How many FDA approved Single-chain FV antibody fragment are there?

17 March 2025

Introduction to Single‐chain FV Antibody Fragments

Single‐chain FV antibody fragments, more commonly referred to as single‐chain variable fragments (scFvs), are recombinant antibody derivatives that consist solely of the variable regions of the heavy (VH) and light (VL) chains connected by a short flexible peptide linker. This design results in a molecule typically weighing around 25–30 kDa, making it far smaller than conventional monoclonal antibodies (mAbs) that also contain the constant regions. The flexibility and compactness of this structure facilitate proper antigen recognition despite the absence of the Fc (crystallizable) region, which is normally responsible for effector functions in full antibodies. The genetic construction of scFvs generally involves techniques such as RT-PCR amplification of the VH and VL regions from B-cell mRNA followed by the assembly of these sequences using linker sequences (e.g., (GGGGS)_3), which not only maintains conformational proximity but also preserves the binding activity of the native antibody. Furthermore, many studies have demonstrated that the proper interdomain orientation and the length of the linker directly influence the stability and solubility of the scFv.

Advantages over Traditional Antibodies 
The hallmark advantage of scFvs is their dramatically reduced size, which enables enhanced tissue penetration and potentially improved pharmacokinetics, especially in dense tissues such as solid tumors. Due to their small molecular size, scFvs can be produced in microbial systems like Escherichia coli, resulting in significantly reduced manufacturing costs and production times when compared with full-length antibodies that require mammalian cell expression systems. In addition, the absence of an Fc region tends to reduce the risk of triggering Fc-mediated adverse effects such as antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). This inherent feature not only minimizes off-target immune activation but also makes scFvs particularly suitable as targeting vehicles or for applications that require rapid clearance from the bloodstream. Another substantial benefit is the ease of genetic manipulation. Researchers can readily fuse scFvs with other molecules such as toxins, drugs, enzymes, or imaging agents to create multifunctional conjugates and bispecific constructs, thereby broadening their clinical and diagnostic applications. Despite their many advantages, challenges do exist—such as stability during storage and the tendency to aggregate—but advances in protein engineering are continually addressing these issues.

FDA Approval Process for Biologics

Overview of FDA Approval Stages 
The pathway to FDA approval for biologics is rigorous and typically involves several key stages. Initially, a new candidate undergoes preclinical development that encompasses both in vitro and in vivo studies. These studies assess pharmacodynamics, pharmacokinetics, toxicity, and the safety profile of the molecule in relevant animal models. Once sufficient preclinical data have been generated, sponsors submit an Investigational New Drug (IND) application to the FDA to seek approval for initiating human clinical trials. 
Clinical development is structured in multiple phases:
- Phase I focuses on evaluating the safety, tolerability, and pharmacokinetics in a small group of healthy volunteers or patients.
- Phase II expands on initial findings and begins mapping the therapeutic dose range while also providing preliminary data on efficacy.
- Phase III involves large-scale studies designed to generate robust evidence on safety and clinical efficacy under controlled conditions relative to standard treatments or placebo. 
After successful completion of these phases, a New Drug Application (NDA) is submitted for a final FDA review. The application includes comprehensive preclinical and clinical data, manufacturing details, and proposed labeling information. The FDA then reviews this body of evidence with a focus on whether the benefits outweigh any associated risks. For biologics, additional emphasis is placed on demonstrating consistency in manufacturing and ensuring that the production process reliably yields a product with the intended physicochemical and functional characteristics.

Specific Requirements for Antibody Fragments 
Antibody fragments such as scFvs have additional considerations in the FDA approval process due to their unique properties and manufacturing processes. Unlike full-length antibodies, scFvs lack the Fc region, which necessitates a focus on demonstrating adequate in vivo stability, proper folding, binding affinity, and specificity through robust analytical and functional assays. 
Specific requirements for scFvs include:
- Analytical Characterization: In-depth characterization is required to ensure that the produced scFv maintains its antigen-binding activity, proper tertiary structure, and minimal aggregation. Techniques such as size-exclusion chromatography, dynamic light scattering, and surface plasmon resonance are often employed in these evaluations. 
- Comparative Studies: Because scFvs are designed to mimic the antigen-binding capabilities of full-length antibodies, comparative studies against a reference molecule (if available) are crucial. This typically means bioequivalence studies that examine both the binding kinetics and the functional inhibition or activation of target molecules. 
- Stability and Storage: Given their smaller size and lack of stabilizing constant regions, additional stability studies are conducted to guarantee that the scFv retains its functional integrity over its proposed shelf life. 
- Manufacturing Consistency: The production of scFvs in microbial systems, while cost-effective, introduces variability related to post-translational modification and folding. The FDA requires rigorous process validation to confirm that each production batch meets stringent quality control parameters.

Current FDA Approved Single‐chain FV Antibody Fragments

List of Approved Products 
When considering the landscape of FDA-approved antibody therapeutics, it is critical to differentiate between various antibody formats. As of the current state of regulatory approvals, only one single-chain FV antibody fragment has achieved FDA approval for therapeutic use. That product is brolucizumab. Marketed under the trade name Beovu, brolucizumab represents the first and, to date, the only FDA-approved scFv molecule. The approval of brolucizumab was based on comprehensive clinical studies that demonstrated its efficacy and safety in the management of neovascular (wet) age-related macular degeneration (AMD)
Brolucizumab's development and subsequent FDA approval highlight a significant milestone in the field of antibody fragment therapeutics. The drug application details for brolucizumab indicate that it was submitted by Novartis Pharmaceuticals Corp., and successfully cleared the FDA review under the Center for Drug Evaluation and Research (CDER). Its unique molecular design as an scFv enables it to achieve high molar doses compared with larger antibody formats, thereby enabling improved targeting and tissue penetration in the intraocular environment.

Indications and Applications 
Brolucizumab (Beovu) is indicated for the treatment of wet AMD—a condition characterized by abnormal blood vessel growth (neovascularization) and leakage beneath the retina, leading to potentially severe vision loss. The therapeutic rationale behind using a single-chain FV antibody fragment for wet AMD is primarily based on its small size, which permits better retinal tissue penetration and rapid binding to vascular endothelial growth factor A (VEGF-A), a key driver of neovascularization. Clinical studies have demonstrated that brolucizumab leads to significant improvements in visual acuity and reductions in retinal fluid accumulation—with dosing intervals that benefit patients by potentially reducing the frequency of intravitreal injections. 
The clinical outcomes observed with brolucizumab underscore several application benefits:
- Enhanced Exposure at the Target Site: The small molecular size of brolucizumab enables it to achieve higher molar dosing, increasing its bioavailability in ocular tissues compared with larger antibody fragments. 
- Rapid Clearance: Although rapid clearance is generally considered a potential drawback for antibody fragments, in the context of intraocular administration for AMD, this feature minimizes systemic exposure and adverse events. 
- Efficacy in Recurrence: Clinical trial data revealed that many patients maintained fluid-free retinal profiles for extended periods after treatment, suggesting long-term benefits in reducing disease recurrence. 
Overall, the success of brolucizumab has set a precedent for the clinical translation of scFv technology and highlights the opportunities for further development of similar antibody fragment-based therapeutics in other indications.

Challenges and Future Directions

Current Challenges in Development 
Despite the successful FDA approval of brolucizumab as a single-chain FV antibody fragment, several challenges remain in the development and clinical adoption of scFv molecules. One of the primary challenges is the inherent instability of these small proteins. In the absence of the stabilizing Fc region, scFvs can be prone to aggregation, misfolding, or rapid degradation in vivo, all of which can compromise therapeutic efficacy. Although brolucizumab was optimized to overcome many of these hurdles, researchers continue to work towards improved designs and formulations to enhance the stability and shelf life of scFvs. 
Another significant challenge is ensuring manufacturing consistency. The microbial expression systems commonly used for scFv production are subject to variations in protein folding and post-translational modifications, which can lead to batch-to-batch variability. This necessitates rigorous quality control measures and process validations to guarantee product homogeneity and safety. 
Furthermore, while the lack of Fc-mediated effector functions minimizes some adverse immune reactions, it also eliminates beneficial functions like ADCC or CDC, which may be desirable in certain therapeutic applications. Thus, there is an ongoing debate and challenge in the field regarding the balance between minimizing immunogenicity and harnessing the immune system for therapeutic benefit. 
Additionally, there is limited clinical experience with scFv-based therapeutics outside the ocular space. The immunogenicity profile, pharmacokinetics, and potential off-target effects of scFvs in systemic applications remain areas where deeper understanding is required. These challenges underscore the need for continued preclinical and clinical research to optimize these molecules for broader therapeutic applications.

Future Prospects and Research Directions 
Looking ahead, the outlook for scFv technology is promising. The FDA approval of brolucizumab has demonstrated that scFv molecules can meet the necessary efficacy and safety standards when properly engineered. Future research is likely to focus on several key areas:
- Engineering for Enhanced Stability: Advances in protein engineering, such as the introduction of novel linker sequences, stabilization mutations, and fusion of scFvs to smaller constant-like domains, are expected to improve the long-term stability and solubility of these molecules. 
- Multispecific and Biparatopic Constructs: Researchers are exploring the design of multispecific antibody fragments that can simultaneously target multiple epitopes. The modular nature of scFvs makes them excellent building blocks for creating bispecific or even trispecific constructs, which could synergistically improve therapeutic outcomes, particularly in oncology and immunotherapy. 
- Improved Manufacturing Techniques: The development of robust microbial expression platforms and refined purification methods could further reduce production costs while ensuring batch-to-batch reproducibility. Automation and high-throughput screening techniques are also being integrated into the manufacturing workflow to expedite the optimization process. 
- Novel Therapeutic Indications: With the continued success of brolucizumab in ocular diseases, there is growing interest in expanding the application of scFvs to other therapeutic areas such as oncology, infectious diseases, and autoimmune disorders. Moreover, the ability to genetically fuse scFvs with other functional domains (e.g., toxins, enzymes, or imaging agents) opens the door to a diverse range of bioconjugates and drug-delivery systems. 
- In Vivo Half-life Extension Strategies: One current limitation of scFvs is their short circulatory half-life, which can lead to rapid clearance. Novel strategies including PEGylation, fusion with albumin-binding domains, or other half-life extension technologies are under active investigation. These modifications aim to prolong therapeutic exposure without compromising the favorable tissue penetration and rapid target engagement associated with scFvs. 
- Regulatory Framework Evolution: As more scFv-based therapeutics progress through clinical trials, lessons learned from the brolucizumab approval process will help refine regulatory guidelines tailored specifically for antibody fragments. This evolution will further streamline the path to approval for future scFv products and might include more flexible criteria regarding immunogenicity, potency, and manufacturing consistency. 
Overall, the combination of technological advances, improved molecular design, and an evolving regulatory landscape promises to further enhance the development and clinical utility of single-chain FV antibody fragments beyond the current sole example of brolucizumab.

Conclusion 
In summary, the current state of FDA-approved single-chain FV antibody fragment therapeutics is represented by a single product: brolucizumab (marketed as Beovu). This milestone achievement demonstrates that scFvs can be engineered to meet complex regulatory requirements, exhibit robust antigen-binding activity, and provide clear clinical benefits, particularly in the treatment of neovascular (wet) age-related macular degeneration. 

The journey from discovery to FDA approval involves an intricate process characterized by multiple preclinical and clinical stages. For scFvs, the challenges of molecular stability, manufacturing consistency, and the balancing act between minimizing immunogenic effects while retaining therapeutic potency are central to both their development and optimization. Despite these hurdles, the inherent advantages of scFvs—including their small size, enhanced tissue penetration, and ease of genetic manipulation—provide a solid foundation for future advances. Consequently, ongoing research is focused on improving stability, expanding biological functions through multispecific constructs, and extending the circulatory half-life of these promising molecules. 

From a broader perspective, the approval of brolucizumab opens the door for further exploration and expansion of scFv-based therapeutics into other disease areas. Innovations in protein engineering, coupled with advanced expression and purification methodologies, stand to reduce production costs and improve therapeutic efficacy. In addition, evolving regulatory frameworks that incorporate the growing body of experience with antibody fragments promise to streamline the approval process for future products. 

In conclusion, while brolucizumab remains the only FDA-approved single-chain FV antibody fragment to date, its success not only validates the therapeutic potential of scFvs but also sets the stage for an exciting era of antibody engineering that could revolutionize the treatment of a range of challenging diseases. The continued integration of scientific innovation, rigorous regulatory oversight, and clinical research is likely to yield a new generation of scFv therapeutics with improved properties and broader applications, thereby expanding the impact of antibody-based therapies well beyond the current landscape.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

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

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

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