How PROTAC Degraders Are Revolutionizing Cancer Therapy
For decades, the epidermal growth factor receptor (EGFR) has been both a beacon of hope and a source of frustration in oncology. As a key driver in numerous cancers—including non-small cell lung cancer (NSCLC), colorectal cancer, and head and neck cancers—EGFR has been the target of some of the most successful precision medicines ever developed.
The journey began with first-generation inhibitors like gefitinib and erlotinib, which delivered impressive responses but eventually succumbed to resistance, primarily through the T790M mutation 3 .
This led to second-generation drugs like afatinib and third-generation solutions like osimertinib, each overcoming previous limitations but ultimately facing new resistance challenges such as the notorious C797S mutation .
The pattern became familiar: each breakthrough eventually met its match in the form of new resistance mutations. This therapeutic arms race has highlighted the fundamental limitation of traditional inhibitors—they merely block EGFR's function temporarily, while cancer cells often find alternative pathways to survive and proliferate.
With approximately 50% of NSCLC patients in Asia harboring EGFR mutations , and global significance of these driver mutations, the medical need for more durable solutions has never been more pressing. The question has shifted from whether we can develop the next inhibitor to whether we can fundamentally change our approach to targeting EGFR altogether.
PROTACs (Proteolysis-Targeting Chimeras) represent a paradigm shift in therapeutic approaches, moving beyond simple inhibition to actual destruction of disease-causing proteins.
These innovative molecules function as cellular demolition crews with a unique triple-element structure:
Recognizes the protein of interest (in this case, EGFR)
Engages the cellular protein degradation machinery
Optimally spaces these two elements for efficient degradation
The magic of PROTACs lies in their catalytic nature—they don't merely block one molecule of EGFR at a time but can facilitate the destruction of multiple EGFR proteins through repeated cycles of binding, ubiquitination, and degradation.
"This technology has created excitement not just in academic circles but also in the pharmaceutical industry, where companies are racing to develop PROTAC-based therapies for various targets, with EGFR being among the most prized."
The field of EGFR degraders represents a convergence of multiple technological domains—medicinal chemistry, structural biology, protein degradation science, and oncology therapeutics. This complexity is reflected in the sophisticated patent strategies being employed to protect these innovations.
Modern patent filings in the EGFR degrader space typically employ a layered claiming strategy that builds defensive networks around core inventions.
Covering the specific degrader molecules with structural variations and modifications.
For treating various cancers and mutation profiles, including combination therapies.
Covering pharmaceutical compositions, delivery systems, and dosage forms.
Protecting manufacturing methods, purification techniques, and scale-up processes.
The high-stakes nature of this competition was highlighted in 2025 when one Chinese biopharmaceutical company challenged another's EGFR-ADC patent in the U.S. through an Inter Partes Review (IPR) proceeding 7 . This case illustrates how companies are willing to engage in complex patent litigation to clear freedom-to-operate space for their EGFR-targeting therapies.
Companies developing EGFR degraders face several strategic patent considerations:
While broad claims are desirable, they face greater scrutiny during examination and potential challenges
With global markets at stake, companies must prioritize key markets like the U.S., Europe, and China
Balancing the need for early priority dates with the desire to include the latest data
The commercial implications are substantial, with potential licensing deals reaching up to $50 billion in combined upfront and milestone payments for promising EGFR-targeted therapies 7 .
Developing effective EGFR degraders requires specialized reagents and tools throughout the discovery and optimization process.
| Reagent Category | Specific Examples | Function in Research | Considerations |
|---|---|---|---|
| EGFR-Binding Warheads | Osimertinib, Afatinib derivatives, Novel inhibitors | Provide target engagement | Must maintain binding against resistance mutations |
| E3 Ligase Ligands | VHL ligands, CRBN modulators, IAP antagonists | Recruit cellular degradation machinery | Tissue distribution and efficiency vary |
| Linker Libraries | PEG-based, alkyl chain, triazole-containing linkers | Connect warhead to E3 recruiter | Length and flexibility critical for ternary complex |
| Cell Line Panels | Ba/F3 EGFR mutants, Patient-derived cells, CNS models | Assess efficacy across mutations | Should include treatment-resistant lines |
| Analytical Tools | Cellular thermal shift assay, Western blotting, RT-qPCR | Confirm degradation and downstream effects | Multiple methods needed for verification |
This toolkit enables researchers to systematically explore structure-activity relationships and optimize degrader properties for improved potency, selectivity, and pharmacokinetic profiles.
Select EGFR mutations and resistance profiles to target
Choose appropriate EGFR inhibitors as binding moieties
Systematically test linker length and composition
Evaluate degradation efficiency and selectivity
Assess efficacy in animal models and toxicology profiles
As EGFR PROTACs progress through development, several challenges and opportunities emerge:
The blood-brain barrier represents a particular challenge for treating NSCLC patients with brain metastases—a common occurrence in EGFR-mutant disease. An ideal EGFR degrader would need sufficient brain penetration to address these lesions 3 .
Additionally, the heterogeneity of tumor populations means that effective degraders must maintain activity against diverse mutation profiles, potentially requiring combination approaches.
The novel mechanism of action of PROTACs raises questions about:
The field is becoming increasingly crowded, with companies pursuing both broad-spectrum EGFR degraders and mutation-selective approaches. This competition is driving rapid innovation but also creating a complex intellectual property landscape.
Companies are investing significant resources in not only developing degraders but also securing strong patent protection across major markets.
Companies in Development
Clinical Candidates
Phase II Trials
Phase III Trials
"The development of EGFR-targeting PROTACs represents more than just another incremental advance in oncology therapeutics—it signifies a fundamental shift in how we approach disease-causing proteins."
By moving beyond simple inhibition to actual elimination of the target, this approach has the potential to overcome limitations that have plagued traditional small molecule therapies for decades.
The remarkable progress in this field, exemplified by the recent clinical approval of and径医药's EGFR PROTAC 1 and the sophisticated patent strategies being employed, highlights the convergence of scientific innovation and strategic commercial thinking.