Peptide Drug Conjugates

Designed for biological research and industrial applications, not intended for individual clinical or medical purposes.

Enhanced SelectivityImproved PharmacokineticsEnhanced Drug DeliveryReduced Drug Dosage

We provide comprehensive Peptide–Drug Conjugate (PDC) development services designed to accelerate your targeted drug discovery and delivery programs. Our end-to-end platform integrates custom peptide design, linker chemistry, conjugation synthesis, and biological validation, delivering fully characterized PDC candidates ready for preclinical or clinical advancement. Whether you are developing oncology therapeutics, precision imaging agents, or targeted delivery systems, we offer flexible, high-quality solutions tailored to your scientific and regulatory goals.

What are Peptide Drug Conjugates?

Peptide-drug conjugates (PDCs), a subset of drug conjugates, are composed of carrier peptides ranging from 5 to 30 amino acid residues, toxic payloads, and linkers that connect the payload to the peptide. PDCs are further broken down into cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs), each having their own differences in the delivery of cytotoxic payloads. The efficacity of PDCs lies in their ability to deliver drugs to targeted cells without affecting healthy cells. The peptide in the PDCs attaches to specific receptors or proteins on the disease-causing cells' surfaces. In this state, the medicine is released to the diseased cells alone, reducing the side effects commonly associated with traditional therapies. Generally, compared to antibody-drug conjugates (ADCs), PDCs have advantages in tumor penetration, ease of synthesis and cost, and reduced off-target effects. Further, as compared to traditional cancer treatments (e.g., chemotherapy and radiation), PDCs have higher specificity for the target cancer with generally less toxic side effects in smaller doses.

Fig. 1 A schematic of a peptide-drug conjugate construct consisting of a homing peptide, linker and payload. The structure of 177Lu-dotatate an FDA approved peptide-drug conjugate. (Cooper, B. M., 2021)

What Problems We Solve

Developing effective targeted drug conjugates involves multiple challenges — and our expertise helps you overcome each of them:

Our PDC Service Offerings

Fig. 3 Mal-K-V-R-PABC small molecule-peptide conjugate Fig. 2 Mal-K-V-R-PABC small molecule-peptide conjugate

Peptide Design & Targeting

  • Identification and design of tumor-homing or tissue-specific peptides, using phage display, AI-assisted modelling and receptor / ligand screening.
  • Optimization of peptide stability, affinity, immunogenicity via modifications (cyclization, D-amino acids, non-natural residues).
  • Detailed target-peptide matching to ensure high specificity for receptors (e.g., integrins, CD13, EGFR, LRP-1) and efficient binding.

Linker & Payload Strategy

  • Custom design of cleavable or non-cleavable linkers, tailored to drug-release mechanism (enzyme-sensitive, acid-sensitive, redox-sensitive) and payload chemistry.
  • Payload selection and integration: cytotoxic small molecules, radionuclides, imaging probes, biologics.
  • Development of conjugation strategy and reaction scheme to preserve bioactivity of both peptide and drug.

Conjugation & Synthesis

  • Solid-phase peptide synthesis (SPPS) to generate high-purity peptide building blocks with custom modifications, such as cyclization, PEGylation, non-natural amino acids, or site-specific reactive groups.
  • Precise conjugation of drug payloads using click chemistry, maleimide-thiol coupling, amide bond formation, or site-directed bioconjugation strategies optimized for stability and bioactivity.
  • Development of conjugation reaction routes that preserve drug potency and peptide-receptor affinity while achieving controlled drug-to-peptide ratios (DPR).
  • Compatibility evaluation between peptides, linkers, and payloads to ensure efficient coupling and minimize by-products.
  • mg- to g-scale PDC production with full traceability and reproducibility to support research and preclinical studies.

Purification & Analytical Characterization

  • Purification of peptide-drug conjugates using high-resolution chromatographic methods (RP-HPLC, SEC, IEX) tailored to peptide length, hydrophobicity, and linker/payload properties.
  • Structural confirmation of conjugates using LC-MS, MS/MS, and NMR, ensuring accurate molecular weight, conjugation position, and integrity of both peptide and drug.
  • Determination of drug loading, purity levels, and residual reagents, ensuring compliance with research or preclinical quality requirements.
  • Stability testing under physiological conditions (pH, serum, enzymatic environments) to verify linker performance and drug-release behavior.
  • Batch reporting with complete certificate of analysis (CoA), chromatograms, spectra, and analytical method details.

In Vitro Functional & Stability Validation

  • Cell-based assays for uptake, receptor binding, internalization, endosomal escape and payload release kinetics.
  • Stability assessment of PDC in plasma, whole blood and tissue homogenates; drug release profiling under defined conditions (pH, enzymatic, redox).
  • Cytotoxicity, efficacy and selectivity testing in target vs control cell lines.

In Vivo Pharmacokinetics, Biodistribution & Efficacy

  • Animal model studies (tumour-bearing, orthotopic, metastatic) to assess biodistribution, tumour targeting, payload release and therapeutic effect.
  • PK/PD profiling: plasma/tissue concentrations of PDC and payload, mass-balance, metabolite identification; bioanalysis tailored for PDC challenges (e.g., low LLOQ, matrix binding).
  • Evaluation of therapeutic index, off-target/toxicity assessment and selection of lead candidates for development.

Peptide Strategies in PDC Development

Peptides play a critical role in Peptide Drug Conjugates (PDCs), functioning as the targeting ligand, delivery enhancer, or therapeutic effector. Advanced peptide engineering significantly improves selectivity, stability, tumor penetration, and payload delivery efficiency. Our platform provides all major classes of targeting and functional peptides, allowing seamless integration into next-generation PDC design and optimization.

Peptide StrategyKey BenefitsCore CharacteristicsIdeal Applications
Tumor-Homing Peptides (THPs) Highly selective targeting; enables precise payload deliveryRecognize tumor-specific receptors or microenvironment markersSolid tumors, metastatic cancer, high-specificity targeting
Cell-Penetrating Peptides (CPPs)Enhances intracellular delivery; overcomes membrane barriersPositively charged or amphipathic sequences; efficient endocytosisDifficult-to-penetrate tumors, intracellular targets, oligonucleotide payloads
Receptor-Specific Peptides Binds defined receptors with high affinityOften derived from natural ligands or optimized sequencesGPCR targets, integrins, RTKs, disease-specific surface proteins
Enzyme-Responsive Peptides Activated or cleaved by disease-specific enzymesCleavable motifs triggered by MMPs, cathepsins, or proteasesTumor microenvironment targeting; conditional activation
pH-Responsive Peptides Preferential activation under acidic tumor conditionsConformational switch or charge change at low pHTumor microenvironment delivery; improved endosomal escape
Peptide Ligands from Phage Display / Screening High screening flexibility; strong optimization potentialSelected via in vitro / in vivo biopanning; customizable specificityAny disease requiring novel or unique targeting ligands
Cyclic PeptidesGreatly improved stability; resistant to proteolysisRigid conformation increases affinity and half-lifeSystemic PDCs, long-circulation applications, harsh biological environments
Stapled / Helical Peptides Enhanced structural stability and cell permeabilityHydrocarbon-stapling maintains α-helix structureIntracellular protein–protein interaction (PPI) targets
Peptide–Antibody Hybrid Ligands (Peptide Mimetics) Combines antibody-level specificity with peptide size advantagesSmaller, more stable, lower immunogenicity than antibodiesHigh-specificity PDCs, where antibody size is limiting
Bioinspired / Natural-Derived Peptides Balanced affinity and safety; often lower immunogenicityDerived from hormones, antimicrobial peptides, or natural ligandsBroad applicability from oncology to inflammation
Computationally Designed Peptides Tunable binding affinity and selectivityAI/ML-guided optimization; de novo sequence designRapid pipeline generation; customized targeting strategies

Linker Strategies for Peptide-Drug Conjugates (PDCs)

The linker is a critical component of every Peptide-Drug Conjugate (PDC), determining how securely the drug is attached to the peptide and how precisely it will be released at the target site. An optimized linker ensures controlled drug release, enhances therapeutic index, and minimizes off-target toxicity, making it essential for developing effective targeted medicines. We provide customized linker design, synthesis, and validation services to ensure optimal stability, controlled drug release, and seamless compatibility across all peptide-drug conjugate (PDC) projects.

Linker TypeTrigger / Release MechanismTypical ApplicationsAdvantages
pH-Sensitive Linkers (e.g., hydrazone, cis-aconityl)Cleaved in acidic environments such as tumors or endosomesTumor-targeted chemotherapy, intracellular drug deliverySelective release inside tumor tissue; minimal release in bloodstream
Enzyme-Cleavable Linkers (e.g., MMP-sensitive, cathepsin B-sensitive, peptide linkers)Cleaved by overexpressed enzymes in tumor microenvironmentOncology PDCs, tumor-penetrating systemsHigh specificity; compatible with a wide range of peptides and drugs
Redox-Sensitive Linkers (e.g., disulfide linkers)Cleaved in high-glutathione intracellular environmentsTargeted cytotoxic drug release inside cancer cellsRapid intracellular release; well-studied mechanism
Non-Cleavable Linkers (e.g., stable thioether bonds)Remain intact; payload is released only after peptide degradationDurable payload delivery, imaging agents, stable conjugatesHigh plasma stability; predictable PK/PD profile
Self-Immolative Linkers Undergo cascade breakdown after initial triggerSmart drug-delivery systems, controlled release PDCsHighly customizable; fine-tuned release kinetics
PEG-Based Linkers Provide steric flexibility and stabilityImproving solubility, serum stability, reducing immunogenicityEnhanced circulation time; improved physicochemical properties

Payload Strategies for Peptide Drug Conjugates (PDCs)

In Peptide Drug Conjugates (PDCs), the payload is the therapeutic engine that ultimately determines potency, selectivity, and clinical performance. A well-engineered payload strategy ensures the ideal balance among efficacy, safety, stability, and controlled release, making it a critical differentiator for next-generation targeted therapies.

Payload StrategyKey BenefitsCore FeaturesIdeal Application Scenarios
High-Potency Cytotoxic Payloads Enables rapid and efficient tumor cell elimination with minimal dosingExtremely potent; designed to minimize off-target toxicity when delivered via peptidesHighly expressed tumor targets; oncology applications requiring strong cytotoxic action
Pathway-Modulating Payloads (e.g., epigenetic modulators) Achieves precise pathway control with moderate toxicityMechanism-driven; suitable for combinational or long-term therapyChronic cancer therapy, drug-resistant tumors, or patient groups sensitive to toxicity
Immunomodulatory Payloads Reprograms the tumor microenvironment to enhance immune responseLow toxicity; strong synergy with immuno-oncology agents"Cold" tumors, immune-evasive cancers, combination immunotherapy
RNA / Oligonucleotide Payloads Targets diseases at the genetic level for highly selective interventionHighly customizable sequences; requires enhanced stability via conjugationGene-driven cancers, precision medicine, rare genetic disorder pipelines
Photo- or Sono-Activated Payloads Allows spatiotemporal control for highly localized therapeutic activityInactive during circulation; activated only by external stimuliLocalized solid tumors, therapies requiring minimal systemic exposure
Dual-Modality Payloads Simultaneously targets multiple disease pathways to reduce resistanceSynergistic multi-mechanism design; modular combination optionsHard-to-treat cancers, multi-pathway disease models
Next-Generation Natural-Product-Derived Payloads Combines high biological potency with improved safety profilesUnique chemical architecture; offers strong innovation valueDifferentiated oncology pipelines, high-value proprietary drug portfolios
Low-Toxicity "Enhanced-Delivery" Payloads Transforms moderately potent molecules into clinically valuable therapeutics through targeted deliveryOptimized for safety; suitable for broader dosing and long-term treatmentMetabolic diseases, inflammatory disorders, non-oncology therapeutic areas

Why Our PDC Development Platform Stands Out

Comprehensive PDC Platform

End-to-end capability covering peptide design, linker chemistry, conjugation, and biological evaluation in one integrated workflow.

Targeted Drug Delivery Expertise

Proven experience in developing tumor-targeting peptides (RGD, NGR, iRGD) for oncology, imaging, and precision therapy applications.

Advanced Linker Design

Expertise in cleavable linkers for controlled drug release, enhancing efficacy while minimizing systemic toxicity.

High-Purity Peptide Synthesis

State-of-the-art solid-phase synthesis and HPLC/MS quality control ensure reproducibility and analytical accuracy.

Integrated Analytical Support

Comprehensive LC-MS, NMR, DLS, and PK/PD testing for precise conjugate characterization and stability evaluation.

Scalable Manufacturing

Flexible production from milligram to gram scale for research and preclinical development.

Rapid Turnaround & Project Transparency

Efficient communication, milestone-based reporting, and real-time project updates shorten development timelines.

Confidentiality & IP Protection

Full intellectual-property security and customizable NDAs to safeguard client innovations.

Collaborative Development Partnership

Support for co-development, technology transfer, and IND-enabling documentation to accelerate market translation.

Our Streamlined PDC Development Workflow

1

Project Assessment & Strategy Design

We evaluate your therapeutic goals, target receptors, and payload requirements to define the optimal peptide-drug conjugation strategy.

2

Targeted Peptide Design & Synthesis

Custom peptides are developed using phage display, AI modeling, or rational design, followed by high-purity SPPS synthesis and analytical confirmation.

3

Linker Engineering & Payload Integration

We design cleavable or stable linkers and select compatible payloads to ensure controlled release, high stability, and efficient conjugation.

4

Conjugation & PDC Construction

Your peptide and drug molecules are conjugated using precision chemistries such as click reactions, maleimide-thiol coupling, or amide linking.

5

Purification & Analytical Characterization

PDCs undergo multi-step purification (HPLC/SEC) and full analytical testing (LC-MS, NMR, stability profiles) to confirm structure and purity.

6

In-Vitro & In-Vivo Validation

We evaluate cellular uptake, drug-release kinetics, cytotoxicity, and in-vivo biodistribution/PK/PD to identify high-value PDC candidates.

Application Fields of Peptide-Drug Conjugate (PDC) Technology

Our PDC technology platform can be applied across multiple therapeutic and research domains.

Each application benefits from targeted delivery, controlled release, and enhanced efficacy while reducing systemic toxicity.

Oncology & Cancer Therapeutics

  • Design of tumor-targeting PDCs using RGD, NGR, or iRGD peptides to bind integrins or CD13 receptors on tumor vasculature.
  • Development of peptide-chemotherapy conjugates for selective cytotoxic delivery to cancer cells.
  • Combination therapy studies integrating PDCs with immune checkpoint inhibitors or radiotherapy.
  • Imaging-guided tumor diagnosis using fluorescent or radiolabeled peptide conjugates.

Targeted Drug Delivery Systems

  • Engineering peptide-functionalized nanoparticles or liposomes for receptor-specific targeting.
  • Conjugation of small-molecule drugs to peptides for tissue- or organ-specific delivery (e.g., liver, lung, or brain).
  • Dual-function systems combining cell-penetrating peptides (CPPs) and therapeutic agents for intracellular delivery.

Central Nervous System (CNS) & Brain Delivery

  • Use of BBB-penetrating peptides (T7, Angiopep-2) to deliver neuroprotective drugs or biologics into the brain.
  • Development of brain-targeted PDCs for neurodegenerative diseases such as Alzheimer's and glioblastoma.
  • Application in neuro-oncology imaging with peptide-based contrast agents.

Imaging & Diagnostics

  • Synthesis of radiolabeled or fluorescent peptide conjugates for tumor and tissue imaging.
  • Dual-modality PDCs combining therapeutic and diagnostic capabilities (theranostics).
  • Custom labeling of peptides for PET, SPECT, or optical imaging applications.

Infectious Disease & Antimicrobial Therapy

  • Peptide-antibiotic conjugates targeting bacterial membranes or biofilms.
  • Delivery of antiviral drugs via receptor-targeting peptides for improved cellular uptake.
  • Use of AMP-based (antimicrobial peptide) conjugates for resistant bacterial strains.

Regenerative Medicine & Tissue Repair

  • Peptide conjugates guiding growth factors or regenerative drugs to damaged tissue.
  • Targeted delivery for angiogenesis and wound healing via RGD or CendR peptides.
  • Controlled local release systems in tissue-engineered scaffolds.

Preclinical Research & Drug Discovery

  • Rapid screening of new PDC candidates using our synthesis and validation platform.
  • Evaluation of pharmacokinetic and pharmacodynamic properties of conjugates.
  • Support for mechanistic studies of receptor binding, internalization, and drug release.

Differences Between PDCs and ADCs

PDCs and ADCs both represent advanced targeted drug delivery modalities, but they differ significantly in size, targeting mechanisms, manufacturability, tissue penetration, safety profile, and development scalability. Understanding these differences is essential for selecting the right modality for specific therapeutic applications.

CategoryPDCs (Peptide Drug Conjugates)ADCs (Antibody–Drug Conjugates)
Molecular Size Small (typically<5 kDa)Large (~150 kDa)
Tumor Penetration Excellent; easily diffuses into solid tumorsLimited due to large size; slower tissue penetration
Targeting Precision High-peptides can target receptors, microenvironment markers, or internalizing motifsHigh-driven by antibody–antigen specificity
Manufacturing Complexity Simple, fully chemical synthesis; scalableComplex biological/chemical hybrid production; costly
Production Cost Low to moderateVery high
Immunogenicity Risk Generally lowHigher; antibodies can trigger immune responses
Circulation Half-Life Shorter; can be extended via modificationLong natural antibody half-life
Payload Capacity Lower payload load; typically 1–2 moleculesHigh drug-to-antibody ratio (DAR up to ~8)
Linker Compatibility Highly flexible with chemical linkersBioconjugation constraints with amino acids/cysteines
Specificity Drivers Peptide sequence, receptor selectivity, tumor homingMonoclonal antibody binding to specific antigens
Biodistribution Broad distribution due to small size; rapid clearanceMore restricted; prolonged systemic presence
Stability Requires optimization (cyclization, stapling)Generally very stable in plasma
Best Use Cases Solid tumors, intracellular targets, rapid iteration pipelines, emerging targetsHematologic cancers, high-antigen-expression tumors
Discovery Speed Fast; peptides can be screened or designed rapidlySlow; antibody discovery, validation, cell line development required
Scalability & Cost Efficiency Excellent; ideal for large-scale or rapid development programsLimited; expensive and resource-intensive

FAQs

1. What is a Peptide–Drug Conjugate (PDC)?

A PDC is a targeted therapeutic composed of a bioactive peptide linked to a drug molecule via a chemical linker. The peptide ensures specific binding to target cells, while the linker controls drug release at the desired site.

Start Your Peptide–Drug Conjugate Project Today

Ready to bring your peptide-based therapeutics from concept to reality?

Our expert scientists specialize in Peptide–Drug Conjugate (PDC) development, offering integrated solutions for targeted drug delivery, oncology therapeutics, CNS applications, and diagnostic imaging. We combine advanced peptide design, linker chemistry, and pharmacological validation to deliver PDC candidates that meet your scientific and regulatory needs. Contact us now to discuss your project — and discover how our custom PDC development services can accelerate your innovation and reduce time-to-market.