1B. Pharm final year, Laddhad College of pharmacy Yelgaon, Buldhana, Maharashtra, India
2M. Pharm, Department of pharmaceutics, Rajarshi Shahu College of Pharmacy, Buldhana, Maharashtra, India
Clinical trials in rare and orphan diseases face unique and complex challenges due to limited patient populations, disease heterogeneity, and constrained economic incentives. This review provides a comprehensive analysis of the key barriers and emerging innovations in the design and execution of clinical trials targeting rare conditions. Regulatory frameworks such as the FDA’s Orphan Drug Act and the European Medicines Agency’s orphan designation process offer incentives like market exclusivity and accelerated approval to spur drug development. However, recruitment and retention difficulties, ethical concerns regarding placebo use, lack of natural history data, and variable disease progression hinder traditional randomized controlled trial designs. To overcome these barriers, adaptive methodologies—such as Bayesian statistics and seamless Phase II/III trials—have been introduced to enhance flexibility and efficiency. Real-world evidence, patient registries, N-of-1 trials, and basket trials provide valuable alternatives when conventional designs are unfeasible. Decentralized and virtual trial models, utilizing telemedicine and wearable devices, are reducing geographic and logistical barriers. Central to these efforts is the role of patient advocacy, which improves recruitment, retention, and trial relevance through community engagement. Global collaborations, including international consortia and harmonized data-sharing frameworks, are essential for maximizing research impact. Looking ahead, the integration of genomics, artificial intelligence, and supportive policy mechanisms will be pivotal in shaping the future landscape of rare disease research. The review emphasizes the need for multi-stakeholder coordination and patient-centric innovation to develop equitable, ethical, and scientifically robust trials for rare and orphan diseases.
Rare diseases are defined differently across regions, primarily based on prevalence thresholds. In the European Union (EU), a disease is considered rare when it affects fewer than 1 in 2,000 individuals [1]. In contrast, the United States Food and Drug Administration (FDA) defines an orphan disease as one affecting fewer than 200,000 people nationwide, equivalent to approximately 6.4 in 10,000 individuals [2]. Similarly, Japan considers a disease rare if it affects fewer than 50,000 people, or about 4 in 10,000 individuals [3]. These differences reflect regional policy frameworks and economic considerations, especially regarding drug development incentives and research priorities. Globally, over 300 million people are estimated to live with one of more than 7,000 identified rare diseases [4]. Despite their low individual prevalence, rare diseases collectively present a significant health burden. Many of these conditions are chronic, progressive, life-threatening, and often genetically inherited, disproportionately affecting pediatric populations [5]. Patients frequently experience delayed diagnoses, misdiagnoses, limited treatment options, and high out-of-pocket healthcare costs. For healthcare systems, the financial burden is compounded by high-cost therapies, long-term care requirements, and the need for specialized medical expertise [6]. Clinical trials are critical to improving diagnosis, understanding pathophysiology, and developing effective therapies for rare diseases. However, challenges such as small and geographically dispersed patient populations, disease heterogeneity, and lack of standardized outcome measures often hinder conventional trial designs [7]. In recent years, innovations including adaptive trial designs, use of real-world data, and patient-centric approaches have been introduced to overcome these obstacles and facilitate therapeutic advancement [8]. This review aims to critically examine the key challenges associated with conducting clinical trials in rare and orphan diseases and to explore the innovative methodologies that are addressing these hurdles. The scope includes regulatory frameworks, recruitment and retention issues, trial design adaptations, patient involvement, and emerging technologies. By providing a comprehensive overview, the review seeks to support researchers, regulators, and pharmaceutical stakeholders in optimizing trial strategies for rare disease populations.
2. Regulatory Framework
2.1Designation of Orphan Drugs by Regulatory Authorities
The recognition and regulation of orphan drugs are pivotal for stimulating drug development in rare diseases. Various global regulatory authorities define and designate orphan drugs using specific prevalence thresholds and criteria. In the United States, the Orphan Drug Act (1983) provides the legal framework under which a drug is designated as “orphan” if it treats a condition affecting fewer than 200,000 individuals in the country, or if it is not expected to recover development costs through U.S. sales [9]. The European Medicines Agency (EMA), through its Committee for Orphan Medicinal Products (COMP), designates a drug as orphan if it targets a life-threatening or chronically debilitating condition affecting not more than 5 in 10,000 people in the European Union [10]. In India, the Central Drugs Standard Control Organization (CDSCO) defines rare diseases in accordance with the National Policy for Rare Diseases (2021). While India lacks a comprehensive orphan drug act, CDSCO recognizes drugs for rare conditions and provides certain regulatory flexibilities for clinical trials and marketing authorization [11].
2.2 Incentives Provided for Orphan Drug Development
To offset the economic challenges associated with rare disease drug development, regulatory agencies offer a range of incentives. In the U.S., these include tax credits for clinical trial costs, exemption from user fees, protocol assistance, and 7 years of market exclusivity upon approval [12]. The EU provides 10 years of market exclusivity, reduced regulatory fees, and access to centralized marketing authorization procedures [13]. In Japan and South Korea, similar benefits include financial support for research, prioritized regulatory reviews, and extended market exclusivity [14]. India’s framework, while less structured, is evolving, and recent policies have indicated the potential for import fee waivers and expedited approvals for rare disease drugs [15].
2.3 Key Regulatory Pathways
Innovative regulatory pathways have been established to expedite the development and approval of therapies targeting rare and serious conditions. This table summarizes key regulatory mechanisms established by the FDA and EMA to expedite the development, approval, or access to drugs, particularly in cases involving serious conditions or rare diseases where traditional clinical trials may not be feasible.
Table:1. Overview of Regulatory Pathways Facilitating Early or Expanded Drug Access in the U.S. and EU
Regulatory Pathway |
Authority |
Key Features |
Purpose / Context |
Accelerated Approval |
FDA |
Approval based on surrogate endpoints likely to predict clinical benefit; post-marketing studies required |
Speeds up access to therapies, especially when conventional trials take too long |
Breakthrough Therapy Designation |
FDA |
Granted for drugs with preliminary evidence of substantial improvement over existing treatments; enables intensive guidance and priority review |
Facilitates faster development and review for promising therapies |
Compassionate Use / Expanded Access Programs |
FDA |
Allows access to investigational drugs for patients with serious/life-threatening conditions outside clinical trials |
Provides options when no comparable alternatives exist |
Conditional Marketing Authorization |
EMA |
Permits early approval based on less comprehensive data for drugs meeting unmet medical needs, with obligation to provide data post-approval |
Helps in early access to drugs for rare or serious conditions where full data is not yet available |
3. Challenges in Conducting Trials For Rare Diseases
3.1 Limited Patient Population
A defining challenge in rare disease clinical trials is the limited and dispersed patient population, which complicates both trial recruitment and retention, often delaying or halting trial progress.
3.1.1 Difficulty in Recruitment and Retention
Recruiting adequate numbers of eligible participants remains a major barrier in rare disease trials. Due to the very nature of rare diseases—by definition affecting a small number of individuals—identifying sufficient participants within a reasonable timeframe is often impractical [20]. Additionally, inclusion criteria can be stringent due to disease heterogeneity, further narrowing the eligible pool [21]. Retention is also problematic. Patients with rare diseases often face debilitating symptoms, mobility issues, or comorbidities, which can discourage continued participation in lengthy or demanding trial protocols [22]. Furthermore, the lack of trial sites in proximity to patients' residences can increase dropout rates due to travel burdens and logistical difficulties [23].
3.1.2 Geographic Dispersion of Patients
Patients affected by rare diseases are typically widely scattered across regions, countries, or continents. This geographic dispersion makes centralized clinical trials unfeasible, leading to increased trial complexity and costs [24]. Establishing multiple trial sites, coordinating regulatory compliance across jurisdictions, and ensuring data consistency become increasingly challenging when patients span multiple health systems and languages [25]. In addition, cross-border trials often face regulatory misalignment, customs restrictions on investigational products, and difficulties in monitoring site performance remotely. These issues collectively contribute to significant delays in trial initiation and completion [26].To address such dispersion, decentralized or virtual trial models are being explored (discussed in Section 4), though they bring their own set of operational and regulatory hurdles.
3.2 Study Design Limitations
3.2.1 Issues with Randomized Controlled Trials (RCTs)
Randomized controlled trials (RCTs) are considered the gold standard in clinical research due to their ability to minimize bias and establish causal relationships. However, applying conventional RCT methodologies to rare and orphan diseases presents significant challenges. The most prominent issue is the infeasibility of enrolling a sufficient number of participants to meet statistical power requirements. Standard RCTs often require large sample sizes, which are unattainable in most rare disease populations due to low prevalence and geographical dispersion [27]. As a result, many trials are underpowered, leading to inconclusive or non-generalizable results [28]. Moreover, disease heterogeneity in rare conditions may necessitate stratification or subgroup analyses, further complicating study design and data interpretation [29]. Additionally, there is often a lack of validated clinical endpoints and biomarkers specific to rare diseases, hindering the development of robust outcome measures [30].
3.2.2 Ethical Concerns in Using Placebo
The use of placebo controls in rare disease trials raises serious ethical concerns, particularly when the disease is severe, progressive, and life-threatening, and when no alternative therapies exist [31]. In such cases, withholding potentially beneficial treatment from patients in the control arm may violate ethical principles of beneficence and non-maleficence. Ethical challenges are further intensified in pediatric trials or those involving neurodegenerative diseases, where disease progression is irreversible and timely intervention is crucial [32]. As a result, regulatory agencies often encourage alternative designs such as historical controls, cross-over designs, or adaptive trials to minimize patient exposure to placebos while preserving scientific validity [33]. These limitations necessitate the development of innovative trial methodologies that can maintain ethical standards and generate meaningful evidence in the context of small and vulnerable patient populations.
3.3 Lack of Disease Understanding
One of the major challenges in conducting clinical trials for rare and orphan diseases is the limited understanding of disease mechanisms and progression. This lack of foundational knowledge significantly hampers the design, execution, and interpretation of clinical research.
3.3.1 Insufficient Natural History Data
A critical barrier in trial design for rare diseases is the scarcity of comprehensive natural history data. Natural history studies provide essential insights into disease onset, progression, and variability, which are fundamental for defining meaningful clinical endpoints and designing appropriate trial protocols. However, due to the low prevalence of patients and often limited longitudinal data, many rare diseases lack well-documented trajectories, making it difficult to determine the optimal timing for interventions or to evaluate the efficacy of a treatment over time (34,35). Without this data, placebo groups in randomized trials may be ethically or practically unfeasible, and alternative designs, such as single-arm trials or historical controls, must be considered, though they come with limitations in scientific rigor and regulatory acceptance (36).
3.3.2 Variability in Clinical Presentation and Progression
Rare diseases often exhibit considerable heterogeneity in their clinical manifestations, even among patients with the same genetic mutation. This phenotypic variability complicates patient stratification, outcome measurement, and biomarker development (37). For instance, differences in age of onset, severity, and affected systems can influence how patients respond to interventions, leading to challenges in identifying suitable inclusion and exclusion criteria. Furthermore, this variability can obscure treatment effects and reduce statistical power, necessitating the use of adaptive trial designs or novel statistical methodologies to address intra-disease diversity (38,39).
3.4 Funding and Economic Challenges
Conducting clinical trials for rare and orphan diseases presents significant economic and financial obstacles. The low prevalence of these conditions inherently limits commercial incentives, while the high cost of trial implementation often exceeds available funding resources.
3.4.1 Limited Commercial Interest
Rare diseases, by definition, affect a small number of individuals, making the market for associated therapies relatively small. This limited market size diminishes the financial return on investment for pharmaceutical companies, which discourages extensive research and development (40). Although regulatory incentives—such as market exclusivity, tax credits, and fee waivers under orphan drug legislations—have stimulated some interest, these measures are not always sufficient to offset the financial risks associated with developing treatments for small patient populations (41). In particular, early-stage research and proof-of-concept trials remain underfunded, often relying on academic institutions, non-profit organizations, or patient advocacy groups to initiate studies (42).
3.4.2 High Cost of Trials
Despite involving fewer patients, clinical trials for rare diseases can be as expensive—or more expensive—than those for common conditions. Recruitment challenges, geographical dispersion of patients, the need for specialized diagnostic tools, and customized treatment protocols contribute to elevated operational costs (43). Furthermore, establishing appropriate clinical endpoints and regulatory-compliant data infrastructure often requires additional investment in natural history studies, biomarker development, and surrogate outcome validation (44). These complexities increase the per-patient cost of trials and create significant financial barriers for both sponsors and investigators. In light of these challenges, innovative funding models and public-private partnerships are being explored to share the economic burden. Collaborative frameworks involving industry, academia, governments, and patient groups are essential for sustaining long-term investment in rare disease research.
4. Innovative Approaches in Trial Design
To address the unique challenges in conducting clinical trials for rare and orphan diseases, researchers and regulators have increasingly turned to innovative trial designs. These novel methodologies aim to enhance trial efficiency, optimize resource use, and generate meaningful results despite small patient populations.
4.1 Adaptive Trial Designs
Adaptive trial designs offer flexible frameworks that allow modifications to key trial parameters based on interim data analyses without compromising the integrity or validity of the study. Such designs are particularly useful in rare disease research where patient populations are limited, and trial timelines are often constrained.
4.1.1 Bayesian Methods
Bayesian statistical methods are gaining traction in rare disease trials due to their ability to incorporate prior knowledge and update probabilities as data accumulate during the study. Unlike traditional frequentist approaches, Bayesian models allow for dynamic decision-making, such as stopping a trial early for efficacy or futility, or modifying dosage levels (45,46). This adaptability can enhance trial efficiency and reduce the number of participants exposed to suboptimal treatments. Additionally, Bayesian approaches facilitate the use of historical controls or external data, which is advantageous in rare disease contexts where randomized controls may be ethically or logistically challenging (47).
4.1.2 Seamless Phase II/III Trials
Seamless Phase II/III trial designs integrate the exploratory and confirmatory phases of drug development into a single continuous protocol. This approach minimizes delays between trial phases and allows early data to inform later stages without the need for new study initiation (48). In rare disease research, seamless designs are particularly beneficial as they reduce the need for separate patient cohorts and streamline regulatory pathways (49). Adaptive features such as sample size re-estimation, dose selection, and endpoint refinement can also be incorporated within seamless frameworks, further enhancing their utility and efficiency (50).The application of adaptive trial designs, particularly Bayesian models and seamless Phase II/III trials, represents a transformative step in rare disease research. These methodologies not only improve the feasibility and ethical acceptability of trials but also increase the likelihood of timely and robust outcomes.
4.2 Use of Real-World Evidence (RWE) and Registries
In the context of rare and orphan diseases, where traditional randomized controlled trials (RCTs) often face feasibility issues due to small and geographically dispersed patient populations, real-world evidence (RWE) and patient registries offer vital complementary tools. These resources can support trial design, regulatory decisions, and post-marketing surveillance by capturing data from actual clinical settings.
4.2.1 Observational Studies
Observational studies, derived from real-world clinical data, provide valuable insights into treatment outcomes, disease progression, and healthcare utilization. In rare diseases, where prospective trial data may be limited or unavailable, well-designed observational studies can help bridge evidence gaps and inform treatment decisions (51). These studies can include cohort studies, case-control studies, or pragmatic trials, all of which reflect routine clinical practice and patient diversity more accurately than RCTs (52). While they may be more susceptible to bias, methodological advances such as propensity score matching and instrumental variable analysis have improved the reliability of these data sources (53). Regulatory agencies, including the FDA and EMA, are increasingly recognizing the potential of RWE to support approval processes, especially for treatments targeting unmet needs in rare conditions (54).
4.2.2 Natural History Databases
Natural history databases are a cornerstone for understanding the baseline course and progression of rare diseases. These longitudinal data repositories help define clinical endpoints, identify prognostic factors, and establish benchmarks for evaluating new treatments (55). In the absence of placebo-controlled trials, natural history data can serve as external comparators to assess treatment effects (56). Furthermore, they facilitate the identification of eligible patients for trials and improve recruitment by centralizing information across institutions and borders. For ultra-rare diseases, where trial feasibility is extremely limited, natural history data may represent the only viable source of clinical evidence (57). Together, observational studies and natural history registries enhance the evidentiary foundation for rare disease research. By integrating these data into clinical trial design and evaluation, stakeholders can optimize resource use and accelerate the development of effective therapies.
4.3 N-of-1 and Basket Trials
In the pursuit of effective clinical trial designs for rare and orphan diseases, non-traditional models such as N-of-1 trials and basket trials have gained prominence. These approaches challenge conventional paradigms by focusing on personalization and molecular targeting, providing feasible alternatives when patient numbers are extremely limited.
4.3.1 Individualized Trial Models (N-of-1 Trials)
N-of-1 trials are a type of single-patient, crossover study where an individual serve as their own control. These trials are particularly suited to rare diseases, where inter-patient variability is high and recruiting sufficient participants for large-scale trials is impractical (58). By alternating between treatment and placebo or between different interventions, the trial can assess treatment efficacy on an individualized basis. This design is especially useful when disease symptoms are stable or reversible, and outcomes can be measured objectively within a short timeframe (59). N-of-1 trials can generate clinically actionable data and may be aggregated across patients to form meta-analyses that inform broader treatment strategies (60). Although traditionally limited to symptomatic treatments, advances in genomics and digital health tools are expanding the applicability of this model to more complex and chronic rare diseases (61).
4.3.2 Grouping by Molecular Targets (Basket Trials)
Basket trials represent a paradigm shift by focusing on shared molecular or genetic alterations rather than traditional disease classifications. In these trials, patients with different disease types but the same molecular aberration are grouped together to evaluate the efficacy of targeted therapies (62). This approach is particularly relevant in rare cancers and genetic disorders, where the same mutation may drive different phenotypes. By leveraging molecular diagnostics, basket trials allow more efficient use of limited patient populations and open new avenues for drug repurposing across rare indications (63). Regulatory bodies have shown increasing flexibility in accepting such trial designs, especially when supported by strong biological rationale and robust biomarker data (64). Incorporating N-of-1 and basket trial designs into the clinical research landscape offers flexible, patient-centered, and innovative solutions that address the inherent constraints of rare disease trials. These models exemplify the shift toward precision medicine, where therapeutic strategies are informed by individual or shared molecular profiles rather than traditional disease categories.
4.4 Decentralized and Virtual Trials
Decentralized and virtual clinical trials are emerging as innovative solutions to the logistical and geographic barriers commonly encountered in rare and orphan disease research. These trial models utilize digital tools and remote technologies to reduce patient burden, increase accessibility, and enhance data collection efficiency.
4.4.1 Use of Telemedicine and Wearable Devices
Telemedicine plays a pivotal role in decentralized trials by enabling remote consultations, virtual follow-ups, and real-time communication between investigators and participants. This is particularly beneficial for patients with rare diseases who often reside far from specialized research centers (65). In addition, wearable devices and mobile health applications allow for continuous monitoring of physiological parameters such as heart rate, movement, and sleep patterns, generating real-time, objective data outside the clinical setting (66). These technologies not only improve data granularity but also empower patients by making participation more convenient and less intrusive (67). Moreover, remote assessments reduce the need for frequent travel, which can be especially taxing for individuals with chronic or progressive rare conditions (68).
4.4.2 Home-Based Monitoring
Home-based monitoring expands the scope of clinical trial participation by allowing patients to undergo diagnostic procedures, treatment administration, and outcome assessments from their own homes. Examples include self-administered medications, home nursing visits for sample collection, and use of remote diagnostic kits (69). This approach enhances patient retention and adherence while preserving the quality and consistency of data collection. In rare diseases where patient populations are limited and geographically dispersed, the ability to include participants regardless of location is a substantial advantage (70). Regulatory agencies are increasingly supportive of decentralized trial components, recognizing their potential to enhance inclusivity and reduce operational costs (71).
5. Role of Patient Advocacy and Community Engagement
Patient advocacy and community engagement play a pivotal role in the successful planning, execution, and dissemination of clinical trials in rare and orphan diseases. Involving patients and advocacy organizations from the earliest stages of clinical research ensures that studies are aligned with the real-world needs, values, and experiences of the affected community.
5.1 Importance of Patient-Centric Approaches
A patient-centric approach shifts the focus of clinical research from solely scientific or regulatory goals to addressing the priorities and quality-of-life concerns of patients and caregivers. For rare diseases, where lived experience is often underrepresented in the medical literature, this approach helps shape relevant endpoints, improve trial design, and enhance the ethical integrity of studies (72). Engaging patients in the co-design of protocols can lead to more realistic inclusion criteria, acceptable visit schedules, and the selection of outcomes that are meaningful to those affected (73). Regulatory authorities, such as the FDA and EMA, have increasingly emphasized the value of patient input in drug development, including through patient-focused drug development (PFDD) initiatives (74).
5.2 Partnership with Rare Disease Organizations
Collaboration with rare disease advocacy groups and foundations is essential in bridging the gap between researchers and the patient population. These organizations often possess in-depth knowledge of the disease, maintain patient registries, and provide access to geographically dispersed individuals who may be eligible for trials (75). In addition, they help disseminate trial information, provide logistical support, and advocate for regulatory and funding pathways that facilitate rare disease research (76). Such partnerships also foster trust between communities and researchers, increasing transparency and shared ownership of the research process.
5.3 Enhancing Patient Recruitment and Retention
Recruitment and retention remain major challenges in rare disease trials due to small population sizes, clinical heterogeneity, and travel burdens. Community engagement can significantly alleviate these challenges. By involving advocacy groups in outreach and education, trials can better reach underserved populations and raise awareness about available opportunities (77). Furthermore, patient-friendly materials, continuous communication, and support services—often coordinated through advocacy partnerships—help maintain participant motivation and reduce dropout rates (78). Culturally sensitive engagement strategies and use of digital platforms further enhance inclusivity and long-term involvement.
CASE STUDIES
Case studies of successful clinical trials in rare diseases provide important insights into overcoming the unique challenges associated with these conditions. By analyzing these examples, researchers and stakeholders can identify best practices, inform future trial designs, and improve patient outcomes.
6.1 Successful Clinical Trials in Rare Diseases
One of the most notable examples of success in rare disease therapeutics is the approval of nusinersen (Spinraza) for spinal muscular atrophy (SMA). SMA is a severe genetic neuromuscular disorder with a high infant mortality rate in its most common form. The ENDEAR trial, a randomized, double-blind, sham-controlled study, demonstrated significant improvement in motor function and survival among infants receiving nusinersen compared to controls (79). Key to the trial’s success was the use of robust natural history data, early collaboration with patient advocacy groups, and an adaptive trial design that allowed the trial to be halted early for efficacy (80). Another landmark achievement is voretigene neparvovec (Luxturna), the first gene therapy approved for an inherited retinal disease (Leber’s congenital amaurosis caused by RPE65 mutations). The pivotal Phase III trial demonstrated significant improvement in functional vision, using a novel, patient-centric primary endpoint: the multi-luminance mobility test (MLMT) (81). The study leveraged well-characterized natural history data, patient engagement in endpoint selection, and rigorous long-term follow-up to assess durability (82). These examples underscore how rare disease trials, despite inherent limitations, can lead to transformative treatments when innovative methodologies and stakeholder collaboration are effectively employed.
6.2 Lessons Learned and Best Practices
This table summarizes critical success factors identified from case studies, highlighting strategies that enhance patient-centricity, regulatory efficiency, and collaborative innovation in rare disease clinical trials.
Table: 2. Key Lessons Learned and Best Practices from Rare Disease Clinical Trial Case Studies
S.No. |
Best Practice / Lesson Learned |
Description |
1 |
Early and continuous patient engagement |
Ensured trial designs were relevant, feasible, and patient-centric. |
2 |
Development of tailored endpoints and biomarkers |
Captured meaningful clinical outcomes, especially where traditional endpoints were not applicable. |
3 |
Use of natural history data and historical controls |
Provided context for interpretation and regulatory review, particularly when placebo use was ethically challenging. |
4 |
Implementation of adaptive trial designs and regulatory flexibility |
Accelerated trial timelines and enabled earlier patient access to critical treatments. |
5 |
Multidisciplinary collaboration (industry, academia, regulators, advocacy groups) |
Facilitated alignment of scientific and patient priorities, improving trial success and impact. |
7. Global Collaboration and Data Sharing
In the field of rare and orphan disease research, global collaboration and data sharing are essential to overcome the inherent challenges of small, dispersed patient populations and limited scientific knowledge. International networks, consortia, and harmonized regulatory frameworks play a central role in accelerating clinical development, ensuring ethical standards, and maximizing the utility of collected data.
7.1 Importance of International Networks and Consortia
International networks such as the International Rare Diseases Research Consortium (IRDiRC) and Orphanet have been instrumental in coordinating global efforts to improve diagnosis, research, and therapy development for rare diseases. IRDiRC, launched in 2011, brings together funding agencies, companies, researchers, and patient advocacy groups with the goal of enabling the development of 1,000 new therapies for rare diseases and improving diagnosis by 2027 (83). Similarly, Orphanet provides a comprehensive, interoperable database of rare diseases and orphan drugs, supporting clinicians, researchers, and policymakers worldwide (84). These initiatives facilitate multi-center clinical trials, reduce duplication of efforts, and foster cross-border collaborations that are critical in rare disease research where national patient numbers are insufficient. Moreover, global consortia enhance the development of disease registries, natural history studies, and patient-reported outcome measures by pooling resources and expertise (85).
7.2 Harmonization of Data and Regulatory Standards
Harmonizing data collection practices, outcome measures, and regulatory standards across countries is crucial to ensure the comparability and usability of clinical data. Lack of standardization in clinical endpoints, diagnostic criteria, and data formats has historically hindered collaboration and data integration in rare disease research. Recent efforts led by the IRDiRC, the European Medicines Agency (EMA), and the U.S. Food and Drug Administration (FDA) have promoted alignment in regulatory guidance and data-sharing policies to address these issues. Initiatives such as Common Data Elements (CDEs) and the Global Rare Disease Registry (GRDR) program have laid the groundwork for interoperable, high-quality datasets that can be reused across studies. Regulatory harmonization also enables faster drug development and approval pathways, especially through mutual recognition procedures, shared review models, and alignment on orphan drug designation criteria. Overall, global collaboration and data harmonization are not only desirable but necessary for the efficient and ethical advancement of clinical research in rare diseases. These efforts ensure that limited resources are maximized and that patients worldwide can benefit from scientific progress (86).
8. Ethical Considerations
Clinical trials in rare and orphan diseases pose distinct ethical challenges due to the vulnerability of the patient population, the urgency of unmet medical needs, and the often-limited availability of alternative treatment options. Ethical frameworks must ensure that trials are both scientifically valid and ethically justifiable, with particular attention to informed consent, equitable access, and post-trial responsibilities.
8.1 Informed Consent in Small Populations
Securing truly informed consent in rare disease trials can be complex. Patients and caregivers may feel pressured to participate due to the lack of available treatments, potentially compromising voluntariness. Moreover, trials often involve children or cognitively impaired individuals, necessitating additional safeguards such as assent procedures and surrogate decision-making. Small patient populations also increase the risk of therapeutic misconception—where participants overestimate the likelihood of personal benefit from experimental treatments. Clear, culturally appropriate communication and robust ethics committee oversight are essential to uphold patient autonomy in these contexts.
8.2 Equity and Access to Experimental Therapies
Equitable access to rare disease trials remains a critical concern. Geographic barriers, socioeconomic status, and limited trial sites often prevent many eligible patients from participating (94). Additionally, trials may prioritize patients with specific genetic mutations or disease subtypes, inadvertently excluding broader patient groups. Ethical trial design must therefore consider diversity and inclusion, ensuring that underrepresented populations are not systematically excluded from potential benefits. Global collaborations and decentralized trial models can help address these disparities and promote broader accessibility (87).
8.3 Post-Trial Access
Post-trial access to investigational therapies is another significant ethical issue in rare disease research. Given the life-threatening nature of many rare conditions and the lack of alternative treatments, denying continued access to a drug that demonstrated benefit during the trial can be unjust. Ethical guidelines, including those by the Declaration of Helsinki, emphasize that sponsors and researchers should have a plan for post-trial provisions when necessary. This is particularly relevant in cases where marketing approval is delayed or not pursued. Sustainable strategies for compassionate use, early access programs, and pricing transparency are needed to ensure patients are not left without care following trial completion (88).
9. Future Directions and Recommendations
As the landscape of rare and orphan disease research continues to evolve, innovative technologies and supportive policies are poised to transform clinical trial methodologies. Looking ahead, the integration of genomics, artificial intelligence (AI), and machine learning (ML)—combined with strategic policy and funding support—can significantly enhance the efficiency, equity, and success of clinical trials for rare diseases.
9.1 Integration of Genomics and Precision Medicine
The application of genomics and precision medicine has already begun to revolutionize the diagnosis and treatment of rare diseases. Next-generation sequencing (NGS) technologies enable the identification of novel pathogenic variants, accelerating the diagnosis of ultra-rare conditions and facilitating patient stratification for clinical trials. Precision medicine approaches allow for targeted interventions based on specific genetic or molecular characteristics, making clinical trials more efficient and relevant. For instance, molecularly guided treatments, such as those used in basket trials, demonstrate the potential for precision approaches in populations with shared mutations across disease phenotypes. To fully realize the benefits, integrating genomic screening into routine clinical care and ensuring the availability of centralized databases for genetic data will be essential (89).
9.2 AI and Machine Learning in Trial Design
Artificial intelligence (AI) and machine learning (ML) offer powerful tools for optimizing clinical trial design and execution. These technologies can be used to identify suitable participants through electronic health records, predict disease progression, and model treatment outcomes using real-world and historical data. AI-driven analytics can also aid in adaptive trial designs, enhance monitoring through digital biomarkers, and automate regulatory reporting processes. In rare diseases, where small sample sizes pose a challenge to traditional statistical methods, AI can assist in building synthetic control arms and improving endpoint sensitivity (. However, the ethical use of AI in clinical research necessitates robust validation, transparency in algorithms, and regulatory oversight (90).
9.3 Policy and Funding Support for Rare Disease Trials
Sustained policy and funding support are crucial for fostering innovation in rare disease clinical research. Orphan drug legislations—such as the U.S. Orphan Drug Act and the EU Regulation on Orphan Medicinal Products—have played a significant role in incentivizing drug development through tax credits, market exclusivity, and regulatory assistance (91). However, the high cost and risk associated with rare disease trials still require targeted public funding and public-private partnerships to ensure long-term sustainability. Future policy efforts should prioritize reducing regulatory burdens, promoting international trial harmonization, and ensuring equitable access to emerging therapies. Investment in global infrastructure, such as shared registries and biobanks, will also support more inclusive and efficient trials (92).
CONCLUSION
Clinical trials for rare and orphan diseases present a unique and pressing set of challenges due to limited patient populations, geographic dispersion, complex regulatory frameworks, and constrained financial resources. These factors have historically hindered drug development and access for individuals living with rare conditions. However, recent advancements in trial design and regulatory flexibility have opened promising avenues for overcoming these hurdles. Adaptive trial designs, Bayesian methods, real-world evidence, and decentralized models have transformed traditional research paradigms by enabling more efficient, inclusive, and ethical studies. Notably, individualized approaches such as N-of-1 and basket trials, along with genomic and molecular profiling, have revolutionized precision medicine in rare diseases. Patient advocacy organizations play a pivotal role in bridging the gap between researchers and affected communities, improving recruitment, retention, and trial relevance through patient-centric approaches. Furthermore, international collaboration and harmonization of data and regulatory practices have enhanced global research efforts, facilitating data sharing and reducing duplication of effort. Ethical considerations remain central to these trials, with emphasis on informed consent, equitable access, and post-trial provisions. As the field evolves, integrating AI, machine learning, and genomic data will be crucial in optimizing trial design and identifying novel therapeutic targets. Moving forward, sustained policy support, collaborative infrastructure, and inclusive research strategies will be essential to ensure that clinical innovation translates into tangible benefits for patients with rare and orphan diseases. Collectively, these advancements underscore a growing commitment to equity, innovation, and patient empowerment in rare disease research.
REFERENCE
Mrudula Dhengale*, Harsha Bathe, Prajakta Hage, Aachal Kawade, Neha Phopse, Nikita Sapkal, Tejal Dharme, Shivshankar Nagrik, Challenges and Innovations in Conducting Clinical Trials in Rare and Orphan Diseases, Int. J. Sci. R. Tech., 2025, 2 (5), 427-441. https://doi.org/10.5281/zenodo.15459246