ad image
Therapeutic Nanobodies and Intrabodies Rewriting the Rules of Target Engagement

Therapeutic Nanobodies and Intrabodies Rewriting the Rules of Target Engagement

May 27, 2025PAO-05-25-NI-10

From camelid-derived nanobodies to cell-penetrating intrabodies, antibody fragments are redefining the boundaries of therapeutic targetability. Their compact size, modularity, and intracellular reach enable interventions once considered out of reach — from neutralizing protein aggregates in neurodegeneration to disrupting viral replication from within. While nanobodies have already made inroads in diagnostics and immunotherapy, the clinical trajectory of intrabodies is just beginning.. As delivery and design technologies mature, nanobodies and intrabodies may emerge as central tools in precision medicine, intracellular immunotherapy, and real-time biosensing.

Introduction: A New Frontier in Antibody Engineering

For decades, monoclonal antibodies (mAbs) have formed the backbone of biologic therapeutics, revolutionizing the treatment of cancer, autoimmune diseases, and infectious conditions. Their high specificity, well-characterized pharmacology, and modular design have made them indispensable in modern medicine. Yet despite their success, traditional mAbs have inherent limitations. Their large molecular size (~150 kDa) restricts tissue penetration and access to certain epitopes, particularly those buried within the cell or located in tightly packed extracellular matrices. Moreover, mAbs are generally ineffective against intracellular targets — proteins that contribute to disease from within the cytosol or nucleus — due to their inability to cross cellular membranes. Their production in mammalian cell systems is also complex and costly, contributing to high therapeutic prices and variable global access.

To address these challenges, researchers have turned to smaller, more versatile antibody fragments. Among the most promising are nanobodies and intrabodies — distinct but complementary formats designed to extend the reach of antibody-based therapies. Nanobodies are derived from the variable regions of heavy-chain-only antibodies found in camelids and cartilaginous fish. These naturally occurring fragments, typically around 15 kDa in size, retain high antigen-binding affinity while exhibiting exceptional thermal and chemical stability. Their compact structure allows them to access conformational epitopes inaccessible to larger antibodies, with the added benefits of simpler expression systems and improved tissue penetration.1,2

Intrabodies, by contrast, are engineered antibody fragments — often derived from single-chain variable fragments (scFvs) or nanobody scaffolds — designed to function within the intracellular environment. They are typically expressed as genes inside the target cell, where they are translated and folded in the cytosol or other organelles. This intracellular orientation enables direct interference with pathogenic proteins involved in neurodegenerative diseases, viral replication, or dysregulated signaling pathways.3,4 Unlike nanobodies, which are often administered as extracellular proteins, intrabodies must overcome challenges related to stability in the reducing cytoplasmic environment and efficient intracellular delivery.

Both formats rely on the modular structure of antibody variable domains — VHHs in nanobodies and VH-VL linkers in scFvs — to mediate high-affinity target binding. While their structural simplicity makes them attractive from a manufacturing standpoint, it is their ability to access difficult targets — whether deeply buried in a tumor microenvironment or hidden inside the cell — that sets them apart. As innovation continues to reshape the therapeutic landscape, nanobodies and intrabodies offer a glimpse into the next generation of biologics: compact, customizable, and capable of targeting what was once considered undruggable.

Nanobodies: Compact and Capable

Origins and Evolution

The discovery of nanobodies traces back to the early 1990s, when researchers identified a unique class of heavy-chain-only antibodies circulating in camelids. Unlike conventional antibodies, which consist of two heavy and two light chains, these molecules lack light chains entirely and rely on a single variable domain — VHH — for antigen recognition. This structural simplicity gave rise to the concept of nanobodies: the isolated VHH domains that retain full binding capability while being only a fraction of the size of traditional mAbs.2

Structurally, nanobodies differ significantly from mAbs in both scale and composition. While mAbs weigh approximately 150 kDa and feature multiple domains requiring complex folding and assembly, nanobodies are around 15 kDa and consist of a single, compact immunoglobulin fold.5 This streamlined format contributes to greater thermal and chemical stability, as well as reduced immunogenicity when properly humanized. Initially, nanobodies found broad use in basic research and imaging, where their small size enabled precise labeling of targets in fixed cells, tissues, and in vivo models. Their high affinity and specificity, combined with ease of recombinant production, soon made them indispensable in diagnostic development as well.1,6

Key Properties

One of the most notable advantages of nanobodies is their exceptional biochemical robustness. They maintain solubility and binding activity across a wide range of temperatures and pH levels, making them well suited for industrial and therapeutic applications requiring harsh conditions or long shelf life.2,5 Their single-domain nature also reduces the risk of aggregation, a common issue in biologic formulations.

Beyond stability, nanobodies offer unique access to epitopes that are poorly accessible to bulkier antibodies. Their slender, convex paratopes enable them to wedge into enzyme active sites, receptor crevices, and viral envelope clefts that would otherwise be occluded. This quality makes them especially valuable in neutralizing conformationally complex or cryptic targets, such as intracellular signaling domains or viral fusion interfaces.7

Additionally, nanobodies are highly amenable to engineering. Their simple structure facilitates genetic fusion to other biologic domains, enzymes, toxins, or carrier systems. They can be multimerized, bispecific, or fused to Fc domains to extend half-life. Their coding sequences are short, allowing expression in microbial systems, viral vectors, or mRNA constructs.8 This modularity opens the door to bespoke designs for targeted therapy, imaging, or intracellular delivery.

Clinical Applications

Nanobodies have advanced from preclinical curiosities to serious therapeutic candidates across a range of disease areas. In oncology, they are being explored for both direct tumor targeting and as components of multifunctional therapies. Nanobody constructs targeting HER2 and EGFR have demonstrated promising activity in preclinical and early clinical settings, particularly when fused with cytotoxic domains or incorporated into CAR-T designs to improve tumor infiltration and specificity.8

In the autoimmune space, nanobodies have shown potential to modulate cytokine signaling, disrupt proinflammatory protein–protein interactions, or deplete autoreactive cell populations in animal models.9 Their small size and rapid tissue penetration make them especially attractive for applications where large antibodies might fail to reach pathogenic cells or mediators.

Perhaps the most dramatic demonstration of nanobody potential came during the COVID-19 pandemic, when they were rapidly developed to neutralize SARS-CoV-2. Their rapid manufacturability, stability under ambient conditions, and ability to bind conserved regions of the viral spike protein made them appealing candidates for low-cost, scalable antiviral therapeutics.10

Finally, nanobodies continue to play a critical role in imaging and diagnostics. Their ability to rapidly localize to tissues and clear from circulation improves contrast in positron emission tomography (PET) and single-photon emission computed tomographyt (SPECT) imaging, and their ease of conjugation to reporter enzymes or fluorophores facilitates use in biosensors and in vitro assays.6

Altogether, nanobodies exemplify the potential of rational antibody miniaturization: preserving the core advantages of mAbs while extending their reach into previously inaccessible biological terrain.

Intrabodies: Therapeutic Tools for Intracellular Targets

Concept and Rationale

Intrabodies are engineered antibody fragments designed to function within the intracellular environment, expanding the reach of biologics beyond cell surface receptors and extracellular ligands. Unlike conventional antibodies, which are too large and structurally dependent on disulfide bonds to remain stable or active inside cells, intrabodies are specifically selected or modified to fold and function in the reducing conditions of the cytoplasm, nucleus, or other subcellular compartments.3

This sets them apart from nanobodies, which, although structurally simple and stable, are most commonly used in extracellular applications or imaging contexts. While nanobodies can occasionally be engineered for intracellular expression, intrabodies are defined by their intended intracellular action and are typically optimized for this function from the outset.

The rationale behind developing intrabodies lies in the therapeutic importance of intracellular targets. Many of the most intractable diseases — neurodegenerative disorders, certain viral infections, and some cancers — are driven by intracellular protein misfolding, toxic aggregates, or dysregulated signaling pathways. These targets remain largely inaccessible to traditional small molecules, which often lack specificity, and to mAbs, which cannot cross the plasma membrane. Intrabodies offer a route to bind, block, or degrade these pathogenic proteins directly at their source.11,12

Mechanisms of Action

Intrabodies can be designed to act through several distinct mechanisms. In their simplest form, they bind to target proteins and block interactions or conformational changes critical to disease processes. For example, intrabodies targeting misfolded alpha-synuclein or tau can prevent aggregation or inhibit their seeding activity, disrupting the pathological spread seen in Parkinson’s and Alzheimer’s diseases.13

Another increasingly popular approach involves fusing intrabodies with degron tags or E3 ligase–recruiting domains, redirecting the bound protein to the proteasome for selective degradation. This targeted protein knockdown strategy is particularly effective for neutralizing toxic gain-of-function mutations or aberrantly active intracellular enzymes.4,14

These mechanisms have been demonstrated across a range of model systems. In neurodegenerative disease, intrabodies have successfully reduced intracellular levels of tau, alpha-synuclein, and huntingtin protein, improving motor function and neuroprotection in animal models.11,12 In infectious disease, intrabodies have shown the ability to interfere with viral protein assembly, processing, or trafficking, blocking replication from within infected cells.10

Intrabody Formats

Intrabodies are typically derived from single-chain variable fragments (scFvs), which consist of the variable heavy and light domains of antibodies connected by a short peptide linker. This format allows for high-affinity binding while maintaining a size and structure conducive to cytosolic expression. More recently, nanobody-based intrabodies have gained popularity due to their inherent stability and lack of light chain dependence, which simplifies folding in the intracellular environment.13

Advanced formats include degron-tagged intrabodies for inducible degradation, as well as tandem constructs that combine binding, signaling, or trafficking functions. The development of large synthetic antibody libraries optimized for cytoplasmic folding has further accelerated the discovery of functional intrabodies. Intracellular screening platforms using yeast or mammalian display systems — are enabling high-throughput selection of binders that maintain functionality under physiological intracellular conditions.15

Together, these formats reflect the growing sophistication of intrabody engineering and the diversity of tools now available to tackle intracellular dysfunction. As expression systems and delivery methods improve, the versatility of intrabodies is likely to rival that of traditional antibodies — bringing the precision of immunotherapy to targets that were, until recently, beyond the reach of biologics.

Technical and Translational Challenges

Despite the remarkable promise of nanobodies and intrabodies, their clinical translation faces several significant technical hurdles, spanning molecular design, delivery strategies, target validation, and immunological considerations. Addressing these barriers is critical to unlocking the full therapeutic potential of these compact antibody fragments.

Stability and Folding in the Cytoplasm

A primary obstacle for intrabodies is the biochemical nature of the intracellular environment. Unlike the oxidizing extracellular space or endoplasmic reticulum, the cytoplasm is a reducing environment that disrupts disulfide bond formation — bonds that are essential for the structural integrity of many antibody domains. Most conventional antibody fragments misfold or aggregate under these conditions, rendering them inactive.3

To overcome this, researchers have employed protein engineering strategies to develop intrabody scaffolds that are inherently disulfide-independent or stabilized by alternative interactions. Humanization efforts have reduced immunogenicity while improving compatibility with host cell machinery. Thermostabilization — through point mutations that enhance hydrophobic packing or hydrogen bonding — has further improved folding efficiency and solubility of intrabody constructs expressed within the cell.5 These optimizations are essential not only for maintaining binding activity but also for preventing unwanted proteotoxic effects due to intracellular aggregation.

Delivery and Expression

Efficient delivery remains a major translational bottleneck, particularly for intrabodies. Because they must act inside the cell, direct administration of protein-based formats is often impractical. Instead, gene delivery systems are typically used to express intrabodies endogenously within target cells. Viral vectors, such as adeno-associated virus (AAV) and lentivirus, are frequently employed owing to their high transduction efficiency and tissue tropism. These approaches have shown promise in preclinical models of neurodegenerative disease and cancer, but raise safety, dosing, and durability concerns for broader clinical use.

Emerging strategies include the delivery of intrabody-encoding mRNA, which offers transient expression without the risks associated with genomic integration. mRNA formulations can be rapidly updated and delivered using lipid nanoparticles, similar to COVID-19 vaccines, making this approach attractive for personalized or time-sensitive applications.15 Additionally, direct delivery of recombinant intrabodies using cell-penetrating peptides or nanocarriers has been explored, though these techniques face challenges related to endosomal escape and target cell specificity.5

Target Specificity and Off-Target Effects

As with all antibody-based therapeutics, ensuring target specificity is crucial to minimize off-target effects and maximize therapeutic efficacy. For intrabodies, which often act on complex intracellular protein–protein interactions or aggregates, this challenge is particularly acute. Advanced screening platforms, including yeast or mammalian cell display, allow for the selection of binders with high affinity and precision. Cell-based functional assays are increasingly used to validate activity in physiologically relevant contexts.6,15

Nonetheless, some intrabody designs risk unintended consequences. Misfolded or overexpressed intrabodies can form aggregates themselves, potentially disrupting proteostasis or activating stress responses. In neurodegenerative models, for instance, intrabodies targeting misfolded proteins have shown occasional exacerbation of proteotoxicity, depending on expression levels and degradation pathways involved.11 These findings underscore the importance of careful dose optimization, degradation control mechanisms, and thorough preclinical validation.

Immunogenicity and Pharmacokinetics

Immunogenicity is an ever-present concern in biologic development. Although nanobodies are smaller and less complex than full-length mAbs, they are still derived from camelid or synthetic scaffolds, which may be recognized as foreign by the human immune system. Humanization strategies — modifying framework residues to resemble human antibody germline sequences — can mitigate this risk. Fusion to human Fc or albumin-binding domains also helps extend serum half-life and reduce immune detection, particularly in therapeutic applications requiring systemic exposure.8

For intrabodies, the immune response may be shaped not only by the scaffold itself but by the mode of expression. Gene-delivered constructs, especially those introduced via viral vectors, may elicit immune recognition of the encoded protein or the delivery vehicle. While this is less of a concern for localized or short-term applications, systemic delivery or chronic diseases could require additional strategies to suppress immune activation or enhance tolerability.3

Together, these challenges reflect the complexity of translating nanobody and intrabody technologies from the bench to the clinic. However, ongoing advances in protein design, delivery science, and immune modulation continue to reduce these barriers, laying the groundwork for broader therapeutic deployment.

Innovation at the Interface: Synthetic Biology and Next-Gen Tools

The convergence of nanobody and intrabody technologies with synthetic biology is rapidly expanding the functional and therapeutic landscape of these compact antibody fragments. No longer confined to static binding or blocking roles, nanobodies and intrabodies are now being integrated into programmable, dynamic systems capable of sensing, responding to, and even rewriting cellular behavior. These innovations are paving the way for next-generation biologics that operate not only with high specificity, but with logic, tunability, and precision control.

One of the most compelling areas of development is the use of nanobody-based biosensors. By fusing nanobodies to fluorescent proteins or enzymatic reporters, researchers have created intracellular sensors capable of visualizing real-time changes in protein conformation, localization, or concentration. These tools have proven invaluable for studying transient signaling events and spatially restricted protein dynamics within live cells.6 More recently, whole-cell screening platforms have employed synthetic libraries of nanobodies to identify binders that respond to specific disease states, enabling the creation of diagnostic systems that activate only in the presence of pathogenic conformers or signaling cues.15

Regulated expression systems further extend the utility of nanobodies and intrabodies by introducing temporal and environmental control. Advances in optogenetics and synthetic promoters have yielded constructs that activate or deactivate intrabody expression in response to light, small molecules, or endogenous transcriptional cues. Such systems can be used to minimize off-target toxicity or allow reversible control over therapeutic activity in vivo.16 This is particularly important for applications in the central nervous system or other sensitive tissues where chronic or unregulated intrabody expression could lead to unintended consequences.

An elegant extension of this concept is the use of degron-tagged intrabodies for inducible protein knockdown. By attaching specific degron motifs — peptide sequences recognized by the ubiquitin-proteasome machinery — to the intrabody scaffold, researchers can direct the degradation of bound target proteins with spatial and temporal precision. These constructs effectively act as programmable degradation switches, enabling targeted removal of misfolded, hyperactive, or toxic proteins inside the cell.14 The approach offers several advantages over conventional RNA interference or gene editing, including faster kinetics, reversibility, and the ability to degrade posttranslationally modified isoforms.

Perhaps most transformative is the emerging synergy between intrabodies and CRISPR technologies. While CRISPR-based genome editing has reshaped the landscape of gene therapy, many disease phenotypes arise not from DNA sequence errors but from aberrant protein interactions or conformations. By combining CRISPR perturbations with intrabody-mediated functional modulation, researchers can now perform phenotypic correction at multiple levels, rewriting gene expression while simultaneously neutralizing dysfunctional proteins. In one notable application, intrabodies were used to guide CRISPR screens toward functionally relevant targets in disease models of protein aggregation, offering a dual-pronged strategy for therapeutic discovery.7

Together, these synthetic biology tools are transforming nanobodies and intrabodies from simple binding agents into dynamic control modules capable of responding to cellular context. As this toolkit grows, it is likely to spawn new therapeutic paradigms — targeted degradation circuits, self-regulating feedback loops, and conditionally activated biologics — that could fundamentally redefine how we treat complex diseases.

Commercial Landscape and Regulatory Trajectory

After years of academic exploration and preclinical validation, nanobody therapeutics are beginning to achieve commercial maturity. The field’s most prominent milestone to date is the approval of caplacizumab, a nanobody-based therapeutic developed by Ablynx and now marketed by Sanofi for acquired thrombotic thrombocytopenic purpura. As the first nanobody approved for clinical use in humans, caplacizumab has validated the platform’s potential, both scientifically and commercially, while setting a precedent for future candidates.2

Building on this success, a number of biotechnology companies have emerged to capitalize on the unique properties of nanobody scaffolds. Ablynx remains a dominant player, but newer entrants, such as VHH Therapeutics and Biolojic Design, are developing diverse applications, from multispecific nanobody constructs to artificial intelligence (AI)-guided binder design. These companies are leveraging the small size, modularity, and high specificity of nanobodies to tackle indications ranging from oncology to autoimmune and infectious diseases. Several are exploring tandem formats, such as bispecific constructs or nanobody-enzyme fusions, to create multifunctional agents tailored to complex pathologies.

The clinical trial landscape for nanobodies continues to expand. As of the most recent data, dozens of nanobody candidates are in various stages of development, including early-phase trials for cancer, respiratory infections, and inflammatory disorders.8,9 In oncology, nanobody-based radiotracers and immunotoxins are being tested for improved tumor targeting and minimal off-target toxicity. In infectious disease, the rapid manufacturability and stability of nanobodies make them appealing for pandemic preparedness, particularly in combination with mRNA delivery or inhalable formulations.

Intrabodies, however, face a more complex regulatory path. Because they operate within cells and are typically delivered through gene or RNA-based vectors, they occupy a regulatory space that overlaps with gene therapy, RNA therapeutics, and biologics. Regulatory frameworks for intracellular biologics are still evolving, and many intrabody candidates are currently confined to preclinical models. Key issues include long-term safety, off-target effects, biodistribution of delivery vehicles, and the potential immunogenicity of intracellular proteins expressed at non-native levels or in non-target tissues.5

Nonetheless, regulatory agencies have signaled a willingness to adapt. Lessons from recent advances in cell and gene therapy — as well as mRNA vaccine deployment — are informing new pathways for evaluating intrabody safety and efficacy. For both nanobodies and intrabodies, successful commercialization will hinge not only on scientific performance but on the ability to navigate complex regulatory requirements, scale manufacturing efficiently, and demonstrate clear clinical benefit in well-chosen indications.

As more candidates enter the clinic and early successes accumulate, investor confidence in nanobody platforms has grown. The next phase of market development will likely involve broader integration with delivery innovations, such as lipid nanoparticles and viral vectors, and closer alignment with synthetic biology and AI-driven design. With a growing number of stakeholders and expanding therapeutic scope, nanobodies and intrabodies are poised to reshape the commercial landscape of targeted biologics.

Future Outlook: Charting the Path from Lab to Clinic

The road from laboratory innovation to clinical adoption is particularly complex for next-generation biologics like nanobodies and intrabodies. Success in this domain will require more than demonstration of binding affinity or mechanistic novelty — it demands evidence demonstrating real-world efficacy, safety, scalability, and versatility across therapeutic areas. To that end, the field is beginning to converge on key benchmarks for translational progress, including well-defined biomarkers, meaningful clinical endpoints, and reproducible target engagement in relevant patient populations.

For nanobodies and intrabodies alike, early-stage programs have frequently focused on rare diseases and niche indications — settings in which unmet need is high, patient populations are well characterized genetically or molecularly, and regulatory flexibility can accelerate development. In these contexts, proof-of-concept trials have shown that small, engineered antibody fragments can achieve pharmacodynamic effects on par with or superior to conventional mAbs, particularly when tissue penetration, rapid clearance, or intracellular delivery are essential. The challenge moving forward will be scaling these successes beyond individual indications to establish platform-based approaches that support rapid design, validation, and manufacturing of new candidates across therapeutic classes.

Several application areas stand out as especially promising. Neurodegeneration remains a frontier where conventional antibody therapeutics have struggled, largely due to the impermeability of the blood–brain barrier and the intracellular nature of pathogenic proteins. Intrabodies capable of blocking or degrading misfolded proteins like tau, alpha-synuclein, or huntingtin offer a compelling alternative, especially when paired with targeted delivery systems or expression in specific neuronal subtypes. Similarly, intracellular infections caused by viruses or parasites present targets that are inaccessible to extracellular therapeutics but vulnerable to engineered intrabodies designed to inhibit replication, assembly, or host-protein hijacking.

Another exciting opportunity lies in immune modulation — both for enhancing antitumor responses and for dampening autoimmunity. Nanobodies can be deployed as tumor-targeting modules in CAR-T cells or as immune checkpoint inhibitors with enhanced tumor penetration. Intrabodies, meanwhile, can selectively inhibit transcription factors, inflammasome components, or other intracellular immune regulators, opening up entirely new avenues for precision immunotherapy.

Crucially, the future of intrabody and nanobody discovery will be increasingly shaped by technological convergence. High-throughput screening methods, directed evolution platforms, and synthetic library design have already accelerated the identification of binders with superior affinity and specificity. The integration of AI into these workflows — particularly for structure prediction, binding site mapping, and in silico affinity maturation — is reducing the time and cost associated with therapeutic design and optimization.7 AI-guided tools can also help predict off-target interactions and immunogenicity risk, informing better candidate selection and safety profiling before reaching animal models or human trials.

Ultimately, the path forward will rely not only on technical breakthroughs, but on the creation of collaborative ecosystems that bring together expertise in protein engineering, delivery science, clinical medicine, and regulatory strategy. As these fields align, nanobodies and intrabodies are well positioned to transition from specialized tools to foundational components of a new therapeutic era — one defined not by molecular size, but by functional precision and biological reach.

Conclusion: Small Antibodies, Big Impact

Nanobodies and intrabodies represent a fundamental shift in how the biopharmaceutical industry approaches previously intractable targets. By enabling direct engagement with cryptic extracellular epitopes and intracellular proteins once considered “undruggable,” these compact antibody formats expand the therapeutic arsenal in both scope and precision. Their emergence signals not merely an incremental improvement over traditional mAbs but the advent of a new class of biologics engineered for agility, specificity, and modular integration with next-generation platforms.

Nanobodies, with their high affinity, stability, and tunable formats, have already demonstrated their value as flexible agents for targeted therapy and diagnostics. Their rapid production, amenability to multimerization, and compatibility with a variety of delivery systems position them as ideal candidates for both standalone therapies and combinatorial modalities, including CAR-T cells, bispecifics, and theranostics. Their clinical progress, including regulatory approvals and a growing pipeline across diverse indications, underscores their readiness for broader adoption.

Intrabodies, meanwhile, offer an entirely new dimension of therapeutic control by functioning within the cell. As precision instruments for modulating protein conformation, localization, or degradation, they have the potential to reshape the treatment of diseases driven by intracellular dysfunction — neurodegeneration, viral infections, and autoimmune conditions among them. Yet realizing this promise will depend on solving persistent challenges around intracellular delivery, in vivo stability, and long-term safety.

To truly unlock the potential of these platforms, concerted investment is needed, not only in protein engineering but in the supporting infrastructure of delivery science, regulatory science, and translational medicine. Collaborative frameworks between biotech developers, clinical researchers, and regulatory bodies will be essential to refine evaluation standards, streamline development pathways, and ensure safe, scalable implementation.

In an era defined by precision, adaptability, and increasing therapeutic complexity, nanobodies and intrabodies offer a blueprint for the next generation of drug development. Compact in form but expansive in capability, they are poised to reshape the contours of what biologics can achieve. The challenge now is not whether they can work — but how fast we can bring them to those who need them most.

References

1. Messer, Anne and David C Butler.Optimizing intracellular antibodies (intrabodies/nanobodies) to treat neurodegenerative disorders.” Neurobiology of Disease. 134: 104619 (2020).

2. Frecot, Desiree I, Theresa Froehlich, and Ulrich Rothbauer.30 years of nanobodies – an ongoing success story of small binders in biological research.” J. Cell Sci. 136: jcs261395 (2023).

3. Gettemans, Jan and Brian De Dobbelaer. Transforming nanobodies into high-precision tools for protein function analysis.Americal Journal of Physiology. 16 Feb. 2021.

4. Böldicke, Thomas.Therapeutic Potential of Intrabodies for Cancer Immunotherapy.: Current Status and Future Directions.” Antibodies. 11: 49 (2022).

5. Joest, Eike, et al.Light-guided intrabodies for on-demand in situ target recognition in human cells.” Chem. Sci. 12: 5787–5795 (2021).

6. Leonard, Bojana, et al.Shaping Nanobodies and Intrabodies against Proteoforms.” Analytical Chemistry. 26 May 2023.

7. Dong, Jie-Xian, et al.A toolbox of nanobodies developed and validated for use as intrabodies and nanoscale immunolabels in mammalian brain neurons.” eLife. 8: e48750 (2019).

8. Zheng, Fang, et al.Applications of nanobodies in brain diseases.” Front. Immunol. 7 Nov. 2022.

9. Jin, Bo-kyung, et al.NANOBODIES®: A Review of Diagnostic and Therapeutic Applications.” Int. J. Mol. Sci. 24: 5994 (2023).

10. Wagner, Teresa R and Ulrich Rothbauer.Nanobodies Right in the Middle: Intrabodies as Toolbox to Visualize and Modulate Antigens in the Living Cell.” Biomolecules. 10: 1701 (2020).

11. Trimmer, James S. “Genetically encoded intrabodies as high-precision tools to visualize and manipulate neuronal function.” Semin. Cell Dev. Biol. 126: 117–124 (2022).

12. Dingus, John G, et al.A general approach for stabilizing nanobodies for intracellular expression.” eLife. 11: e68253 (2022).

13. Chatterjee, Diptaman, et al. Proteasome-targeted nanobodies alleviate pathology and functional decline in an α-synuclein-based Parkinson’s disease model.” npj Parkinson’s Disease. 4: 25 (2018).

14. D'Brant, Lianna, et al.Fully Human Bifunctional Intrabodies Achieve Graded Reduction of Intracellular Tau and Rescue Survival of MAPT Mutation iPSC-derived Neurons.” bioRxiv. 1 Jun. 2024.

15. Joshi, Mansi, Pranay Dey, and Abhijit De.Recent advancements in targeted protein knockdown technologies—emerging paradigms for targeted therapy.” Explor. Target. Antitumor Ther. 4: 1227–1248 ( 2023).

16. Rizk, Soha S., et al.Nanobodies in the fight against infectious diseases: repurposing nature's tiny weapons.” World Journal of Microbiology and Biotechnology. 40: 209 (2024).