Your cart

Your cart is empty

Peptide Drug Conjugates: Overcoming ADC Limitations in Tumor Targeting

Introduction to Peptide Drug Conjugates

Chemotherapy has long been a cornerstone of cancer treatment, yet its lack of selectivity and frequent emergence of resistance significantly limit its long-term efficacy. To address these challenges, drug conjugates were developed as modular systems consisting of a targeting moiety, a linker, and a cytotoxic payload, enabling selective delivery of therapeutic agents to tumor cells. Among these, antibody-drug conjugates (ADCs) represent the most successful modality to date, with more than a dozen approved products for both hematological malignancies and solid tumors.

However, the limited tumor penetration, high production cost, significant risk of off-site toxicity and limited payload choices of ADCs continue to constrain their broader application. These limitations have inspired the development of alternative modalities such as peptide-drug conjugates (PDCs). PDC share the same fundamental design as ADCs but use short homing peptides as the targeting moiety instead of antibodies. These peptides can selectively recognize receptors overexpressed in tumors, offering advantages such as easier synthesis, enhanced tissue penetration, and lower immunotoxicity.

Figure 1. From antibody drug conjugate to peptide drug conjugate.

As illustrated above, the clinical challenges faced by ADCs open the door for PDCs as a complementary approach. The table below summarizes the key differences across critical factors between ADCs and PDCs, from molecular properties and manufacturing costs to overall clinical potential.

Table 1. The comparison of ADCs and PDCs

Properties

ADC

PDC

Molecular weight

>150,000 Da

~2,000 – 20,000 Da

Pharmacokinetic

Long half-life; nonspecific liver/reticuloendothelial system uptake and risk of hepatotoxicity

 

Short half-life; rapid renal clearance and reduced bone marrow/liver toxicity

Tumor penetration

Limited

 

Yes

Immunogenicity

Some are high

 

Low

Cancer targets

Limited to cell surface receptors

Variety of targets located on cell surface or intracellular protein

 

Chemical synthesis

No

 

Yes

Manufacture cost

High (relatively difficult to manufacture and costly to produce and qualify)

 

Low (It can be expressed in situ or chemically synthesized for simple production and easy scale-up)

Quality control complexity

High (heterogeneity, advanced analytical methods)

Low (homogeneity, simpler analytical methods)

 

The Structure of Peptide-Drug Conjugates

Like ADCs, PDCs are modular constructs composed of three components: a homing peptide, a linker, and a cytotoxic payload. Each part plays a distinct role in ensuring selective delivery and therapeutic activity.

Figure 2. Schematic structure of peptide drug conjugate.

Homing Peptide

In a PDC, the peptide is the key part that guides the drug to its target. These short amino acid chains can recognize receptors or signals that are unusually abundant in tumors, allowing the drug to home in on diseased tissue instead of circulating everywhere. Compared with antibodies, peptides are smaller, easier to synthesize, and highly versatile in their design, making them a promising alternative for targeted delivery.

Depending on their function and destination, homing peptides used in PDCs can be grouped into several categories.

1.     Targeting Peptides

These peptides provide the core specificity for PDCs. They bind to receptors that are frequently overexpressed in specific tissues or cells.

·       Tumor Targeting Peptides: These bind directly to receptors on cancer cells or the tumor’s blood vessels, providing specificity for malignant tissues.

·       Hormone or Ligand-Derived Peptides: These are a specialized class of targeting peptides derived from natural hormones or ligands, retaining their receptor specificity for diseases like hormone-driven cancers.

 

2. Penetrating Peptides

These peptides enhance drug delivery by helping the PDC cross biological barriers or enter cells. 

·       Cell-Penetrating Peptides (CPPs): CPPs are able to efficiently cross cell membrances, promoting the intracellular delivery of conjugated drugs. While they often lack tissue specificity, they are valuable for enhancing cellular uptake.

·       Brain-Penetrating Peptides: These peptides are specifically designed to cross the blood-brain barrier, expanding the potential of PDCs to treat neurological disorders and brain cancers.

·       Tumor-Penetrating Peptides: This newer class of peptides not only targets tumors but also facilitates drug penetration into the dense tumor stroma, allowing for deeper distribution.

 

2.   3. Self-Assembling Peptides

Beyond their targeting function, certain peptides are designed to self-assemble into nanostructures, improving drug stability and enabling controlled release. These peptides act as both a targeting moiety and a delivery vehicle.

Table 2. Peptides in peptide drug conjugates.

Function

Category

Representative Peptides

Target

Application

Targeting

Tumor-targeting

RGD, NGR, LS-7, GE11

Integrins, CD13, CD133, CD-47, EGFR

 

Solid tumors, angiogenesis

 

Hormone or ligand-derived

GnRH analogs,

Somatostatin analogs

 

SST2, GnRH receptor

Prostate, breast, neuroendocrine cancers

Penetrating

Cell-penetrating

TAT, Penetratin

Cell membrane (non-specific)

 

Enhances intracellular delivery

 

Brain-penetrating

Angiopep-2, RVG

BBB transport receptors

 

Brain tumors, CNS disorders

 

Tumor-penetrating

iRGD, LyP-1

Integrins + NRP-1, lymphatic markers

 

Enhances drug distribution within solid tumors

 

Self-assembling

Self-assembling

Amphiphilic peptides

Self-assembly motifs

Stability, controlled release

 

 

Improving the Stability and Pharmacokinetics of Peptides

While peptides provide excellent flexibility and tumor-targeting capability, their clinical translation faces important pharmacokinetic challenges. Compared with larger biomolecules such as antibodies, peptides generally exhibit: 1) short half-life due to rapid degradation by blood proteases. 2) Fast renal clearance, as peptides under ~25 kDa are readily filtered by the kidney glomeruli. 3) Low oral bioavailability, which makes oral dosing impractical in most cases. These limitations often require frequent dosing and restrict the broader application of PDCs. To overcome these hurdles, several strategies have been developed to enhance peptide stability, bioavailability, and circulation time. 

1. Cyclization and Peptide Stapling

Cyclization locks a peptide into a defined secondary structure (α-helix, β-sheet), improving both binding affinity and protease resistance. Common approaches include head-to-tail, head-to-side-chain, and side-chain-to-tail cyclization, which help restrict unwanted conformational changes during circulation. More advanced stapling techniques, such as one or two component peptide stapling, further stabilize peptides and have already produced clinical candidates like ALRN-6924. Bicyclic peptides, stabilized by multiple cross-links, offer an especially favorable “blood-to-tumor ratio,” enabling higher dosing without increased systemic toxicity.

Figure 3. Cyclization and peptide stapling methods.

2.  Amino Acids Modifications

Substituting natural L-amino acids with D-amino acids can reduce enzymatic degradation and extend half-life, as demonstrated by the improved stability of octreotide. Incorporating unnatural amino acids (UAAs) can fine-tune receptor binding and improve metabolic resistance. However, these modifications must be balanced carefully, as they may alter peptide folding or interactions.

Figure 4. Two general amino acids modification methods.

 3. Conjugation with Macromolecules

Attaching peptides to larger macromolecules can significantly prolong their circulation. Among these, PEGylation improves solubility and reduces renal clearance, as seen with HM-3, whose half-life increased nearly six-fold after PEG modification. Albumin-binding strategies also extend exposure by exploiting serum albumin’s long half-life, enabling convenient dosing schedules in drugs like liraglutide and semaglutide. Beyond these, other carriers such as fatty acid, polysialic acid (PSA), and hydroxyethyl starch (HES) have been explored to further enhance peptide stability and therapeutic potential. 

Figure 5. Conjugation with macromolecules methods.

4.   Formulation and Dosage Form Innovations

Innovative formulations also play a key role in improving peptide stability and bioavailability. pH-sensitive coatings can protect peptides from stomach acid and release them later in the intestine, while penetration enhancers facilitate transport across epithelial barriers to increase absorption. For injectable products, long-acting depots such as Sandostatin LAR, which encapsulates octreotide in PLGA microspheres, provide sustained drug release and reduce the frequency of dosing.

 

Figure 6. Formulation and dosage innovation methods.

Linkers

Just as in ADCs, the linker in a PDC is much more than a simple connector. It determines when and where the toxic payload is released. An effective linker must balance two requirements: remain stable in systemic circulation to prevent premature release, and trigger efficient cleavage once the conjugate reaches the tumor microenvironment or is internalized into target cells. Linkers used in PDCs can be broadly categorized into several classes.

  1. Non-cleavable linkers

    These linkers form a permanent bond between the peptide and the payload. Drug release occurs only after the entire conjugate is degraded inside the cell. Non-cleavable linkers, such as thioethers, oximes, and triazoles, provide strong stability in circulation and reduce the risk of premature release, but they may limit flexibility in controlling when the payload becomes active.

      2. Enzyme-sensitive linkers

    These linkers are designed to be cleaved by enzymes overexpressed in tumors, such as cathepsins or matrix metalloproteinases (MMPs). This technology uses specific peptide sequences like Val-Cit and Val-Ala.

     3.  pH-sensitive linkers

    Exploiting the slightly acidic environment of tumors and endosomes, pH-sensitive linkers release the payload under low-pH conditions while remaining intact in the bloodstream. This approach minimizes systemic toxicity and enhances tumor selectivity.

     4. Reduction-sensitive linkers

    Tumor cells often have higher intracellular glutathione (GSH) levels than normal tissues. Linkers containing disulfide bonds can be cleaved in this reducing environment, releasing the payload once inside the cancer cell.

    Table 3. Types of linkers in PDC and representative examples

    Linker types

    Representative examples

    Non-cleavable linkers

     

     

    Enzyme-sensitive linkers

     

     

    pH-sensitive linkers

     

     

    Reduction-sensitive linkers

     

     

    Compared with ADCs, PDC linkers are typically shorter and simpler, reflecting the smaller size of peptides. In many cases, linkers are also tailored to improve the overall stability of the conjugate, such as incorporating PEG chains to reduce proteolysis and extend circulation time. Importantly, the linker must be engineered so that conjugation does not disrupt the peptide’s binding affinity or the cytotoxicity of the payload.

     

    Payloads

    The payload is the effector component of a PDC, responsible for delivering the therapeutic impact once the conjugate reaches its target. While the peptide guides the conjugate to diseased tissue and the linker controls release, it is ultimately the payload that determines the cytotoxic potency, therapeutic scope, and safety profile of the drug. Payloads in PDCs can be categorized into several main classes depending on their therapeutic function.

    1.     Small targeted drugs

    Molecularly targeted inhibitors (e.g., kinase inhibitors, HDAC inhibitors) have better tumor selectivity than conventional chemotherapeutics but still suffer from resistance and systemic side effects. By conjugating them to peptides, their delivery can be refined, increasing efficacy while lowering nonspecific toxicity.

    2.     Chemotherapeutic drugs

    Traditional cytotoxins such as doxorubicin (DOX), paclitaxel (PTX), camptothecin (CPT), and platinum-based agents remain the most widely used payloads. Conjugation with peptides helps to overcome their inherent drawbacks such as poor solubility, short half-life, drug resistance, and system toxicity, by enhancing tumor targeting and improving their safety profile.

    3.     Radionuclides

    Radioactive isotopes serve both diagnostic and therapeutic purposes. Clinically approved examples include 111In-DTPA-octreotide (Octreoscan) for neuroendocrine tumor imaging, and 177Lu-DOTATATE, which prolongs progression-free survival in patients with neuroendocrine cancers. Other radionuclides such as 68Ga, 64Cu, 18F, 123I, and 99mTc are also used in preclinical and clinical studies.

    4.     Anti-cancer peptides

    Certain peptides have intrinsic cytotoxic or pro-apoptotic activity, such as lytic peptides and the KLA pro-apoptotic peptide. When combined with tumor-targeting or cell-penetrating peptides, these payloads can achieve potent anti-tumor effects with improved safety, as shown in VEGFR-targeting peptide conjugates like QR-KLU.

    5.     Biomacromolecules

    Protein-based drugs and nucleic acids are promising payloads for difficult-to-treat diseases but face challenges in stability and delivery. Examples include NGR-TNF, which improves tumor selectivity while reducing systemic toxicity. Despite their potential, poor pharmacological properties limit their direct clinical translation, and many are now being studies in nanoparticle formulations.

    6.     Gas molecules

    Therapeutic gases such as NO, H2S, O2, CO, and SO2 are being explored for cancer therapy because they cause few side effects and rarely induce resistance. Linking them to peptides allows controlled release and offers new ways to tackle multidrug resistance and tumor relapse, though this approach is still in its early stages.

     

    Selecting a payload for PDCs requires balancing multiple factors. First, the payload must be highly potent, as only a limited number of molecules are delivered to each tumor cell. Second, it must be chemically compatible with peptide conjugation, retaining its anti-tumor activity without disrupting the peptide’s binding affinity. Third, the payload should offer an acceptable safety margin, with the targeting peptide improving selectivity while minimizing systemic toxicity. Finally, favorable drug-like properties such as small molecular weight, adequate stability, and resistance to premature degradation are critical for ensuring clinical success.

     

    Conclusion

    Peptide-drug conjugates are emerging as a complementary modality to antibody-drug conjugates, offering advantages such as better tumor penetration, lower immunogenicity, and easier synthesis. However, challenges remain, including rapid clearance, limited stability, and the need for finely tuned linkers and payloads. Future progress will depend on innovations in peptide design, linker chemistry, and payload diversity. Among these, smart linkers that combine stability in circulation with precise drug release are especially critical.

    At PrecisePEG, we specialize in advanced linker technologies, from PEG-based linkers that enhance stability to both non-cleavable and cleavable linkers optimized for both ADCs and PDCs.

    Explore our offerings at www.precisepeg.com or contact with us at sales@precisepeg.com for custom solutions.

     

    Reference

    1.     Chen, F.; Yu, L.; Miao, Y.; Liu, X.; Yu, Z.; Wei, M. Peptide–drug conjugates (PDCs): a novel trend of research and development on targeted therapy, hype or hope? Acta Pharm. Sin. B 2023, 13, 498–516. https://doi.org/10.1016/j.apsb.2022.07.020.

    2.     Alas, M.; Saghaeidehkordi, A.; Kaur, K. Peptide–Drug Conjugates with Different Linkers for Cancer Therapy. J. Med. Chem. 2021, 64, 216–232. https://doi.org/10.1021/acs.jmedchem.0c01530.

    3.     Cooper, B. M.; Iegre, J.; O' Donovan, D. H.; Ölwegård Halvarsson, M.; Spring, D. R. Peptides as a platform for targeted therapeutics for cancer: peptide–drug conjugates (PDCs). Chem. Soc. Rev. 2021, 50, 1480–1494. https://doi.org/10.1039/D0CS00556H.

    4.     Rizvi, S. F. A.; Zhang, L.; Zhang, H.; Fang, Q. Peptide-Drug Conjugates: Design, Chemistry, and Drug Delivery System as a Novel Cancer Theranostic. ACS Pharmacol. Transl. Sci. 2024, 7, 309–334. https://doi.org/10.1021/acsptsci.3c00269.

    5.     Wang, D.; Yin, F.; Li, Z.; Zhang, Y.; Shi, C. Current progress and remaining challenges of peptide–drug conjugates (PDCs): next generation of antibody-drug conjugates (ADCs)? J. Nanobiotechnol. 2025, 23, 305. https://doi.org/10.1186/s12951-025-03277-2.

     

     

     

     

Previous
Back to Advancing Innovation, Research, and News at Precise PEG