Skip to main content

Molecular Display Systems in Drug Discovery: Principles, Platforms, and Applications

Introduction

Modern drug discovery is driven by the identification of high-affinity ligands that modulate disease-relevant targets with precision. To accelerate this process, a suite of molecular display systems has been developed that enable rapid screening of vast molecular libraries against proteins, nucleic acids, and even complex cellular surfaces. Technologies such as phage display [1,2], mRNA display [3,4], and DNA-encoded library (DEL) screening [5-8] have revolutionized early-stage hit identification, allowing researchers to interrogate binding interactions with unprecedented throughput and chemical diversity.

These platform technologies exploit the fundamental principle of genotype-phenotype linkage, allowing researchers to identify and amplify molecules with desired binding characteristics from among billions—or even trillions—of variants. As the biopharmaceutical industry shifts towards increasingly complex and previously intractable targets, molecular display platforms are playing a critical role in enabling efficient, scalable, and cost-effective ligand discovery.

Principles of Molecular Display Systems

All molecular display systems rely on the linkage of a genotype (nucleic acid sequence) to a phenotype (displayed molecule). This linkage enables selection for function—typically high-affinity binding to a target—followed by amplification of the encoding sequence for downstream analysis and iterative enrichment.

Key attributes of effective display platforms include:

  • Library Size and Diversity: Display libraries routinely exceed 10810¹³ unique entities, enhancing the probability of identifying rare high-affinity binders.
  • Selection Fidelity: Iterative rounds of selection (biopanning) enrich for ligands with favorable binding kinetics and specificities.
  • Compatibility with Downstream Assays: Selected hits can be rapidly synthesized, expressed, or rescreened in orthogonal formats.

Phage Display Technology

Phage display screening is among the most established molecular display platforms, leveraging filamentous bacteriophages (typically M13) to present peptides or proteins on their coat proteins, most commonly pIII or pVIII [1]. In a phage display library, each phage particle presents a variant of a peptide or antibody fragment on its surface and carries the corresponding DNA internally.

Applications and Advantages

Phage display technology has been widely used for:

  • Antibody engineering: The development of humanized monoclonal antibodies such as adalimumab (Humira) [2].
  • Peptide ligand discovery: Identification of binding motifs against receptors, enzymes, and cell-surface proteins.
  • Epitope mapping and vaccine design

The main advantages include:

  • Direct genotype-phenotype coupling
  • Ease of library construction and amplification
  • Robust selection against purified and immobilized targets

Limitations

Despite its strengths, phage display is generally limited to peptides and proteins and has restricted chemical diversity due to reliance on ribosomal synthesis.

mRNA Display: Expanding Chemical and Structural Diversity

mRNA display is a cell-free, in vitro translation-based system that couples a synthetic peptide or protein to its encoding mRNA via a covalent linker, typically involving puromycin[3]. This enables the screening of libraries with up to 10¹³ unique members, far exceeding what is feasible with in vivo systems.

Advantages of mRNA Display

  • Greater library size due to in vitro amplification
  • Incorporation of some non-natural amino acids and chemical modifications
  • Utility in macrocyclic peptide screening and PPI modulation[4]

This platform is particularly useful for discovering ligands against challenging targets such as protein–protein interaction (PPI) surfaces, and its flexibility allows for the evolution of high-stability, high-affinity binders.

DNA-Encoded Library (DEL) Technology

DNA-encoded library technology combines the chemical diversity of small molecule synthesis with the scalability of nucleic acid tagging. In DEL screening, small molecules are covalently linked to DNA barcodes that encode their synthetic history. Libraries of 10⁶ to 10¹² compounds can be pooled and screened simultaneously against a target protein under near-physiological conditions[5].

Principles of DEL Screening

  • Split-and-pool synthesis generates highly diverse libraries[6].
  • DNA tags allow PCR-based enrichment and next-generation sequencing (NGS) readout.
  • Enables affinity-based selection of small molecules with high specificity.

Benefits Over Traditional HTS

  • Higher throughput with lower reagent consumption
  • Miniaturized, parallelizable workflows
  • Hit identification and structure elucidation from a single screen[7]

DEL is especially powerful for identifying starting points in small molecule drug discovery, particularly when integrated with AI/ML-driven predictive modeling [8] and biophysical validation tools.

Choosing the Right Molecular Display Platform

Selecting the optimal display technology depends on several parameters:
Parameter Phage Display mRNA Display DEL Technology
Molecular class Peptides, proteins Peptides, macrocycles Small molecules and peptides
Library size 10⁹–10¹¹ 10¹²–10¹³ 10⁶–10¹²
Chemical diversity Limited Moderate to high Very high
In vitro compatibility Partial Full Full
Target types Proteins, cells Proteins, PPIs Proteins, enzymatic targets, molecular glues, PPIs

For early hit identification in small molecule programs, DEL screening is increasingly favored due to its compatibility with automated workflows and downstream medicinal chemistry.

Applications Across Therapeutic Modalities

Small Molecule Discovery

DEL screening enables rapid identification of lead-like molecules with drug-like properties, serving as a starting point for optimization campaigns[5].

Peptide and Macrocycle Therapeutics

mRNA display and phage display support the discovery of constrained peptides and macrocycles that can target flat or dynamic protein surfaces[4].

Target Deconvolution

Display-based hits can be used in chemoproteomic workflows to deconvolute biological targets of phenotypic hits.

Mechanism of Action Studies

Enrichment kinetics and hit validation from molecular display platforms provide mechanistic insight and support target engagement studies.

Trends in Commercial Use and Collaboration

As display technologies become more complex, many pharmaceutical companies are partnering with contract research organizations (CROs) and platform providers. This outsourcing model facilitates:

  • Access to proprietary libraries
  • Custom synthesis of target-specific libraries
  • Data interpretation and hit triaging support

Moreover, the integration of display technologies with structure-based drug design, cryo-EM, and computational modelling is accelerating lead optimization cycles. Integration with machine learning (ML) tools is further enhancing the predictive power of DEL screening and is paving the way for AI-augmented hit selection [8].

Our DEL Screening Platform: Accelerating Ligand Discovery

At Vipergen, we specialize in custom DNA-encoded library screening solutions tailored to the unique needs of biopharmaceutical discovery teams. Our platform features:

  • Modular DEL library design, including privileged scaffolds and in house designed chemotypes
  • Validated selection workflows for diverse protein targets, including integral membrane proteins and selection in living cells
  • Identification of hits and families with instant Structure Activity Relationship (SAR) and cheminformatics-based clustering
  • Instant selectivity and specificity of hits by multiplexed screen against target and anti-targets

Whether you’re pursuing novel oncology targets, GPCR ligands, or tool compounds for target validation, our DEL screening services offer a powerful and flexible path to high-quality chemical matter.

Learn more about our DEL capabilities or contact our discovery team to discuss custom library screening projects. 

Conclusion

Molecular display systems such as phage display, mRNA display, and DNA-encoded library screening have transformed how researchers discover bioactive ligands and therapeutic leads. By linking molecular recognition to genetic information, these platforms enable efficient exploration of chemical and structural space far beyond the capabilities of traditional high-throughput screening.

As these technologies continue to evolve—with improvements in library design, selection methodologies, and computational analysis—they are poised to play a central role in the next generation of precision drug discovery.

References

  1. McCafferty J, Griffiths AD, Winter G, Chiswell DJ. Phage antibodies: filamentous phage displaying antibody variable domains. Nature. 1990;348(6301):552–554. https://doi.org/10.1038/348552a0
  2. Winter G, Griffiths AD, Hawkins RE, Hoogenboom HR. Making antibodies by phage display technology. Annu Rev Immunol. 1994;12:433–455. https://doi.org/10.1146/annurev.iy.12.040194.002245
  3. Roberts RW, Szostak JW. RNA–peptide fusions for the in vitro selection of peptides and proteins. Proc Natl Acad Sci USA. 1997;94(23):12297–12302. https://doi.org/10.1073/pnas.94.23.12297
  4. Huang Y, Wiedmann MM, Suga H. RNA Display Methods for the Discovery of Bioactive Macrocycles. Chem Rev. 2019;119(17):10360–10391. https://doi.org/10.1021/acs.chemrev.8b00430
  5. Peterson AA, Liu DR. Small-molecule discovery through DNA-encoded libraries. Nat Rev Drug Discov. 2023;22(9):699–722. https://doi.org/10.1038/s41573-023-00713-6
  6. Favalli N, Bassi G, Scheuermann J, Neri D. DNA-encoded chemical libraries – achievements and remaining challenges. FEBS Lett. 2018;592(17):2168-2180. https://doi.org/10.1002/1873-3468.13068
  7. Mason JW, Wang Y, et al. DNA-encoded library-enabled discovery of proximity-inducing small molecules. Nat Chem Biol. 2024;20:170–179. https://doi.org/10.1038/s41589-023-01458-4
  8. McCloskey K, Sigel EA, et al. Machine learning on DNA-encoded libraries: A new paradigm for hit-finding. J. Med. Chem. 2020;63(16):8857-8866.  https://doi.org/10.1021/acs.jmedchem.0c00452

Do you have an inquiry?

Contact us today and explore partnership opportunities.

Related Services

Service
Small molecule drug discovery for even hard-to-drug targets – identify inhibitors, binders and modulators
Molecular Glue Direct
PPI Inhibitor Direct
Integral membrane proteins
Specificity Direct – multiplexed screening of target and anti-targets
Express – optimized for fast turn – around-time
Snap – easy, fast, and affordable