Synthetic Peptide Libraries: Unlocking New Possibilities in Immunotherapy and Drug Development

This blog provides an in-depth look at peptide libraries, their design strategies, and their crucial role in drug discovery, immunotherapy, and personalized therapies. From identifying neoantigens for cancer vaccines to advancing T-cell and B-cell screening, we explore how peptide libraries are shaping the future of peptide-based therapeutics.
1. Introduction to Peptide Libraries
Peptide libraries are powerful tools in modern biotechnology, comprising vast collections of systematically designed peptides. These libraries can facilitate protein interaction studies as well as accelerate drug development. Created through advanced chemical synthesis or molecular cloning, peptide libraries encompass diverse amino acid sequences, unlocking new possibilities in immunotherapy, precision medicine, and targeted therapeutics [1].
1.1. Different Types of Peptide Libraries
Peptide libraries can be broadly categorized into chemical and biological types.
Chemical Peptide Libraries:
- Linear peptide libraries: These consist of peptides with linear sequences and an open-chain structure. They are valuable tools in immunotherapy, vaccine development, and targeted drug discovery, particularly for epitope mapping and protein interaction studies [2].
- Cyclic peptide libraries: These contain peptides with cyclic structures, with enhanced stability, target binding affinity, and resistance to enzymatic degradation. Due to these advantages, cyclic peptides are widely preferred in therapeutic development [3].
- Combinatorial peptide libraries: These libraries generate large, diverse sets of molecules by systematically combining different building blocks (e.g., amino acids, chemical fragments, or nucleotides). They enable high-throughput screening for peptides with optimized properties and can be used for drug discovery applications [4].
Directed evolution techniques creating Biological Peptide Libraries:
Biological peptide libraries, such as phage display libraries from phage display, are constructed using molecular biology techniques to identify high-affinity peptide binders. These technologies facilitate the screening of very large numbers of peptides (up to 10¹⁰ variants) and are widely used in drug development and vaccine design [5].
2. Applications of Peptide Libraries
Peptide libraries serve as powerful tools in multiple domains, enabling the discovery and optimization of peptides for therapeutic applications.
2.1. Molecular Binding and Epitope Mapping
This is a fundamental approach in understanding molecular interactions, particularly in immunology, drug discovery, and protein research. It can identify key binding regions within proteins, leading to the development of targeted therapeutics and vaccines.
- Peptide-protein, peptide-ligand, and peptide-receptor interactions: Peptide libraries help identify high-affinity binders that can act as inhibitors or modulators of disease-associated protein-protein interactions (PPIs) [6]. Since PPIs often involve large, flat interaction surfaces, they are difficult to target with traditional small molecules. Peptide libraries provide an effective alternative by identifying short peptides that can selectively disrupt or stabilize these interactions. Many diseases, including cancer, neurodegenerative disorders, and autoimmune conditions, result from dysregulated PPIs, making peptide-based modulators attractive therapeutic candidates [7]. For example, stapled peptides have been developed to block protein interactions in cancer, such as MDM2-p53, restoring tumor suppressor function [8].
- Epitope mapping for vaccine and antibody development: Overlapping peptide and alanine scanning libraries can allow for the identification of immunogenic epitopes capable of eliciting robust and specific immune responses. By systematically testing peptide fragments derived from a target antigen, both linear and conformational epitopes can be determined that contribute to protective immunity [9]. Epitope mapping is particularly important for designing peptide-based vaccines and neoantigen-based cancer vaccines. This ensures that selected epitopes not only generate strong T-cell and B-cell responses but also minimize the risk of immune evasion by pathogens or tumors.
- Drug discovery and small-molecule targeting: Peptide libraries can facilitate the development of therapeutic peptides, peptide-mimetic drugs, and small-molecule inhibitors that can modulate key biological pathways involved in disease progression. In early-stage drug discovery, positional scanning and cyclic peptide libraries are widely used to screen and optimize lead compounds [10].
- One of the key applications of peptide libraries in drug discovery is the design of peptide-based drugs that mimic natural ligands. By screening peptide libraries, one can identify peptides that competitively bind to receptors, either blocking or activating downstream signalling pathways. For example, peptide-based inhibitors of PPIs are being developed to target critical disease-related pathways in cancer [11].
- Additionally, peptide libraries are instrumental in small-molecule drug targeting by identifying minimal binding motifs within peptides that can be converted into non-peptide small-molecule drugs. This rational drug design approach bridges the gap between biologics and traditional small-molecule therapeutics, leading to the discovery of new drug classes with enhanced specificity and efficacy.
2.2. Cancer Immunotherapy
For developing innovative cancer therapies, peptide libraries play a pivotal role in identifying effective epitopes and monitoring therapy efficacy.
2.2.1. T- Cell and B- Cell Screening
Peptide libraries are widely used to screen and identify T-cell and B-cell epitopes, facilitating the development of targeted immunotherapies and vaccines.
- CD8+ T- cell epitope identification (MHC-I restricted peptides): Peptide libraries are used to identify epitopes recognized by CD8+ T- cells, which play a key role in eliminating virus-infected or cancerous cells. These epitopes are typically 8–1 amino acids long and must bind to MHC class I molecules for presentation to T- cells [12].
- CD4+ T cell epitope identification (MHC-II restricted peptides): CD4+ T- cells coordinate immune responses by stimulating antibody production and enhancing cytotoxic T- cell activity. Identifying MHC class II-restricted epitopes (12-25 amino acids) helps to develop vaccines that elicit robust and long-lasting immune memory [12].
- T-cell receptor (TCR)-peptide interaction studies: This approach is critical in designing TCR-engineered T-cell therapies, where synthetic peptides are used to enhance TCR affinity and selectivity for tumor-associated antigens. Incorporating structural modifications, including constrained peptides and peptidomimetics, helps to optimize the interaction between engineered TCRs and peptide-MHC complexes, boosting the therapeutic potential [13].
- B-cell epitope mapping and antibody optimization: Peptide libraries help to understand how antibodies recognize antigens, particularly through alanine-scanning and overlapping peptide libraries, by systematically identifying linear and conformational epitopes targeted by neutralizing antibodies. Advanced strategies, such as glycosylation and chemical modifications, further enhance peptide-antibody interactions. This enables the refinement of antibody affinity and specificity, ensuring more potent and durable therapeutic effects [14].
- Identification of neoantigens: Peptide libraries can be used to identify Neoantigens by systematically screening mutated peptides for their immunogenic potential. These libraries contain overlapping peptides derived from tumor-specific mutations and are used to assess which peptides can be recognized by the immune system. Once potential neoantigens are identified, synthetic peptide libraries help validate their immunogenicity by determining whether they can activate T-cell responses [15].
- Library diversity for broad immune coverage: In vaccine development and immunotherapy, a diverse peptide library ensures that a wide range of epitopes is explored, increasing the likelihood of identifying peptides that bind to MHC molecules and trigger a broad immune response. The diversity also allows for the identification of peptides that bind to both MHC class I and class II molecules, ensuring activation of both cytotoxic and helper T-cells, thereby enhancing the overall efficacy and durability of the immune response.
2.2.2. Monitoring T-Cell Responses
Accurate assessment of T-cell responses is essential for optimizing vaccine efficacy and evaluating immune-based therapies. Immunomonitoring provides deep insights into T-cell activation, functional polarization, and long-term immunological memory [16, 17].
- ELISpot assay: It enables the precise quantification of cytokine-secreting T cells at a single-cell resolution. By detecting IFN-γ, IL-2, and other cytokines, ELISpot measures the magnitude and quality of antigen-specific T-cell responses. A high frequency of IFN-γ-secreting cells correlates with robust CTL activation, critical for anti-tumor immunity.
- Intracellular cytokine staining (ICS) and flow cytometry: These allow multi-parametric analysis of T-cell subsets based on cytokine expression, activation markers, and differentiation status. ICS distinguishes between effector and memory T-cell populations, providing insight into the longevity and effectiveness of immune responses. Flow cytometric analysis of polyfunctional T cells (expressing multiple cytokines) is a key predictor of vaccine-induced protective immunity.
- MHC tetramer assay: It utilizes fluorescent-labeled peptide-MHC complexes to directly stain and quantify antigen-specific T-cell populations. This high-sensitivity method enables the tracking of immune responses over time, distinguishing between naive, activated, and memory T-cell subsets. By identifying dominant T-cell clones, researchers can evaluate immune persistence and correlate specific T-cell populations with clinical outcomes in cancer immunotherapy.
3. Strategies to Design Peptide Libraries
The design of peptide libraries requires careful consideration of the experimental goals and the type of function desired. Below is an overview of key peptide library design strategies, their applications, and relevant scientific findings.
3.1. Overlapping Peptide Libraries
These are constructed by designing peptides that sequentially overlap with one another, typically by 8-12 amino acids. This overlap ensures comprehensive coverage of a target protein sequence, increasing the likelihood of identifying biologically relevant sequences. By systematically shifting the sequence window across the target protein, researchers can identify the specific regions critical for protein-protein interactions or receptor binding [2].
3.2. Randomized Peptide Libraries
These are constructed by varying the amino acids at each position within a peptide sequence in a completely random manner, generating a highly diverse set of peptides. This randomness ensures that a broad range of sequences is explored, increasing the likelihood of identifying peptides with high binding affinity or specific biological activity. These libraries are particularly valuable for discovering novel peptide structures and functions that may not exist in natural sequences [18].
3.3. Alanine Scanning Libraries
These are designed to systematically study the functional importance of individual amino acids within a peptide or protein sequence. In this approach, each amino acid is sequentially replaced with alanine, which is chosen due to its small, non-reactive methyl side chain (-CH₃). This substitution minimizes steric hindrance while preserving the peptide backbone structure, allowing researchers to assess the role of each residue in biological activity. By evaluating changes in peptide binding affinity or biological function, one can identify critical residues involved in antibody-antigen interactions (epitope mapping) and enzyme-substrate or receptor-ligand binding [19].
3.4. Positional Scanning Peptide Libraries
In this approach, each position in the peptide sequence is systematically varied while keeping the other positions constant. For example, in a peptide of fixed length, one position is held constant with a specific amino acid, while all other positions are randomly varied. This process is repeated for each position in the sequence, helping to identify the most favorable amino acid substitutions. This allows researchers to determine which amino acids at specific positions enhance or reduce biological activity, such as binding to a receptor or enzyme. These libraries are widely used in epitope mapping and peptide optimization [20].
3.5. Truncation Peptide Libraries
These are designed to systematically shorten peptides by removing amino acids from the N-terminal, C-terminal or both ends of a sequence. This approach helps to identify the core functional peptide sequence required for biological activity and reduces synthesis costs by identifying the shortest effective peptide sequence. Truncation libraries are often used in conjunction with alanine scanning libraries within peptide research workflows to refine peptide design. They are widely applied in epitope mapping and in receptor-ligand studies [21].
3.6. Scrambled Peptide Libraries
These are created by rearranging the amino acids of a known peptide sequence in different orders while keeping the overall composition the same. This helps to determine whether a peptide’s biological activity depends on its exact sequence or just its general properties, such as charge and hydrophobicity. If a scrambled peptide retains activity, it suggests that the function is influenced more by its physicochemical characteristics rather than a specific sequence. These libraries are commonly used as negative controls in experiments to confirm that the original peptide’s effect is truly sequence dependent. They also help in optimizing peptide design for drug development by identifying alternative sequences that maintain function but may have better stability or reduced immunogenicity [22].
3.7. Cyclic and Modified Peptide Libraries
Cyclic peptide libraries are created by chemically linking the N- and C-termini of peptides, forming a covalent ring structure. This cyclization offers several key advantages [3, 7]:
- Enhanced stability: Cyclization increases resistance to enzymatic degradation, as proteolytic enzymes are unable to cleave the peptide. This extends its half-life in biological systems, reducing the frequency of administration and ensuring stability in circulation.
- Improved binding affinity: The cyclization process induces a more rigid conformation in the peptide, which can enhance the specificity and affinity for its target receptors or proteins.
- Increased target selectivity: The fixed structure of cyclic peptides allows for a precise fit to binding sites, improving target selectivity and reducing off-target effects, thereby enhancing therapeutic efficacy.
3.8 Modifications
Modified peptide libraries incorporate various chemical alterations to the peptide structure to improve drug-like properties. Common modifications include [8]:
- Incorporation of non-natural amino acids: Non-natural amino acids, such as D-amino acids (the mirror image of natural L-amino acids), which can be integrated into peptides to improve their stability and resistance to proteolytic degradation. This also allows the design of peptides with novel functionalities, such as improved binding or enzymatic activity.
- Backbone modifications: Altering the peptide backbone (e.g., using β-turn mimetics or peptoid backbones) can enhance resistance to proteases and provide better structural integrity.
- Side-chain substitutions: Modifying the side chains of peptides can influence their hydrophobicity or hydrophilicity, improving interactions with specific biological targets. These modifications can also enhance cell membrane penetration, facilitating the delivery of peptides to intracellular targets. Additionally, modifications like PEGylation (adding polyethylene glycol) can improve solubility, extend circulation time and optimize the pharmacokinetic profile of the peptide.
3.9. Computational and AI-Assisted Design of Peptide Libraries
In recent years, computational methods and artificial intelligence (AI) have become powerful tools in the design and optimization of peptide libraries. Here’s how computational and AI-assisted approaches are transforming peptide library design [23, 24]:
- Peptide structure prediction: Computational methods are used to predict the three-dimensional structure of peptides based on their amino acid sequence. Algorithms can simulate how peptides interact with their targets, providing insights into the most effective structures for target binding.
- Predicting binding regions: AI and machine learning models can be used to predict sequence patterns that correlate with high binding affinity, allowing for the rapid design of potent peptides without the need for time-consuming wet-lab screening.
- Virtual screening: Computational platforms can simulate the interaction of a large library of peptides with target proteins, enabling high-throughput virtual screening. This allows one to evaluate and prioritize peptides for experimental testing based on predicted binding properties and biological activity, significantly speeding up the drug discovery process.
- De Novo peptide design: AI-powered algorithms can generate entirely new peptide sequences with optimized properties. By learning from existing peptide libraries, AI can design novel sequences that have a higher chance of success in specific therapeutic applications.
- Optimizing peptide stability and bioavailability: AI and computational models can predict how modifications, such as backbone alterations or incorporation of non-natural amino acids, will impact peptide stability, bioavailability, and pharmacokinetic properties. This enables the design of peptides that are more resistant to degradation, have improved circulation time, or penetrate biological membranes more effectively.
- Computational and AI-assisted approaches dramatically reduce the time and cost associated with traditional peptide library development. By narrowing down the most promising candidates before experimental testing, these technologies streamline the peptide discovery process, leading to faster development of peptide-based therapeutics.
4. Peptide Library Synthesis: Techniques and Advances
Peptide library synthesis has evolved beyond conventional linear approaches, incorporating chemical innovations that enhance sequence diversity, structural complexity and screening efficiency. These advancements enable the rapid discovery of functional peptides for drug development and immunotherapy.
4.1. Solid-Phase Peptide Synthesis (SPPS) and Functional Modifications
SPPS remains the gold standard method for peptide library synthesis, allowing stepwise addition of amino acids to a growing peptide chain anchored to a solid resin. This method enables rapid parallel synthesis of multiple peptides and is compatible with various chemical modifications. The efficiency of SPPS has been enhanced through optimized coupling reagents and automated synthesis platforms, reducing aggregation and synthesis failures [25].
- N-terminus and C-terminus modifications: SPPS allows the introduction of non-natural amino acids, fluorescent tags, biotinylation, or PEGylation, improving stability and functionality.
- Cyclization strategies: Head-to-tail cyclization or disulfide bond formation during SPPS enhances structural rigidity and proteolytic resistance, making peptides more suitable for therapeutic applications. Also, introducing lactam bridges or ring-closing metathesis, generates constrained structures that mimic protein secondary structures, increasing target affinity and selectivity.
- Post-synthetic functionalization: Advanced methodologies integrate orthogonal protecting groups, allowing site-specific modifications such as PEGylation, fluorophore labeling and bioconjugation with nanoparticles or drug payloads for targeted delivery.
4.2. Combinatorial and Split-Pool Synthesis for High-Diversity Libraries
Combinatorial synthesis techniques have drastically increased the chemical diversity of peptide libraries, enabling the rapid screening of millions of sequences in a single experiment [26].
- Mix-and-split methodology: A high-throughput approach where resin-bound peptides are divided, coupled with different amino acids, and recombined in iterative cycles, generating vast sequence diversity.
- One-Bead-One-Compound (OBOC) libraries: Each bead carries a unique peptide sequence, facilitating high-throughput screening against target molecules, including enzymes, receptors, and antibodies.
- Encoded peptide libraries: DNA or barcode tagging of peptides during synthesis allows rapid identification of high-affinity binders without requiring extensive sequencing efforts.
4.3. Advancements in Peptide Library Synthesis
Technological innovations have improved peptide library screening efficiency and broadened their applicability.
- Tag-free screening platforms: Traditional peptide screening relies on fluorescent or affinity tags, which may interfere with native binding properties. Recent advancements enable label-free, mass spectrometry-based or biophysical detection methods for unbiased screening [1].
- Cyclic peptide synthesis and macrocycle libraries: Advances in cyclization chemistries, including lactam bridges, disulfide bonds and stapled peptides, have led to the development of macrocyclic peptide libraries with enhanced protease resistance, membrane permeability and drug-like properties.
- Automated high-throughput synthesis: Robotics and microfluidic platforms now facilitate parallel synthesis of thousands of peptides with precise modifications.
The continuous evolution of peptide library synthesis techniques enables the discovery of novel therapeutics, epitope mapping strategies and functional peptides for diverse biological applications.
5. Challenges and Future of Peptide Libraries
Despite the transformative potential of peptide libraries in drug discovery, immunotherapy, and molecular targeting, several challenges remain that impact their scalability, stability, and functional application. Addressing these limitations through emerging technologies will define the future of peptide-based therapeutics.
5.1. Overcoming Limitations
- Scalability and cost limitations: The synthesis of large peptide libraries, particularly for high-throughput screening, remains expensive and
resource-intensive. The scalability of peptide libraries is further hindered by synthesis inefficiencies associated with long peptide sequences, aggregation and post-synthetic modifications. Automated synthesis platforms are being developed to mitigate these limitations and enhance cost-effectiveness. - Peptide stability and cell permeability: Peptides often suffer from rapid degradation due to enzymatic cleavage in biological systems, limiting their therapeutic utility. Additionally, their hydrophilic nature and lack of passive membrane permeability make intracellular targeting difficult. Structural modifications such as cyclization and hydrocarbon stapling have improved protease resistance and bioavailability. Conjugation strategies, including cell-penetrating peptides (CPPs) and lipid-based modifications, are being explored to facilitate intracellular delivery.
- Difficulty in identifying functional hits: Screening peptide libraries for biologically relevant hits remains a major challenge. Many peptides identified in vitro fail in vivo due to poor pharmacokinetics, weak binding affinity, or lack of specificity. The integration of biophysical techniques and next-generation sequencing of peptide hits is improving the identification of functional candidates.
6. Conclusion
Peptide libraries have become indispensable tools in drug discovery, vaccine development, and immunotherapy, offering unique advantages in targeting complex biomolecular interactions. Despite challenges related to scalability, stability, and functional screening, advances in synthesis techniques, computational modeling, and structural modifications are driving innovation in the field. Researchers and industry leaders must invest in AI-driven design, next-generation screening platforms, and novel peptides to accelerate vaccine development and therapeutic advancements.
Bibliography
[1] Bruce, A. et al. (2024) ‘A tag-free platform for synthesis and screening of cyclic peptide libraries’, Angewandte Chemie, 63(21), pp. 1-19
[2] Iaculli, D., and Ballet, S. (2024) ‘Peptide libraries: From epitope mapping to in-depth high-throughput analysis’, Trends in Pharmacological Sciences, 45(7), pp. 579-582
[3] Ji, X., Nielsen, A. L., Heinis, C. (2024) ‘Cyclic peptides for drug development’, Angewandte Chemie, 63(10), pp. 1-15
[4] Liu, R., Li, X., Lam, K. S. (2017) ‘Combinatorial chemistry in drug discovery’, Current Opinion in Chemical Biology, 38, pp. 117-126
[5] Bazan, J., Całkosiński, I., Gamain, A. (2012) ‘Phage-display – A powerful technique for immunotherapy’, Human Vaccines & Immunotherapeutics, 8(12), pp. 1817-1828
[6] Vincenzi, M., Mercurio, F. A., Leone, M. (2024) ‘Virtual screening of peptide libraries: The search for peptide-based therapeutics using computational tools’, International Journal of Molecular Sciences, 25(3), pp. 1-44
[7] Cheng, J. et al. (2023) ‘Stabilized cyclic peptides as modulators of protein-protein interactions: promising strategies and biological evaluation’, RSC Medicinal Chemistry, 14(12), pp. 2496-2508
[8] Chandramohan, A. et al. (2024) ‘Design-rules for stapled peptides with in vivo activity and their application to Mdm2/X antagonists’, Nature Communications, 15(1), pp. 1-19
[9] Gershoni, J. et al. (2012) ‘Epitope mapping’, BioDrugs, 21(3), pp. 145-156
[10] Bhat, A., Roberts, L. R., Dwyer, J. J. (2015) ‘Lead discovery and optimization strategies for peptide macrocycles’, European Journal of Medicinal Chemistry, 94, pp. 471-479
[11] Wang, X., Ni, D., Liu, Y., Lu, S. (2021) ‘Rational design of peptide-based inhibitors disrupting protein-protein interactions’, Frontiers in Chemistry, 9, pp. 1-15
[12] Melssen, M. M., Fisher, C. T., Slingluff, C. L., and Melief, C. J. M. (2022) ‘Peptide emulsions in incomplete Freund's adjuvant create effective nurseries promoting egress of systemic CD4+ and CD8+ T cells for immunotherapy of cancer’, Journal for ImmunoTherapy of Cancer, 10(9), pp. 1-15
[13] Jones, H. F. et al. (2021) ‘Empirical and rational design of T cell receptor-based immunotherapies’, Frontiers in Immunology, 11, pp. 1-21
[14] Potocnakova, L., Bhide, M., Pulzova, L. B. (2016) ‘An introduction to B-cell epitope mapping and in silico epitope prediction’, Journal of Immunology Research, 2016, pp. 1-11
[15] Saxena, M. et al. (2025) ‘PGV001, a multi-peptide personalized neoantigen vaccine platform: Phase I study in patients with solid and hematological malignancies in the adjuvant setting’, Cancer Discovery, pp. 1-37
[16] Choueiri, T. K. et al. (2025) ‘A neoantigen vaccine generates antitumour immunity in renal cell carcinoma’, Nature, pp. 1-9
[17] Schreiber, R. D. et al. (2019) ‘MHC-II neoantigens shape tumor immunity and response to immunotherapy’, Nature, 574, pp. 696-701
[18] Kubo, T. (2017) ‘Random peptide library for ligand and drug discovery’, in Cruz, L., and Luo, S. (eds.) Toxins and Drug Discovery. Springer, Dordrecht, pp. 207-230
[19] Morrison, K. L., and Weiss, G. A. (2001) ‘Combinatorial alanine-scanning’, Current Opinion in Chemical Biology, 5(3), pp. 302-307
[20] Dooley, C.T., and Houghten, R.A. (1998) ‘Synthesis and screening of positional scanning combinatorial libraries’, in Cabilly, S. (ed.). Combinatorial Peptide Library Protocols. Methods in Molecular Biology, Humana Press, 87, pp. 13-24
[21] Kumar, E. A., et al. (2012) ‘Peptide truncation leads to a twist and an unusual increase in affinity for casitas B-lineage lymphoma tyrosine kinase binding domain’, Journal of Medicinal Chemistry, 55(7), pp. 3583-3587
[22] Farkas, Ö., Lajkó, E., Kalabay, M., Enyedi, K. N. (2022) ‘How to scramble? A ‘wise way’ to achieve a useful control peptide sequence’, SSRN Electric Journal, pp. 1-18
[23] Raja, R. et al. (2025) ‘Immunogenic cryptic peptides dominate the antigenic landscape of ovarian cancer’, Science Advances, 11(8), pp. 1-15
[24] Goles, M. et al. (2024) ‘Peptide-based drug discovery through artificial intelligence: towards an autonomous design of therapeutic peptides’, Briefings in Bioinformatics, 25(4), pp. 1-15
[25] Mäde, V., Els-Heindl, S., Beck-Sickinger, A. G. (2014) ‘Automated solid-phase peptide synthesis to obtain therapeutic peptides’, Beilstein Journal of Organic Chemistry. 10, pp. 1197-1212.
[26] Kofoed, J., Reymond, J-L. (2007) ‘A general method for designing combinatorial peptide libraries decodable by amino acid analysis’, Journal of Combinatorial Chemistry. 9(6), pp. 1046-1052
Peptide bestellen
Wenn Sie auf Bestellen klicken öffnet sich ein Formular, in welches Sie Ihre Sequenz(en) direkt eingeben können. Alternativ können Sie auch ein Dokument mit Ihren Peptid - oder Konjugatsequenzen hochladen