The 505(b)(2) New Drug Application (NDA) provides a streamlined route for drug approval, making it a popular choice among many clients seeking FDA approval. Established by the Hatch-Waxman Amendments of 1984, this pathway refers to a specific section of the Federal Food, Drug, and Cosmetic Act.
Its inception aimed to eliminate the redundancy of conducting studies on drugs that have already received approval. In simpler terms, the 505(b)(2) provision allows the FDA to consider pre-existing data when reviewing an application, even if the applicant didn’t develop this data.
Under this application, complete safety and effectiveness reports are mandatory. However, the 505(b)(2) provision permits the inclusion of data from external studies, specifically those about the safety and efficacy of the active ingredient. This exception accelerates the approval process and reduces associated costs when contrasted with the traditional 505(b)(1) pathway.
As a result, applicants can achieve FDA approval more efficiently and at a reduced cost while bringing innovative, high-value products to the market.
This guide introduces and explains the 505(b)(2) new drug application process's intricacies, benefits, and strategies for successfully navigating this regulatory pathway.
Need expert 505(b)(2) regulatory support? We can help.The FDA Group provides a versatile range of services to assist clients in crafting top-tier, compliant NDA submissions. We are equipped to accommodate diverse needs—from managing the complete NDA process to aiding in publishing and submission or reviewing specific documents. Our team of consultants comprises former FDA reviewers and skilled professionals, including medical writers and project managers, all dedicated to supporting every aspect of your NDA preparation. Reach out to us to explore how we can help you bring your product to market and keep it there. |
What is the 505(b)(2)?
The 505(b)(2) provisions were designed to avoid unnecessary duplication of studies already performed on previously approved ("reference" or "listed") drugs.
A 505(b)(2) NDA contains complete safety and effectiveness reports but allows at least some of the information required for approval to come from studies not conducted by or for the applicant. This can include safety and efficacy information on the active ingredient from studies in the public domain or the FDA's previous findings.
Drug candidates with 505(b)(2) potential
The 505(b)(2) pathway is ideal for various types of drug innovations, including:
|
These new products often contain well-understood active ingredients present in existing, approved drug products. Companies need only create a bridge between what is already known about the previously approved reference drug and the novel drug product or indication.
A similar regulatory approval route in Europe is the hybrid procedure based on Article 10 of Directive 2001/83/EC.
The Three 505 Regulatory Pathways at a Glance
To better understand the 505(b)(2) pathway, it's helpful to compare it with other regulatory routes.
The FDA recognizes three primary pathways for the approval of new drugs and abbreviated new drug applications (ANDA): the 505(b)(1) NDA, 505(j) ANDA, and 505(b)(2) NDA. Each pathway serves a distinct purpose in the drug approval process.
505(b)(1) NDAThe 505(b)(1) NDA is a full application where data is predominantly obtained from studies conducted by and for the sponsor. Every study conducted under this pathway is specifically tailored for the drug by the sponsor and serves as the foundational data for FDA approval. This pathway is the most resource-intensive and time-consuming of the three. |
505(b)(2) NDAThe 505(b)(2) NDA is essentially a hybrid between an ANDA (505(j)) and a full NDA (505(b)(1)), combineing aspects of both the full NDA and ANDA. This pathway is ideal for modified or improved versions of existing innovator drugs, leading to the creation of a distinct drug product with its own exclusivity rights. The process incorporates pre-existing data and new findings to facilitate a more efficient approval process. In Europe, a similar process is known as the Hybrid application, reflecting its combined nature. |
505(j) ANDAThe 505(j) ANDA pathway is appropriate for drug products that are the same as approved products (generics). This pathway becomes relevant when a patented innovator drug is approaching its expiration. The central criterion for approval through this pathway is demonstrating bioequivalence to the innovator product. A food effect study is typically required for oral dosage forms to ensure similar efficacy and safety profiles. |
The Benefits of the 505(b)(2) Pathway
The 505(b)(2) pathway offers several high-level advantages:
|
The 505(b)(2) pathway offers pharmaceutical companies a strategic advantage by reducing the time and financial investment typically required for the conventional full NDA. This approach bypasses the need for numerous nonclinical studies and extensive safety and efficacy tests.
This pathway specifically benefits new drugs similar to already approved drugs but have slight variations in formulation or administration routes. Before the Waxman/Hatch Act, approval for such drugs depended on extensive literature-based arguments provided by the applicant to prove the safety and efficacy of the new product.
Applicants can now reference the safety and efficacy data of the original innovator drug without needing a right of reference. This is possible because the API remains unchanged, implying inherent similarities in safety and efficacy between the new and original products.
While some differences in formulation or administration might require additional clinical studies for 505(b)(2) applicants, these are typically less extensive than what’s required for original innovator drugs. In many cases, a bioequivalence study suffices to demonstrate the similarity between the two drugs, leading to FDA approval. In instances where additional clinical studies are needed, the requirements are generally less stringent.
Again, the pathway also presents an opportunity for market exclusivity ranging from 3 to 7 years.
Navigating the 505(b)(2) Submission Process
The 505(b)(2) submission journey involves four crucial steps: candidate identification, candidate assessment, product planning, and the pre-IND meeting.
1. Candidate identification
Success in the 505(b)(2) pathway begins with identifying products that have:
- Documented market differentiation
- Low development risk
- High-profit potential
It typically involves a few nonclinical studies.
During Phase 1, it's essential to understand that the new product’s pharmacokinetic (PK) profile need not mirror the innovator product exactly but should be as favorable, ensuring low development risk and distinct market positioning.
Ideal 505(b)(2) candidates include:
Potential types of 505(b)(2)s include:
|
2. Feasibility—candidate assessment
This phase is critical for minimizing costly mistakes and validating the product concept’s value to investors. Candidates should undergo a rigorous evaluation process that examines their scientific, medical, regulatory, and commercial viability. This thorough assessment ensures that the candidate has a solid foundation and the potential for successful market entry and sustainability.
Before embarking on development, it's crucial to assess candidates thoroughly. Consider the following aspects:
Scientific Viability
Medical Viability
Regulatory Viability
Commercial Viability
|
3. Product Planning
At this stage, developers should strategically incorporate existing data to optimize their development plans. This often involves evaluating potential market exclusivity opportunities, with options ranging from orphan drug exclusivity to new chemical entity (NCE) exclusivity and others. Even within the 505(b)(2) pathway, there's a possibility to secure NCE exclusivity, typically associated with 505(b)(1) drugs.
Unlike the traditional 505(b)(1) pathway, which can take up to 15 years and cost millions or even a billion dollars, the 505(b)(2) pathway offers a faster and less expensive route to market.
Key differences in the product planning phase include:
- Leveraging existing data: Look for ways to incorporate data from the public domain and the FDA's previous findings to reduce the size, scope, timeline, and cost of development.
- Exclusivity potential: Products approved under 505(b)(2) may qualify for various types of market exclusivity:
- Orphan drug exclusivity (7 years)
- New chemical entity exclusivity (5 years)
- "Other" exclusivity (3 years for a "change" if certain criteria are met)
- Pediatric exclusivity (6 months added to existing patents/exclusivity)
4. The Pre-IND Phase
The pre-IND phase for a 505(b)(2) product differs significantly from that of a 505(b)(1) product:
- Order of steps: The process begins with the pre-IND meeting, followed by formulation development and studies (if necessary), and then the IND filing.
- Goals of the pre-IND meeting: The objective is to gain FDA input and concurrence on the proposed studies, CMC strategy, and clinical research plans to minimize the required new studies.
- Number and type of studies: Some development programs may conduct bridging studies that preclude the need for extensive nonclinical or clinical studies.
- Timing of CMC work: Clinical trial materials for Phase I studies must be representative of the commercial manufacturing process, including packaging. The three stability batches for shelf-life determinations are often prepared at this time.
- Timing of studies: 505(b)(2) development plans often allow for simultaneous and parallel clinical studies, significantly shortening the overall time to market.
Initiating the pre-IND meeting with the FDA is a pivotal step in the 505(b)(2) pathway, distinguished from the 505(b)(1) process in several ways, including the order of steps and the extent of required studies.
This meeting aims to secure FDA input and agreement on the planned studies and strategies, streamlining the approval process and enhancing investor attractiveness. Using public or previous FDA data can also reduce the need for additional studies, accelerating the timeline and reducing costs.
Let's dig into the details of the pre-IND meeting in a bit more clarifying detail.
Goals of the pre-IND meeting
The pre-IND meeting is a crucial step in the 505(b)(2) pathway, serving several important purposes:
- Initiate the regulatory process: This meeting marks the formal beginning of the 505(b)(2) process, setting the stage for subsequent development steps.
- Secure FDA guidance: Obtain input and agreement from the FDA on planned studies, development strategies, and data requirements specific to the 505(b)(2) pathway.
- Discuss existing data utilization: Explore how to leverage public data or previous FDA findings, a key advantage of the 505(b)(2) pathway.
- Outline study requirements: Determine which studies are necessary, often fewer than in the 505(b)(1) pathway, to demonstrate safety and efficacy.
- Plan formulation development: Discuss the approach to formulation, considering how it may differ from the reference listed drug.
- Address regulatory strategy: Develop a clear regulatory strategy that aligns with 505(b)(2) requirements and optimizes the approval timeline.
- Enhance investor appeal: By clarifying the development path and potential time and cost savings, the meeting can make the project more attractive to investors.
- Identify potential challenges: Proactively discuss any unique aspects of the drug candidate that may require special consideration in the 505(b)(2) context.
Chronology of actions and steps
The 505(b)(2) process begins with the pre-IND meeting, unlike the 505(b)(1), which starts with formulation development. It then proceeds to formulation development and any necessary additional studies. The process concludes with the IND filing.
Required studies
Utilization of public data or previous FDA findings is a hallmark of the 505(b)(2) pathway. This allows for bridging studies, reducing the need for extensive clinical or nonclinical studies, a requirement in the 505(b)(1) pathway.
CMC timing
For Phase 1 studies, clinical trial materials should mirror the commercial manufacturing process, including packaging, in the 505(b)(2) pathway. Preparation of three stability batches for shelf-life determination is required early on, necessitating significant CMC work before study initiation. Contact us for specialized CMC or other consulting support.
Study timing
The 505(b)(2) pathway’s reliance on existing data enables simultaneous initiation and parallel development of clinical studies. It’s possible to commence a Phase 3 study before completing all Phase 1 studies and without a Phase 2 study, leading to cost and time savings.
Pre-IND meetings: A few keys to successPre-IND meetings are crucial for 505(b)(2) programs. Here are some tips for a successful meeting:
|
Qualifying for the 505(b)(2) Pathway
Again, the 505(b)(2) pathway allows applicants to leverage existing public literature or the FDA’s prior safety and effectiveness findings for an approved drug. It often applies to changes in already approved drugs or the introduction of an NCE without a right of reference to previous studies.
Key categories include:
- New Chemical Entities: When relying on published studies that the applicant can’t reference directly.
- Modifications to Approved Drugs: When the new product is a variation of an approved drug and integrates the safety and efficacy data of the original, complemented by additional information supporting the modification. (Most 505(b)(2) NDAs fit this second category, including minor alterations not classifiable as a generic drug or extensive modifications that still refer to the original drug’s safety or efficacy data.)
Some examples of qualifications include:
- Changes in dosage form, strength, or formulation.
- Alterations in the route of administration, dosing regimen, or indication.
- A combination of the above changes.
Generally speaking, a product with such changes qualifies for the 505(b)(2) pathway if it establishes a scientific bridge to the approved “listed drug,” especially when the applicant lacks a right of reference to the original approval data.
A Few 505(b)(2) Program Development Best Practices
Navigating the 505(b)(2) development pathway can be intricate, yet it unveils significant opportunities for sponsors aiming to expedite the drug approval process. The initial step involves meticulously comparing the new and innovator drugs, focusing intensely on their PK attributes.
To harness the full potential of the 505(b)(2) approach, a well-crafted "PK bridge" is instrumental. This bridge is a comprehensive compilation of data, drawing parallels between the in vivo performance of the new drug and the innovator drug. This strategy's pivotal component is understanding and outlining these parallels, ensuring that the development and approval process is streamlined and cost-effective.
Specifically, a well-rounded PK development plan for oral products that encompasses studies on bioequivalence, bioavailability, multiple-dose PK, and food effect can substantially satisfy the minimum PK requisites. This strategic focus propels the regulatory approval journey and curtails associated costs, optimizing the value proposition of the 505(b)(2) development program.
Below are a few other 505(b)(2) program development best practices we've developed through regulatory support engagements.
1. Conduct a comprehensive literature review and gap analysis before finalizing your development strategy.Many sponsors underestimate the importance of a thorough literature review. A strategic review can uncover valuable data that may reduce the need for certain studies, saving time and resources. It can also identify potential pitfalls or challenges that might not be immediately apparent. We recommend:
|
2. Involve commercialization experts from the very beginning of program development.While regulatory success is crucial, commercial viability determines the ultimate success of a product. Early input from commercial experts can shape the development program to ensure that the final product meets market needs and has a clear path to profitability. We recommend:
|
3. Develop a comprehensive patent strategy in parallel with your regulatory strategy.The 505(b)(2) pathway often involves products that improve or modify existing drugs. A strong patent strategy can provide additional protection and market exclusivity beyond what regulatory exclusivity offers. We recommend:
|
4. Develop a robust bridging strategy that goes beyond just pharmacokinetics.While, as we mentioned above, pharmacokinetic bridging is often a key component of 505(b)(2) programs, a comprehensive bridging strategy should also consider other factors such as pharmacodynamics, safety, and efficacy. We recommend:
|
5. Strategically utilize all available FDA meeting types, not just the pre-IND meeting.While the pre-IND meeting is crucial, other meeting types (e.g., Type C meetings, end-of-Phase 2 meetings) can provide valuable opportunities to align with the FDA throughout development. We recommend:
|
The Critical Role of Regulatory Consultants in the 505(b)(2) Process
Navigating the 505(b)(2) pathway can be complex and challenging, even for experienced pharmaceutical companies. This is where regulatory consultants play a crucial role, offering expertise and guidance throughout the process.
Let's explore how these professionals add value at various stages of 505(b)(2) drug development.
Initial assessment and strategy developmentRegulatory consultants begin by evaluating the suitability of a product for the 505(b)(2) pathway. They conduct thorough assessments of the proposed product, considering its scientific, medical, and commercial viability. Their expertise allows them to identify potential reference listed drugs (RLDs) and evaluate the extent to which existing data can be leveraged. Based on this analysis, consultants develop a tailored regulatory strategy that aligns with the sponsor's goals and resources, setting the foundation for a successful development program. Gap analysis and development planningOnce the initial strategy is in place, regulatory consultants perform comprehensive literature reviews and gap analyses. This crucial step helps identify what data already exists and what additional studies may be needed. Consultants then recommend specific studies to bridge these data gaps efficiently, helping to optimize the development timeline and budget. Their experience allows them to prioritize development activities, ensuring that resources are allocated effectively and that the development plan is both comprehensive and efficient. Liasing FDA interactionsRegulatory consultants are instrumental in managing interactions with the FDA. They develop clear, concise meeting packages that communicate the sponsor's position and prepare sponsors for FDA meetings by anticipating potential questions and concerns. Consultants often attend these meetings to provide expert support and clarification. Following the meetings, they help interpret FDA feedback and adjust development plans accordingly. This expertise in FDA communications can significantly smooth the regulatory process and avoid potential delays or misunderstandings. CMC strategy and implementationCMC is a critical component of any drug application. Regulatory consultants advise on formulation development strategies and help establish appropriate specifications for drug substances and products. They ensure that CMC activities align with regulatory expectations and Good Manufacturing Practice (GMP) requirements. This expertise can be particularly valuable in 505(b)(2) applications, where formulation changes or new delivery methods may be key differentiators for the product. NDA preparation and submissionWhen it comes time to prepare and submit the NDA, regulatory consultants play a crucial role in managing the process. They coordinate the compilation of all necessary data and documents, ensuring the NDA is complete, well-organized, and compliant with FDA requirements. Consultants develop a clear and compelling narrative throughout the application, effectively presenting the product's story to regulators. Their attention to detail and understanding of FDA expectations can significantly enhance the quality of the submission. Post-submission supportAfter the NDA is submitted, regulatory consultants continue to provide valuable support. They manage FDA interactions during the application review, responding to information requests and clarifying questions promptly and effectively. If required, consultants prepare sponsors for potential advisory committee meetings, helping to develop presentations and anticipate potential questions. They may also assist in developing post-approval commitments if needed, ensuring a smooth path to approval and market launch. Special considerationsRegulatory consultants provide expertise on special topics that may arise in 505(b)(2) development. This could include guidance on orphan drug designation, pediatric studies, or combination products. They assist with patent certifications and exclusivity considerations, navigating the complex intersection of patent law and FDA regulations. Consultants also help resolve regulatory challenges unique to 505(b)(2) products, drawing on their deep understanding of this pathway and experience with similar products. |
When to Engage an Expert Regulatory Consultant with 505(b)(2) Experience
Firms should seek a 505(b)(2) regulatory consultant when internal expertise, resources, or experience is limited.
As we've demonstrated time and again, consultants can be instrumental in complex developments, regulatory challenges, risk mitigation, strategic planning, FDA engagement, and post-approval support. Their role is pivotal in optimizing the submission process, reducing errors, and ensuring timely approval.
Below are a few key questions to consider that all speak to the need for a skilled regulatory consultant. Contact us to start the conversation.
8 questions to consider for 505(b)(2) consulting support
|
Contact The FDA Group for Expert 505(b)(2) Regulatory Support
The FDA Group offers exclusive access to a large staff of seasoned senior scientists and regulatory professionals proficient in both the strategic and operational facets of 505(b)(2) development programs. We are adept at combining regulatory insights with a broad spectrum of expertise in scientific, clinical, and nonclinical domains, ensuring a customized and effective pathway for each 505(b)(2) initiative.
Our track record includes active participation in numerous 505(b)(2) programs. We pride ourselves on offering comprehensive support, as exemplified in a case where we facilitated an end-to-end 505(b)(2) submission. Our role extended to managing FDA interactions and serving as the primary contact for regulatory affairs, ensuring seamless communication and coordination.
Reach out to us to explore how we can help you bring your product to market and keep it there.
International Auditing Project with Worldwide Consultant Resourcing
Learn how The FDA Group partnered with a healthcare product Company to support a series of international Quality Audits to assess both Good Manufacturing Practices (GMP) and Good Clinical Practices (GCP) throughout the United States, Europe, Asia, and the Middle East.