What Small molecule-drug conjugates are being developed?

17 March 2025
Introduction to Small Molecule‑Drug Conjugates

Small molecule‑drug conjugates (SMDCs) represent an innovative class of targeted therapeutic agents designed to combine the advantageous properties of small molecules with the precision of targeting moieties. Unlike antibody‑drug conjugates (ADCs), which use large immunoglobulins for the selective delivery of potent cytotoxic drugs, SMDCs employ small molecule ligands as the targeting elements. This approach has attracted increasing attention in recent years because of the potentially simpler manufacturing processes, reduced immunogenicity, and improved tumor penetration associated with smaller chemical entities. In what follows, we discuss SMDCs in a general‑specific‑general structure—starting with their definition and historical context, then explaining current developments and innovations from various perspectives, and finally broadening the discussion to applications, benefits, challenges, and future directions.

Definition and Mechanism of Action

SMDCs are hybrid molecules that consist of three core components: a small molecule targeting ligand, a chemical linker, and a potent cytotoxic payload. The targeting ligand is usually selected for its high affinity toward a particular biomarker found predominantly on diseased tissue (e.g., a tumor‑specific antigen). The chemical linker is designed to be stable during circulation but to undergo controlled cleavage in response to triggers in the tumor microenvironment—such as low pH, high glutathione levels, or specific protease activity—thereby releasing the cytotoxic agent selectively at the disease site. The payload, typically a highly potent chemotherapeutic drug, exerts its cell‑killing activity once liberated inside the target cell.

The mechanism of action for an SMDC starts with the ligand binding to its specific target expressed on the surface of a cell. On binding, the conjugate is internalized through receptor‑mediated endocytosis. Once inside the cell, the linker undergoes cleavage triggered by specific conditions—for instance, acidic endosomal pH or tumor‑associated enzymes—resulting in the release of the active small molecule drug. This precise release mechanism minimizes systemic exposure to the cytotoxic payload and maximizes the therapeutic index, thereby reducing off‑target toxicity compared to conventional chemotherapies.

Historical Development and Evolution

The concept of targeted drug delivery can be traced back several decades, evolving in parallel with the development of monoclonal antibodies and other targeting moieties. Early work on antibody–drug conjugates in the 1980s and 1990s laid the foundation for selectively delivering cytotoxic agents; however, the recognition of limitations such as heterogeneity, immunogenicity, and poor tissue penetration motivated researchers to explore alternative carriers. Small molecule ligands offered an attractive alternative. Unlike antibodies, small molecules can be chemically synthesized in a more controlled manner with reproducible structures, rendering them more conducive to large‑scale production and allowing for the fine‑tuning of binding and pharmacokinetic properties.

Over time, advances in medicinal chemistry, chemical biology, and linker technology have enabled the design of SMDCs that not only mimic the targeting ability of ADCs but also address some of their inherent challenges. Early SMDC prototypes relied on the established prodrug concept, while more recent research has focused on incorporating sophisticated cleavable linkers—such as disulfide bonds and pH‑sensitive hydrazine linkers—to ensure controlled release of the payload only at the tumor site. This evolution has been guided by improvements in our understanding of the tumor microenvironment and the molecular mechanisms that control receptor‑mediated endocytosis, thereby driving continuous optimization of SMDC designs.

Current Developments in Small Molecule‑Drug Conjugates

Recent advances in SMDC research have resulted in a variety of new constructs that are being developed by academic laboratories and industry alike. These developments span novel linker chemistries, innovative payloads, and dual‑functionality platforms that combine diagnostic and therapeutic capabilities (theranostics). In addition, strategic innovations have been aimed at improving the stability, specificity, and overall pharmacokinetic properties of SMDCs.

Leading Research and Development Entities

A number of research groups and companies are actively involved in the development of SMDCs. Early academic work has demonstrated the feasibility of constructing SMDCs by conjugating small molecule ligands to cytotoxic drugs via specially designed linkers. For instance, one study reported the design of new thioester‑linked maytansinoid conjugates, where the thioester linkage helped to prevent premature release of the payload during systemic circulation. Such advancements underline the importance of integrating medicinal chemistry with pharmacological considerations.

Leading biopharmaceutical companies are also investing resources into SMDC platforms to capitalize on their potential advantages. There is growing interest among companies that have previously focused on ADC technologies to expand their pipelines to include SMDCs. These entities are leveraging established expertise in linker and payload conjugation chemistries while shifting toward targeting ligands that are chemically synthesized and have low molecular weight. Furthermore, collaborations between academic groups and industry partners are increasingly being reported, with promising candidates emerging in pre‑clinical studies. This trend is fueling a renaissance in the excitement surrounding targeted chemotherapeutics that use small molecule conjugation strategies.

Additionally, several academic laboratories are exploring innovative targeting moieties. For example, studies on targeting phosphatidylserine (a marker present in the tumor microenvironment) using zinc‑dipicolylamine as the ligand have yielded promising SMDCs. Other research highlights include the development of SMDCs targeting carbonic anhydrase IX for renal cell carcinoma and PSMA‑targeted conjugates for prostate cancer treatment. These initiatives demonstrate that SMDCs are being tailored to a variety of cancer types and are being optimized for their specific target environments.

Recent Innovations and Breakthroughs

Recent years have witnessed significant breakthroughs in the design and synthesis of SMDCs. Some of the most notable innovations include:

Advanced Cleavable Linkers:
Innovations such as pH‑sensitive linkers and redox‑responsive disulfide bonds have enabled researchers to create SMDCs with precise control over drug release. For example, pH‑labile linkers that release payloads at the lower pH levels found in endosomes, or disulfide bonds that are cleaved by high glutathione concentrations in tumor cells, have been successfully employed. These advances result in improved stability in systemic circulation while ensuring rapid release within target cells.

Novel Payload Strategies:
New cytotoxic payloads are being incorporated into SMDC constructs, including maytansinoids, auristatins, and SN38 derivatives. Thioester‑linked maytansinoid conjugates have been optimized for in vitro cytotoxicity and favorable pharmacokinetics, with studies demonstrating promising tumor regression in triple‑negative breast cancer xenograft models. Other payloads, such as DM1 (a potent antimicrotubule agent), have been chemically linked to small molecule targeting units such as PSMA‑binding ligands for prostate cancer treatment.

Theranostic and Dual‑Function Platforms:
Researchers are exploring SMDCs with theranostic capabilities—that is, conjugates that combine therapeutic activity with imaging functionality. For instance, one innovative design described a theranostic SMDC for prostate cancer that integrated a polyethylene glycol (PEG) scaffold with a chelating moiety for positron emission tomography (PET) imaging, a PSMA‑specific ligand, and a cytotoxic drug (DM1) for chemotherapy. This dual‑function approach not only allows for targeted therapy but also for non‑invasive imaging to monitor biodistribution and tumor uptake in real time.

Enhanced Delivery through Structural Optimization:
Efforts have been made to improve tissue penetration and overall pharmacokinetics by optimizing the chemical structure of the small molecule ligands and linkers. SMDCs benefit from their lower molecular weight compared to ADCs, allowing for rapid tumor infiltration and clearance from non‑target tissues. This structural optimization may lead to reduced off‑target toxicity and improved safety profiles. For example, targeting ligands designed for carbonic anhydrase IX have shown rapid internalization and effective tumor suppression in renal cell carcinoma models.

Innovative Conjugation Techniques:
The development of reliable and modular conjugation chemistries has been instrumental in SMDC advancement. Techniques such as click chemistry have been applied to reliably attach therapeutic payloads to small molecule targeting moieties, ensuring high conversion rates and precise control over the stoichiometry of the drug conjugates. These advancements in conjugation methodology significantly contribute to the reproducibility and scalability of SMDC production for clinical applications.

Applications and Potential Benefits

The applications of SMDCs are being explored across a broad spectrum of therapeutic areas—most notably oncology, but also in emerging fields such as immunotherapy and targeted drug delivery for other diseases. SMDCs are developed to address several unmet clinical needs by effectively delivering potent payloads while minimizing systemic toxicity.

Therapeutic Areas and Indications

Cancer Therapy:
Cancer remains the most common target for SMDC development. Given the aggressive nature of many cancers and the frequent development of drug resistance to conventional chemotherapy, targeted delivery systems such as SMDCs are highly attractive. Specific SMDC candidates include:
- Phosphatidylserine-Targeted Conjugates: SMDCs employing zinc‑dipicolylamine targeting phosphatidylserine have demonstrated significant antitumor efficacy across a range of cancer models such as pancreas, prostate, colon, liver, breast, and glioblastoma.
- Carbonic Anhydrase IX Targeting: A conjugate designed for the treatment of carbonic anhydrase IX expressing tumors has demonstrated preferential accumulation and potent antitumor activity in renal cell carcinoma models.
- Prostate-Specific Membrane Antigen (PSMA) Targeted SMDCs: Various constructs have been developed for prostate cancer treatment, including theranostic agents that combine PET imaging with a cytotoxic payload, thereby enabling real-time tracking of drug distribution and effective tumor suppression.

Immunotherapy:
While most SMDC efforts have focused on cytotoxic payload delivery for cancer therapy, there is also growing interest in combining SMDC technology with immunomodulatory agents. For example, some approaches integrate toll-like receptor (TLR) agonists into SMDC formulations to provoke an immune response and create a “cold-to-hot” tumor conversion that may work synergistically with other immunotherapies.

Other Therapeutic Indications:
Beyond oncology, the modular design of SMDCs is being considered for other diseases where targeted delivery may enhance efficacy and reduce side effects. Although the majority of current efforts are concentrated in cancer therapy, the generality of the conjugation approach allows expansion into other areas such as anti-inflammatory or anti-infective therapies. In addition, some research has examined SMDCs for applications in regenerative medicine and even targeted delivery in chronic diseases, leveraging the ease of tuning the pharmacokinetic properties of these agents.

Advantages Over Traditional Therapies

SMDCs offer several potential advantages compared to traditional chemotherapy and even ADCs:

Improved Tumor Penetration:
The small size of SMDC constructs enables deeper tissue penetration, particularly in solid tumors, which often have dense and heterogeneous structures that restrict the passage of larger molecules such as monoclonal antibodies. This advantage can translate into more effective drug delivery and higher localized concentrations of the cytotoxic payload.

Enhanced Pharmacokinetics and Safety Profile:
SMDCs are engineered to have a precise drug-to-ligand ratio and a controlled release mechanism. This design minimizes premature payload release during circulation, thereby reducing systemic toxicity. Moreover, the faster clearance from non‑target tissues further improves the safety profile relative to conventional chemotherapeutics.

Lower Immunogenicity:
As SMDCs are built upon low molecular weight targeting ligands, they are less likely to evoke immune responses compared to biologics, which are often associated with immunogenicity concerns. This reduction in immunogenicity can improve patient tolerability and allow for repeated dosing regimens.

Scalability and Manufacturing Efficiency:
The synthetic routes employed in SMDC production are typically more straightforward and reproducible than those used for protein-based therapeutics. This chemical approach allows for scalable production with consistent quality control, potentially reducing costs and accelerating translation from bench to clinic.

Modularity and Flexibility:
One of the most significant benefits of SMDCs is the modularity inherent in their design. Researchers can swap out targeting moieties, linkers, or payloads to tailor the conjugate to specific indications or to improve performance as needed. Dual‑functional or theranostic SMDCs, for instance, combine therapeutic and diagnostic features within the same molecule, offering unparalleled versatility in personalized medicine.

Challenges and Future Prospects

Despite the many potential benefits, the development of SMDCs still faces several challenges. Addressing these hurdles is critical for the translation of promising pre‑clinical candidates into safe and effective clinical therapeutics.

Current Challenges in Development

Linker Stability and Premature Release:
One of the most critical aspects of SMDC development is ensuring that the linker, which connects the targeting ligand to the cytotoxic payload, remains stable during systemic circulation but is efficiently cleaved upon reaching the target site. Inappropriate linker stability can result in premature release of the drug, leading to off‑target toxicity and reduced efficacy. Although recent innovations—such as disulfide bonds and pH‑sensitive linkers—have improved these properties, ensuring absolute control over the release process remains a challenge.

Heterogeneity and Precise Stoichiometry:
While SMDCs benefit from the relatively straightforward synthesis of small molecules compared to antibodies, achieving a uniform and precise conjugation ratio between the ligand and the drug is still essential. Variability in the drug-to-ligand ratio could lead to differences in pharmacokinetics, efficacy, and safety. Advanced chemical synthesis and purification techniques are being developed to overcome this hurdle; however, further progress is needed to guarantee the production of homogeneous and reproducible SMDC batches.

Target Specificity and Off-Target Effects:
The success of SMDCs is contingent upon the targeting ligand’s affinity and specificity for markers overexpressed in diseased tissues. Although several targets have been validated in pre‑clinical models—such as PSMA for prostate cancer, carbonic anhydrase IX for renal cancer, and phosphatidylserine as a universal tumor marker—variability in target expression across patients and potential cross‑reactivity with normal tissues may limit therapeutic windows. Therefore, careful target selection and validation remain ongoing priorities to minimize off‑target toxicity.

Pharmacokinetics and Tissue Penetration:
Although a smaller molecular size confers some advantages in tissue penetration, it can also lead to rapid clearance from systemic circulation. Balancing these two opposing aspects—enhanced tumor penetration versus sufficient time in circulation for target accumulation—is a significant formulation challenge. Structural modifications, such as incorporating PEG scaffolds, can be used to optimize pharmacokinetics, yet they must be finely tuned to avoid compromising the ligand’s targeting efficiency or payload release kinetics.

Evaluation and Regulatory Challenges:
Given that SMDCs are relatively new compared to traditional small molecule drugs and ADCs, standardized pre‑clinical models and regulatory guidelines for their evaluation are still evolving. This situation can slow the clinical development process, as safety and efficacy endpoints may not be fully defined initially. Collaborative efforts between regulatory bodies, academia, and industry are required to establish robust guidelines and expedite the clinical translation of these novel agents.

Future Research Directions and Opportunities

Looking forward, there are several promising directions that could help overcome these challenges and further enhance the therapeutic potential of SMDCs:

Next‑Generation Linker Technologies:
Continued research into novel linkers that respond more precisely to the unique conditions within the tumor microenvironment will likely yield even more effective SMDC designs. Efforts aimed at optimizing disulfide bonds, hydrazone linkers, and even enzyme‑cleavable peptides are expected to advance, ensuring that payload release occurs only when and where it is needed.

Advances in Target Validation and Biomarker Discovery:
Improving the selection of appropriate targets by harnessing cutting‑edge techniques in genomics and proteomics can help identify tumor‑specific markers. The development of companion diagnostics to assess target expression in patients will facilitate personalized SMDC therapies, ensuring that only those individuals with the proper biomarker profile receive the treatment.

Modular and Dual‑Functional Conjugate Platforms:
The future of SMDCs may involve the design of multifunctional conjugates that not only deliver a cytotoxic drug but also include diagnostic or immunomodulatory elements within a single molecule. The successful incorporation of imaging agents into SMDCs, as demonstrated in theranostic constructs for prostate cancer, is one promising avenue. These dual‑function platforms will allow clinicians to monitor drug distribution in real time, optimize dosing strategies, and potentially combine therapy with targeted immunomodulation.

Improved Conjugation Chemistry and Manufacturing Processes:
The continued development of robust chemical methodologies such as click chemistry for high‑yield and site‑specific conjugation will further improve the homogeneity and scalability of SMDC production. Advances in chemical synthesis and purification technologies will help to standardize the production of SMDCs, reducing batch‑to‑batch variability and facilitating regulatory approval.

Exploration Beyond Oncology:
Although the majority of SMDC research focuses on cancer, the potential applications of these conjugates extend into other therapeutic areas. Research into SMDCs for autoimmune diseases, infectious diseases, and regenerative medicine is emerging. The modularity of SMDC design means that once proven in oncology, similar strategies could be adapted to target dysregulated pathways in other pathologies, thereby broadening their therapeutic impact.

Computational Modeling and Systems Pharmacology:
Leveraging computational methods to predict pharmacokinetic behavior, target engagement, and the dynamics of linker cleavage can help optimize SMDC design before in‑vitro testing begins. Systems pharmacology approaches that integrate in silico ADME/T modeling, target fishing, and network pharmacology analyses are expected to play an increasingly important role in tailoring SMDCs to the complex interactions within the human body.

Clinical Trials and Collaborative Networks:
Increased collaboration between academic research centers, biotech companies, and pharmaceutical giants will likely accelerate the progression of SMDCs from pre‑clinical studies to clinical trials. Establishing multicenter clinical trials and standardized endpoints specifically for SMDCs will help build a stronger data set for evaluating safety and efficacy, ultimately paving the way for regulatory approvals and commercialization.

Conclusion

In summary, small molecule‑drug conjugates are being developed as a promising alternative to traditional chemotherapy and antibody‑drug conjugates. SMDCs are composed of a targeting small molecule, an intelligent linker engineered to respond to the unique conditions of the tumor microenvironment, and a potent cytotoxic payload capable of inducing cell death upon release. Their historical evolution—from the early days of prodrug strategies to the incorporation of advanced cleavable linkers and theranostic elements—demonstrates the rapid pace of innovation in this field. Current developments highlight cutting‑edge research from both academic laboratories and leading biopharmaceutical companies, with breakthroughs in linker stability, payload selection, and modular conjugation techniques pushing the boundaries of what is achievable. Applications in cancer therapy, particularly for targets such as PSMA, carbonic anhydrase IX, and phosphatidylserine, attest to the potential of SMDCs to deliver improved therapeutic efficacy with reduced systemic toxicity. Advantages such as improved tumor penetration, lower immunogenicity, and manufacturing efficiencies further elevate the promise of these agents.

Nonetheless, challenges remain, including the need to balance linker stability with rapid release, achieving precise stoichiometry in the conjugates, ensuring target specificity, and optimizing pharmacokinetics. Future research directions are focused on developing next‑generation linkers, integrating dual‑functional platforms that combine therapy with diagnostic capability, enhancing computational modeling to fine‑tune designs, and expanding the application of SMDC technology beyond oncology. Collaborative efforts among industry, academia, and regulatory bodies are essential to overcome these hurdles and to accelerate the clinical translation of SMDCs.

In conclusion, the field of small molecule‑drug conjugates is at an exciting crossroads. With extensive research addressing key technical challenges and the accumulation of promising pre‑clinical data, SMDCs represent a dynamic and evolving area of therapeutic innovation. Their potential to revolutionize targeted therapy—by delivering potent drugs specifically to diseased tissue with minimal side effects—could transform treatment paradigms not only in oncology but across a spectrum of challenging diseases. The ongoing research and collaborative momentum in this field underscore the optimism that SMDCs will be integral to the next generation of precision medicines, ultimately improving patient outcomes and expanding the therapeutic arsenal available to clinicians.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.