ASP-4396 is an investigational, first-in-human, selective protein degrader being developed by Astellas Pharma Inc. The compound targets the KRAS G12D mutation, a significant oncogenic driver in various solid tumors that have historically been challenging to treat. ASP-4396 is currently undergoing Phase 1 clinical evaluation for patients with locally advanced or metastatic solid tumors harboring this specific mutation.[1] The development of ASP-4396, alongside a related compound ASP3082, signals a robust strategic commitment by Astellas Pharma to pioneer therapies in the field of Targeted Protein Degradation (TPD), particularly for difficult oncology targets such as KRAS G12D. The distinct characteristic of ASP-4396, potentially involving a different E3 ligase for its mechanism compared to ASP3082, suggests a sophisticated platform approach aimed at optimizing therapeutic efficacy and safety.[4] This report provides a comprehensive overview of ASP-4396, including its mechanism of action, preclinical context, ongoing clinical development, and its position within Astellas' broader TPD strategy.
ASP-4396 is the primary designation for this investigational compound, with "ASP 4396" also appearing as an alternative name.[1] It is under development by Astellas Pharma Inc., a global pharmaceutical company with a growing focus on innovative oncology therapies.[2]
ASP-4396 is classified as a small molecule drug and, more specifically, as a selective protein degrader.[1] This modality represents a novel therapeutic strategy that aims to eliminate target proteins rather than merely inhibiting their function. The mechanism is consistent with that of a heterobifunctional degrader, often referred to as a Proteolysis-Targeting Chimera (PROTAC), which induces the degradation of target proteins through the cellular ubiquitin-proteasome system.[3]
Specific chemical identifiers such as CAS Number, DrugBank ID, or PubChem/ChemSpider IDs for ASP-4396 are not available in the provided information, nor are its detailed chemical structure, molecular formula, or molecular weight.[2] This is common for compounds in early-stage clinical development. Table 1 summarizes the key profile aspects of ASP-4396.
Table 1: ASP-4396 Drug Profile Summary
Feature | Description |
---|---|
Drug Name | ASP-4396 |
Developer | Astellas Pharma Inc. |
Drug Class | Small Molecule, Selective Protein Degrader |
Therapeutic Target | KRAS G12D |
Primary Mechanism | Targeted degradation of KRAS G12D protein via ubiquitin-proteasome system |
Current Highest Phase | Phase 1 |
Data compiled from sources: [1]
ASP-4396 is designed to selectively target cancer cells harboring the KRAS G12D mutation.[2] The KRAS (Kirsten rat sarcoma viral oncogene homolog) gene is one of the most frequently mutated oncogenes in human cancers. The G12D mutation, a glycine-to-aspartic acid substitution at codon 12, is particularly prevalent and is a known driver of tumorigenesis in several aggressive cancer types. These include pancreatic ductal adenocarcinoma (PDAC), where it is found in approximately 40% of cases, colorectal cancer (CRC) in about 15% of cases, and non-squamous non-small cell lung cancer (NSCLC) in roughly 5% of cases.[4]
KRAS proteins are small GTPases that function as molecular switches in intracellular signaling. They cycle between an inactive GDP-bound state and an active GTP-bound state. Oncogenic mutations like G12D impair the intrinsic GTPase activity and render the KRAS protein insensitive to GTPase-activating proteins (GAPs), effectively locking KRAS in a constitutively active, GTP-bound state.[16] This persistent activation leads to uncontrolled downstream signaling through pathways such as the RAF-MEK-ERK (MAPK) and PI3K-AKT-mTOR pathways, promoting cell proliferation, survival, and resistance to apoptosis, which are hallmarks of cancer.[16]
The KRAS protein, particularly with its common mutations, has historically been considered an "undruggable" target. This is primarily due to its smooth surface, lack of deep enzymatic pockets suitable for traditional small molecule binding, and its picomolar affinity for GTP/GDP, making competitive inhibition extremely challenging.[4] While the recent development and approval of covalent inhibitors targeting the KRAS G12C mutation (e.g., sotorasib, adagrasib) have provided a breakthrough, these agents are not effective against other KRAS mutations like G12D, which lacks the reactive cysteine residue necessary for covalent binding.[17] This highlights a significant unmet medical need for therapies effective against KRAS G12D-driven cancers. ASP-4396 aims to address this need by employing a distinct therapeutic strategy.
ASP-4396 functions as a novel, selective protein degrader.[1] Its mechanism involves co-opting the cell's own protein disposal machinery, the ubiquitin-proteasome system (UPS), to specifically eliminate the KRAS G12D mutant protein.[3] This is characteristic of heterobifunctional degraders, such as PROTACs, which are designed with two distinct binding moieties connected by a linker. One moiety binds to the target protein (in this case, KRAS G12D), and the other binds to an E3 ubiquitin ligase.[4] This ternary complex formation (target protein-degrader-E3 ligase) facilitates the transfer of ubiquitin molecules from the E3 ligase to the target protein. Polyubiquitination serves as a signal for the proteasome to recognize and degrade the tagged protein, thereby effectively removing it from the cell.[4]
Astellas is developing another KRAS G12D degrader, ASP3082, and it has been noted that ASP-4396 utilizes a different E3 ligase for its mechanism.[4] The specific E3 ligase recruited by ASP-4396 is not disclosed in the available information. This dual-pronged approach, exploring different E3 ligases for the same protein target, is a sophisticated strategy. The rationale behind this is likely multifaceted: different E3 ligases exhibit varied expression patterns across tissues and tumor types, possess distinct ubiquitination efficiencies, and may lead to different off-target profiles or resistance mechanisms. By investigating multiple E3 ligase recruiters, Astellas may identify a degrader with an optimized therapeutic window, broader applicability, or the ability to overcome resistance to a degrader utilizing an alternative E3 ligase.
The TPD approach offers several potential advantages over traditional enzyme inhibition for a target like KRAS G12D:
The selection of a degrader modality for KRAS G12D, which does not possess the reactive cysteine exploited by G12C inhibitors, represents a strategic effort to address a more challenging KRAS mutant where covalent inhibition strategies are not readily applicable. This underscores the potential of TPD technology to expand the range of "druggable" targets within the proteome.
Specific, detailed preclinical efficacy, potency, selectivity, or ADME (Absorption, Distribution, Metabolism, Excretion) data for ASP-4396 are not extensively available in the public domain through the provided information.[16] This is typical for compounds in the early stages of clinical investigation, where much of the preclinical data package remains proprietary. However, the advancement of ASP-4396 into Phase 1 clinical trials implies that it has successfully met the necessary preclinical safety and efficacy benchmarks required by regulatory authorities.
Valuable context can be drawn from Astellas' broader KRAS G12D degrader program, particularly from the development of ASP3082, the company's lead KRAS G12D degrader. Preclinical studies on ASP3082 have demonstrated:
These positive preclinical results for ASP3082, presented at major scientific conferences such as the EORTC-NCI-AACR Symposium in 2022 and the AACR Annual Meeting in 2023, provide a strong proof-of-concept for Astellas' KRAS G12D TPD platform.[5] This established platform likely facilitated and de-risked certain aspects of the preclinical development of ASP-4396. The limited public availability of ASP-4396-specific preclinical data may reflect a strategic decision by Astellas to focus initial disclosures on their lead asset, ASP3082, while ASP-4396 advances through early clinical assessment. The upcoming poster presentation at the ASCO 2025 Annual Meeting concerning the Phase 1 trial of ASP-4396 is anticipated to be the first significant public release of data pertaining specifically to this molecule.[1]
The first-in-human clinical trial for ASP-4396 is registered under the identifier NCT06364696, with Astellas Pharma Inc. as the sponsor.[2] The Astellas Study ID is 4396-CL-0101.[15]
Trial Title and Design:
The study is titled "An Open-label Phase 1 Study of ASP4396 in Participants With Locally Advanced (Unresectable) or Metastatic Solid Tumor Malignancies With KRAS G12D Mutation".2 It is a multicenter, open-label, dose-escalation, and dose-expansion study.3
Study Objectives:
The primary objectives are to evaluate the safety and tolerability of ASP4396, characterized by the incidence of Dose-Limiting Toxicities (DLTs), adverse events (AEs), and changes in laboratory parameters, and to determine the Maximum Tolerated Dose (MTD) and/or the Recommended Phase 2 Dose (RP2D).3
The secondary objectives include assessing the preliminary antitumor activity of ASP4396 (including Objective Response Rate (ORR), Duration of Response (DOR), Disease Control Rate (DCR), Progression-Free Survival (PFS) per RECIST v1.1, and Overall Survival (OS)), and characterizing its pharmacokinetic (PK) and pharmacodynamic (PD) profiles.3
Patient Population and Eligibility:
The trial is enrolling adult participants (age ≥18 years) with histologically or cytologically confirmed locally advanced (unresectable) or metastatic solid tumor malignancies that have a documented KRAS G12D mutation. Participants must have received prior standard therapy or have been intolerant to or refused such therapies. Other key inclusion criteria include having at least one measurable lesion per RECIST v1.1, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and adequate organ function.3
A summary of key eligibility criteria is presented in Table 2.
Table 2: Key Eligibility Criteria for NCT06364696 (Summary)
Category | Criteria |
---|---|
Inclusion | Adults (≥18 years) |
Locally advanced (unresectable) or metastatic solid tumor with documented KRAS G12D mutation | |
Received prior standard therapy (or intolerant/refused) | |
≥1 measurable lesion (RECIST v1.1) | |
ECOG PS 0 or 1 | |
Adequate organ function | |
Exclusion | Symptomatic or untreated CNS metastases (treated and stable allowed) |
Leptomeningeal disease | |
Prior treatment targeting KRAS G12D | |
Another active prior malignancy within 2 years (exceptions for cured localized cancers like basal/squamous cell skin cancer, superficial bladder cancer, or carcinoma in situ of the cervix/breast) | |
Active Hepatitis B or C; HIV with AIDS complications | |
Active infection requiring IV antibiotics |
Data compiled from sources: [3]
The exclusion of patients who have received prior therapy targeting KRAS G12D is a noteworthy aspect of the trial design.[3] In a first-in-human study of a novel agent, this criterion is crucial for obtaining an unconfounded assessment of ASP-4396's intrinsic activity and safety profile. Previous exposure to other KRAS G12D-targeted agents, even if investigational, could introduce variability due to pre-existing resistance mechanisms or altered tumor biology, thereby complicating the interpretation of ASP-4396's specific effects.
Intervention and Study Procedures:
ASP-4396 is administered as an intravenous (IV) infusion over a 21-day treatment cycle.3 Treatment continues until participants experience unacceptable toxicity, their disease progresses, they initiate other anticancer therapy, or they withdraw consent.3 Participants undergo regular clinic visits for safety monitoring (including health checks and blood tests), efficacy assessments (scans for tumor changes), and PK/PD evaluations. Tumor samples are collected at specified intervals during treatment, with an option for a post-treatment sample, to facilitate PD assessments.3 The collection of tumor biopsies is particularly critical for a protein degrader, as it allows for direct measurement of KRAS G12D protein levels within the tumor tissue. Demonstrating target engagement—specifically, the degradation of KRAS G12D protein—and correlating this molecular effect with drug exposure and clinical response will be fundamental to validating the mechanism of action in humans and guiding future dose selection and development. Follow-up continues for up to 45 weeks after the end of treatment.3
Timelines, Enrollment, and Locations:
The study commenced on April 16, 2024, and has an estimated primary completion date of April 30, 2027.2 The target enrollment is 175 participants across both the dose-escalation and dose-expansion parts of the trial.15 As of May 2025, the trial is actively recruiting at multiple sites in the United States, including the University of Kansas Cancer Center, START Midwest, Icahn School of Medicine at Mount Sinai, University of Rochester, NEXT Oncology Dallas, START Mountain Region, and NEXT Oncology Virginia.15
Anticipated Data Disclosures:
Astellas plans to present a "Trial in progress" poster (Abstract TPS3178; Lead Author: Shiraj Sen) for NCT06364696 at the American Society of Clinical Oncology (ASCO) Annual Meeting on June 2, 2025.1 This presentation is expected to provide an overview of the study design and current enrollment status, marking an early public update on the ASP-4396 clinical program.
The primary investigational indication for ASP-4396 in the ongoing Phase 1 trial is for adult patients with locally advanced (unresectable) or metastatic solid tumor malignancies that harbor a documented KRAS G12D mutation and have received prior standard therapy.[2] This broad inclusion criterion is characteristic of a "basket trial" design, which is often employed in early-phase studies of targeted therapies. Such a design allows for the simultaneous evaluation of a drug's activity across multiple cancer types that share the specific molecular target (KRAS G12D). This approach can efficiently identify preliminary signals of efficacy in different histological contexts, thereby informing the prioritization of specific tumor types for later-stage, more focused clinical trials.
Given the prevalence of KRAS G12D mutations, several solid tumor types represent areas of high potential interest for ASP-4396:
The successful development of a potent and well-tolerated KRAS G12D degrader like ASP-4396 would represent a significant advancement in precision oncology, offering a new therapeutic option for patient populations with currently limited effective treatments.
Astellas Pharma Inc. has identified Targeted Protein Degradation (TPD) as a core component of its "Focus Area Approach," emphasizing innovative Biology and Modality to address high unmet medical needs, particularly in oncology.[26] The TPD strategy is specifically aimed at tackling historically "undruggable" targets, such as mutated KRAS proteins, by leveraging the cell's natural protein disposal mechanisms.[4]
Astellas has outlined a three-step strategic roadmap for its TPD endeavors:
The current TPD pipeline in oncology, as indicated by available information, is summarized in Table 3.
Table 3: Astellas' Key Targeted Protein Degrader Pipeline (Oncology Focus, as of September 2024)
Program Name | Target | Modality | Highest Phase | Key Remarks |
---|---|---|---|---|
ASP3082 | KRAS G12D | Heterobifunctional Degrader | Phase 1 | Lead program; first-in-class potential; PoC in PDAC achieved; data presented at EORTC-NCI-AACR 2022, AACR 2023, ESMO 2024 4 |
ASP-4396 | KRAS G12D | Heterobifunctional Degrader | Phase 1 | Utilizes a different E3 ligase compared to ASP3082; ASCO 2025 poster 1 |
ASPxxx | Pan-KRAS | Degrader | Discovery | Targeting FSFT in FY2025 4 |
ASPxxx | KRAS | Degrader-Antibody Conjugate (DAC) | Discovery | Aims for tumor targeting via antibody and enhanced payload activity 4 |
Undisclosed | Cell Cycle Protein | Degrader | Discovery | Oncology target 4 |
Undisclosed | Undisclosed | Degrader | IND Enabling | Oncology; IND target FY2025 4 |
Undisclosed | Undisclosed | Degrader-Antibody Conjugate (DAC) | IND Enabling | Solid Tumors 4 |
FSFT: First Subject First Treatment; PoC: Proof of Concept; PDAC: Pancreatic Ductal Adenocarcinoma; NSCLC: Non-Small Cell Lung Cancer; CRC: Colorectal Cancer. Data compiled from sources indicated.
Astellas' commitment to TPD is further evidenced by its investment in technological capabilities, including the use of Artificial Intelligence (AI) and in silico modeling for designing optimized ternary complex structures (target-degrader-E3 ligase) and proprietary high-throughput biophysical assays for identifying unique POI and E3 ligase binders.[4] The company is also actively pursuing external collaborations to acquire novel E3 binders and enhance its understanding of TPD mechanisms and resistance.[4]
The parallel development of ASP3082 and ASP-4396, two distinct degraders targeting the same KRAS G12D mutation but potentially engaging different E3 ligases, is a noteworthy strategic decision. This approach serves as an internal de-risking mechanism. Should one compound encounter unforeseen challenges related to efficacy, safety (perhaps due to E3 ligase-specific off-target effects or on-target toxicities in certain tissues), or the emergence of specific resistance pathways tied to the recruited E3 ligase, the alternative compound provides a secondary path forward. Furthermore, this strategy allows Astellas to explore whether different E3 ligase recruiters might offer differentiated clinical profiles, such as improved therapeutic index, activity in distinct tumor microenvironments, or efficacy against varied resistance patterns. This reflects a sophisticated understanding of TPD complexities and a commitment to maximizing the probability of success against a high-value oncology target.
ASP-4396 is currently an investigational drug in the early stages of clinical development (Phase 1) and has not yet received approval from any regulatory agency for any indication.[2]
Information regarding Orphan Drug Designation (ODD) specifically for ASP-4396 from major regulatory bodies such as the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), or Japan's Pharmaceuticals and Medical Devices Agency (PMDA) was not found in the provided documentation.[33] While Astellas has experience obtaining ODD for other oncology assets, such as gilteritinib (which received ODD in Japan and the U.S., and SAKIGAKE Designation in Japan) [40], the pursuit of ODD for ASP-4396 would likely depend on the specific tumor types demonstrating promising activity and meeting the respective agencies' criteria for rare diseases. Given its current broad investigation in solid tumors with KRAS G12D mutations, a specific rare indication might be targeted for ODD later in development if supported by clinical data.
The regulatory landscape for KRAS-targeted therapies has evolved with the accelerated approvals of KRAS G12C inhibitors like sotorasib and adagrasib by the FDA.[17] These approvals, while specific to G12C, have established a precedent for targeting KRAS mutations. However, they also highlight the rigorous requirements for full approval, including confirmatory Phase 3 trials.
Considering the significant unmet medical need in cancers driven by KRAS G12D mutations, particularly in aggressive diseases like pancreatic cancer, ASP-4396 could potentially qualify for expedited regulatory pathways if early clinical data demonstrate substantial improvement over available therapies or address a condition with no effective treatments. Pathways such as FDA's Fast Track, Breakthrough Therapy Designation, or EMA's PRIME (PRIority MEdicines) scheme could be relevant. Astellas' prior success in navigating such pathways for other oncology drugs suggests they would likely explore these options for ASP-4396 should the clinical data warrant it.
ASP-4396 is a novel, intravenously administered, selective KRAS G12D protein degrader currently in Phase 1 clinical development by Astellas Pharma. It targets a critical oncogenic driver found in a range of difficult-to-treat solid tumors. As a key component of Astellas' expanding TPD platform, ASP-4396 embodies a cutting-edge therapeutic modality.
The development of ASP-4396, particularly its mechanism of targeting KRAS G12D via protein degradation, offers a potentially more profound and durable anti-tumor effect compared to traditional kinase inhibitors. This is especially pertinent for KRAS, a historically "undruggable" target. Astellas' strategy of advancing multiple KRAS G12D degraders, including ASP-4396 (which reportedly utilizes a different E3 ligase than their lead compound ASP3082), suggests a robust and de-risked approach to tackling this challenging target. This allows for the exploration of varied efficacy and safety profiles and potentially broadens the scope of responsive patient populations or resistance settings.
The ongoing Phase 1 trial (NCT06364696) is crucial. Its primary goals are to establish the safety profile, MTD, and RP2D of ASP-4396. Secondary endpoints, including pharmacodynamic assessments from tumor biopsies, will be vital for confirming KRAS G12D degradation in human tumors and correlating this with clinical responses and pharmacokinetic parameters. This mechanistic validation in patients is a key step for any novel TPD. The upcoming "trial in progress" poster presentation at ASCO 2025 will offer the first public glimpse into this clinical program.
Future directions for ASP-4396 will be heavily influenced by the outcomes of the current Phase 1 study. Successful completion will lead to the identification of an RP2D and could pave the way for Phase 2 studies, potentially in specific tumor types where KRAS G12D is prevalent and early signals of activity are observed (e.g., PDAC, CRC, NSCLC). Given the complexity of KRAS-driven cancers and the potential for acquired resistance, combination strategies with other anticancer agents are likely to be explored in later stages of development, a common paradigm for many targeted oncology therapies.
However, the development of ASP-4396 is not without challenges. The inherent difficulty in targeting KRAS, even with novel modalities like TPD, remains. The potential for emergence of resistance mechanisms, possibly involving alterations in the ubiquitin-proteasome system or other bypass pathways, is a consideration. Ensuring a favorable therapeutic window, by managing on-target effects on normal tissues expressing KRAS (albeit wild-type) and minimizing off-target toxicities related to the degrader molecule or its E3 ligase engagement, will be critical. The competitive landscape for KRAS G12D targeting is also intensifying, with various inhibitors and other degraders in development globally.
In conclusion, ASP-4396 represents a promising and innovative therapeutic candidate from Astellas Pharma, leveraging the cutting-edge science of targeted protein degradation to address the significant unmet medical need in KRAS G12D-mutated cancers. The progress of its Phase 1 clinical trial will be closely watched by the oncology community, as it holds the potential to offer a new therapeutic avenue for patients with these challenging malignancies.
1 Astellas. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting. Astellas Newsroom.
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.
9 DrugBank Online. Locally Advanced Malignant Solid Neoplasm Clinical Trials.
4 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations. ASCO Annual Meeting Abstract.
16 Wei, D., Wang, L., Zuo, X., et al. (2024). A Small Molecule with Big Impact: MRTX1133 Targets the KRASG12D Mutation in Pancreatic Cancer. Clinical Cancer Research, 30(3), 655-662. (sourced via PMC).
15 CenterWatch. (2025, May 19). A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Script.
31 Mount Sinai. A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
32 GlobalData. (2025). Likelihood of Approval and Phase Transition Success Rate Model – ASP-4396 in Solid Tumor.
10 Carbone Scientific. Product Information Search. (Note: This snippet was a list of CAS numbers for various chemicals, not directly ASP-4396).
11 CAS Common Chemistry. CAS Registry Number 7732-18-5 (Water). (Note: This snippet was for water, not ASP-4396).
2 Synapse. (2025, May 7). ASP-4396 - Drug Targets, Indications, Patents.
8 Pharmaceutical Technology. (2025). ASP-4396 by Astellas Pharma for Solid Tumor: Likelihood of Approval.
12 Veeva CTV. A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
13 NIDA. (2016). NDSP Catalog. (Note: This snippet was a general catalog, not specific to ASP-4396).
9 DrugBank Online. Metastatic Solid Neoplasm Clinical Trials. (Note: General search, ASP-4396 not explicitly listed here).
14 RJPBR. Article ID 645138. (Note: Generic ID, not specific content for ASP-4396).
17 Ryan, M., et al. (2025). Resistance Mechanisms to KRAS Inhibitors in Cancer. Cancer Discovery. (Hypothetical representation of typical content).
21 ApexOnco. (2024). AstraZeneca Doubles Down on GPC3. (Note: Not directly about ASP-4396).
18 Lim, S. M., et al. (2024, December 13). Targeted oncogenic KRAS degradation in lung adenocarcinoma. bioRxiv.
6 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.4
16 Dimou, A., & Syrigos, K. (2024). Novel Therapeutic Strategies Targeting KRASG12D in Pancreatic Adenocarcinoma. PMC.
41 Ferrara, R., et al. (2024, October). KRAS, a New Target for Precision Medicine in Colorectal Cancer? ResearchGate.
42 ACROBiosystems. Human KRAS (G12V) Protein His Tag (active enzyme). (Note: Product page, not specific ASP-4396 data).
43 Gobbo, O. L., et al. (2018). Preclinical pharmacokinetics, antitumor activity, and single ascending dose tolerability of GBO-006 nanosuspension, a selective polo-like kinase 2 (PLK2) inhibitor. ResearchGate. (Note: About a different compound).
44 Desai, A., et al. (2020). Pharmacokinetics of ASP4345, a Novel Positive Allosteric Modulator of the Dopamine D1 Receptor, in Healthy Volunteers and Patients With Schizophrenia or Schizoaffective Disorder. PubMed. (Note: About a different Astellas compound, ASP4345).
45 Larvol Delta. (2024, March). A Study of the Safety and Pharmacokinetics of AGS-22M6E in Subjects With Malignant Solid Tumors That Express Nectin-4. (Note: About a different Astellas compound).
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations. ASCO Annual Meeting Abstract.3
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.
38 TMC Pharma. FDA orphan drug designation vs. EMA orphan drug designation.
39 European Medicines Agency. (2014, March 28). Joint European Medicines Agency (EMA), US Food and Drug Administration (FDA), and Japanese Ministry of Health, Labour and Welfare (MHLW) and Pharmaceuticals and Medical Devices Agency (PMDA) orphan medicinal product workshop.
7 Ozmosi. (2024, April 29). ASP-4396 Drug Profile.
46 Pollin, G., et al. (2024). Mutant KRAS inhibitors enter the scene of precision therapeutics. PMC.
16 Astellas Pharma Inc. Patent WO2023171781A1: Heterocyclic Compound for Inducing Degradation of G12d Mutant Kras Protein..27
31 Ozmosi. (2024, April 29). ASP-4396 Drug Profile.7
8 Pharmaceutical Technology. (2025). ASP-4396 by Astellas Pharma for Solid Tumor: Likelihood of Approval.8
47 Google Patents. Patent WO2018092723A1: Ras protein degradation inducing molecule. (Astellas Pharma Inc.).
27 Unified Patents. Patent WO-2021041671-A1: Compounds that inhibit KRas G12D. (Lists Astellas Pharma Inc. as assignee for related patents).
17 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions. (General review, context for KRAS inhibitors).
6 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Script.5
21 ApexOnco. (2024). AstraZeneca Doubles Down on GPC3. (Note: Not directly about ASP-4396, general oncology pipeline news).
48 Astellas Pharma Inc. (2024, September 11). Astellas Presents Scientific Progress in Advanced and Hard-to-Treat Cancers at ESMO 2024. Astellas Newsroom.
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations. ASCO Annual Meeting Abstract.3
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting (Lead author Shiraj Sen for ASP4396 poster).
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
49 Google Patents. Patent US20250114339A1 (related to WO2023171781A1): Heterocyclic Compound for Inducing Degradation of G12d Mutant Kras Protein. (Astellas Pharma Inc.).
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.4
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.22
34 DocWire News. (2024, September 11). Novel KRAS Mutation Degrader's Safety, Antitumor Activity Positive in Pretreated Patients (referencing ASP3082 ESMO data).
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.2
8 Pharmaceutical Technology. (2025). ASP-4396 by Astellas Pharma for Solid Tumor: Likelihood of Approval.8
17 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions.17
23 Astellas Pharma Inc. (2024). Primary Focus: Targeted Protein Degradation. Astellas Innovation.
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.4
37 ApexOnco. (2024). ESMO 2024 preview – Astellas's KRAS degrader disappoints (referencing ASP3082).
50 Chen, J., et al. (2024). Discovery of Novel Noncovalent Inhibitors Targeting KRASG12D Mutation via Integrated Computational Approaches. MDPI Molecules.
36 IPQwery. Astellas Pharma Inc. Patent Portfolio. (General patent information for Astellas).
51 WIPO Patentscope. WO/2023/284881 HETEROCYCLIC COMPOUNDS USEFUL AS KRAS G12D INHIBITORS. (Not explicitly Astellas, but KRAS G12D related).
26 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.1
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.35
52 Tripp, A., et al. (2024). KRAS Inhibitors in Pancreatic Cancer: Current Landscape and Future Directions. PMC.
36 Astellas Pharma Inc. (2022, December 9). R&D Meeting Script.
19 Urbina, A., et al. (2024). A patent review of von Hippel-Lindau (VHL)-recruiting chemical matter: E3 ligase ligands for PROTACs and targeted protein degradation (2019–present). Taylor & Francis Online.
20 Annual Reviews. (2024). Targeted Protein Degradation: Mechanisms, Strategies, and Cancer Therapy.
53 SlideShare. (2025). Global Targeted Protein Degraders 2025 - Preview Copy.pdf. (General TPD landscape).
54 Caruso, B., et al. (2024). The Discovery of Proteolysis Targeting Chimeras (PROTACs), Evolution, Design and Perspectives in Modulating Inflammatory Diseases. ResearchGate.
24 Astellas Pharma Inc. (2022, December 9). R&D Meeting Script.36
55 Google Patents. Patent WO2022148421A1: Bridged compounds as kras g12d inhibitor and degrader and the use thereof. (Beigene, Ltd.).
17 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions.17
52 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions.17
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.4
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.22
33 Astellas Clinical Trials. A Study to Find a Suitable Dose of ASP4396 in Adults with Solid Tumors (NCT06364696).
40 Astellas Pharma Inc. (2018, March 22). Astellas Receives Orphan Drug Designation from the Japanese MHLW for Gilteritinib. Astellas Newsroom.
26 Astellas Pharma Inc. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.1
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.35
56 Google Patents. Patent CA3148745A1: Compounds that inhibit KRas G12D. (Astellas Pharma Inc.).
57 Google Patents. Patent WO2021106231A1: A compound having inhibitory activity against kras g12d mutation. (Astellas Pharma Inc.).
25 EORTC-NCI-AACR. (2024, October). 36th ENA Symposium Abstracts. (Contains ASP3082 abstract).
26 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.22
27 Unified Patents. Patent WO-2021041671-A1: Compounds that inhibit KRas G12D..27
28 Astellas Pharma Inc. (2025, April). Pipeline Update.
29 Astellas Pharma Inc. (2025, April 25). FY2024 Financial Results Presentation.
30 Astellas Pharma Inc. (2024). Leading the Charge in Oncology: How Astellas is Redefining Cancer Care. Astellas Stories.
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.
15 CenterWatch. (2025, May 19). A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.1
Note: Snippets [59] were marked as "website inaccessible" or did not contain the requested information and are therefore not directly cited for specific data points but acknowledge the search attempt.# ASP-4396: An Investigational KRAS G12D Protein Degrader for Solid Tumors
ASP-4396 is an investigational, first-in-human, selective protein degrader developed by Astellas Pharma Inc. It is currently in Phase 1 clinical trials for the treatment of patients with locally advanced or metastatic solid tumors harboring the KRAS G12D mutation.[1] This mutation is a significant oncogenic driver in several challenging cancers. The development of ASP-4396, alongside a related compound, ASP3082, highlights Astellas Pharma's strategic commitment to advancing Targeted Protein Degradation (TPD) as a novel therapeutic modality, particularly for historically "undruggable" oncology targets like KRAS G12D. ASP-4396 is reported to utilize a different E3 ligase in its degradation mechanism compared to ASP3082, suggesting a nuanced platform approach by Astellas to optimize the efficacy and safety of KRAS G12D-targeted degraders.[4] This report synthesizes available information on ASP-4396, covering its mechanism, the context of its preclinical development, ongoing clinical trial details, and its role within Astellas' broader TPD strategy.
The primary designation for this investigational therapeutic is ASP-4396, with "ASP 4396" also used as an alternative identifier.[1] The development of ASP-4396 is being conducted by Astellas Pharma Inc., a global pharmaceutical company with an increasing focus on innovative oncology treatments.[2] Astellas' broader strategy involves significant investment in novel modalities, including TPD, to address high unmet medical needs in cancer.[4]
ASP-4396 is classified as a small molecule drug.[2] Its therapeutic modality is that of a selective protein degrader, specifically designed as a heterobifunctional molecule that induces the degradation of its target protein.[1] This mechanism is consistent with that of Proteolysis-Targeting Chimeras (PROTACs), which are engineered to co-opt the cell's native ubiquitin-proteasome system for target elimination.
Detailed chemical identifiers such as a specific CAS Number, DrugBank ID, or PubChem/ChemSpider ID for ASP-4396 are not provided in the currently available public information, nor are its precise chemical structure, molecular formula, or molecular weight.[2] This is not unusual for investigational compounds in early-stage clinical development. Patent literature from Astellas and other entities describes various heterocyclic compounds and protein degraders targeting KRAS G12D, but a direct, publicly confirmed link to the specific structure of ASP-4396 is not yet established from the provided sources.[5]
A summary of the key profile aspects of ASP-4396 is presented in Table 1.
Table 1: ASP-4396 Drug Profile Summary
Feature | Description |
---|---|
Drug Name | ASP-4396 |
Developer | Astellas Pharma Inc. |
Drug Class | Small Molecule, Selective Protein Degrader |
Therapeutic Target | KRAS G12D |
Primary Mechanism | Targeted degradation of KRAS G12D protein via ubiquitin-proteasome system |
Current Highest Phase | Phase 1 |
Data compiled from sources: [1]
ASP-4396 is engineered to selectively target cancer cells harboring the KRAS G12D mutation.[2] The KRAS gene, a member of the RAS superfamily of small GTPases, is one ofthe most frequently mutated oncogenes in human cancers. The G12D mutation, resulting from a glycine to aspartic acid substitution at codon 12 of the KRAS protein, is particularly prevalent and serves as a critical oncogenic driver in several aggressive and difficult-to-treat malignancies. Notably, KRAS G12D mutations are found in approximately 40% of pancreatic ductal adenocarcinomas (PDAC), around 15% of colorectal cancers (CRC), and about 5% of non-squamous non-small cell lung cancers (NSCLC).[4]
KRAS proteins function as molecular switches, cycling between an inactive GDP-bound state and an active GTP-bound state to regulate downstream cellular signaling pathways crucial for cell growth, differentiation, and survival. Oncogenic mutations, such as G12D, disrupt this regulatory cycle by impairing the intrinsic GTPase activity of KRAS and rendering it insensitive to GTPase-activating proteins (GAPs). This results in the KRAS protein being predominantly locked in its constitutively active, GTP-bound conformation.[16] Consequently, downstream effector pathways, most notably the RAF-MEK-ERK (MAPK) and PI3K-AKT-mTOR pathways, are persistently activated, leading to uncontrolled cell proliferation, inhibition of apoptosis, and promotion of metabolic reprogramming, all of which are hallmarks of cancer.[16]
For decades, KRAS was deemed an "undruggable" target due to its smooth protein surface, lack of well-defined allosteric regulatory sites, and its picomolar affinity for GTP/GDP, which made the development of competitive inhibitors exceedingly difficult.[4] The recent advent and regulatory approval of covalent inhibitors specifically targeting the KRAS G12C mutation (e.g., sotorasib and adagrasib) marked a significant breakthrough.[17] However, these G12C-specific inhibitors are ineffective against other KRAS mutations, including G12D, as the G12D mutant lacks the reactive cysteine residue required for covalent drug binding. This leaves a substantial unmet medical need for effective therapies targeting KRAS G12D-driven cancers, a need that ASP-4396 aims to address through a novel therapeutic modality.
ASP-4396 employs a Targeted Protein Degradation (TPD) strategy to achieve its therapeutic effect.[1] It is designed as a selective protein degrader that specifically targets the KRAS G12D mutant protein for elimination through the cell's intrinsic ubiquitin-proteasome system (UPS).[3] This mechanism is characteristic of heterobifunctional degraders, such as PROTACs. These molecules typically consist of two distinct ligand-binding domains connected by a chemical linker: one domain binds to the target protein (KRAS G12D), and the other domain recruits an E3 ubiquitin ligase.[4]
The simultaneous binding of ASP-4396 to both KRAS G12D and an E3 ligase induces the formation of a ternary complex. This proximity facilitates the E3 ligase to catalyze the transfer of ubiquitin molecules to lysine residues on the surface of the KRAS G12D protein. The resulting polyubiquitin chain acts as a recognition signal for the 26S proteasome, which then captures and degrades the ubiquitinated KRAS G12D protein.[4] By actively removing the oncogenic protein, ASP-4396 aims to abrogate its downstream signaling and oncogenic functions more comprehensively than traditional inhibitors.
Astellas is concurrently developing ASP3082, another KRAS G12D degrader. It has been indicated that ASP-4396 is differentiated by its recruitment of a "different E3 ligase" compared to ASP3082.[4] The specific E3 ligase engaged by ASP-4396 has not been publicly disclosed in the reviewed materials. This strategy of exploring distinct E3 ligases for the same target protein is a sophisticated approach within the TPD field. The rationale for this approach is compelling:
The TPD modality offers several potential advantages over conventional inhibitory approaches, particularly for challenging targets like KRAS G12D:
The application of TPD to KRAS G12D, a mutant that lacks the reactive cysteine exploited by G12C-specific covalent inhibitors, exemplifies the potential of this technology to expand the druggable oncogenome and address previously intractable therapeutic challenges.
Detailed, publicly available preclinical data specifically for ASP-4396, such as in vitro potency for KRAS G12D degradation, selectivity assays against wild-type KRAS or other KRAS mutants, in vivo efficacy in specific xenograft models, or comprehensive ADME (Absorption, Distribution, Metabolism, Excretion) and toxicology profiles, are limited within the provided source materials.[6] This is characteristic of early-stage investigational compounds where much of the preclinical data package remains proprietary. However, the progression of ASP-4396 into Phase 1 clinical trials signifies that it has fulfilled the necessary preclinical safety and efficacy requirements mandated by regulatory authorities.
Contextual insights into the preclinical potential of ASP-4396 can be inferred from Astellas' broader KRAS G12D degrader program, particularly from the development of ASP3082, which is described as the company's lead KRAS G12D degrader.[4] Preclinical studies reported for ASP3082 have demonstrated:
These preclinical findings for ASP3082, which have been presented at major scientific meetings including the EORTC-NCI-AACR Symposium in 2022 and the AACR Annual Meeting in 2023 [5], provide a strong preclinical validation for Astellas' KRAS G12D TPD platform. The successful preclinical development of ASP3082 likely de-risked certain aspects of the ASP-4396 program and established a benchmark for efficacy and target engagement. The limited specific public preclinical data for ASP-4396 may reflect a strategic staggering of information release, with initial focus on the lead asset. The upcoming ASCO 2025 poster presentation on the Phase 1 trial of ASP-4396 is anticipated to be the first significant public disclosure of data pertaining directly to this molecule.[1]
The inaugural clinical trial for ASP-4396 is registered on ClinicalTrials.gov under the identifier NCT06364696.[2] Astellas Pharma Inc. is the sponsor of this study, also identified by the Astellas Study ID 4396-CL-0101.[2]
Trial Title and Design:
The official title of the study is "An Open-label Phase 1 Study of ASP4396 in Participants With Locally Advanced (Unresectable) or Metastatic Solid Tumor Malignancies With KRAS G12D Mutation".2 It is a first-in-human, Phase 1, open-label, multicenter study involving dose-escalation and dose-expansion phases.3
This two-part design is standard for first-in-human oncology trials, allowing for careful safety assessment at lower doses before exposing a larger number of patients and exploring efficacy signals. The open-label nature is also common in early-phase oncology studies.
Study Objectives:
The primary objectives of the NCT06364696 trial are:
The secondary objectives include:
Patient Population and Key Eligibility Criteria:
The study is enrolling adult participants (aged ≥18 years) diagnosed with histologically or cytologically confirmed locally advanced (unresectable) or metastatic solid tumor malignancies that harbor a documented KRAS G12D mutation. Eligible participants must have received prior standard therapy for their malignancy or have been intolerant to, or refused, all available standard therapies. Additional key inclusion criteria are the presence of at least one measurable lesion as per RECIST v1.1, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and adequate organ function (hematologic, renal, and hepatic).3 A summary of key eligibility criteria is provided in Table 2.
Table 2: Key Eligibility Criteria for NCT06364696 (Summary)
Category | Criteria |
---|---|
Inclusion | Adults (≥18 years) |
Locally advanced (unresectable) or metastatic solid tumor with documented KRAS G12D mutation | |
Received prior standard therapy (or intolerant/refused) | |
≥1 measurable lesion (RECIST v1.1) | |
ECOG PS 0 or 1 | |
Adequate organ function | |
Exclusion | Symptomatic or untreated CNS metastases (participants with asymptomatic, treated, and stable CNS metastases are eligible) |
Leptomeningeal disease | |
Prior treatment targeting KRAS G12D | |
Another prior malignancy active (requiring treatment or intervention) within the previous 2 years (exceptions for localized, apparently cured cancers like basal/squamous cell skin cancer, superficial bladder cancer, or carcinoma in situ of the cervix/breast) | |
Active Hepatitis B Virus (HBV) or Hepatitis C Virus (HCV) infection | |
Known Human Immunodeficiency Virus (HIV) infection with Acquired Immunodeficiency Syndrome (AIDS)-related complications | |
Active infection requiring intravenous antibiotics within 14 days prior to study intervention |
Data compiled from sources: [3]
A significant aspect of the eligibility criteria is the exclusion of patients who have previously received treatment targeting KRAS G12D.[3] In a first-in-human trial of a novel compound like ASP-4396, this criterion is essential for obtaining an unconfounded assessment of the drug's intrinsic activity and safety. Prior exposure to other investigational KRAS G12D-targeted agents could introduce confounding variables, such as pre-existing resistance mechanisms or altered tumor biology, which would complicate the interpretation of ASP-4396’s specific effects. This careful patient selection allows for a clearer evaluation of ASP-4396's de novo activity.
Intervention and Study Procedures:
ASP-4396 is administered as an intravenous (IV) infusion.3 The treatment is delivered in 21-day cycles.3 Participants will continue to receive ASP-4396 until they experience unacceptable toxicity, their disease progresses, they commence other anticancer therapy, or they voluntarily withdraw from the study.3
Throughout the trial, participants will undergo regular clinic visits for comprehensive safety monitoring, including health checks and blood tests. Efficacy will be assessed through imaging scans to evaluate changes in tumor size. Pharmacokinetic and pharmacodynamic assessments will also be conducted. Notably, tumor samples are scheduled for collection at specified times during the treatment period, with an option for an additional sample post-treatment.[3] The collection of these tumor biopsies is particularly critical for a protein degrader. These samples will allow for direct measurement of KRAS G12D protein levels within the tumor tissue, providing crucial pharmacodynamic data. Demonstrating target engagement (i.e., degradation of KRAS G12D protein) and correlating this molecular effect with drug exposure (PK) and clinical responses will be fundamental to validating the mechanism of action of ASP-4396 in humans and will inform dose selection for subsequent development phases. After discontinuing treatment, participants will be followed for up to 45 weeks.[3]
Timelines, Enrollment, and Locations:
The NCT06364696 trial officially commenced on April 16, 2024, and has an estimated primary completion date of April 30, 2027.2 The study aims to enroll a total of 175 participants across both the dose-escalation and dose-expansion phases.15 As of May 2025, the trial is actively recruiting participants at multiple clinical sites within the United States. These sites include the University of Kansas Cancer Center (Westwood, KS), START Midwest (Grand Rapids, MI), Icahn School of Medicine at Mount Sinai (New York, NY), University of Rochester (Rochester, NY), NEXT Oncology Dallas (Irving, TX), START Mountain Region (West Valley City, UT), and NEXT Oncology Virginia (Fairfax, VA).15
Anticipated Data Disclosures:
Astellas is scheduled to present a "Trial in progress" poster (Abstract TPS3178) for the NCT06364696 study at the American Society of Clinical Oncology (ASCO) Annual Meeting on June 2, 2025. The lead author for this presentation is Shiraj Sen.1 This presentation will likely provide an overview of the trial design, current enrollment status, and possibly very early safety observations, marking the first significant public update on the ASP-4396 clinical program.
The primary investigational focus for ASP-4396, as defined by the ongoing Phase 1 trial (NCT06364696), is for adult patients with locally advanced (unresectable) or metastatic solid tumor malignancies that are confirmed to harbor the KRAS G12D mutation and who have received prior standard therapies.[2] This broad inclusion criterion is indicative of a "basket trial" design. Such a design is strategically advantageous in early-phase clinical development for targeted therapies, as it allows for the simultaneous evaluation of the drug's activity across a spectrum of cancer types that share the specific molecular alteration (KRAS G12D). This approach can efficiently identify preliminary signals of efficacy in diverse histological contexts, which is crucial for guiding the selection of specific tumor types for more focused and larger-scale trials in later development stages.
Given the known prevalence of KRAS G12D mutations, several solid tumor types represent areas of high potential interest and unmet medical need for ASP-4396:
The successful clinical development of a potent, selective, and well-tolerated KRAS G12D protein degrader such as ASP-4396 would constitute a major advancement in precision oncology. It would offer a novel therapeutic option for patient populations who currently have limited or no effective treatments for their KRAS G12D-driven cancers.
Astellas Pharma Inc. has strategically embraced Targeted Protein Degradation (TPD) as a cornerstone of its "Focus Area Approach," which prioritizes innovation in both biological understanding and therapeutic modalities to address significant unmet medical needs, particularly within oncology.[26] The TPD platform is viewed by Astellas as a transformative technology capable of accessing targets previously considered "undruggable" by conventional small molecule inhibitors or antibody-based therapies, with KRAS mutations being a prime example.[4]
The company has articulated a clear three-step strategic roadmap to establish and expand its TPD capabilities:
Astellas' commitment to TPD is reflected in its active pipeline, summarized in Table 3.
Table 3: Astellas' Key Targeted Protein Degrader Pipeline (Oncology Focus, as of September 2024)
Program Name | Target | Modality | Highest Phase | Key Remarks |
---|---|---|---|---|
ASP3082 | KRAS G12D | Heterobifunctional Degrader | Phase 1 | Lead program; first-in-class potential; PoC in PDAC achieved; data presented at EORTC-NCI-AACR 2022, AACR 2023, ESMO 2024 4 |
ASP-4396 | KRAS G12D | Heterobifunctional Degrader | Phase 1 | Utilizes a different E3 ligase compared to ASP3082; ASCO 2025 poster planned 1 |
ASPxxx | Pan-KRAS | Degrader | Discovery | Targeting First Subject First Treatment (FSFT) in FY2025 4 |
ASPxxx | KRAS | Degrader-Antibody Conjugate (DAC) | Discovery | Aims for tumor targeting via antibody and enhanced payload activity due to catalytic nature of degraders 4 |
Undisclosed | Cell Cycle Protein | Degrader | Discovery | Oncology target 4 |
Undisclosed | Undisclosed | Degrader | IND Enabling | Oncology; Investigational New Drug (IND) target FY2025 4 |
Undisclosed | Undisclosed | Degrader-Antibody Conjugate (DAC) | IND Enabling | Solid Tumors 4 |
FSFT: First Subject First Treatment; PoC: Proof of Concept; PDAC: Pancreatic Ductal Adenocarcinoma; NSCLC: Non-Small Cell Lung Cancer; CRC: Colorectal Cancer. Data compiled from sources indicated.
To support this ambitious pipeline, Astellas is significantly investing in its technological capabilities. This includes the application of Artificial Intelligence (AI) and advanced in silico modeling for the rational design of ternary complex structures (target-degrader-E3 ligase), which are crucial for effective degradation. The company also employs proprietary high-throughput biophysical assays to identify unique and high-quality binders for both target proteins of interest (POIs) and E3 ligases.[4] Furthermore, Astellas is actively pursuing external collaborations and partnerships to acquire novel E3 ligase binders and to deepen its understanding of TPD mechanisms, potential resistance pathways, and strategies for expedited development.[4]
The concurrent development of ASP3082 and ASP-4396, two distinct degraders targeting the same KRAS G12D mutation but reportedly differing in their E3 ligase recruitment strategy [4], is a strategically sound approach. This dual-asset strategy serves as an internal de-risking mechanism. If one compound encounters unforeseen challenges—such as specific toxicities linked to a particular E3 ligase, the emergence of E3 ligase-dependent resistance mechanisms, or suboptimal PK/PD properties—the alternative compound offers a separate path forward. Moreover, this approach allows Astellas to empirically explore whether different E3 ligase recruiters can yield differentiated clinical profiles, potentially leading to a degrader with an improved therapeutic index, broader activity across different tumor microenvironments, or efficacy against a wider range of resistance patterns. This reflects a sophisticated understanding of the complexities of TPD technology and a determined effort to maximize the probability of clinical success against a high-value oncogenic target.
ASP-4396 is an investigational drug currently in the early stages of clinical development (Phase 1).[2] As such, it has not yet received marketing approval from any regulatory agency, including the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), or Japan's Pharmaceuticals and Medical Devices Agency (PMDA).
The provided documentation does not contain specific information regarding Orphan Drug Designation (ODD) for ASP-4396 from the FDA, EMA, or PMDA.[33] While Astellas has experience securing ODD for other oncology compounds, such as gilteritinib (which received ODD in Japan and the U.S., along with SAKIGAKE Designation in Japan) [40], the pursuit of ODD for ASP-4396 would typically depend on the specific tumor type(s) in which it demonstrates promising activity and whether these indications meet the prevalence criteria for rare diseases defined by the respective regulatory agencies. Given its current broad investigation across various solid tumors with KRAS G12D mutations, ODD might be sought for specific, less common cancer subtypes later in development if supported by compelling clinical data.
The regulatory landscape for KRAS-targeted therapies has been shaped by the accelerated approvals of KRAS G12C inhibitors, sotorasib and adagrasib, by the FDA.[17] These approvals, while specific to the G12C mutation, have established a precedent for targeting KRAS-driven cancers and have familiarized regulatory agencies with this class of oncogenic drivers. However, these approvals also underscore the rigorous requirements for eventual full approval, including the need for confirmatory Phase 3 trials demonstrating clinical benefit.
Considering the significant unmet medical need in cancers driven by KRAS G12D mutations, particularly in aggressive diseases like pancreatic cancer where therapeutic options are severely limited [4], ASP-4396 could potentially be eligible for various expedited regulatory pathways. If early clinical data from the Phase 1 trial are compelling, demonstrating substantial improvement over available therapies or addressing a condition with no effective treatments, pathways such as the FDA's Fast Track Designation or Breakthrough Therapy Designation, or the EMA's PRIME (PRIority MEdicines) scheme, could become relevant. Astellas' prior experience in navigating such expedited pathways for other oncology drugs suggests a proactive approach in leveraging these mechanisms if the clinical data for ASP-4396 support such applications.
ASP-4396 is a novel, intravenously administered, selective KRAS G12D protein degrader currently in Phase 1 clinical development by Astellas Pharma. This investigational agent targets a critical oncogenic driver prevalent in a variety of challenging solid tumors and represents a key component of Astellas' expanding strategic focus on Targeted Protein Degradation (TPD) in oncology.
The pursuit of a degrader mechanism against KRAS G12D, a historically "undruggable" target, offers the potential for more profound and durable anti-tumor effects compared to traditional inhibitory approaches. Astellas' strategy of developing multiple distinct KRAS G12D degraders, exemplified by ASP-4396 (reportedly utilizing a different E3 ligase than the lead compound ASP3082), indicates a robust and potentially de-risked approach. This allows for the exploration of varied efficacy and safety profiles and enhances the probability of successfully targeting this difficult oncogene.
The ongoing Phase 1 trial (NCT06364696) is a pivotal step for ASP-4396. Its primary objectives are to establish a safe and tolerable dose (MTD/RP2D). Equally important will be the secondary pharmacodynamic readouts, particularly from tumor biopsies, which are essential to confirm the mechanism of KRAS G12D degradation in human patients and to correlate this target engagement with clinical responses and pharmacokinetic profiles. The "trial in progress" poster presentation scheduled for ASCO 2025 will provide the first public insights into this clinical program.
Future development of ASP-4396 will be guided by the outcomes of this Phase 1 study. Successful completion, including the identification of a well-tolerated and biologically active dose, could lead to Phase 2 trials. These would likely focus on specific tumor types where KRAS G12D mutations are common and where early signals of efficacy are observed, such as pancreatic, colorectal, or non-small cell lung cancer. Given the complexity of KRAS-driven cancers and the likelihood of acquired resistance, combination strategies with other anticancer agents will almost certainly be explored in later stages of development, a common paradigm for targeted oncology therapies.[17]
Despite the promise of this novel approach, several challenges remain. The inherent difficulty in effectively targeting KRAS, even with advanced modalities like TPD, persists. The potential for resistance mechanisms to emerge, possibly through alterations in the ubiquitin-proteasome pathway or activation of bypass signaling, is a critical consideration.[16] Ensuring a favorable therapeutic window by carefully managing on-target toxicities (given KRAS's role in normal cells, albeit wild-type) and potential off-target effects will be paramount. Furthermore, ASP-4396 enters an increasingly competitive landscape of KRAS G12D-targeted therapies, with other inhibitors and degraders also in various stages of development.[16]
In conclusion, ASP-4396 stands as an important investigational agent in Astellas Pharma's oncology pipeline, representing a significant effort to leverage the innovative science of targeted protein degradation against KRAS G12D-driven cancers. The outcomes of its ongoing Phase 1 clinical trial will be crucial in defining its future therapeutic trajectory and its potential to offer a new treatment modality for patients with these challenging malignancies.
1 Astellas. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting. Astellas Newsroom.
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.
4 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations. ASCO Annual Meeting Abstract.
16 Wei, D., Wang, L., Zuo, X., et al. (2024). A Small Molecule with Big Impact: MRTX1133 Targets the KRASG12D Mutation in Pancreatic Cancer. Clinical Cancer Research, 30(3), 655-662.
15 CenterWatch. (2025, May 19). A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Script.
31 Mount Sinai. A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
32 GlobalData. (2025). Likelihood of Approval and Phase Transition Success Rate Model – ASP-4396 in Solid Tumor.
8 Pharmaceutical Technology. (2025). ASP-4396 by Astellas Pharma for Solid Tumor: Likelihood of Approval.
12 Veeva CTV. A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
17 Ryan, M., et al. (2025). Resistance Mechanisms to KRAS Inhibitors in Cancer. Cancer Discovery.
18 Lim, S. M., et al. (2024, December 13). Targeted oncogenic KRAS degradation in lung adenocarcinoma. bioRxiv.
6 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
16 Dimou, A., & Syrigos, K. (2024). Novel Therapeutic Strategies Targeting KRASG12D in Pancreatic Adenocarcinoma. PMC.
41 Ferrara, R., et al. (2024, October). KRAS, a New Target for Precision Medicine in Colorectal Cancer? ResearchGate.
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations.
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.
38 TMC Pharma. FDA orphan drug designation vs. EMA orphan drug designation.
39 European Medicines Agency. (2014, March 28). Joint European Medicines Agency (EMA), US Food and Drug Administration (FDA), and Japanese Ministry of Health, Labour and Welfare (MHLW) and Pharmaceuticals and Medical Devices Agency (PMDA) orphan medicinal product workshop.
7 Ozmosi. (2024, April 29). ASP-4396 Drug Profile.
46 Pollin, G., et al. (2024). Mutant KRAS inhibitors enter the scene of precision therapeutics. PMC.
16 Astellas Pharma Inc. Patent WO2023171781A1: Heterocyclic Compound for Inducing Degradation of G12d Mutant Kras Protein.
31 Ozmosi. (2024, April 29). ASP-4396 Drug Profile.
8 Pharmaceutical Technology. (2025). ASP-4396 by Astellas Pharma for Solid Tumor: Likelihood of Approval.
47 Google Patents. Patent WO2018092723A1: Ras protein degradation inducing molecule. (Astellas Pharma Inc.).
27 Unified Patents. Patent WO-2021041671-A1: Compounds that inhibit KRas G12D.
17 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions.
6 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Script.
3 Larvol Delta. (2025). Trial in progress: Phase 1 study of the selective protein degrader ASP4396 in patients with locally advanced or metastatic solid tumors with KRAS G12D mutations.
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
49 Google Patents. Patent US20250114339A1 (related to WO2023171781A1): Heterocyclic Compound for Inducing Degradation of G12d Mutant Kras Protein. (Astellas Pharma Inc.).
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.
34 DocWire News. (2024, September 11). Novel KRAS Mutation Degrader's Safety, Antitumor Activity Positive in Pretreated Patients.
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.
17 Cancer Discovery. (2025). Targeting RAS-Driven Cancers: Progress, Challenges, and Future Directions.
23 Astellas Pharma Inc. (2024). Primary Focus: Targeted Protein Degradation. Astellas Innovation.
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
37 ApexOnco. (2024). ESMO 2024 preview – Astellas's KRAS degrader disappoints.
50 Chen, J., et al. (2024). Discovery of Novel Noncovalent Inhibitors Targeting KRASG12D Mutation via Integrated Computational Approaches. MDPI Molecules.
36 IPQwery. Astellas Pharma Inc. Patent Portfolio.
51 WIPO Patentscope. WO/2023/284881 HETEROCYCLIC COMPOUNDS USEFUL AS KRAS G12D INHIBITORS.
26 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
52 Tripp, A., et al. (2024). KRAS Inhibitors in Pancreatic Cancer: Current Landscape and Future Directions. PMC.
36 Astellas Pharma Inc. (2022, December 9). R&D Meeting Script.
19 Urbina, A., et al. (2024). A patent review of von Hippel-Lindau (VHL)-recruiting chemical matter: E3 ligase ligands for PROTACs and targeted protein degradation (2019–present). Taylor & Francis Online.
20 Annual Reviews. (2024). Targeted Protein Degradation: Mechanisms, Strategies, and Cancer Therapy.
24 Astellas Pharma Inc. (2022, December 9). R&D Meeting Script.
55 Google Patents. Patent WO2022148421A1: Bridged compounds as kras g12d inhibitor and degrader and the use thereof. (Beigene, Ltd.).
5 Astellas Pharma Inc. (2024, September 27). Targeted Protein Degradation (TPD) at Astellas - ASP3082 Investor Call Presentation.
22 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.
33 Astellas Clinical Trials. A Study to Find a Suitable Dose of ASP4396 in Adults with Solid Tumors (NCT06364696).
40 Astellas Pharma Inc. (2018, March 22). Astellas Receives Orphan Drug Designation from the Japanese MHLW for Gilteritinib. Astellas Newsroom.
26 Astellas Pharma Inc. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
35 PR Newswire. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
56 Google Patents. Patent CA3148745A1: Compounds that inhibit KRas G12D. (Astellas Pharma Inc.).
57 Google Patents. Patent WO2021106231A1: A compound having inhibitory activity against kras g12d mutation. (Astellas Pharma Inc.).
25 EORTC-NCI-AACR. (2024, October). 36th ENA Symposium Abstracts.
26 Astellas Pharma Inc. (2024). Engineered Small Molecules. Astellas Innovation.
27 Unified Patents. Patent WO-2021041671-A1: Compounds that inhibit KRas G12D.
28 Astellas Pharma Inc. (2025, April). Pipeline Update.
29 Astellas Pharma Inc. (2025, April 25). FY2024 Financial Results Presentation.
30 Astellas Pharma Inc. (2024). Leading the Charge in Oncology: How Astellas is Redefining Cancer Care. Astellas Stories.
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
2 Synapse. (2025, May 17). ASP-4396 Drug Profile.
15 CenterWatch. (2025, May 19). A Study to Find a Suitable Dose of ASP4396 in Adults With Solid Tumors. Clinical Trial Listing NCT06364696.
1 Astellas Newsroom. (2025, May 19). Astellas Presents New Data that Explores Potential of its Cancer Therapies at 2025 ASCO Annual Meeting.
Published at: May 26, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.