1 Executive Summary
2 Market Analysis
3 Marketing Strategy
4 Product Development Plan
5 Skills, Capabilities and Stakeholders
6 Long-term Impacts
Reference
Approximately 10 million individuals worldwide are living with Parkinson’s disease —a chronic, progressive neurological disorder characterized by the gradual loss of dopamine-producing neurons in the brain (Parkinson’s Foundation). This dopamine deficiency primarily leads to motor symptoms such as tremors, rigidity, and bradykinesia, as well as non-motor complications like depression, anxiety, and sleep disturbances. These symptoms significantly impair patient’s ability to perform daily tasks and engage in social interactions, leading to a substantial decline in quality of life. Despite existing treatments, available medications often lose effectiveness over time or produce adverse effects. Therefore, ongoing research into more effective and safer pharmacological therapies remains a pressing need.
Current treatments primarily rely on dopamine replacement therapies, including dopamine precursors as L-DOPA, that could cross blood-brain barrier and changed into dopamine. However, this therapy brings some side effects that negatively impacts the intestinal function of most medicine takers, potentially leading to the deceleration of intestinal motility, causing constipation or abdominal bloating in patients. Also, slowed gastrointestinal motility could hinder the absorption of L-DOPA, interference with drug efficacy. Therefore, developing alternative therapies or adjuvant strategies that can effectively supplement dopamine while minimizing gastrointestinal side effects has become an important direction in current Parkinson's disease treatment research.
Existing therapy options have side effects that prevent patients from receiving effective treatment. Also, weakened gut function leads to a decline in quality of life of patients that receive this medical approach, hindering their gastrointestinal tract from working effectively.
Our team aims to minimize the side effects of current therapy. By colonizing probiotics in the intestines, the precursor of dopamine could be generated in the gut and transferred to the brain in a stable manner. After the dopamine precursor reaches the brain, it can be transformed into dopamine, which complements the lack of dopamine in patient’s brains. In addition, the additional supply of probiotics could adjust the gut flora structure, enhance intestinal function, and reduce the occurrence of gastrointestinal problems, thereby exerting a positive effect (Chandrasekaran et al.).
We are developing a probiotic-based therapy for Parkinson’s disease. By engineering probiotics that can stably colonize the intestines, we aim to create a living drug delivery system that continuously produces dopamine precursors in the gut. These precursors can be transported to the brain, where they are converted into dopamine to relieve dopamine deficiency in patients. The final product will be delivered in the form of an oral capsule, making the treatment non-invasive and easy to use.
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders globally, currently affecting over 10 million people and growing in prevalence due to global population aging. The economic burden of PD is substantial, with costs associated with treatment, care, and productivity loss exceeding USD 50 billion annually in the United States alone.
The global market for Parkinson’s disease therapeutics was valued at approximately USD 5.7 billion in 2023, and is projected to reach USD 8.9 billion by 2030, growing at a compound annual growth rate (CAGR) of 6.5%. Among all available pharmacological treatments, L-DOPA (levodopa) remains the most widely used and clinically effective drug for managing motor symptoms of PD, often considered the “gold standard” of treatment. It is prescribed to over 60% of all diagnosed PD patients, representing a highly significant therapeutic segment.
However, conventional L-DOPA production relies heavily on chemical synthesis, which is resource-intensive, expensive, and environmentally harmful due to the use of hazardous reagents and complex purification steps. In response to growing concerns around sustainability and scalability in pharmaceutical manufacturing, there is rising interest in biotechnological alternatives, particularly biosynthetic production using engineered microorganisms. This emerging subsector aligns with broader trends in green chemistry, personalized medicine, and decentralized drug production.
Recent studies and market analyses estimate that the market potential for biosynthetically produced L-DOPA could reach USD 1.2 billion by 2030, particularly as healthcare systems seek lower-cost and cleaner production methods for essential medicines. Our project aims to enter this niche by developing an engineered microbial system capable of producing L-DOPA in a more efficient, cost-effective, and sustainable manner. In the longer term, our system may be adaptable for on-demand localized production, offering major logistical and economic advantages, especially in low-resource settings or regions with fragile supply chains.
To estimate our Serviceable Obtainable Market (SOM), we take a conservative approach: assuming our solution initially targets just 1% of early- to mid-stage PD patients in high-income countries (e.g., North America, Europe, East Asia), and each patient requires L-DOPA equivalent to USD 1,500/year, this represents an obtainable market of approximately USD 15-20 million annually. As clinical validation, regulatory approval, and manufacturing scale advance, we project significant expansion into broader patient populations and geographic markets.
By addressing a critical unmet need at the intersection of synthetic biology, neurodegenerative medicine, and sustainable manufacturing, our project is well-positioned to capture value in a rapidly evolving therapeutic landscape.
Figure 1 The Summary of Policies
In order to promote the development of pharmaceuticals, China has set up many policies on pharmaceuticals to encourage pharmaceutical enterprises to increase their investment in research and development, and to support the research and development and industrialisation of innovative medicines, which provides a favourable policy environment for enterprises to innovate. We have compiled the relevant policies in the table above.
First, the biomedical economy operates on a substantial scale. The 2024 Government Work Report identified biomedicine as a key strategic sector, and according to the 14th Five-Year Plan Biological Economy Development Plan, the total scale of China’s bio-economy is projected to reach 22 trillion by 2025, with biomedicine accounting for over 40% of this total.
Second, per capita healthcare spending continues to rise. Growth in medical expenditures has outpaced GDP growth, reaching 6,200 per person in 2023. This figure is expected to exceed 8,000 by 2025, driven by increasing health awareness and expanded consumer purchasing power—factors that are likely to further stimulate pharmaceutical industry development.
Third, innovative drugs are entering a phase of accelerated returns. China’s outbound licensing deals for innovative drugs surpassed $40 billion in 2023 and exceeded $50 billion in 2024, reflecting growing global recognition and commercial maturity.
In summary, the expanding biomedical economy, coupled with rising personal health investment, provides strong financial underpinnings for sustained growth and innovation in the biomedicine sector.
By the end of 2023, China’s population aged 60 and above had reached 297 million, accounting for 21.1% of the total population (Xinhua News Agency, 2024.11). Population aging has become a long-term fundamental national condition, providing sustained and stable growth potential for the pharmaceutical market.
At the same time, the continuous rise in per capita healthcare spending reflects the public’s increasing attention to health and growing medical demands. With improved consumption capacity, people are not only more willing but also better able to allocate resources toward healthcare, thereby further expanding the pharmaceutical market.
The technical support for drug discovery and development is mainly reflected in the following aspects:
1. The development of artificial intelligence. The traditional drug discovery model of high investment, long cycle and low output has seriously constrained the output of new drugs. In recent years, data from market analysis companies show that AI will save more than $70 billion in the field of drug discovery by 2028 (Science and Technology Daily, 2025.1).AI shows its role in all aspects of drug discovery by virtue of its superior data processing ability, pattern recognition ability, and generalised prediction ability.
2. Application of big data. Firstly, at the early stage of research, searching and integrating biomedical data and combining it with AI can quickly predict data such as drug activity and shorten the time of research and development. Secondly, in pharmaceutical production, big data can record key parameters in real time, such as temperature, humidity, etc., so as to realise the precise regulation of key data in the production process and improve the quality and efficiency of production. Finally, in the later stage of drug clinical trials, big data can match eligible patients by analysing health records, etc., and at the same time be able to detect data and analyse results in real time.
Overall, the development of artificial intelligence and big data provides powerful technical support for biopharmaceutical and medical development.
Figure 2 Porter's Five Forces
Porter’s Five Forces helps to analyze the competitive environment of an industry and understand the profitability and competitive pressure of a product or enterprise in the market. Through in-depth analysis of these five dimensions, we are able to gain insight into the profit potential of the target market, identify key risks, and formulate business strategies accordingly to enhance the competitive barriers and commercial feasibility of our products.
In the Parkinson’s disease treatment market, suppliers have moderate bargaining power overall. Although raw materials such as active pharmaceutical ingredients (API), key chemical intermediates, fermentation strains, special enzymes and drug delivery devices rely on specialized suppliers, there are multiple alternative sources in the market, allowing us to effectively suppress price pressure through long-term contracts.
In addition, the cost of switching suppliers is relatively controllable. In practice, many pharmaceutical companies choose to cooperate with CDMO (Contract Development and Manufacturing Organization) to produce and avoid the risks of relying on a single manufacturer by diversifying the supply chain.
In the B2B2C model of our Parkinson’s probiotic therapy, buyers consist of hospitals and pharmacies at the B2B level, and patients at the B2C end.
B2B: In the B2B part of our model, institutional buyers—such as hospitals, doctors, pharmacies, and insurers—hold significant bargaining power. Hospitals control procurement channels and treatment formularies, meaning only therapies backed by strong clinical data and regulatory approval can be adopted into mainstream care. Doctors serve as key decision-makers, and their willingness to prescribe a new treatment is directly tied to its demonstrated efficacy and safety. Furthermore, pharmacies tend to favor lower-cost alternatives and may resist high pricing unless the product demonstrates clear cost-benefit advantages over traditional therapies.
B2C: On the consumer side, they are influenced heavily by physician’s recommendations but still play a role in treatment acceptance and adherence. Their power lies in brand perception, ease of use, and trust in the therapy’s outcomes. For an oral probiotic-based therapy, patient-friendly features such as non-invasiveness, minimal side effects, and affordability can drive strong consumer preference. While their individual bargaining power may be low, collective consumer feedback (via social media, patient advocacy groups, etc.) can shape the market landscape over time and influence institutional buyer’s decisions.
Although the Parkinson's disease treatment market is attractive due to its huge demand, new entrants face many high barriers, and the overall threat is low.
First, the R&D cost and approval cycle are extremely high: it takes an average of 10-15 years and billions of dollars, about 2.0-2.6 billion US dollars to develop a new therapy, and the failure rate of clinical trials is as high as more than 90%, and the final approval rate of new drugs is less than 10%, among which the success rate of neurological drugs is even lower, only about 8-15%. Secondly, the regulatory barriers are strict. Regulatory agencies such as the FDA and EMA are more cautious about CNS therapies, such as for Parkinson’s disease. The approval process is long and complicated, often resulting in rejections of early candidate drugs.
In terms of market channels, mature giants have a strong sales network, distribution system and medical insurance reimbursement system, and it is difficult for new entrants to quickly establish coverage.
Finally, it takes tens of millions of dollars to build a production line that meets GMP standards, and a single Phase III clinical trial can cost tens of millions of dollars (admin). Therefore, it is difficult for small and medium-sized or start-ups to participate in this market on a large scale in the short term.
In the Parkinson’s disease treatment market, the threat of substitutes is generally considered low to moderate. The existing mainstream treatments are still centered on oral drugs. Although they have certain side effects, they are widely adopted due to their stable efficacy and low cost. On the other hand, invasive treatment techniques such as deep brain stimulation (DBS) have significant effects in some advanced patients, but the surgery is risky and expensive.
In addition, an increasing amount of patients are trying auxiliary and alternative therapies, including complementary therapies such as exercise, physical therapy, acupuncture, and nutritional intervention. However, at present, these methods are mostly aimed at improving the quality of life, lack strong clinical evidence, and cannot replace drugs or surgery as the main treatment for some patients. Meanwhile, cutting-edge experimental therapies such as metabolic copper and L-line protein delivery have achieved certain results in animal models but are still in the preclinical or early clinical stages and are difficult to replace existing solutions.
According to the industry report, the leading companies in the Parkinson’s disease treatment market include: Cerevel Therapeutics, Novartis AG, Teva Pharmaceutical Industries Ltd., Merck & Co., Inc., GlaxoSmithKline Plc. (GSK), AbbVie, Inc., H. Lundbeck A/S, Amneal Pharmaceuticals LLC, Supernus Pharmaceuticals, Inc. These companies own the dominant market share within the Parkinson’s disease treatment market. The global Parkinson’s disease treatment market has reached approximately US $5.6 billion in 2024 and is expected to continue to grow at a compound annual rate of more than 5%.
Figure 3 Size of Parkinson's Disease Treatment Market (2020-2030)
It covers a variety of treatment methods, including traditional oral drugs, MAO-B inhibitors, inhalation administration, subcutaneous injection pumps, etc. Market leaders have established high-entry barriers through patent protection, clinical scale and integrated supply chain, making it difficult for small and medium-sized or new entrants to shake the industry’s position. In addition, with the aging of the population and the improvement of diagnostic technology, the market size continues to expand, prompting companies to increase their investment in treatment innovation and differentiated layout, thereby further intensifying the game between existing competitors.
Figure 4 SWOT Analysis
Traditional methods of synthesizing levodopa involve chemical synthesis that often relies on toxic chemicals and generates harmful by-products. In contrast, engineering probiotic to biosynthesize levodopa in the gut provides a more eco-friendly alternative. This aligns with global trends and sustainable pharmaceutical development. As a result, this method reduces the use of harmful chemicals and minimizes environmental impact.
The gut microbiome can be improved through taking probiotics. Probiotics can fix imbalances in gut bacteria, increase the number of good bacteria to produce helpful substances like short-chain fatty acids and reduce gut damage and body inflammation, which are all relevant to early stages of Parkinson’s.
Long-term use of Levodopa and other Parkinson’s medicines often causes uncomfortable effects such as stomach issues, constipation and drug absorption. Research shows that combining probiotics and prebiotics with these drugs can help reduce these problems. For example, a study in the Chinese Medical Journal reported that after 12 weeks of taking a mixture of Lactobacillus and Bifidobacterium, 60 PD patients improved in both motor symptoms and digestive issues (Chinese medical journal). Another analysis found that patients who drank fermented milk containing probiotics and prebiotic fiber had significantly more complete bowel movements than those who took a placebo.
Cold chain transportation means keeping the product cold all the time during shipping and storage. This needs special tools like fridges and cold trucks, which cost a lot.
Each person has a unique composition of gut flora, which means the product's efficacy may differ from person to person.
With the growth of global aging, the number of Parkinson's patients all over the world is increasing day by day, so the demand for better and safer treatments is growing.
More people now care about gut health and how it affects the body and mind. Social media, books, and videos have helped spread this idea. As a result, people are more likely to accept probiotic products.
After COVID-19, more people became open to trying new health treatments, like natural products and probiotics. This makes it easier for new ideas such as engineered probiotics.
There are already many probiotic and health food products on the market. It can be hard for a new product to get attention or win people’s trust, unless it shows clear benefits or strong.
Products that use live bacteria must pass strict safety checks before they can be sold. This can take a lot of time, money, and effort, which makes it harder to bring new probiotic treatments to market quickly.
In the management of Parkinson’s disease (PD), treatment modalities can be broadly classified into pharmacological therapies and surgical or device-based interventions, such as Deep Brain Stimulation (DBS). While pharmacological treatments are generally regarded as the first line of management, surgical approaches serve as critical options, particularly for patients in advanced stages. A meaningful comparison between these strategies requires consideration of several key indicators, including therapeutic effectiveness, safety, cost, accessibility, patient acceptance, and future potential.
Pharmacological treatments, particularly dopamine replacement therapies like levodopa, are highly effective in the early to moderate stages of PD, offering rapid symptom relief. However, their long-term use is often associated with motor complications such as fluctuations and dyskinesia.
In contrast, surgical interventions like DBS show superior efficacy in controlling motor symptoms in advanced PD, especially in patients who no longer respond consistently to medications. DBS provides customizable, continuous symptom management and can significantly reduce the need for medications, thereby mitigating drug-induced side effects.
Pharmacological approaches are non-invasive and generally associated with fewer immediate risks. However, certain drugs may lead to behavioral side effects, including impulse control disorders or hallucinations.
Surgical treatments, on the other hand, inherently carry procedural risks such as bleeding, infection, or electrode misplacement. Though rare, complications may lead to lasting neurological effects. Despite these risks, DBS is considered safe when performed in experienced centers, and its reversible nature offers an additional layer of safety compared to permanent interventions.
Surgical treatments like DBS involve high initial expenses, with total costs in the United States ranging from $53,200 to $100,000. While this upfront investment may be offset over time by reduced medication use, it remains a significant barrier for many patients.
In contrast, pharmacological therapies are generally more accessible financially in the short term, though the cumulative cost of lifelong medication use can be substantial, especially in healthcare systems where drug costs are not fully covered.
Pharmacological treatments are widely available and easily administered, even in low-resource settings. They are often the only viable option in regions lacking specialized care.
Surgical therapies, however, are restricted to well-equipped medical centers and require a high level of technical expertise. Accessibility is further limited by a shortage of specialists trained in surgical referrals and inconsistent outcomes in low-volume treatment centers. Regulatory hurdles and insurance coverage issues also restrict the availability of newer technologies like focused ultrasound (FUS).
Willingness to adopt a treatment also varies significantly between the two approaches. Medication is generally well accepted due to its familiarity and non-invasive nature.
Despite its benefits, patient hesitation remains a barrier to surgical options due to fears surrounding brain surgery and mismatched expectations about results. While DBS effectively alleviates motor symptoms, some patients may opt for non-invasive alternatives like disease-modifying drugs (e.g., alpha-synuclein antibodies), gene therapies, or stem cell treatments, which are perceived as lower-risk options. Misinformation or unrealistic expectations may further hinder acceptance, underscoring the need for improved patient education and psychological support.
Pharmacological research is rapidly evolving, with experimental therapies such as alpha-synuclein antibodies, gene editing, and stem cell transplantation offering the possibility of disease modification rather than mere symptom management.
In parallel, advancements in surgical techniques may improve accuracy and minimize complications. Research into earlier surgical intervention and combination therapies (e.g., DBS with gene therapy) could expand treatment efficacy. Additionally, improved physician and patient education may increase adoption by setting realistic expectations for outcomes.
Our product is an enteric-coated microcapsule containing engineered probiotics that produce dopamine precursors (L-dopa) directly in the gut. Compared to traditional pharmacological formulations, the enteric coating improves gastrointestinal tolerance, enhances drug delivery, and ensures that live probiotics survive stomach acid to reach the intestines. This non-invasive, sustainable, and easy-to-use format also makes long-term patient adherence more feasible, providing a unique edge over invasive procedures and conventional drugs.
We adopt a cost-plus pricing strategy initially to maintain affordability and encourage early adoption in clinical settings. As the product gains market acceptance and regulatory approval, we plan to incrementally increase the markup percentage, reflecting the growing value of clinical evidence, brand trust, and differentiated therapeutic outcomes.
The product will be distributed through hospitals, pharmacies, and online platforms. Our initial focus is on neurologist-recommended hospital channels, expanding later into community pharmacies and certified e-commerce partners to ensure broad access across both clinical and consumer healthcare settings.
We will leverage academic and clinical credibility to promote the product. Channels include:
Outreach to Parkinson’s disease (PD) patient communities via online platforms and offline education sessions to enhance awareness and acceptance
Hospitals represent a primary institutional customer segment for probiotic-based combination therapies. These formulations offer multiple clinical and operational advantages: they reduce adverse drug reactions, improve therapeutic outcomes, and enhance overall care quality for Parkinson’s disease (PD) patients. Moreover, they support hospitals in advancing research capabilities in neurodegenerative disease management.
Importantly, probiotic therapies contribute to increased patient satisfaction by minimizing side effects, thereby reducing the frequency of follow-up visits and the need for prescription modifications. This alleviates physician workload and streamlines clinical workflows.
Additionally, such therapies can lower total healthcare expenditures by mitigating common PD-related gastrointestinal complications, such as constipation and delayed gastric emptying—conditions that often necessitate secondary treatments like enemas or prokinetic agents. The gut-protective properties of probiotics can reduce the incidence of these complications, resulting in fewer hospitalizations and more efficient use of medical resources.
The core target population includes patients with a confirmed diagnosis of idiopathic Parkinson’s disease, primarily at Hoehn-Yahr stages 1–3. These patients commonly present with:
Early-stage patients seeking adjunctive therapies to delay disease progression or improve treatment responsiveness, particularly those not yet on or poorly responding to levodopa.
Long-term patients experiencing side effects from conventional medications—especially gastrointestinal discomfort—who may be more receptive to novel treatment modalities.
High-risk individuals with a family history of PD or pathogenic gene mutations, particularly those also presenting with intestinal dysbiosis. These individuals are often proactive in researching new interventions as preventive options.
An increasingly health-aware population is turning to probiotics as part of daily preventive health practices. This demographic sees probiotic interventions as a credible strategy for PD prevention, underpinned by emerging biomedical evidence.
Our approach leverages the emerging opportunity for early intervention in Parkinson’s disease (PD). Many prodromal symptoms, such as reduced sense of smell, chronic constipation, and REM sleep behavior disorder, can appear years before motor symptoms develop. This early phase offers a critical window for microbiome-targeted prevention.
Figure 5 Business Canvas of Prodopa
We designed two plasmid modules for L-DOPA biosynthesis. Module I enhances precursor supply by overexpressing tktA, aroF, and aroE. Module II includes tyrA, hpaB, and hpaC to convert chorismate into L-DOPA. All genes were His-tagged for detection. Plasmids were constructed using standard cloning techniques and verified via sequencing.
We constructed three versions of Module II, each carrying a tyrA gene from a different source. These were co-transformed with Module I into E. coli Nissle to generate three production strains. Each strain differed only in the tyrA variant. Plasmid verification ensured correct assembly for downstream functional comparison.
We conducted functional testing of the three engineered strains. Protein expression was assessed using SDS-PAGE or Western blot targeting the His-tagged enzymes. Shake flask fermentation was performed to evaluate L-DOPA production under identical conditions. The best-performing tyrA source was selected for further optimization based on yield.
To further improve tyrA expression, we designed and replaced the ribosome binding site with two synthetic variants. Each modified Module II plasmid was transformed with Module I to generate new strains. Fermentation assays compared L-DOPA output across RBS variants, identifying the most efficient design for translation initiation.
We tested the optimal strain under different initial glucose concentrations (20, 40, 60, and 80 g/L) in shake flask fermentations. This phase aimed to explore the upper limit of L-DOPA production and evaluate how substrate availability affects yield. Results will guide future optimization and inform fed-batch fermentation strategies.
We expect that in the next 20 years, the ProDopa project will go through a complete life cycle from technology verification, industrialization, rapid growth to strategic exit. The first five years are the product development and technology verification stage, focusing on completing the transformation from laboratory scale to pilot scale, establishing a stable engineering strain system, and obtaining key data support through preclinical research. During this period, we will also simultaneously advance patent applications to ensure that core technology protection is obtained in major global markets (such as China, the United States, and Europe).
The sixth to tenth years are the commercial expansion stage. It is expected that after obtaining the necessary approvals, we will officially launch the ProDopa product and cooperate with CDMO to establish a cold chain production and supply system to further expand production capacity to meet initial market demand. This stage will focus on the layout of North America, Europe, and East Asia, where Parkinson's patients are concentrated, and strive to promote the company's listing on the capital market around the eighth year to provide financial support for subsequent global expansion.
The eleventh to fifteenth years are the steady-state development stage. We will continue to promote product optimization and indication expansion, such as adjuvant treatment options for other dopamine deficiency-related diseases. At the same time, we will also expand our product pipeline and develop the next generation of gene regulation or delivery systems to improve the precision and safety of treatment. During this period, we plan to increase market penetration and further reduce the manufacturing cost of single products by establishing strategic alliances with large pharmaceutical companies.
In the 16th to 20th years, that is, the late stage of the product life cycle, we will enter a mature transformation period. As the core patents gradually expire, we will evaluate whether to continue to operate independently or seek a strategic exit. If the market and policy environment is suitable, we will sell core production technology and market assets to leading pharmaceutical suppliers in the industry at an acceptable price, realize capital returns and promote the widespread application of ProDopa technology around the world. At the same time, the company will retain the core R&D team and pipeline accumulation to explore the development and transformation of the next generation of synthetic biology technology platforms.
This financial forecast is based on the enteric-coated probiotic levodopa capsules for Parkinson’s treatment, covering a five-year projection of sales and costs. It follows a bottom-up estimation approach, detailed as follows:
Figure 6 Simplified Financial Statement of ProDopa
(All amounts are in Chinese yuan.)
Product pricing is adjusted annually to reflect increased market acceptance and added value—from 250 per box in the first year to 350 per box in the fifth year.
Initial sales volume is set at 8,000 boxes, with an annual growth rate of 50%, reaching 40,500 boxes by year five.
Corresponding sales revenue grows from 2 million in the first year to 14.175 million in the fifth year, demonstrating strong growth momentum.
Includes factory and equipment investments. A one-time investment of 1.5 million is made in the first year, with an additional 500,000 in equipment upgrades in the second year.
Long-term fixed costs are phased in during the first three years, with no further capital expenditures afterward.
Comprise labor, office, utilities, and promotion expenses.
Labor costs increase from 300,000 in the first year to 400,000 in later years, reflecting business expansion and increased staffing.
Marketing costs remain high to support market education and brand penetration—300,000 in the first two years, followed by 250,000 in the remaining years.
Overall annual fixed costs are controlled between 750,000 and 840,000 throughout the five years.
Unit variable costs (including material and supply chain costs) decreased from 180/box in year one to 120/box from year three onward, indicating improved production efficiency through economies of scale.
Annual variable costs grow from 1.44 million in the first year to 4.86 million in the fifth year, in line with rising sales volume.
Total cost increases from 3.69 million in year one to 5.7 million in year five.
Net income improves significantly over the years: a loss of 1.69 million in the first year narrows to 470,000 in the second year. Profitability is achieved in year three with a net profit of 2.4 million.
Years four and five show strong profitability, with net income of 4.56 million and 8.475 million, respectively, highlighting efficient scaling and strong margin performance.
Our project has achieved strong financial performance and continues to grow steadily. We have significantly increased our revenue by selling more products and adjusting pricing appropriately over time. At the same time, we have effectively managed our costs through continuous improvements in operational efficiency.
Although we experienced a loss in the first year, we successfully achieved profitability by the third year and have maintained steady earnings growth since then. We are confident in our sustainable business model and believe it holds strong potential for future expansion.
The financing channels are limited.For instance, in the early stage of medical drug research,a large amount of funds is invested for experiment ,and in the later stage, funds are also needed for marketing or continuous improvement and updates. Therefore,only financing methods that can provide a large amount of funds simultaneously can be chosen.The demand for funds is huge and the returns from this new technology are uncertain.Therefore,investors such as banks may not be willing to take the risk of providing a large amount of funds.
Gong Youran, Sun Lirong, Wang Deyuan, Fang Zipeng, Nicholas Gong, Li Jiachen, Ma Xuanyu, Li Zihan, Su Xiaolang, Zhou Zhenghang, Li Mengjia, Yu Mingzhuo, Sun Yiqiao,
Chen Hanhan, Su Kexin, Li Jiafeng, Guo Qianjing, Ma Sijin, Liu Junge, Xing Zishu, Chai Zhihan, Wang Jinchen, Qian Shiyu, Wang Lujia, Zhang Jiale, Yang Shuangrui, Ye Yunzhi, Guo Jingfei, Chen Jiawei, Zhang Yiyang, Li Qijia, Lu Zongmiao, Wang Zixuan, Peng Tianke, He Zixuan, Chen Xingyu.
Figure 7 Stakeholder Analysis of ProDopa
Engineered probiotics are designed to colonize the human gut and continuously release low levels of dopamine, providing stable and long-term relief from both motor and non-motor symptoms associated with Parkinson’s disease. By offering sustained symptom control, this approach reduces the need for frequent medication and lowers the overall treatment burden, thereby significantly improving patient’s quality of life and supporting functional independence. Furthermore, leveraging individual gut microbiome profiles allows for personalized treatment strategies that enhance therapeutic efficacy and compatibility. Additionally, these probiotics may offer preventive benefits or slow disease progression through ongoing neuromodulation.
The adoption of engineered probiotic therapies supports the growth of the biotechnology and precision medicine sectors, fostering innovation and contributing to economic development. By decreasing the reliance on conventional medications and reducing hospital visits and long-term care requirements, this approach helps alleviate pressure on healthcare systems and reduces the overall treatment costs for Parkinson’s disease. Moreover, increasing public awareness of microbiome-based therapies may improve health literacy and encourage earlier interventions, potentially contributing to a reduction in the incidence of Parkinson’s.
The production of probiotic-based therapies predominantly relies on microbial fermentation, which is generally more energy-efficient and generates less hazardous waste compared to traditional chemical synthesis methods. This environmentally sustainable manufacturing process not only supports greener pharmaceutical production but also aligns with increasingly stringent environmental regulations and corporate sustainability goals.
The use of engineered bacterial therapies presents certain risks for patients. If dopamine release is not adequately controlled, it may result in side effects such as anxiety, impulsive behaviors, or even hallucinations. Furthermore, should adverse events occur, the long-term colonization of engineered bacteria within the gut could pose challenges for reversal or eradication, potentially leading to sustained health complications.
The introduction of engineered bacterial strains raises public health concerns, particularly regarding environmental contamination. If these microorganisms are inadvertently released, they could spread within communities and increase public health risks, including potential antibiotic resistance transfer or unintended ecological consequences.
There is a tangible risk that engineered bacteria could enter natural ecosystems through wastewater or improper waste disposal. This introduction may disrupt indigenous microbial communities and undermine ecological balance. Additionally, biosecurity risks during manufacturing, transportation, or disposal processes require stringent oversight to prevent accidental environmental release.