Barcelona, Spain – A significant shift is underway in the treatment of non-small cell lung cancer (NSCLC), driven by innovative antibody-drug conjugates (ADCs). These agents, combining the precision of antibodies with the potency of cytotoxic drugs, are providing fresh therapeutic avenues for patients, as detailed at the 2025 World Conference on Lung Cancer.
Understanding the Architecture of Antibody-Drug Conjugates
Table of Contents
- 1. Understanding the Architecture of Antibody-Drug Conjugates
- 2. The Critical Role of the Payload
- 3. How Antibody-Drug Conjugates Work
- 4. Biomarker-Driven vs. Broad-spectrum ADCs
- 5. Recent Approvals and Clinical Trial Data
- 6. Sacituzumab Tirumotecan: A New Option for EGFR-Mutated NSCLC
- 7. Challenges and Future Directions
- 8. Looking Ahead: The Future of ADCs
- 9. Frequently Asked Questions about Antibody-Drug Conjugates
- 10. What are the key components of an antibody-drug conjugate (ADC) and how does each contribute to its function?
- 11. Antibody-Drug Conjugates in Advanced Non-Small Cell Lung Cancer: Expanding Therapeutic Roles and opportunities
- 12. Understanding Antibody-Drug Conjugates (ADCs)
- 13. Current ADC Landscape in NSCLC Treatment
- 14. Mechanisms of Action & How adcs Overcome Resistance
- 15. Biomarker Selection and Patient Stratification
- 16. Clinical Trial updates & Future Directions
giannis Mountzios, MD, MSc, PhD, a leading medical oncologist from the Henry Dunant Hospital Center in Athens, Greece, outlined the basic structure of ADCs. These complex molecules comprise three essential components: the antibody, the linker, and the payload. The antibody, typically an IgG1 monoclonal antibody, is engineered for exceptional target specificity, guiding the ADC directly to cancer cells and also activating immune responses.
The linker component is critical, determining whether the payload is released inside cancer cells only (non-cleavable) or both inside and outside the cell (cleavable).This distinction significantly impacts how the drug behaves in the body. the payload, a potent cytotoxic molecule, disrupts cellular processes – DNA structure or tubulin polymerization – leading to cancer cell death. Newer ADC designs include bispecific and biparatopic agents, expanding targeting capabilities, as well as immune-stimulating and protein-degrader ADCs.
The Critical Role of the Payload
The payload’s characteristics generally define an ADC’s therapeutic strength and potential toxicity. These payloads fall into three main groups: antimicrotubule agents, DNA-cleaving agents, and topoisomerase inhibitors. mountzios likened payload design to spacecraft engineering, where the payload’s size and weight influence the overall ADC structure and behavior. As an example, sacituzumab govitecan and datopotamab deruxtecan, both targeting TROP2, demonstrate differing potencies due to variations in their payloads and drug-to-antibody ratios.
How Antibody-Drug Conjugates Work
The process begins with the antibody binding to a specific target on the cancer cell’s surface.The ADC is then absorbed into the cell, where the payload is released via lysosomal degradation. This released payload then interferes with vital cellular functions, ultimately causing cell death. Importantly, the antibody itself can contribute to anti-cancer activity through immune mechanisms, enhancing the overall therapeutic effect. Some payloads can also diffuse into neighboring cells, offering a ‘bystander effect’ useful for cancers with uneven biomarker expression, but also carrying a risk of off-target toxicity.
Biomarker-Driven vs. Broad-spectrum ADCs
ADCs are categorized as either biomarker-selected or biomarker-agnostic. Biomarker-selected ADCs, such as telisotuzumab vedotin for MET overexpression and trastuzumab deruxtecan for HER2 mutations, require the presence of a specific biomarker for effectiveness.Conversely, biomarker-agnostic ADCs, like those targeting Trop2 or Her3, demonstrate efficacy across a wider patient population, irrespective of biomarker levels. This distinction is crucial for tailoring treatment strategies.
Recent Approvals and Clinical Trial Data
Recent years have witnessed several key approvals. Trastuzumab deruxtecan received accelerated FDA approval in August 2022 for HER2-mutant NSCLC, demonstrating over 50% objective response rates and a median overall survival approaching 18 months. Telisotuzumab vedotin gained accelerated approval in May 2025 for patients with high c-Met protein overexpression, while datopotamab deruxtecan followed in June 2025 for pre-treated EGFR-mutant NSCLC. Sacituzumab tirumotecan was also approved in China in March 2025 for EGFR-positive NSCLC following promising Phase 2 trial results.
Though, progress isn’t always linear. The HERTHENA-Lung02 study of patritumab deruxtecan was halted after failing to show a significant overall survival benefit, highlighting the challenges in ADC development.
Sacituzumab Tirumotecan: A New Option for EGFR-Mutated NSCLC
Results from the phase 2 OptiTROP-Lung03 trial demonstrated that sacituzumab tirumotecan significantly improved progression-free survival in patients with pretreated EGFR-mutated NSCLC compared to docetaxel, with a median progression-free survival of 6.9 months versus 2.8 months. These encouraging results have broadened the treatment landscape for this patient subset.
Challenges and Future Directions
Despite significant progress, challenges remain. Ensuring target selectivity to minimize off-tumor effects is crucial, as is addressing potential resistance mechanisms.Combination therapies – ADCs with immunotherapies or chemotherapy – are being actively investigated, but require careful management to avoid overlapping toxicities.
“We have key challenges remaining,” Mountzios stated. “improving target selection, identifying robust biomarkers, reducing off-tumor toxicity, and determining optimal combination strategies are paramount.”
Did You Know? ADCs represent a convergence of biological and chemical engineering, requiring precise control over antibody specificity, linker stability, and payload potency.
Pro Tip: Biomarker testing is crucial for identifying patients most likely to benefit from biomarker-selected ADCs.
What role do you foresee for ADCs in personalized cancer treatment? How can we best manage the potential toxicities associated with these powerful therapies?
Looking Ahead: The Future of ADCs
The field of ADCs is rapidly evolving, with ongoing research focused on novel payloads, linkers, and antibody designs. Next-generation ADCs aim to overcome current limitations, enhance efficacy, and expand the range of treatable cancers. The development of more elegant biomarkers will also play a vital role in optimizing patient selection and maximizing treatment outcomes.
| ADC | Target | FDA Approval Date | NSCLC Indication |
|---|---|---|---|
| Trastuzumab Deruxtecan | HER2 | August 11, 2022 | HER2-mutant NSCLC |
| Telisotuzumab Vedotin | MET | May 14, 2025 | High c-Met overexpression |
| Datopotamab deruxtecan | EGFR | June 23, 2025 | EGFR-mutant NSCLC |
Frequently Asked Questions about Antibody-Drug Conjugates
- What are antibody-drug conjugates? ADCs are a targeted cancer therapy that combines the specificity of antibodies with the cell-killing power of chemotherapy drugs.
- How effective are ADCs in NSCLC? ADCs have shown promising results in NSCLC, particularly in patients with specific biomarkers like HER2 or EGFR mutations.
- What are the common side effects of ADCs? Common side effects can include nausea, fatigue, and low blood cell counts. More serious side effects can occur and require careful monitoring.
- Are ADCs suitable for all NSCLC patients? ADCs are most effective for patients whose tumors express the target biomarker.
- What is the future of ADC research? Future research is focused on developing more selective and potent ADCs with fewer side effects.
- How do biomarker-agnostic ADCs differ from biomarker-selected ADCs? Biomarker-agnostic ADCs can work across a wider population, while biomarker-selected ones need specific biomarkers present.
- What role does the linker play in ADC effectiveness? The linker determines how and where the payload is released, impacting both efficacy and toxicity.
Share your thoughts and experiences with cancer treatment in the comments below!
What are the key components of an antibody-drug conjugate (ADC) and how does each contribute to its function?
Antibody-Drug Conjugates in Advanced Non-Small Cell Lung Cancer: Expanding Therapeutic Roles and opportunities
Understanding Antibody-Drug Conjugates (ADCs)
Antibody-drug conjugates (ADCs) represent a significant advancement in targeted cancer therapy, particularly in the treatment of advanced non-small cell lung cancer (NSCLC). These complex molecules combine the specificity of monoclonal antibodies with the potent cytotoxic activity of chemotherapy drugs. Essentially, ADCs act as “guided missiles,” delivering chemotherapy directly to cancer cells while minimizing damage to healthy tissues. this targeted approach is crucial in NSCLC, where systemic chemotherapy often comes with debilitating side effects.
Key components of an ADC include:
Antibody: Selectively binds to a specific antigen highly expressed on cancer cells.
Linker: Chemically connects the antibody and the cytotoxic drug. linker stability is critical – it must remain intact in circulation but release the drug inside the cancer cell.
Payload (Cytotoxic Drug): The potent agent that kills the cancer cell. Common payloads include microtubule inhibitors (like monomethyl auristatin E – MMAE) and DNA damaging agents.
Current ADC Landscape in NSCLC Treatment
Currently, several ADCs are approved or in clinical trials for NSCLC. The most prominent example is sacituzumab govitecan, approved for metastatic NSCLC whose tumors express high levels of Trop-2. This ADC has demonstrated significant clinical benefit in patients who have progressed after platinum-based chemotherapy and PD-1/PD-L1 inhibitors.
Other adcs under inquiry target different antigens, including:
HER2: While traditionally associated with breast cancer, HER2 overexpression is found in a subset of NSCLC patients. Several HER2-targeted ADCs are in advancement.
EGFR: Mutations in EGFR are common in NSCLC. ADCs targeting EGFR are being explored, particularly for patients who develop resistance to EGFR tyrosine kinase inhibitors (TKIs).
DLL3: A Notch ligand frequently expressed in small cell lung cancer (SCLC) and some NSCLC subtypes.
CEACAM5: Expressed in a variety of solid tumors, including NSCLC.
Mechanisms of Action & How adcs Overcome Resistance
ADCs exert their anti-cancer effects through several mechanisms:
- Direct Cell Killing: Upon binding to the target antigen, the ADC is internalized into the cancer cell. The linker is cleaved (either by enzymes within the cell or through pH changes), releasing the cytotoxic payload, which then induces cell death.
- Bystander Effect: Some released payloads can diffuse out of the targeted cell and kill neighboring cancer cells, even if they don’t express the target antigen. This is particularly beneficial in heterogeneous tumors.
- Immune Stimulation: ADC treatment can trigger an immune response against the cancer cells, further enhancing anti-tumor activity.
Overcoming resistance to conventional therapies is a major challenge in NSCLC.ADCs offer potential solutions:
Targeting Option Pathways: ADCs can target antigens different from those targeted by TKIs or immunotherapy, providing a treatment option for patients who have developed resistance.
Circumventing Efflux Pumps: Some cancer cells develop resistance by pumping chemotherapy drugs out of the cell. ADCs can bypass these efflux pumps by delivering the drug directly into the cell.
Addressing Tumor Heterogeneity: The bystander effect can help overcome resistance caused by variations in antigen expression within the tumor.
Biomarker Selection and Patient Stratification
Identifying the right patients for ADC therapy is crucial. Biomarker testing plays a vital role in patient selection.
Trop-2 Expression: For sacituzumab govitecan, high Trop-2 expression is a key biomarker for predicting response. immunohistochemistry (IHC) is used to assess Trop-2 levels in tumor samples.
HER2 Status: Patients with HER2-positive NSCLC are candidates for HER2-targeted ADCs. IHC and fluorescence in situ hybridization (FISH) are used to determine HER2 status.
PD-L1 Expression: While not a direct biomarker for ADC response, PD-L1 expression can influence treatment decisions, as ADCs might potentially be used in combination with immunotherapy.
Developing more robust and reliable biomarkers is an ongoing area of research. Liquid biopsies,analyzing circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs),are emerging as promising tools for monitoring ADC response and detecting resistance mechanisms.
Clinical Trial updates & Future Directions
Numerous clinical trials are evaluating adcs in various NSCLC settings. Recent data from trials are showing promising results, particularly in heavily pre-treated patients.
Future research directions include:
Novel Antibody Targets: Identifying new antigens specifically expressed on NSCLC cells.
improved Linker Technology: Developing more stable and cleavable linkers to optimize payload delivery.
next-Generation Payloads: Exploring more potent and selective cytotoxic drugs.
* Combination Therapies: Investigating the synergistic effects of adcs with