding is also split among industry, US federal agencies, and disease‐targeted foundations, which provide critical capital needed to sustain long-term clinical trials in PD.
Furthermore, international strategic partnerships—such as those involving Parkinson’s Foundation and Parkinson’s UK—aim to accelerate drug development by combining funding, clinical trial design expertise, and patient registry data to identify those patients most likely to progress rapidly and, therefore, be optimal candidates for trials of novel agents.

Emerging Drugs for Parkinson's Disease

Promising Drug Candidates
A wide variety of novel therapeutic candidates are currently in development for Parkinson’s disease. The emerging drugs can be grouped into several categories based on their primary mechanism of action and therapeutic aim. Below, we present some of the most promising candidates based on recent synapse‐indexed research:

• Dopaminergic strategies remain at the forefront, with novel dopamine agonists and compounds designed to optimize dopamine delivery or receptor stimulation. Agents such as Tavapadon and P2B001 are in development with the aim of providing more consistent dopaminergic signaling while reducing motor fluctuations and dyskinesias. These drugs are designed to have improved pharmacokinetic profiles and may also address nonmotor symptoms through modulation of additional receptor pathways.

• MAO-B inhibitors continue to evolve. Although selegiline and rasagiline have provided symptomatic relief for decades, next-generation reversible and highly selective MAO-B inhibitors are in the pipeline. These agents aim to reduce oxidative stress and improve mitochondrial function, while also serving as neuroprotectants to possibly slow disease progression.

• Non-dopaminergic therapies target alternative signaling pathways that contribute to neurodegeneration. Istradefylline, an adenosine A2A receptor antagonist, has emerged as a promising drug candidate for improving “off” time in PD patients. Digital and clinical trial data have supported its efficacy in reducing motor fluctuations when used adjunctively with levodopa. Additionally, novel compounds aimed at antagonizing serotonin 2A/2C receptors, such as pimavanserin, have gained approval for treating psychosis in Parkinson’s disease, and further studies are evaluating their neuroprotective potential.

• Immunotherapies are a novel frontier in PD drug development. Given that misfolded α‑synuclein aggregation plays a pivotal role in PD, passive and active immunotherapeutic strategies are being pursued. Monoclonal antibodies designed to clear α‑synuclein aggregates (for example, prasinezumab) have already reached advanced-phase trials, though some have encountered setbacks in primary endpoints. Efforts continue with updated formulations and improved target engagement strategies, as reported in recent synapse‐indexed papers that highlight the challenges and opportunities of targeting α‑synuclein directly.

• LRRK2 inhibitors represent another promising class. Mutations in the LRRK2 gene are one of the most common genetic causes of PD. Inhibitors targeting the kinase activity of LRRK2 have shown promise in preclinical models; however, clinical translation has been challenging due to concerns of lung toxicity in primate studies. Novel delivery systems and antisense oligonucleotide approaches are under investigation to reduce these side effects while effectively modulating LRRK2 function.

• Neurotrophic factors and gene therapy are also being actively explored. Glial cell line–derived neurotrophic factor (GDNF) and related GDNF family ligands have long been considered potentially disease-modifying treatments due to their ability to support dopaminergic neuron survival. Despite mixed results in clinical trials to date, improved delivery methods (for example, via intracerebral infusion or viral vectors) and modified molecules are in development to overcome earlier limitations. In parallel, gene therapy approaches that restore key dopaminergic genes through viral vector delivery are under clinical evaluation in early-stage trials.

• Cell-based therapies, including transplantation of dopaminergic neurons derived from stem cells, are emerging as a promising approach for replacing lost cells and partially restoring motor function. While these therapies are still in the clinical testing phase and face significant regulatory and technical challenges, early results have been encouraging.

• Lastly, repurposing strategies are significant pathways for drug development in PD. Drugs approved for other indications (such as certain antihypertensive agents, anti-inflammatory drugs, and even some natural compounds) are being evaluated for their potential to slow disease progression. For example, molecules targeting mitochondrial function and oxidative stress—such as coenzyme Q10 analogs and novel antioxidants—are being reexamined from a PD perspective through innovative approaches like Mendelian randomization. Additionally, multiple studies have considered the repurposing of anti-inflammatory agents and even nonpharmacological compounds (e.g., certain nutraceuticals) as adjuncts to current therapy.

Mechanisms of Action
The emerging drug candidates for PD work via diverse mechanisms, which can broadly be divided into symptomatic improvement, neuroprotection, and disease modification. For example:
• Dopamine-centric strategies directly increase dopamine levels (via levodopa analogs, dopamine agonists) or prevent its breakdown (via MAO-B, COMT inhibitors), thus alleviating the motor symptoms directly. Their mechanisms are well understood and supported by decades of clinical use; however, their benefit is largely symptomatic rather than curative.

• Non-dopaminergic agents such as adenosine A2A receptor antagonists (istradefylline) work by modulating other neurotransmitter systems which normally interact with the dopaminergic system. By disinhibiting dopaminergic activity in the basal ganglia, these agents help to extend the therapeutic effects of dopamine replacement, with a mechanism that is both complementary and synergistic to traditional therapies.

• Immunotherapy with monoclonal antibodies against α‑synuclein seeks to interrupt the process of protein aggregation and propagation across neurons. This approach targets the very mechanism that underlies the evolution of PD pathology, offering the potential for true disease modification if the clearance of misfolded protein results in preserved neural function.

• LRRK2 inhibitors aim to down-regulate the pathogenic kinase activity associated with mutant forms of LRRK2, which can impair cellular trafficking, mitochondrial function, and autophagy. These inhibitors are designed to normalize LRRK2 signaling and thereby reduce neurodegenerative cascades.

• Neurotrophic factor therapies such as GDNF and its analogs enhance the survival and growth of dopaminergic neurons by activating receptors that signal through pathways promoting cell survival, anti-apoptosis, and neuroplasticity. Gene therapy strategies aim to deliver these factors directly to the affected brain regions, thereby offering a targeted, potentially long-term solution.

• Cell therapies and stem-cell–derived neuron transplantations use the body’s own regenerative capacities to replace lost neurons, potentially restoring dopaminergic circuitry. Their mechanisms depend on the integration and functional maturation of transplanted cells, which in turn require precise control of differentiation and survival signals in vivo.

• Repurposed drugs that target mitochondrial dysfunction, oxidative stress, and neuroinflammation work by interfering with the cellular pathways that cause progressive neuronal damage. For instance, compounds that improve mitochondrial bioenergetics or scavenge reactive oxygen species may slow neurodegeneration; similarly, anti-inflammatory agents can mitigate microglial overactivation which is thought to exacerbate neuron loss.

These various mechanisms—sometimes overlapping—demonstrate a multi-targeted strategy for PD drug development. Importantly, while symptomatic agents improve quality of life in the short term, the ultimate goal is to develop therapies that modify the disease course by preserving neuronal health and function.

Challenges and Future Directions

Development Challenges
Despite the richness of potential targets and drug classes, PD drug development faces several formidable challenges. One major difficulty arises from the slow progression and heterogeneous clinical presentation of Parkinson’s disease, which necessitates long-term trials with sensitive endpoints to detect disease-modifying effects. In many cases, patients may not show measurable progression over the duration of a typical clinical trial, diluting the statistical power of the study.
Another challenge is the translation from preclinical animal models to human clinical trials. Many candidate drugs show promise in toxin-induced models such as the MPTP model, yet these models do not entirely capture the complex pathophysiology of human PD—particularly the interplay of genetic, environmental, and age-related factors. Consequently, a number of compounds that were promising in preclinical studies have failed in human trials.
Furthermore, targeting proteins like α‑synuclein or LRRK2 presents unique pharmacological challenges. For instance, while immunotherapies that target α‑synuclein are conceptually appealing, their clinical efficacy has been inconsistent, and some trials have failed to reach their primary endpoints despite demonstrating target engagement. LRRK2 inhibitors, meanwhile, have been hampered by off-target toxicity issues (e.g., lung toxicity observed in primate studies) that demand the development of more sophisticated delivery methods or alternative molecular strategies.
Regulatory hurdles and funding constraints further complicate the field. With PD being a slowly progressive disease, the costs and time required to conduct sufficiently powered Phase 3 trials are high, and this risk profile often makes large pharmaceutical companies hesitant to invest in novel disease-modifying therapies without substantial preclinical evidence of benefit.
In addition, the need for reliable biomarkers—both for diagnostic purposes and for tracking disease progression—is acute. Traditional clinical endpoints such as the Unified Parkinson’s Disease Rating Scale (UPDRS) are subjective and may not capture subtle yet clinically meaningful changes over time, hindering the assessment of treatment efficacy.

Future Prospects and Innovations
Looking ahead, several innovative approaches and emerging technologies promise to overcome many of the current challenges and accelerate the development of effective therapies for PD. One promising direction is the integration of digital health technologies and digital biomarkers into clinical trials. These tools can capture continuous, objective data on motor and nonmotor symptoms in real-world settings, enabling more sensitive detection of treatment effects and allowing for shorter, more efficient trials.
Advances in genomics and personalized medicine are also expected to play an important role. With the advent of large-scale genetic studies, novel drug targets are being validated through approaches such as Mendelian randomization, which can help to prioritize candidates with strong genetic support—an approach that has already identified several promising targets in PD research.
In parallel, next-generation gene therapy and antisense oligonucleotide strategies are being refined to precisely modulate the expression of risk genes (such as LRRK2) and neuroprotective factors. For example, improved viral vector systems and localized delivery approaches are currently under development to safely and efficiently administer neurotrophic factors like GDNF directly into the brain.
Cell-based therapies also hold significant promise. With advances in stem cell biology and induced pluripotent stem cell technology, researchers are now better able to generate functional dopaminergic neurons for transplantation. Early-phase clinical trials have shown encouraging results in terms of cell survival, integration, and functional improvement, suggesting that these therapies may eventually offer a genuine restorative treatment for PD.
Moreover, multi-target and combination therapies are likely to take center stage in future PD treatment. Given that PD pathogenesis is multifactorial—involving dopaminergic deficits, protein aggregation, oxidative stress, and neuroinflammation—a combination approach that simultaneously addresses several pathogenic pathways may be necessary to achieve meaningful disease modification. Innovative clinical trial designs that allow drugs with complementary mechanisms to be tested together are already being conceptualized and may significantly alter the therapeutic landscape.
Finally, the future of PD drug development will also be shaped by improved patient stratification. Using advanced imaging, genetic profiling, and even machine learning tools to predict disease progression and treatment response, researchers can design more targeted clinical trials. These approaches would allow for the enrollment of patients who are most likely to benefit from a given therapy, thereby increasing the chances of detecting a clinical benefit.
In summary, although challenges abound—from the inadequacy of current animal models to the need for reliable biomarkers and the inherent difficulties of long-duration trials—the future remains promising. By integrating technological innovations, multi-targeted approaches, and precision medicine strategies, the next generation of PD therapies could offer improved symptomatic relief and, crucially, true disease modification.

Conclusion
In conclusion, the current landscape of drug development for Parkinson’s disease is both diverse and rapidly evolving. Our overview has shown that while PD remains a multifactorial disease with both motor and nonmotor manifestations, the limitations of existing symptomatic treatments have spurred an intense global effort to identify and develop drugs that not only mitigate symptoms but also modify disease progression. The drug development pipeline includes a range of phases—from early preclinical investigations to large-scale, long-duration Phase 3 trials—and is driven by key players including global pharmaceutical companies, emerging biotech firms, academic institutions, and strategic partnerships that collectively push forward innovation in PD therapeutics.

Emerging drug candidates span multiple classes and mechanisms of action. Promising candidates include novel dopaminergic agents (e.g., Tavapadon, P2B001), advanced MAO-B inhibitors, non-dopaminergic agents such as istradefylline and pimavanserin, immunotherapies targeting α‑synuclein, LRRK2 inhibitors, neurotrophic factors (GDNF and its analogs) and both cell- and gene-based therapies. Each of these targets a distinct aspect of PD pathogenesis—from symptomatic relief to neuroprotection and disease modification—with mechanisms focusing on dopamine replacement, protein clearance, modulation of inflammatory pathways, and neuronal survival.

Despite significant advances, challenges remain. The slow, heterogeneous progression of PD, limitations in current preclinical models, difficulties in translation from animal studies to human trials, regulatory hurdles, funding constraints, and the lack of reliable progression biomarkers all combine to create a complex drug development environment. However, the integration of digital health tools, personalized medicine approaches, combination therapies, and improved patient stratification tools offer a hopeful path forward.

Overall, the future of PD drug development depends on a continued commitment from diverse stakeholders—researchers, clinicians, industry players, and patient advocacy groups—to innovate and collaborate. With recent innovations in molecular biology, advanced clinical trial design, and digital monitoring, there is renewed optimism that effective, disease–modifying therapies will be developed over the coming decades. The coming era may finally shift Parkinson’s disease treatment from mere symptomatic management toward genuinely transformative therapies that slow, halt, or even reverse disease progression.

This comprehensive approach—examining basic disease mechanisms to sophisticated digital integration—sets the stage for a future with a richer and more effective drug arsenal for Parkinson's disease. Each novel candidate and innovative strategy represents not only a potential breakthrough for improving patient quality of life but also a significant step in understanding and ultimately conquering the complex neurodegenerative process of PD.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.