Thank you, Jason. In this section, I will walk you through the design of ALX2004 and the rationale behind its mechanism of action. As Jason alluded to, EGFR known and validated target as monoclonal antibody, has been hard to crack a successful ADC, primarily due to payload classes as illustrated in these 3 examples. EGFR remains one of the most validated and clinically actionable targets in oncology. Monoclonal antibodies like cetuximab and panitumumab have demonstrated meaningful clinical activity, but translating this success into effective ADCs has proven exceptionally challenging.
Historically, the primary barrier has been the choice of payload. Earlier EGFR-targeted ADCs use extremely potent or highly toxic payload classes, for example, tubulin inhibitors and DNA cross-linking agents such as MMAF or PBD, with toxicities prevented dose escalation to therapeutically meaningful levels. These experiences have shaped our thinking and informed the design of ALX2004. ALX2004 was intentionally engineered to overcome the historical liabilities of EGFR-targeted ADCs by integrating modern, best-in-class design features across 3 critical dimensions: linker payload, antibody and immunomodulatory effects.
On the linker payload front, we prioritized the topoisomerase 1 inhibitor payload for having the greatest potential to achieve a clinically meaningful therapeutic window with an EGFR-targeted ADC and paired it with stable tumor-selective cleavable linker. This ensures efficient tumor cell killing, both direct and bystander, while minimizing off-target toxicity in healthy tissues. With respect to immunomodulatory effects, the release payload triggers immunogenic cell death that can lead to modulation of adaptive immune response, while ALX2004 Fc engagement can lead to activation of innate immunity thus potentially amplifying long-term tumor control.
On the antibody front, the EGFR-targeting antibody retains intrinsic antitumor activity, including inhibition of EGFR signaling and, as just mentioned, engagement of innate immunity through functional Fc domain. Together, these features allow ALX2004 to deliver potent antitumor activity across EGFR-expressing cancers with a significantly broadened therapeutic window compared to legacy ADCs. In the following section, I'm going to dive a bit deeper into each of these properties.
Next slide. Our goal was to maintain potent killing of EGFR-positive tumor cells while maximizing the bystander effect to eliminate EGFR-negative cells within heterogeneous tumors. For the payload, we developed a proprietary topoisomerase 1 inhibitor with potent cytotoxic activity. It induces both direct tumor cell killing and a bystander effect that's effectively targeting neighboring EGFR-negative cells. Importantly, the payload also triggers immunogenic cell death, thereby priming an antitumor immune response.
Regarding the linker payload architecture, we focus on tumor-selective delivery. We stabilize linker payload deconjugation to increase stability in circulation and release payload specifically in the tumor microenvironment. This reduces systemic toxicity associated with off-target release of potent cytotoxin. On the linker payload front, we use native cysteine conjugation for simplicity and scalability but enhance it with proprietary chemistry to increase the plasma stability of the conjugate. The result was targeted payload release at the tumor site and reduced systemic exposure.
Next slide. This slide summarizes the robust cytotoxicity profiling we conducted. The top panel shows the in vitro screening results for 65 novel ALX payloads, each synthesized and tested across 8 tumor cell lines and benchmarked against payload of approved Topoisomerase 1 ADCs, Trodelvy's SN-38, and Enhertu's DXd. Each dashmark on the X-axis corresponds to a unique proprietary ALX payload. 14 payload candidates were selected for testing at full ADCs based on their payload properties, for example, cytotoxicity and permeability.
The lower panel depicts payload candidates converted into linker payloads then conjugated to 3 solid tumor targeting antibodies and tested as full ADCs on 6 tumor cell lines, and again, benchmarked against DXd linker payload. Each dashmark on the X-axis corresponds to unique proprietary ALX linker payload. ALX2004 linker payloads emerged as the most consistent and potent across antibody combinations, showing comparable activity to deruxtecan, the clinically validated DXd payload.
Next slide. We stabilized linker payload deconjugation to increase stability in circulation and increased release of payload specifically in the tumor microenvironment. Off-tumor deconjugation of linker payloads from their antibody remains a challenge even in the current generation of ADCs, which may lead to increased toxicity. To benchmark our linker performance, we conducted a head-to-head comparison with deruxtecan in nonhuman primates. We conjugated the ALX linker payload to trastuzumab in order to make a direct comparison to trastuzumab deruxtecan. We then compare drug to antibody ratios over time after administration in nonhuman primates. Our linker payload demonstrated superior stability, maintaining a higher conjugation level throughout circulation, as you can see on the graph. These data strongly suggests that ALX-2004 will deliver more payload to tumors while limiting exposure to healthy tissues. This is central to our strategy of improving the therapeutic index through intelligent design.
Next slide. The goal of our rigorous linker payload selection process was to match or exceed the activity of the DXd linker payload, both in terms of direct cell killing and the bystander effect. The bystander effect is an important mechanism of cell killing in solid tumors. The ADC is internalized in a target-expressing tumor cell. Payload released within the cell directly kills the cell, and the payload is able to kill non-target expressing neighboring tumor cells through the bystander effect. First, we synthesize an ADC using the ALX-2004 antibody plus the DXd linker payload in order to make a direct comparison of the activity of the 2 linker payload platforms. Second, we compared ALX2004 to DXd ADC, consisting of our antibody conjugated to the DXd linker payload in several EGFR-expressing mouse models, including a model specifically for bystander effect.
Across multiple CDX models with varying levels of EGFR expression, ALX2004, shown in purple bars on the graph, demonstrated equivalent or superior tumor eradication as shown by the percent of mice in which tumors were completely eradicated. Importantly, in comparison of the antigen homogeneous model on the left side of this figure and the bystander effect model containing both EGFR high and EGFR ultra-low cells shown on the right side of the graph, ALX2004 outperformed the DXd comparator, again, achieving a higher rate of tumor eradication. This suggests enhanced bystander activity, consistent with the design goal of tumor selective and membrane permeable payload delivery. We attribute improved activity to combination of improved bystander effect and improved linker stability compared to deruxtecan.
In addition to direct cytotoxicity, ALX2004 is designed to harness the immune system. The payload induces immunogenic cell death, releasing signals that can activate an adaptive antitumor immune response. Further, ALX2004 Fc region mediates antibody-dependent cell-mediated cytotoxicity and antibody-dependent cellular phagocytosis, augmenting tumor clearance by engaging innate immune effector cells.
This slide illustrates ALX2004's ability to induce calreticulin expression, a key marker of immunogenic cell death in EGFR-positive cells. Cells were treated with ALX2004, ALX2004 payload as an unconjugated small molecule and compared to the control, a nontargeted ADC and the ALX2004 naked antibody. After treatment with ALX2004 and its payload as small molecule, surface levels of calreticulin are significantly elevated and much higher than controls, indicating that ALX2004 retains the immunogenic cell death activity that is characteristic of the topoisomerase 1 inhibitor payload class. Similar results were observed for other immunogenic cell death biomarkers, including HMGB1 and ATP. These findings support ALX2004's ability to trigger adaptive immune engagement.
Antibody drug conjugation can inhibit the Fc-mediated antibody-dependent activity of the ADC due to steric hindrance from the 8 payloads attached to the antibody. We verified that ALX2004 maintains Fc-mediated immune effector activity. In vitro ADCC and ADCP assays confirmed robust dose-dependent activity in EGFR high cells with no measurable effect in EGFR ultra-low cells. This confirms that ALX2004 Fc engages immune effector cells, but only when EGFR is present.
The antibody component of ALX2004 was designed to both block EGFR signaling and bind a unique epitope distinct from approved anti-EGFR antibodies. This design may overcome resistance developed to approved EGFR-targeted antibody therapy. We also tuned the antibody's affinity to maximize tumor uptake while minimizing binding to normal EGFR expressing tissues, thus increasing the therapeutic index.
To maximize likelihood of the ADC success in clinic, we designed an antibody backbone with a widest therapeutic window. ALX2004 was selected for binding epitope different and approved antibodies, thus allowing for first-in-human study in patients with resistances to approved therapies due to EGFR mutations. In addition, its affinity was selected for higher tumor uptake and lower tissue uptake based on published studies done by others relating antibody affinity and the distribution. In order to test whether we created a potential safety benefit for activity, we tested a series of antibodies with different epitopes and affinity gaining 500 fold. In mouse models, ADCs showed similar activity, suggesting there was no activity penalty for designing a potentially safer ADC through affinity tuning. The antibody selected for ALX2004 offered a balance between tumor activity and safety.
This slide demonstrates that ALX2004 inhibits EGFR tyrosine kinase activity in a dose-dependent manner in EGFR overexpressing cells. This further contributes to its antitumor mechanism beyond payload-mediated cytotoxicity.
I will now touch on ALX2004 in vivo efficacy in mouse models and its GLP toxicity evaluation in nonhuman primates. ALX2004 showed tumor suppression activity across a panel of xenograft models, representing a broad spectrum of cancer types and EGFR expression levels. Notably, ALX2004 was effective in models harboring KRAS, BRAF and p53 mutations.
ALX2004 showed excellent tumor suppression activity, especially for topoisomerase 1 payload class down to single 1 milligram per kilogram dose level, leading to complete tumor eradication in several models. Notably, complete tumor growth inhibition was seen at all dose levels, even with tumors having H-score of 100, for example, pancreatic models shown here. These results confirm the broad applicability of ALX2004 in targeting EGFR-positive malignancies.
In a patient-derived colorectal organoid xenograft model, ALX2004 showed dose-dependent tumor suppression. These data further support its translational potential in human tumors with low EGFR expression. PDX had an H-score of 60.
Our 6-week repeat dose with 6-week recovery period GLP toxicology study evaluated ALX2004 in nonhuman primates. All findings were minimal to moderate and fully recoverable. The GLP toxicology supports our design selection and safety margin for clinical use.
And next, our Chief Medical Officer, Dr.