MedPath

D3S-001 Advanced Drug Monograph

Published:Jun 16, 2025

Generic Name

D3S-001

D3S-001 (Elisrasib): A Comprehensive Analysis of a Next-Generation KRAS G12C Inhibitor with Best-in-Class Potential

Executive Summary

D3S-001, also known by its International Nonproprietary Name (INN) elisrasib, is an investigational, orally bioavailable, next-generation covalent inhibitor of the Kirsten rat sarcoma viral oncogene homolog (KRAS) G12C mutation. Developed by D3 Bio, a global biotechnology company headquartered in China, D3S-001 is engineered to address the fundamental limitations of first-generation KRAS G12C inhibitors and has demonstrated a compelling clinical profile that positions it as a potential best-in-class therapeutic agent.[1]

The primary differentiating feature of D3S-001 lies in its distinct mechanism of action, characterized by exceptionally rapid and complete target engagement (TE) kinetics. This is driven by a significantly higher association rate constant (kon​) compared to its predecessors, sotorasib and adagrasib.[4] This kinetic superiority enables D3S-001 to effectively overcome the dynamic nucleotide cycling between the inactive, guanosine diphosphate (GDP)-bound state and the active, guanosine triphosphate (GTP)-bound state of the KRAS protein. This cycling represents a key escape mechanism that limits the efficacy of first-generation agents, particularly in the presence of upstream growth factor signaling.[5] By "trapping" the KRAS G12C protein in its inactive state more efficiently, D3S-001 achieves a more profound and durable shutdown of the oncogenic MAPK pathway.

This superior mechanism has translated directly into remarkable clinical efficacy in the ongoing Phase 1/2 trial (NCT05410145). In patients with KRAS G12C-mutant solid tumors who were naïve to prior G12C inhibitor therapy, D3S-001 demonstrated a confirmed overall response rate (ORR) of 73.5% across multiple tumor types. This includes a 66.7% ORR in non-small cell lung cancer (NSCLC), a 75.0% ORR in pancreatic ductal adenocarcinoma (PDAC), and a potentially practice-changing 88.9% ORR as a monotherapy in colorectal cancer (CRC)—a setting where first-generation inhibitors exhibit minimal single-agent activity.[6]

Critically, D3S-001 has provided direct clinical evidence of its ability to overcome acquired resistance. In a cohort of 20 NSCLC patients who had previously progressed on first-generation KRAS G12C inhibitors, D3S-001 monotherapy achieved a 30.0% ORR and an 80.0% disease control rate (DCR), with a median duration of response (DOR) of 8.2 months.[8] Responses were observed in patients with known resistance mechanisms, such as KRAS G12C amplification, further validating its advanced mechanistic profile.[8] Furthermore, the drug has demonstrated the ability to penetrate the central nervous system (CNS) and induce intracranial tumor shrinkage in patients with brain metastases, addressing another significant unmet need.[5]

The safety and tolerability profile of D3S-001 appears favorable and manageable. The most common treatment-related adverse events (TRAEs) are low-grade gastrointestinal effects, such as nausea and diarrhea.[9] The incidence of severe (Grade ≥3) TRAEs is low, and importantly, no cases of interstitial lung disease (ILD), a toxicity of concern for this class, have been reported.[9] In the Phase 1a dose-escalation study, no maximum tolerated dose (MTD) was reached, and a recommended Phase 2 dose (RP2D) of 600 mg once daily has been established for further investigation.[6]

Strategically, D3S-001's unique mechanism, which translates into a superior preclinical and clinical profile, addresses the primary liabilities of the first-generation KRAS G12C inhibitors. Its potent monotherapy activity in CRC, proven efficacy in the resistant setting, and promising CNS activity position it as a potential best-in-class agent. Backed by a well-funded and experienced developer in D3 Bio, D3S-001 is poised to become a cornerstone therapy for KRAS G12C-driven cancers, with a clear development path in multiple high-value indications.

I. The Unmet Need and Scientific Rationale for a Next-Generation KRAS G12C Inhibitor

The "Undruggable" Target Becomes Actionable

For decades, the KRAS oncogene was considered the "white whale" of cancer drug development. As the most frequently mutated oncogene in human cancers, it plays a central role in driving the growth of some of the most lethal malignancies, including approximately 32% of lung adenocarcinomas, 45% of colorectal cancers, and over 90% of pancreatic ductal adenocarcinomas.[12] The KRAS protein functions as a molecular switch, cycling between an active GTP-bound ("on") state and an inactive GDP-bound ("off") state.[15] Mutations, most commonly at codons 12, 13, or 61, lock the protein in its active state, leading to constitutive signaling through downstream pathways like the MAPK pathway (RAF-MEK-ERK), which promotes relentless cell proliferation and survival.[12]

The historical difficulty in targeting KRAS stemmed from its molecular structure. Unlike kinases, which have well-defined ATP-binding pockets amenable to small-molecule inhibition, KRAS has a smooth surface and a picomolar affinity for its natural ligand, GTP, making the development of competitive inhibitors exceedingly challenging.[10]

First-Generation Inhibitors: A Breakthrough with Limitations

This "undruggable" paradigm was shattered by the discovery that the specific KRAS G12C mutation, where glycine is substituted by cysteine at codon 12, creates a unique therapeutic window. The cysteine residue provides a reactive handle for covalent inhibitors to bind to, and this binding induces a cryptic allosteric pocket (the switch-II pocket) that is only accessible when KRAS is in its inactive, GDP-bound state.[12] This groundbreaking insight led to the development and eventual U.S. Food and Drug Administration (FDA) approval of the first-generation KRAS G12C inhibitors: sotorasib (Lumakras) and adagrasib (Krazati).[12] Their approval for patients with previously treated KRAS G12C-mutant NSCLC represented a monumental achievement in precision oncology.[19]

Despite this breakthrough, the clinical performance of these first-generation agents has been marked by significant limitations that have tempered initial enthusiasm. The depth and duration of clinical responses have been modest, especially when compared to the transformative efficacy of targeted therapies for other oncogenic drivers like EGFR or ALK.[6] In the pivotal CodeBreaK 200 trial, sotorasib demonstrated a median progression-free survival (PFS) of just 5.6 months in second-line NSCLC.[19]

This modest efficacy is particularly pronounced in colorectal cancer. As monotherapies, both sotorasib and adagrasib have shown minimal clinical activity in KRAS G12C-mutant CRC.[22] This is largely attributed to robust feedback reactivation of the MAPK pathway, driven by upstream signaling from receptors like the epidermal growth factor receptor (EGFR), which is highly active in CRC.[17] This biological reality has necessitated a combination approach, and the current standard of care for previously treated KRAS G12C CRC is a G12C inhibitor paired with an anti-EGFR antibody, such as sotorasib with panitumumab or adagrasib with cetuximab.[13]

Furthermore, even in responsive tumors, the development of acquired resistance is common and occurs through diverse and heterogeneous mechanisms. These include on-target secondary mutations in the KRAS gene itself (e.g., in the switch-II pocket) that prevent drug binding, as well as off-target mechanisms like the activation of parallel signaling pathways (bypass tracks) or amplification of the KRAS G12C allele, which overwhelms the inhibitor.[10]

The Scientific Imperative for a Superior Agent

The collective limitations of the first-generation inhibitors created a clear and urgent clinical need for a next-generation agent capable of achieving more profound, durable, and broader target inhibition. The central scientific hypothesis guiding the development of agents like D3S-001 is that the key vulnerability of sotorasib and adagrasib is their inability to fully counteract the dynamic "cycling" of the KRAS protein.[4]

The chain of events that limits first-generation inhibitors can be understood as follows:

  1. The inhibitors bind to the inactive, GDP-bound form of KRAS G12C.[12]
  2. However, cancer cells are constantly being stimulated by growth factors in the tumor microenvironment, which activate upstream receptors like EGFR.[6]
  3. This upstream signaling activates guanine nucleotide exchange factors (GEFs), which promote the exchange of GDP for GTP on the KRAS protein.[15]
  4. This shifts the equilibrium towards the active, GTP-bound "on" state of KRAS, a conformation to which the first-generation drugs cannot bind. This active KRAS continues to drive oncogenic signaling, representing a fundamental escape mechanism.[6]
  5. Therefore, an inhibitor's success depends not just on its affinity for the target (potency), but on the speed at which it can bind and "trap" the KRAS G12C protein in its inactive state before it has a chance to be reactivated. This race against reactivation is a battle of kinetics.

This understanding established the scientific imperative for an inhibitor that was not merely more potent, but kinetically superior. The goal was to design a molecule that could engage its target so rapidly and completely that it could effectively shut down the KRAS G12C oncogene, even in the face of strong compensatory signaling. It is precisely this scientific rationale that D3S-001 was engineered to fulfill.[4]

II. Molecular Profile and Preclinical Validation of D3S-001

Chemical and Pharmaceutical Profile

D3S-001 is an orally administered small molecule drug developed by D3 Bio.[28] Its INN is elisrasib.[30] The compound's identity and key properties are summarized in Table 1.

Table 1: Chemical and Pharmaceutical Profile of D3S-001 (Elisrasib)

| Property | Value | Source Snippet(s) |

| :--- | :--- | :--- |

| International Nonproprietary Name (INN) | Elisrasib | 30 |

| Chemical Formula | C32​H35​F6​N7​O3​ | 30 |

| Molecular Weight | 679.7 g/mol | 30 |

| IUPAC Name | 2-((S)-4-((S)-7-(3-amino-2-fluoro-5-methyl-6-(trifluoromethyl)phenyl)-2-(((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methoxy)-7,8-dihydro-5H-pyrano[4,3-d]pyrimidin-4-yl)-1-(2-fluoroacryloyl)piperazin-2-yl)acetonitrile | 30 |

| CAS Number | 2706637-43-4 | 30 |

| DrugBank ID | DB19008 | 30 |

| NCI Thesaurus Code | C188231 | 30 |

| ChEMBL ID | CHEMBL5314913 | 30 |

| InChIKey | JOLORSRKBHXPFT-PNGHJTAWSA-N | 30 |

| SMILES | CC1=CC(=C(C(=C1C(F)(F)F)[C@@H]2CC3=C(CO2)C(=NC(=N3)OC[C@@]45CCCN4CC@@HF)N6CCN(C@HCC#N)C(=O)C(=C)F)F)N | 30 |

A Differentiated Mechanism of Action: The Kinetic Advantage

The core of D3S-001's innovation lies in its molecular mechanism, which was specifically engineered to enhance target engagement and overcome the kinetic liabilities of its predecessors.[5]

Superior Covalent Potency and Target Engagement (TE) Kinetics

Preclinical biochemical and cellular assays have consistently demonstrated that D3S-001 possesses substantially improved covalent potency and significantly faster target engagement kinetics compared to both sotorasib and adagrasib.[5] This superiority is not just a marginal improvement but a leap of one to two orders of magnitude in key parameters.

A critical detail in its design is that this enhanced potency is primarily driven by a much faster association rate constant (kon​)—the speed at which the drug binds to its target pocket—rather than an increase in the rate of chemical reaction (kinact​). This is a hallmark of sophisticated medicinal chemistry, as it signifies a more refined and optimized fit to the target protein, which enhances binding speed and affinity without increasing the risk of indiscriminate, off-target reactions with other proteins. This contributes to both its superior efficacy and its clean safety profile.[4]

Overcoming Nucleotide Cycling

This kinetic advantage has a direct and profound functional consequence: it allows D3S-001 to overcome the challenge of nucleotide cycling. In cellular assays, the inhibitory activity of sotorasib and adagrasib was significantly compromised when cells were stimulated with growth factors like EGF or HGF, which drive KRAS into its active, drug-resistant state. In stark contrast, the target engagement kinetics of D3S-001 were nearly unaffected by these same stimuli.[4] This unique feature allows D3S-001 to effectively deplete the pool of cellular active (GTP-bound) KRAS even at low nanomolar concentrations, achieving a much deeper and more sustained shutdown of the oncogenic signal.[4] The quantitative difference in performance is stark, as detailed in Table 2.

Table 2: Comparative Analysis of Preclinical Potency and Target Engagement Kinetics

| Parameter | D3S-001 | Adagrasib | Sotorasib | Source Snippet(s) |

| :--- | :--- | :--- | :--- | :--- |

| Cellular Active KRAS Inhibition IC50 | 0.6 nM | 78 nM | 35 nM | 15 |

| Covalent Potency (kinact​/KI​) | 1.43×106M−1s−1 | 7.14×104M−1s−1 | 2.04×104M−1s−1 | 15 |

| Cellular TE t1/2​ | 5.8 minutes | 34 minutes | 44 minutes | 15 |

| Effect of EGF on TE | Nearly unaffected | Compromised by >55% | Compromised by >55% | 6 |

Comprehensive Preclinical Validation

The mechanistic superiority of D3S-001 observed in biochemical and cellular assays translates into a robust and differentiated preclinical activity profile.

In Vitro and In Vivo Potency: In a broad panel of KRAS G12C-mutant cancer cell lines, D3S-001 consistently demonstrated single-digit nanomolar IC50 values for inhibiting downstream signaling (p-ERK) and cell proliferation. Importantly, it showed high selectivity, with no anti-proliferative effects on non-KRAS G12C mutant cell lines, underscoring its precision.[18] In animal xenograft models, D3S-001 induced potent, dose-dependent anti-tumor effects, leading to tumor regression at doses as low as 10 mg/kg and achieving durable complete remissions in more challenging syngeneic models.[18] Head-to-head comparisons in cell line-derived (CDX) and patient-derived xenograft (PDX) models of NSCLC, CRC, and pancreatic cancer consistently showed that D3S-001 produced more robust anti-tumor responses than sotorasib or adagrasib.[5]

CNS Penetration and Intracranial Activity: A critical point of differentiation for D3S-001 is its designed ability to cross the blood-brain barrier.[8] This property was confirmed in preclinical studies, which is of immense clinical relevance as brain metastases are a frequent and devastating complication of NSCLC. In mouse models of brain metastases, treatment with D3S-001 resulted in durable intracranial tumor regression, a finding that stands in contrast to the more limited CNS activity of its predecessors and suggests a significant potential advantage in a patient population with high unmet need.[5]

Overcoming Acquired Resistance: The ability of D3S-001 to overcome resistance was validated preclinically. In xenograft models derived from tumors that had developed acquired resistance to sotorasib, including models with confirmed KRAS G12C gene amplification, D3S-001 treatment led to robust tumor regression and delayed disease progression. In the same models, sotorasib and adagrasib had no effect.[5] This provides a powerful preclinical rationale for the drug's use in patients who have already failed a first-generation inhibitor.

Combination Synergy: Recognizing that combination therapy is the future of cancer treatment, preclinical studies also explored D3S-001's potential as a partner agent. When combined with an anti-PD-1 antibody in a syngeneic tumor model, the combination achieved an impressive 70% durable complete remission rate. Furthermore, mice that achieved complete remission were protected from tumor re-growth upon rechallenge, suggesting the induction of a powerful and lasting memory T-cell response.[18] This provides a strong scientific basis for the clinical exploration of D3S-001 in combination with immunotherapy.

The preclinical data package for D3S-001 presents an exceptionally coherent and compelling narrative. There is a clear, evidence-based line connecting its specific molecular design (optimizing kon​ for rapid TE) to its functional advantage in biochemical assays (overcoming nucleotide cycling), which in turn translates to superior efficacy in a wide range of challenging preclinical models (head-to-head comparisons, CNS metastases, acquired resistance). This logical progression from "bench to bedside" provides a high degree of scientific validation and de-risks the asset, as the "why" behind its promising clinical activity is well-understood.

III. The D3S-001-100 Global Clinical Development Program (NCT05410145)

The clinical evaluation of D3S-001 is being conducted under a comprehensive and strategically designed global study, identified by the clinical trial number NCT05410145 and the protocol code D3S-001-100.[29] This first-in-human (FIH), multicenter, open-label, Phase 1/2 trial is structured to rapidly and efficiently assess the drug's safety, pharmacokinetics, and efficacy across multiple settings.

Trial Design and Methodology

The D3S-001-100 study employs a multi-part, adaptive design that allows for the simultaneous investigation of the drug as both a monotherapy and a combination agent. This structure is highly efficient, generating a wealth of data to inform a multifaceted development and regulatory strategy much faster than a traditional, sequential approach.

  • Part 1 (Phase 1a): Monotherapy Dose Escalation. This initial part of the trial enrolled patients with various advanced KRAS G12C-mutant solid tumors. Its primary goal was to establish the safety, tolerability, and pharmacokinetic profile of D3S-001 across a range of escalating doses (from 50 mg to 900 mg once daily) and to determine the maximum tolerated dose (MTD) and the recommended Phase 2 dose (RP2D).[6]
  • Part 2 (Phase 1b/2): Monotherapy Dose Expansion. Following the determination of the RP2D, this part of the study involves enrolling larger cohorts of patients into specific disease- and line-of-therapy-defined groups to further evaluate efficacy. The most prominent and widely reported of these is a cohort of NSCLC patients whose disease had progressed following treatment with a prior KRAS G12C inhibitor. This cohort was strategically designed to provide rapid proof-of-concept for D3S-001's key differentiating feature: its ability to overcome acquired resistance.[6]
  • Part 3: Combination Therapy. This forward-looking part of the trial is designed to explore the future of D3S-001 as a backbone therapy. It evaluates D3S-001 in combination with other standard-of-care and targeted anti-cancer agents, including the anti-PD-1 immunotherapy pembrolizumab, the anti-EGFR antibody cetuximab, and standard platinum-based chemotherapy regimens (cisplatin or carboplatin plus pemetrexed).[28] This allows D3 Bio to get a head start on planning pivotal Phase 3 trials for multiple indications, such as first-line NSCLC and second-line CRC.

The trial's primary objectives focus on safety and tolerability, including the incidence of adverse events (AEs) and dose-limiting toxicities (DLTs), and the determination of the MTD and RP2D.[28] Secondary objectives include a full characterization of the drug's pharmacokinetics and preliminary efficacy, as measured by standard RECIST v1.1 criteria such as ORR, DOR, PFS, and DCR.[6] Eligible patients are adults with histologically confirmed, locally advanced or metastatic solid tumors with a documented KRAS p.G12C mutation and measurable disease.[29]

Pharmacokinetics (PK) and Dose Selection

D3S-001 is administered orally once daily in 21-day treatment cycles.[28] The Phase 1a dose-escalation study evaluated doses ranging from 50 mg to 900 mg once daily.[27] The drug exhibited predictable, dose-dependent pharmacokinetics with a terminal plasma half-life (

t1/2​) of approximately 11 hours, which is supportive of a once-daily dosing schedule.[6]

Crucially, no MTD was reached during dose escalation, indicating a wide therapeutic window and a favorable safety profile even at high doses.[5] Based on an integrated analysis of the safety, tolerability, pharmacokinetic, and preliminary efficacy data, the RP2D was established as

600 mg once daily.[6] Pharmacodynamic data further supported this dose, showing that daily doses of less than 600 mg were sufficient to achieve complete suppression of the mutant KRAS G12C protein in patient plasma.[11]

Pharmacodynamics (PD): The Role of ctDNA as a Predictive Biomarker

A key feature of the D3S-001-100 trial is the intensive use of liquid biopsies to monitor circulating tumor DNA (ctDNA) as an exploratory endpoint.[10] This has proven to be a powerful tool for demonstrating early on-target activity and predicting clinical outcomes.

Treatment with D3S-001 was shown to induce a rapid and sustained reduction in the KRAS G12C mutant allele frequency (MAF) in the blood, with significant declines observed as early as Day 8 of the first treatment cycle.[5] This molecular response serves as a robust pharmacodynamic biomarker, providing an early signal that the drug is effectively engaging its target and inhibiting the oncogenic driver in patients.

More importantly, a strong correlation has been established between this early molecular response and subsequent clinical benefit. For example, in the G12Ci-resistant NSCLC cohort, all six patients who achieved a radiological response were ctDNA-positive at baseline, and five of them achieved complete clearance of the G12C MAF from their blood.[10] In the G12Ci-naïve population, patients who achieved 100% clearance of their G12C ctDNA had a remarkable 90% confirmed partial response rate and a significantly lower risk of disease progression compared to those with incomplete clearance (HR 0.2, p=0.005).[27]

The ability to use ctDNA clearance as an early predictor of efficacy is a significant advantage for agile clinical development. It allows investigators to get an early read on whether the drug is active in a given patient or cohort, long before traditional imaging endpoints mature after several months of treatment. This can help guide dose selection, identify promising patient subgroups for expansion, and increase overall confidence in the asset's potential for investors and potential partners, thereby de-risking further development.

IV. Comprehensive Clinical Efficacy Analysis

The clinical data emerging from the NCT05410145 trial have been consistently impressive, demonstrating high response rates in both treatment-naïve and heavily pretreated patient populations. The results validate the preclinical hypothesis that D3S-001's superior kinetic profile would translate into superior clinical activity.

Activity in KRAS G12C Inhibitor (G12Ci)-Naïve Solid Tumors (Phase 1a)

In the dose-escalation part of the study, D3S-001 demonstrated compelling anti-tumor activity across a range of KRAS G12C-mutant solid tumors in patients who had not previously received a G12C inhibitor. Among 34 evaluable patients in this population, D3S-001 achieved a confirmed ORR of 73.5%.[5] The disease control rate was over 97%, indicating that nearly all patients experienced clinical benefit in the form of tumor shrinkage or disease stabilization.[11] The responses also appeared to be durable, with a reported six-month DOR rate of 70%.[11]

The efficacy was notable across all major KRAS G12C-driven tumor types, as summarized in Table 3.

Table 3: Summary of Efficacy in G12Ci-Naïve Patients by Tumor Type (NCT05410145, Part 1a)

| Tumor Type | Number of Patients (n) | Confirmed Objective Response Rate (ORR) | Disease Control Rate (DCR) | Source Snippet(s) |

| :--- | :--- | :--- | :--- | :--- |

| Overall | 34 (evaluable) | 73.5% | >97% | 6 |

| NSCLC | 21 | 66.7% | Not Reported | 6 |

| CRC | 9 | 88.9% | Not Reported | 6 |

| PDAC | 4 | 75.0% | Not Reported | 6 |

The most striking of these results is the 88.9% ORR in patients with CRC. This is a potential game-changer. As previously discussed, first-generation KRAS G12C inhibitors have very low single-agent activity in CRC (ORRs typically below 20%) due to the strong EGFR-mediated feedback resistance mechanism inherent to this tumor type.[22] The current standard of care therefore requires a combination with an anti-EGFR antibody to achieve response rates in the range of 30-40%.[13] D3S-001's ability to achieve such a high response rate as a monotherapy suggests that its target inhibition is so profound and rapid that it can effectively shut down the oncogenic pathway before this feedback loop can be established. This has profound strategic implications, as a highly effective oral monotherapy would be vastly preferable to a combination therapy involving intravenous infusions in terms of patient convenience, safety, and cost. If the monotherapy is this effective, the planned combination with cetuximab could set an unprecedented efficacy bar for this disease.[28]

Overcoming Acquired Resistance: Efficacy in G12Ci-Pretreated NSCLC (Phase 1b/2)

The ultimate test for a next-generation inhibitor is its ability to work where its predecessors have failed. The dose-expansion cohort enrolling 20 NSCLC patients who had progressed on prior G12C inhibitors (including sotorasib and adagrasib) was designed to answer this question directly.[8] The results, presented at the AACR 2025 meeting (abstract CT266), provide clear clinical evidence that D3S-001 can overcome acquired resistance.[10] The key efficacy results for this cohort are detailed in Table 4.

Table 4: Efficacy Results in the G12Ci-Resistant NSCLC Cohort (NCT05410145, Part 1b/2)

| Efficacy Endpoint | Result | Note/Subgroup | Source Snippet(s) |

| :--- | :--- | :--- | :--- |

| Overall Response Rate (ORR) | 30.0% (6/20) | Confirmed Partial Responses | 8 |

| Disease Control Rate (DCR) | 80.0% (16/20) | PR + Stable Disease | 8 |

| Tumor Shrinkage | 60.0% (12/20) | Any reduction in tumor size | 8 |

| Median Duration of Response (DOR) | 8.2 months | 95% CI: 1.5-not calculable | 9 |

| ORR in ctDNA+ population | 43% (6/14) | All 6 responders were ctDNA+ at baseline | 8 |

| Response in KRAS G12C Amplification | 2 PR, 1 SD (n=3) | Activity in a known resistance mechanism | 8 |

This 30% ORR with a durable median response of 8.2 months is a highly meaningful clinical benefit for this refractory patient population, which currently has no effective targeted options and must often revert to less effective and more toxic chemotherapy.[9] This result validates the drug's mechanism and provides a clear, data-driven path toward a potential accelerated approval in the third-line or later setting for KRAS G12C-mutant NSCLC.

The subgroup analyses provide the biological rationale for this efficacy. The observation of responses in patients with KRAS G12C amplification—a mechanism where the cancer cell produces more of the target protein to overwhelm the drug—directly supports the hypothesis that D3S-001's superior potency and target engagement can overcome this resistance strategy.[8] Furthermore, the strong correlation between ctDNA clearance and response in this cohort reinforces the drug's potent on-target activity.[8] Conversely, patients who did not respond and had immediate disease progression were found to have baseline alterations known to confer resistance, such as secondary mutations in the drug-binding pocket (e.g., KRAS R68S) or activation of bypass pathways (e.g., BRAF V600E), which aligns perfectly with the preclinical findings and helps to define the patient populations most, and least, likely to benefit.[10]

Intracranial Efficacy: Clinical Evidence of CNS Activity

Confirming the promise shown in preclinical models, the clinical trial has generated evidence of D3S-001's activity in the central nervous system. It has been reported that a proportion of patients with brain metastases experienced intracranial tumor shrinkage or achieved stable intracranial disease upon treatment with D3S-001.[5] This is a critical finding, as brain metastases are a major cause of morbidity and mortality in NSCLC, and many systemic therapies penetrate the CNS poorly. An effective, oral agent that can treat both systemic and intracranial disease would represent a significant therapeutic advance and address a major unmet need for these patients.

V. Safety and Tolerability Profile

A successful therapeutic agent must balance high efficacy with a manageable safety profile. Across the Phase 1/2 clinical trial, D3S-001 has demonstrated favorable tolerability, which is crucial for its potential use as both a monotherapy and a combination backbone agent.[8]

Overall Assessment and Common Adverse Events

The safety profile of D3S-001 is characterized primarily by predictable and manageable treatment-related adverse events (TRAEs). The most frequently reported side effects are gastrointestinal in nature, including nausea, vomiting, and diarrhea, which were predominantly mild to moderate (Grade 1 or 2) in severity.[9] Liver dysfunction, manifesting as elevated liver enzymes (transaminases), has also been identified as a notable toxicity, but these events have also been primarily low-grade and manageable.[9]

Severe (Grade ≥3) TRAEs and Discontinuations

The rate of severe TRAEs has been low, underscoring the drug's good tolerability. In the G12Ci-naïve dose-escalation cohort (n=42), Grade 3 TRAEs were reported in 16.7% of patients.[6] In the more heavily pretreated G12Ci-resistant NSCLC cohort (n=20), the rate of Grade 3 TRAEs was even lower at 10%.[10] Specific Grade 3 events that have been mentioned include an increase in lipase levels in one patient.[9]

Critically, no Grade 4 (life-threatening) or Grade 5 (fatal) TRAEs have been reported in the trial, a testament to the drug's favorable safety window.[6] While dose modifications due to TRAEs were necessary in some patients (26.8% in one analysis), permanent treatment discontinuations due to AEs have been rare.[41]

Adverse Events of Special Interest

A key safety differentiator for D3S-001 is the absence of certain severe toxicities that are a concern for other kinase inhibitors. Most importantly, it has been explicitly stated that no patients have developed interstitial lung disease (ILD) or pneumonitis.[9] This is a significant advantage, as ILD/pneumonitis is a known, albeit infrequent, risk with the approved KRAS G12C inhibitor adagrasib, with a reported incidence of 4.1% and one fatal case in its pooled safety data.[42] The clean safety profile of D3S-001, particularly the absence of severe pulmonary toxicity, makes it a more attractive candidate for long-term administration and, crucially, for use in combination therapies where overlapping toxicities can be a dose-limiting factor. A summary of the key safety findings is presented in Table 5.

Table 5: Summary of Treatment-Related Adverse Events (NCT05410145)

| Adverse Event | Any Grade (%) | Grade ≥3 (%) | Key Notes | Source Snippet(s) |

| :--- | :--- | :--- | :--- | :--- |

| All TRAEs (Resistant NSCLC) | 90% (18/20) | 10% (2/20) | Generally well-tolerated | 10 |

| All TRAEs (Naïve Cohort) | 73.2% (30/41) | 14.6% (6/41) | Favorable safety profile | 41 |

| Nausea / Vomiting | Common | Primarily Low Grade | Most frequent AEs | 9 |

| Diarrhea | Common | Primarily Low Grade | Manageable GI toxicity | 11 |

| Liver Dysfunction | Notable | Primarily Low Grade | ALT/AST increase observed | 9 |

| Increased Lipase | Not specified | One Grade 3 case reported | Specific Grade 3 AE noted | 9 |

| Interstitial Lung Disease (ILD) | 0% | 0% | Significant safety advantage | 9 |

VI. Developer, Intellectual Property, and Competitive Landscape

Developer Profile: D3 Bio

D3S-001 is the lead asset of D3 Bio, a privately held, clinical-stage biotechnology company with a global focus.[2] Founded between 2018 and 2020, the company is headquartered in Shanghai and Wuxi, China, and is dedicated to developing innovative medicines in oncology and immunology.[1]

D3 Bio's stated R&D strategy is notably differentiated. Rather than beginning with laboratory science, the company's approach starts by identifying the greatest unmet clinical and market needs. These clinical insights are then used to guide the discovery and development process, a philosophy encapsulated in its name: "Development leading to Discovery that then leads back to more Development".[2]

The company is led by a seasoned team with a strong track record in the pharmaceutical industry. The co-founder, Chairman, and CEO is Dr. George Chen, an oncologist by training who has held senior leadership roles in R&D at major global pharmaceutical companies, including AstraZeneca and Johnson & Johnson.[45] The management team is credited with experience in delivering multiple first-in-class and blockbuster medications.[45]

D3 Bio is well-capitalized, having secured substantial funding from a syndicate of world-class investors. The company launched with a US200millionSeriesAfinancingroundinNovember2020andsubsequentlyclosedaUS62 million Series A+ round in April 2024, specifically to accelerate the development of its oncology pipeline, with a focus on D3S-001.[45] The participation of sophisticated investors such as Boyu Capital, Matrix Partners China, Hongshan (formerly Sequoia Capital China), the Singaporean sovereign wealth fund Temasek, WuXi AppTec's Corporate Venture Fund, and the leading European life sciences firm Medicxi provides strong external validation of the company's strategy and the quality of its assets.[1]

The company's pipeline demonstrates a clear strategic focus. D3S-001 is the lead "backbone" asset. It is complemented by D3S-002, an ERK1/2 inhibitor also in Phase 1 development.[44] This provides a rational, in-house combination strategy to tackle MAPK pathway-mediated resistance, a known escape mechanism for KRAS inhibitors, as demonstrated in preclinical studies.[15] The pipeline also includes D3L-001, a CD47 x HER2 bispecific antibody, indicating broader ambitions in immuno-oncology.[44]

Intellectual Property (IP) and Regulatory Status

D3S-001 is protected by intellectual property covering pyrimidoheterocyclic compounds. The patent WO2021180181 has been cited in reference to the chemical matter of D3S-001.[32] A comprehensive Freedom to Operate (FTO) analysis would require a detailed examination of this patent's claims and geographic coverage, the full text of which was not available in the provided research materials, representing a key data gap for a complete due diligence assessment.[49]

From a regulatory perspective, D3S-001 has garnered significant support from the U.S. FDA, which has granted it multiple designations that can expedite its development and review. These include:

  • Fast Track Designation (FTD) for the treatment of colorectal cancer with a KRAS G12C mutation (granted November 2022).[1]
  • Fast Track Designation (FTD) for the treatment of late-line non-small cell lung cancer.[5]
  • Orphan Drug Designation (ODD) for the treatment of pancreatic cancer.[5]

These designations underscore the agency's recognition of D3S-001's potential to address serious unmet medical needs.

Competitive Landscape and Strategic Positioning

The KRAS inhibitor space is one of the most dynamic and competitive areas in oncology. D3S-001 enters a landscape with established players, a rapidly evolving standard of care, and a robust pipeline of next-generation challengers.

Current Standard of Care (SoC):

  • NSCLC (Second-Line and Beyond): The established SoC is targeted therapy with the first-generation inhibitors sotorasib or adagrasib.[12] D3S-001's path here involves demonstrating superiority over these agents or, more strategically, proving its value in the post-sotorasib/adagrasib resistance setting.
  • CRC (Second-Line and Beyond): The SoC is combination therapy with a G12C inhibitor plus an anti-EGFR antibody (sotorasib + panitumumab or adagrasib + cetuximab).[13] D3S-001's remarkable monotherapy activity presents a direct challenge to this paradigm.
  • PDAC: The SoC remains multi-agent chemotherapy regimens like FOLFIRINOX or gemcitabine/nab-paclitaxel.[51] KRAS G12C inhibitors are an option in later lines but have shown only modest activity, leaving a significant opportunity for a more effective agent like D3S-001.[14]

Next-Generation Pipeline: The field is crowded with companies seeking to improve upon the first-generation agents. Key next-generation G12C inhibitors in late-stage clinical development include divarasib (GDC-6036) from Roche, olomorasib from Eli Lilly, and MK-1084 from Merck & Co., all of which have shown promising data.[54] The competitive landscape also includes pan-KRAS inhibitors and agents targeting other mutations like G12D and G12V, though these are at an earlier stage of development.[58]

D3S-001's current data package appears highly competitive against this backdrop. While it is earlier in its development timeline than some competitors, its unique combination of high monotherapy efficacy in CRC, proven activity in the resistant setting, and CNS penetration gives it multiple avenues to establish a best-in-class profile. A summary comparison is provided in Table 6.

Table 6: Competitive Landscape of Key KRAS G12C Inhibitors

| Drug (Company) | Development Stage | NSCLC ORR (2L+) | CRC ORR (Monotherapy) | CRC ORR (Combo w/ anti-EGFR) | G12Ci-Resistant NSCLC ORR | Key Safety Notes |

| :--- | :--- | :--- | :--- | :--- | :--- | :--- |

| D3S-001 (D3 Bio) | Phase 2 | 66.7% (naïve) | 88.9% | Planned | 30.0% | Favorable; No ILD reported |

| Sotorasib (Amgen) | Approved | 36-41% | ~10% | 26% | Limited activity | Liver toxicity |

| Adagrasib (BMS) | Approved | 43% | ~20% | 34-46% | Limited activity | GI toxicity, ILD/pneumonitis risk |

| Divarasib (Roche) | Phase 3 | 53.4% | Not Reported | Noteworthy activity | Activity reported | Generally well-tolerated |

| Olomorasib (Lilly) | Phase 1/2 | 70% (1L) | Not Reported | 42% | Activity reported | Generally well-tolerated |

| MK-1084 (Merck) | Phase 1/3 | Not Reported | 38% | 46% | Not Reported | Generally well-tolerated |

Note: ORR data are from different trials and patient populations and are not directly comparable. Data for D3S-001 in 2L+ NSCLC is not available; naïve data is shown for context.

Data Sources:.6

The primary strategic challenge for D3 Bio will be navigating this competitive environment. The company's development timeline is behind some key competitors. Therefore, its success will depend on designing and executing pivotal trials that can either accelerate its path to market (e.g., via an accelerated approval in the resistant setting, for which it has FTD) or generate a data package so compelling (e.g., monotherapy superiority in CRC) that it can displace earlier market entrants.

VII. Future Directions and Strategic Outlook

The robust data package for D3S-001 provides a strong foundation for a multifaceted development strategy aimed at establishing it as a cornerstone therapy for KRAS G12C-mutant cancers. The future of KRAS inhibition universally points toward combination therapies to deepen initial responses, prolong durability, and overcome the inevitable emergence of resistance.[12] D3S-001 is exceptionally well-positioned for this future, given its potent monotherapy backbone and favorable safety profile, which provides a wider therapeutic window for combining with other agents.

The Path Forward: Combination Therapies

The ongoing NCT05410145 trial is already exploring several key combination strategies:

  • Combination with Anti-EGFR (Cetuximab): This is a logical and high-potential strategy for CRC. Given D3S-001's unprecedented 88.9% ORR as a monotherapy in this setting, the addition of cetuximab to block the EGFR feedback loop could potentially yield even higher response rates and greater durability, setting a new and formidable standard of care.[28]
  • Combination with Anti-PD-1 (Pembrolizumab): This combination is the key to unlocking the largest commercial opportunity: the first-line treatment of KRAS G12C-mutant NSCLC.[28] The current SoC in this setting is often chemo-immunotherapy. A highly effective, all-oral targeted therapy plus immunotherapy combination could offer a chemotherapy-sparing regimen with superior efficacy and tolerability. The preclinical data showing synergy and the induction of a memory T-cell response with an anti-PD-1 antibody provides a strong rationale for this approach.[18]
  • Combination with Downstream Inhibitors (e.g., D3S-002): D3 Bio's internal pipeline includes an ERK1/2 inhibitor, D3S-002.[44] This provides a rational, vertical inhibition strategy to tackle resistance mediated by reactivation of the MAPK pathway downstream of KRAS. Preclinical studies have already shown that combining D3S-001 with an ERK inhibitor can significantly prolong survival in models of acquired resistance.[15]

Market Positioning and Potential

D3S-001 has multiple potential paths to market, each with significant value:

  • Initial Market Entry (Resistant NSCLC): The most direct and fastest path to approval is likely in the second-line or later setting for NSCLC patients who have progressed on a first-generation G12C inhibitor. The 30% ORR in this population fills a clear unmet need, and the FDA Fast Track Designation for late-line NSCLC supports an accelerated approval strategy.[5]
  • Major Opportunity in CRC: The potent monotherapy activity could fundamentally disrupt the current G12Ci + anti-EGFR combination paradigm. A pivotal trial demonstrating superiority or even non-inferiority of D3S-001 monotherapy compared to the combination SoC would be practice-changing and could capture a significant market share.
  • First-Line NSCLC: Success in the planned combination with immunotherapy would position D3S-001 in the largest and most lucrative market segment for KRAS G12C inhibitors. This will be a highly competitive area, but a regimen that improves upon chemo-immunotherapy would be a major commercial success.
  • Pancreatic Cancer: While based on a small number of patients (n=4), the 75% ORR is exceptionally promising in a disease with notoriously poor outcomes and few effective treatments.[8] The Orphan Drug Designation provides incentives for further development, and success in this indication would be a major clinical advance.[5]

Key Questions and Data Gaps

Despite the highly promising data, several key questions remain that will be critical for the continued development and ultimate valuation of D3S-001:

  • Intellectual Property: What are the full claims, scope, and expiration dates of the patent portfolio covering D3S-001, particularly WO2021180181? A complete FTO analysis is essential.
  • Durability Data: What will the mature Duration of Response (DOR) and Progression-Free Survival (PFS) data look like from the G12Ci-naïve cohorts, especially in CRC and PDAC? Demonstrating durability will be key to confirming the initial high response rates.
  • Combination Safety: How will the toxicity profile of the combination therapies (e.g., with cetuximab or pembrolizumab) compare to monotherapy? Managing overlapping toxicities will be critical for the success of these regimens.
  • Pivotal Trial Strategy: What are the specific designs, timelines, and comparator arms for D3 Bio's planned Phase 3 pivotal trials? The company's ability to execute these trials efficiently and strategically will determine its competitive standing.

VIII. Conclusion

D3S-001 (elisrasib) has emerged as a formidable next-generation KRAS G12C inhibitor with a data package that strongly supports its potential to become a best-in-class therapeutic. Its development is founded on a sophisticated understanding of the kinetic limitations of its predecessors, and its molecular design successfully addresses these core liabilities.

The drug's differentiated mechanism—characterized by exceptionally rapid and complete target engagement that overcomes nucleotide cycling—is not merely a theoretical advantage. It has been shown to translate directly into a superior preclinical profile and, most importantly, into compelling clinical efficacy. The results from the NCT05410145 trial are remarkable for their breadth and depth, demonstrating high response rates in treatment-naïve patients and, critically, meaningful and durable activity in patients who have already developed resistance to first-generation inhibitors.

The standout monotherapy efficacy in colorectal cancer, the proven ability to overcome resistance in non-small cell lung cancer, and the promising activity in the central nervous system represent three distinct and highly valuable pillars of its clinical profile. These achievements, coupled with a favorable and manageable safety profile, position D3S-001 as a highly attractive agent for both monotherapy and combination strategies.

Developed by D3 Bio, a well-funded and strategically focused company with experienced leadership, D3S-001 is poised to become a major contender in the dynamic and competitive KRAS inhibitor landscape. While challenges related to development timelines in a crowded field remain, the strength of the data suggests that D3S-001 has the potential to significantly raise the bar for efficacy and become a future cornerstone therapy for patients with KRAS G12C-driven cancers. The execution of its late-stage clinical and regulatory strategy will be critical in realizing this immense potential.

Works cited

  1. D3 Bio's D3S-001 Receives U.S. FDA Fast Track Designation for the Treatment of Colorectal Cancer with KRAS G12C mutation, accessed June 16, 2025, https://www.d3bio.com/press-releases/d3-bios-d3s-001-receives-u-s-fda-fast-track-designation-for-the-treatment-of-colorectal-cancer-with-kras-g12c-mutation
  2. D3 Bio, Inc, accessed June 16, 2025, https://www.d3bio.com/
  3. D3 Bio (Wuxi) Co. Ltd. - Page 1 - BioWorld, accessed June 16, 2025, https://www.bioworld.com/keywords/52451-d3-bio-wuxi-co-ltd
  4. D3S-001, a KRAS G12C Inhibitor with Rapid Target Engagement Kinetics, Overcomes Nucleotide Cycling, and Demonstrates Robust Preclinical and Clinical Activities - PMC - PubMed Central, accessed June 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11372373/
  5. D3 Bio's D3S-001 Demonstrates Significantly Improved Covalent Potency in Depleting Cellular Active KRAS in Pre-Clinical and Clinical Studies in KRAS G12C-Mediated Cancers - Business Wire, accessed June 16, 2025, https://www.businesswire.com/news/home/20241125088779/en/D3-Bios-D3S-001-Demonstrates-Significantly-Improved-Covalent-Potency-in-Depleting-Cellular-Active-KRAS-in-Pre-Clinical-and-Clinical-Studies-in-KRAS-G12C-Mediated-Cancers
  6. D3S-001 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 16, 2025, https://synapse.patsnap.com/drug/4460f7c4a2f344a4abb9a463f4773736
  7. D3S-001, a KRAS G12C Inhibitor with Rapid Target Engagement Kinetics, Overcomes Nucleotide Cycling, and Demonstrates Robust Preclinical and Clinical Activities - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/38717075/
  8. D3 Bio Announces Nature Medicine Publication and AACR 2025 Presentation Highlighting D3S-001 as a Next-Generation KRAS G12C Inhibitor with Best-in-Class Potential - BioSpace, accessed June 16, 2025, https://www.biospace.com/press-releases/d3-bio-announces-nature-medicine-publication-and-aacr-2025-presentation-highlighting-d3s-001-as-a-next-generation-kras-g12c-inhibitor-with-best-in-class-potential
  9. Dr Loong on the Efficacy of D3S-001 in KRAS G12C Inhibitor–Resistant NSCLC - OncLive, accessed June 16, 2025, https://www.onclive.com/view/dr-loong-on-the-efficacy-of-d3s-001-in-kras-g12c-inhibitor-resistant-nsclc
  10. Abstract CT266: D3S-001, a next generation GDP-bound KRAS G12C inhibitor, as monotherapy in KRAS G12C inhibitor resistant non-small cell lung cancer, accessed June 16, 2025, https://aacrjournals.org/cancerres/article/85/8_Supplement_2/CT266/761474/Abstract-CT266-D3S-001-a-next-generation-GDP-bound
  11. CUHK-led multinational study shows new China-developed cancer drug D3S-001 is effective in treating solid tumours, including lung, colorectal and pancreatic cancer, accessed June 16, 2025, https://www.cpr.cuhk.edu.hk/en/press/cuhk-led-multinational-study-shows-new-china-developed-cancer-drug-d3s-001-is-effective-in-treating-solid-tumours-including-lung-colorectal-and-pancreatic-cancer/
  12. Management of KRAS-Mutated Non–Small Cell Lung Cancer - Hematology & Oncology, accessed June 16, 2025, https://www.hematologyandoncology.net/archives/march-2024/management-of-kras-mutated-non-small-cell-lung-cancer/
  13. Targeted therapy meets immunotherapy: a trial for metastatic KRAS G12C-mutant CRC, accessed June 16, 2025, https://fightcolorectalcancer.org/blog/targeted-therapy-meets-immunotherapy/
  14. KRAS Mutations and Pancreatic Cancer, accessed June 16, 2025, https://pancan.org/facing-pancreatic-cancer/kras-mutations/
  15. D3S-001, a KRAS G12C inhibitor with rapid target ... - AACR Journals, accessed June 16, 2025, https://aacrjournals.org/cancerdiscovery/article-pdf/doi/10.1158/2159-8290.CD-24-0006/3451530/cd-24-0006.pdf
  16. Immunotherapy in advanced, KRAS G12C-mutant non-small-cell lung cancer: current strategies and future directions - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/40093982/
  17. KRAS G12C inhibitor combination therapies: current evidence and challenge - Frontiers, accessed June 16, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2024.1380584/full
  18. Preclinical characterization of D3S-001, a highly potent, selective, and differentiated covalent inhibitor of KRAS G12C. - ASCO Publications, accessed June 16, 2025, https://ascopubs.org/doi/10.1200/JCO.2022.40.16_suppl.e15102
  19. Sotorasib A New Standard of Care for Second- or Third-Line Treatment of KRAS G12C–Mutated NSCLC - The ASCO Post, accessed June 16, 2025, https://ascopost.com/issues/november-10-2022/sotorasib-a-new-standard-of-care-for-second-or-third-line-treatment-of-kras-g12c-mutated-nsclc/
  20. KRAS and Lung Cancer, accessed June 16, 2025, https://www.lung.org/lung-health-diseases/lung-disease-lookup/lung-cancer/symptoms-diagnosis/biomarker-testing/kras
  21. Sotorasib Superiority Vs Standard of Care Docetaxel in Previously Treated KRAS G12C–Mutant NSCLC - CancerNetwork, accessed June 16, 2025, https://www.cancernetwork.com/view/sotorasib-superiority-vs-standard-of-care-docetaxel-in-previously-treated-kras-g12c-mutant-nsclc
  22. (PDF) D3S-001, a KRAS G12C Inhibitor with Rapid Target Engagement Kinetics, Overcomes Nucleotide Cycling, and Demonstrates Robust Preclinical and Clinical Activities - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/380430716_D3S-001_a_KRAS_G12C_inhibitor_with_rapid_target_engagement_kinetics_overcomes_nucleotide_cycling_and_demonstrates_robust_preclinical_and_clinical_activities
  23. Assessment of KRAS inhibitors for colorectal cancer - Semantic Scholar, accessed June 16, 2025, https://pdfs.semanticscholar.org/e7f4/630645764aea21af8acc39782a3540446b99.pdf
  24. Adagrasib Plus Cetuximab in KRAS G12C–Mutated Colorectal Cancer - The ASCO Post, accessed June 16, 2025, https://ascopost.com/news/april-2024/adagrasib-plus-cetuximab-in-kras-g12c-mutated-colorectal-cancer/
  25. Sotorasib Combination Approved for KRAS G12C-mutant Colorectal Cancer, accessed June 16, 2025, https://www.aacr.org/patients-caregivers/progress-against-cancer/sotorasib-combination-approved-for-kras-g12c-mutant-colorectal-cancer/
  26. FDA approves sotorasib with panitumumab for KRAS G12C-mutated colorectal cancer, accessed June 16, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-sotorasib-panitumumab-kras-g12c-mutated-colorectal-cancer
  27. Exploratory ctDNA analyses from first-in-human phase I trial of D3S-001 in patients with advanced solid tumor harboring a KRAS G12C mutation - ESMO Asia Congress 2024 | OncologyPRO, accessed June 16, 2025, https://oncologypro.esmo.org/meeting-resources/esmo-asia-congress-2024/exploratory-ctdna-analyses-from-first-in-human-phase-i-trial-of-d3s-001-in-patients-with-advanced-solid-tumor-harboring-a-kras-g12c-mutation
  28. D3S-001: A Promising New Treatment for Advanced Solid Tumors with KRAS G12C Mutation - Clinical trials, accessed June 16, 2025, https://clinicaltrials.eu/inn/d3s-001/
  29. A Study of D3S 001 Monotherapy or Combination Therapy in Subjects With Advanced Solid Tumors With a KRAS p.G12C Mutation - NCI, accessed June 16, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2023-06728
  30. D3S-001 | C32H35F6N7O3 | CID 167251905 - PubChem, accessed June 16, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/167251905
  31. D3S-001 | CAS#2706637-43-4 | KRAS G12C inhibitor | MedKoo, accessed June 16, 2025, https://www.medkoo.com/products/58551
  32. (Rac)-D3S-001 | Racemate of D3S-001 - MedchemExpress.com, accessed June 16, 2025, https://www.medchemexpress.com/rac-d3s-001.html
  33. D3 Bio Announces Oral Presentation of Clinical Data for D3S-001 at the 2025 AACR Annual Meeting, accessed June 16, 2025, https://www.d3bio.com/press-releases/d3-bio-announces-oral-presentation-of-clinical-data-for-d3s-001-at-the-2025-aacr-annual-meeting
  34. D3 Bio Announces Oral Presentation of the Clinical Data of Its Lead Program, D3S-001, at the ESMO Congress 2024, accessed June 16, 2025, https://www.d3bio.com/press-releases/d3-bio-announces-oral-presentation-of-the-clinical-data-of-its-lead-program-d3s-001-at-the-esmo-congress-2024
  35. Abstract 6503: D3S-001, a second-generation GDP-bound KRAS G12C inhibitor, overcomes nucleotide cycling and demonstrates robust preclinical and clinical activities | Request PDF - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/379215275_Abstract_6503_D3S-001_a_second-generation_GDP-bound_KRAS_G12C_inhibitor_overcomes_nucleotide_cycling_and_demonstrates_robust_preclinical_and_clinical_activities
  36. Abstract CT266: D3S-001, a next generation GDP-bound KRAS G12C inhibitor, as monotherapy in KRAS G12C inhibitor resistant non-small cell lung cancer | Request PDF - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/391166203_Abstract_CT266_D3S-001_a_next_generation_GDP-bound_KRAS_G12C_inhibitor_as_monotherapy_in_KRAS_G12C_inhibitor_resistant_non-small_cell_lung_cancer
  37. D3S-001 in advanced solid tumors with KRAS mutations: a phase 1 trial - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/391281387_D3S-001_in_advanced_solid_tumors_with_KRAS_mutations_a_phase_1_trial
  38. CUHK-led multinational study shows new China-developed cancer drug D3S-001 is effective in treating solid tumours, including lung, colorectal and pancreatic cancer, accessed June 16, 2025, https://www.med.cuhk.edu.hk/press-releases/cuhk-led-multinational-study-shows-new-china-developed-cancer-drug-d3s-001-is-effective-in-treating-solid-tumours-including-lung-colorectal-and-pancreatic-cancer
  39. 2023-508517-16-00 - Clinical Trials in the European Union - EMA, accessed June 16, 2025, https://euclinicaltrials.eu/ctis-public/view/2023-508517-16-00
  40. A Study of D3S 001 Monotherapy or Combination Therapy in Subjects With Advanced Solid Tumors With a KRAS p.G12C Mutation | Clinical Research Trial Listing - CenterWatch, accessed June 16, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT05410145/a-study-of-d3s-001-as-monotherapy-in-subjects-with-advanced-solid-tumors-with-a-kras-pg12c-mutation
  41. Next-generation KRAS-G12C inhibitor D3S-001 shows promise - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/391620511_Next-generation_KRAS-G12C_inhibitor_D3S-001_shows_promise
  42. KRAZATI (adagrasib) Demonstrated Statistically Significant Improvement in Progression-Free Survival in Patients with Pretreated Locally Advanced or Metastatic KRAS G12C -Mutated Non-Small Cell Lung Cancer - Business Wire, accessed June 16, 2025, https://www.businesswire.com/news/home/20240531926564/en/KRAZATI-adagrasib-Demonstrated-Statistically-Significant-Improvement-in-Progression-Free-Survival-in-Patients-with-Pretreated-Locally-Advanced-or-Metastatic-KRASG12C-Mutated-Non-Small-Cell-Lung-Cancer
  43. D3 Bio 2025 Company Profile: Valuation, Funding & Investors | PitchBook, accessed June 16, 2025, https://pitchbook.com/profiles/company/453472-39
  44. D3 Bio (Wuxi) Co. Ltd - Drug pipelines, Patents, Clinical trials - Patsnap Synapse, accessed June 16, 2025, https://synapse.patsnap.com/organization/fc8cfa14bb2e582a049ffa1e0ee1066c
  45. D3 Bio Launches with US$200 Million Investment to Develop Precision Medicines Addressing Unmet Patient Needs in Oncology & Immunology - Business Wire, accessed June 16, 2025, https://www.businesswire.com/news/home/20201117005519/en/D3-Bio-Launches-with-US%24200-Million-Investment-to-Develop-Precision-Medicines-Addressing-Unmet-Patient-Needs-in-Oncology-Immunology
  46. D3 Bio Launches with $200 Million to Advance Precision Medicine in Oncology and Immunology Space - GeneOnline, accessed June 16, 2025, https://www.geneonline.com/d3-bio-launches-with-200-million-to-advance-precision-medicine-in-oncology-and-immunology/
  47. D3 Bio - 2025 Company Profile, Funding & Competitors - Tracxn, accessed June 16, 2025, https://tracxn.com/d/companies/d3-bio/__xYbLrLG9qyGJiIEWcRnMz1NLIt8PeTQ5iKxI8gpN_W4
  48. D3 Bio Completes Series A+ Round to Advance Innovative Oncology Pipeline, accessed June 16, 2025, https://www.prnewswire.com/news-releases/d3-bio-completes-series-a-round-to-advance-innovative-oncology-pipeline-302110142.html
  49. accessed January 1, 1970, https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021180181
  50. patents.google.com, accessed June 16, 2025, https://patents.google.com/patent/WO2021180181A1/en
  51. KRAS G12C-mutated pancreatic cancer: Clinical outcomes based on chemotherapeutic regimen. - ASCO Publications, accessed June 16, 2025, https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.4150
  52. Evaluation of KRAS inhibitor-directed therapies for pancreatic cancer treatment - PMC, accessed June 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11116577/
  53. Treatment Innovations in Pancreatic Cancer: Putting Patient Priorities First, accessed June 16, 2025, https://ascopubs.org/doi/10.1200/EDBK-25-473204
  54. The Future of KRAS Targeting Cancer Therapies Beyond G12C - DelveInsight, accessed June 16, 2025, https://www.delveinsight.com/blog/kras-inhibitors-beyond-g12c
  55. Merck Announces MK-1084, an Investigational KRAS G12C Inhibitor, Shows Antitumor Activity in Phase 1 Trial of Patients With Advanced Colorectal Cancer and Non-Small Cell Lung Cancer Whose Tumors Harbor KRAS G12C Mutations, accessed June 16, 2025, https://www.merck.com/news/merck-announces-mk-1084-an-investigational-kras-g12c-inhibitor-shows-antitumor-activity-in-phase-1-trial-of-patients-with-advanced-colorectal-cancer-and-non-small-cell-lung-cancer-whose-tumors-harbo/
  56. ASCO 2025 preview – KRAS challengers assemble | ApexOnco - Oncology Pipeline, accessed June 16, 2025, https://www.oncologypipeline.com/apexonco/asco-2025-preview-kras-challengers-assemble
  57. KRAS Inhibitors Market Set to Exceed $2 Billion by 2030 with Next-Generation Therapies Advancing - MedPath, accessed June 16, 2025, https://trial.medpath.com/news/d1cac3e2c595f5a7/kras-inhibitors-market-set-to-exceed-2-billion-by-2030-with-next-generation-therapies-advancing
  58. KRAS Inhibitors - Redx Pharma, accessed June 16, 2025, https://www.redxpharma.com/our-pipeline/kras-inhibitors/
  59. Press Release Quanta Announces IND Clearance by US FDA for QTX3544, an Oral G12V-Preferring Multi-KRAS Inhibitor, and Other Pipeline Updates, accessed June 16, 2025, https://www.quantatx.com/news/010825/
  60. Targeting KRAS - PMC - PubMed Central, accessed June 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11845254/
  61. Current status of KRAS G12C inhibitors in NSCLC and the potential for combination with anti-PD-(L)1 therapy: a systematic review - Frontiers, accessed June 16, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2025.1509173/full
  62. Current status of KRAS G12C inhibitors in NSCLC and the potential for combination with anti-PD-(L)1 therapy: a systematic review - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/40303413/
  63. First-Line Therapy in Patients With KRAS-Mutated Non–Small Cell Lung Cancer, accessed June 16, 2025, https://dailynews.ascopubs.org/do/first-line-therapy-patients-kras--mutated-non-x2013-small-cell-lung-cancer

Published at: June 16, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.