ASP-8731, also identified by the synonyms ML-0207 and HRS-6719, was an orally administered small molecule inhibitor of BTB domain and CNC homolog 1 (BACH1) under development for the treatment of sickle cell disease (SCD). Originating from Mitobridge, Inc., later acquired by Astellas Pharma Global Development, Inc., ASP-8731 aimed to address SCD pathophysiology by derepressing NRF2-mediated pathways. This was hypothesized to increase the expression of antioxidant genes and, critically, fetal hemoglobin (HbF), thereby mitigating oxidative stress, inflammation, vaso-occlusion, and anemia associated with SCD.
Preclinical studies, encompassing in vitro experiments with human cell lines (including SCD patient-derived cells) and in vivo assessments in the Townes-SS humanized mouse model of SCD, demonstrated promising activity. ASP-8731 was shown to upregulate antioxidant markers, reduce inflammatory mediators, decrease vaso-occlusion, and induce HbF expression, both alone and in combination with hydroxyurea. These findings provided a strong rationale for clinical investigation.
Astellas initiated a Phase 1 clinical trial (NCT05167526) in March 2022 to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of ASP-8731 in healthy adult volunteers. Despite receiving Orphan Drug Designation from the U.S. FDA for sickle cell anemia in September 2022, the Phase 1 study was terminated prematurely in March 2023 because "protocol defined stopping criteria" were met. The specific adverse events or findings that triggered these criteria have not been publicly disclosed. Consequently, Astellas officially discontinued the development of ASP-8731 for SCD in April 2023.
The discontinuation of ASP-8731, particularly after promising preclinical results and early regulatory support, underscores the significant challenges in translating preclinical findings into safe and effective human therapies. While the specific reasons for its failure remain unelucidated from the available information, the outcome provides a cautionary data point for the development of BACH1 inhibitors. Nevertheless, the BACH1 pathway continues to be an area of active research for SCD and other diseases, with other companies advancing their own candidates, potentially with differentiated properties.
The compound at the center of this report is an investigational drug known by several identifiers, primarily ASP-8731, but also as ML-0207 and, as specified in the inquiry, HRS-6719.[1] The use of multiple synonyms is a common occurrence in the pharmaceutical development lifecycle, reflecting different stages of research, internal coding systems, and the eventual assignment of official non-proprietary names. For clarity and consistency with the bulk of the available literature, ASP-8731 will be the primary identifier used herein, with acknowledgments of its synonyms. ASP-8731 was developed as an orally administered small molecule drug.[1]
ASP-8731 was originally discovered and developed by Mitobridge, Inc..[1] Mitobridge was subsequently acquired by Astellas Pharma Global Development, Inc. (hereafter referred to as Astellas) in 2017.[1] The acquisition involved a significant upfront payment of $225 million, signaling a substantial investment by Astellas and a strong belief in the potential of Mitobridge's pipeline, which included ASP-8731 and focused on mitochondrial biology.[1] Following the acquisition, Astellas assumed responsibility for the continued development of ASP-8731.[1] The subsequent discontinuation of ASP-8731 after this considerable investment highlights the inherent financial risks and scientific uncertainties in pharmaceutical acquisitions, where not all acquired assets ultimately achieve their therapeutic or commercial potential.
ASP-8731 is identified by the Chemical Abstract Service (CAS) Number 2488255-42-9.[3] Its molecular formula is C20H21N5O4, corresponding to a molecular weight of approximately 395.41 g/mol.[3] Physically, ASP-8731 is described as a solid substance, off-white to light yellow in color.[3] The International Union of Pure and Applied Chemistry (IUPAC) name for the compound is 2-(benzo[d]oxazol-2-ylamino)-N-(2-(2-hydroxyethoxy)ethyl)-1-methyl-1H-benzo[d]imidazole-5-carboxamide.[4] Information regarding its solubility and various formulation protocols for research purposes, including solvents like DMSO, PEG300, Tween-80, and saline, further characterizes it as an experimental compound primarily intended for laboratory and clinical investigation.[3]
Table 1 provides a consolidated overview of the key identifiers and the development trajectory of HRS-6719 (ASP-8731/ML-0207).
Table 1: HRS-6719 (ASP-8731/ML-0207) - Key Identifiers and Development Snapshot
Characteristic | Detail | Reference(s) |
---|---|---|
Synonyms | HRS-6719, ASP-8731, ML-0207 | 1 |
CAS Number | 2488255-42-9 | 3 |
Molecular Formula | C20H21N5O4 | 3 |
Molecular Weight | ~395.41 g/mol | 3 |
Originator | Mitobridge, Inc. | 1 |
Developer | Astellas Pharma Global Development, Inc. | 1 |
Target | BACH1 (BTB domain and CNC homolog 1) | 1 |
Mechanism of Action | BACH1 inhibitor | 1 |
Highest Development Phase | Phase 1 | 1 |
Primary Therapeutic Area | Anemia, Sickle Cell | 1 |
Current Status | Discontinued | 1 |
The therapeutic rationale for ASP-8731 in sickle cell disease (SCD) centered on its ability to inhibit BTB domain and CNC homolog 1 (BACH1), a key transcriptional repressor involved in cellular responses to oxidative stress and globin gene regulation.[1]
SCD is a genetic disorder characterized by the production of abnormal hemoglobin S (HbS), which polymerizes upon deoxygenation. This polymerization leads to red blood cell sickling, chronic hemolysis, and the release of substantial amounts of free heme into circulation.[7] Free heme is a potent pro-oxidant and pro-inflammatory molecule, contributing significantly to the hallmark features of SCD, including oxidative stress, chronic inflammation, and vaso-occlusion, which in turn cause painful crises and progressive organ damage.[1]
BACH1 functions as a transcriptional repressor. Interestingly, heme itself can bind to BACH1, leading to the derepression of certain genes.[6] A critical target of BACH1 repression is the nuclear factor erythroid 2-related factor 2 (NRF2).[6] NRF2 is a master regulator of antioxidant responses; its activation leads to the transcription of a battery of cytoprotective genes, including heme oxygenase-1 (HMOX1), glutathione reductase (GR), solute carrier family 7 member 11 (SLC7A11), and NAD(P)H quinone dehydrogenase 1 (NQO1).[6] Furthermore, NRF2 activation in erythroid cells promotes the expression of gamma-globin genes (HBG1 and HBG2), which are components of fetal hemoglobin (HbF).[6] An increase in HbF levels is known to ameliorate the severity of SCD by interfering with HbS polymerization and reducing the proportion of HbS within red blood cells.[6]
ASP-8731 was developed as a selective, small molecule inhibitor of BACH1.[1] The intended mechanism of action was to directly inhibit BACH1, thereby relieving its repressive effects on NRF2-mediated gene transcription.[6] This derepression was expected to achieve two primary therapeutic benefits in SCD:
The therapeutic hypothesis was that by simultaneously addressing oxidative stress/inflammation and inducing HbF, ASP-8731 could lead to a reduction in hemolysis, inflammation, the frequency and severity of vaso-occlusive crises, and an overall improvement in the clinical status of patients with SCD.[1] This dual mechanism, targeting multiple facets of SCD pathology, presented an attractive strategy compared to therapies focusing on a single pathogenic pathway.
However, BACH1 is widely expressed across various mammalian tissues and plays roles in diverse biological processes beyond erythropoiesis and oxidative stress response, including heme homeostasis, immune system development, and cell cycle regulation.[9] Systemic inhibition of such a broadly functional protein by an oral drug like ASP-8731, while potentially beneficial in the context of SCD, carries the risk of unintended physiological consequences in other tissues. The long-term impact of altering this intricate balance involving heme, BACH1, and NRF2 systemically would necessitate careful evaluation, and potential off-target or on-target but undesirable systemic effects could have contributed to the "protocol defined stopping criteria" encountered in the clinical trial.
A substantial body of preclinical work was conducted to evaluate the potential of ASP-8731 in models of SCD, providing the foundational evidence for its progression into clinical trials. These studies spanned in vitro cellular assays and in vivo experiments using a humanized mouse model of SCD.
In vitro experiments using various cell types demonstrated ASP-8731's ability to modulate pathways relevant to SCD:
The Townes-SS mouse model, which expresses human α-globin and human βS-globin and thus recapitulates many features of human SCD, was used for in vivo validation:
Collectively, the preclinical data painted a compelling picture for ASP-8731. It consistently demonstrated the ability to modulate multiple pathways implicated in SCD pathophysiology, including the upregulation of antioxidant defenses, reduction of inflammatory markers, amelioration of vaso-occlusion, and, importantly, the induction of HbF.[1] The positive results across in vitro human cell systems (including SCD patient-derived cells and HU-non-responder cells) and the in vivo humanized SCD mouse model built a strong scientific rationale for advancing ASP-8731 into clinical development.[6] The potential for combination therapy with HU also suggested a flexible clinical development path. While these studies heavily emphasized efficacy markers, comprehensive details regarding preclinical Good Laboratory Practice (GLP) toxicology or extensive safety pharmacology studies were not extensively detailed in the available information. This focus on efficacy, common in early research publications, means that any potential safety liabilities identified in subsequent dedicated toxicology studies, which are crucial before human testing, are not apparent from this dataset.
Table 2 summarizes the key preclinical findings for ASP-8731 in SCD models.
Table 2: Summary of Key Preclinical Findings for ASP-8731 in Sickle Cell Disease Models
Model System | Key Parameters Assessed | Key Outcomes/Effects of ASP-8731 | Comparison/Combination with Hydroxyurea (HU) | Reference(s) |
---|---|---|---|---|
HepG2 liver cells (human) | HMOX1, FTH1 mRNA | Increased | N/A | 1 |
Pulmonary Artery Endothelial Cells (human) | VCAM1 mRNA (TNF-α induced), Glutathione (hemin induced) | Decreased VCAM1, Blocked glutathione decrease | N/A | 1 |
CD34+ erythroid cells (human, SCD) | % F-cells, HBG mRNA, HBA mRNA, NRF2 target genes | Increased (~2-fold for F-cells), Increased HBG/HBA, Increased NRF2 targets (e.g., HMOX1) | Similar F-cell induction to HU; Additive/synergistic effect with HU on F-cells; Effective in HU non-responder cells | 6 |
Townes-SS mice (SCD model) | Hepatic HO-1, ICAM-1, NF-$\kappa$B p-p65; WBC counts | Increased HO-1; Decreased ICAM-1, NF-$\kappa$B p-p65, WBC counts | Effects observed similar to or in conjunction with HU | 6 |
Townes-SS mice (SCD model) | Heme-induced microvascular stasis | Decreased (dose-responsive) | Combination with HU more effective than HU alone | 1 |
Townes-SS mice (SCD model) | γ-globin expression, % F-cells, Blood A-γ globin | Increased | N/A | 6 |
Following the encouraging preclinical results, Astellas advanced ASP-8731 into clinical development with a Phase 1 study designed to assess its safety and characteristics in humans.
The sole clinical trial identified for ASP-8731 is registered on ClinicalTrials.gov with the identifier NCT05167526.[1] The Astellas internal study ID for this trial was 8731-CL-0101.[2] The full title of the study was "A Phase 1 Combined Single and Multiple Ascending Oral Dose Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of ASP8731 in Healthy Adult Participants, Including an Assessment of a Food Effect".[1] The trial was conducted primarily in the USA, and no EudraCT (European Union Drug Regulating Authorities Clinical Trials Database) ID was listed, indicating it was likely not conducted within the European Economic Area.[10]
The NCT05167526 trial was a Phase 1 study.[1] Its primary purpose, often categorized as "Basic Science" in some databases for early-phase trials, was to determine a suitable dose range for ASP-8731 and to meticulously evaluate its safety profile and any medical problems arising at each dose level in healthy individuals.[10]
The study employed a combined single ascending dose (SAD) and multiple ascending dose (MAD) design, a standard approach for first-in-human trials to gradually increase exposure and monitor for dose-limiting toxicities.[1] The trial was double-blinded, meaning neither the participants nor the investigators knew who was receiving ASP-8731 versus a placebo or control.[10] Key objectives included the evaluation of safety, tolerability, pharmacokinetics (PK; how the body absorbs, distributes, metabolizes, and excretes the drug), and pharmacodynamics (PD; the drug's effect on the body) of orally administered ASP-8731.[1] An assessment of the effect of food on the drug's absorption was also part of the study design.[1] Primary endpoints focused on safety and tolerability, encompassing the incidence and nature of adverse events (AEs) and various laboratory abnormalities.[12] One source mentioned eight primary endpoints related to these safety assessments, though specific details of these measures are not provided in the available documents.[12]
The trial was conducted in healthy adult volunteers.[1] The planned enrollment for the study was 36 participants.[10] However, the study was terminated "well short of its enrollment target".[1] While one source states "Participants were not enrolled (for withdrawn studies), however if participants were enrolled, they are no longer being examined or treated" [10], the context from other documents strongly suggests that at least some participants were enrolled and dosed before the trial was halted.[1]
The NCT05167526 trial is officially listed as terminated.[1] The study was active between March 2022 and November 2022.[10] Astellas Pharma formally terminated the Phase 1 trial on March 1, 2023, and it was stated that the study "stopped early and will not start again".[10]
The explicitly stated reason for this premature termination was that "protocol defined stopping criteria" were met.[1] Such criteria in a Phase 1 healthy volunteer study are typically pre-specified safety or tolerability thresholds. If these thresholds are breached (e.g., due to the occurrence of serious adverse events, specific patterns of AEs, or unacceptable laboratory changes), the protocol mandates halting the study. The termination of a Phase 1 study in healthy volunteers due to such criteria strongly points towards an unacceptable safety or tolerability profile of ASP-8731 itself at the doses tested, rather than a lack of efficacy, which is generally not a primary assessment in this type of early-phase study for a drug intended for a chronic disease. The fact that the study was a SAD/MAD design and was terminated well short of its enrollment target may imply that these stopping criteria were encountered at an early stage of dose escalation, possibly during the SAD cohorts or early into the MAD cohorts. This would have prevented the full exploration of the planned dose range and characterization of the drug's profile.
Crucially, the specific nature of the "protocol defined stopping criteria" – i.e., the precise adverse events, pharmacokinetic issues, or other safety signals that led to the trial's cessation – is not detailed in the provided research materials. This absence of specific information represents a significant gap in understanding the exact reasons for ASP-8731's clinical failure and limits the ability to draw detailed conclusions about its human toxicity profile from the available data.
Table 3 provides an overview of the key details of the NCT05167526 clinical trial.
Table 3: Overview of Clinical Trial NCT05167526
Parameter | Detail | Reference(s) |
---|---|---|
ClinicalTrials.gov ID | NCT05167526 | 1 |
Astellas Study ID | 8731-CL-0101 | 2 |
Full Title | A Phase 1 Combined Single and Multiple Ascending Oral Dose Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of ASP8731 in Healthy Adult Participants, Including an Assessment of a Food Effect | 1 |
Phase | Phase 1 | 1 |
Study Design | Combined SAD/MAD, Double-blind, Oral | 1 |
Participant Type | Healthy Adult Volunteers | 1 |
Planned Enrollment | 36 | 10 |
Key Objectives | Evaluate Safety, Tolerability, Pharmacokinetics (PK), Pharmacodynamics (PD), Food Effect | 1 |
Primary Outcome Measures | Incidence of Adverse Events, Laboratory abnormalities (specific measures not detailed) | 12 |
Start Date | March 2022 | 10 |
End Date (Actual Period) | November 2022 | 10 |
Sponsor | Astellas Pharma Global Development, Inc. | 1 |
Status | Terminated | 1 |
Stated Reason for Termination | "Protocol defined stopping criteria were met" (specifics not provided) | 1 |
Despite its early-stage development, ASP-8731 did receive a notable regulatory designation in the United States.
ASP-8731 was granted Orphan Drug Designation by the U.S. Food and Drug Administration (FDA) for the indication of Sickle Cell Anemia.[13] This designation was officially conferred on September 1, 2022.[13] Orphan Drug Designation is a status assigned by regulatory authorities to drugs intended for the treatment, diagnosis, or prevention of rare diseases or conditions affecting a small patient population. The purpose of this designation is to incentivize pharmaceutical companies to develop products for these underserved markets by offering benefits such as extended market exclusivity post-approval, tax credits for clinical research, waiver of certain regulatory fees, and dedicated regulatory assistance and guidance (e.g., protocol assistance).[14]
The timing of this designation is noteworthy: it was granted in September 2022, while the Phase 1 trial NCT05167526 was ongoing (March 2022 - November 2022) and just a few months before the trial's early cessation and subsequent formal termination announcement in March 2023.[10] This juxtaposition suggests that, up until late 2022, there was continued regulatory support and an ongoing commitment from Astellas towards the development of ASP-8731 for SCD. The fact that development was halted despite this favorable regulatory status implies that the issues encountered in the Phase 1 trial were sufficiently acute and serious to outweigh the potential benefits and incentives associated with the orphan drug pathway.
The granting of Orphan Drug Designation typically relies on a strong preclinical rationale demonstrating the drug's potential to address a significant unmet medical need in a rare disease. The subsequent clinical failure of ASP-8731, even with this designation, serves as a stark reminder of the high attrition rates inherent in drug development. Regulatory incentives can facilitate and support the development process but do not guarantee success; the ultimate determinants remain the demonstration of an acceptable safety profile and clinical efficacy in human subjects. No information was found in the provided materials regarding an application for or granting of orphan drug status by the European Medicines Agency (EMA) or other regulatory bodies outside the USA.
The development of ASP-8731 for sickle cell disease followed a path from promising discovery to an early clinical-stage halt.
The trajectory of ASP-8731 can be summarized by the following key milestones:
The relatively short timeframe from the optimistic presentation of preclinical data and initiation of the Phase 1 trial to its early termination suggests that the clinical issues encountered were significant and decisive, leading to a rapid halt in development.
The highest phase of development reached by ASP-8731 was Phase 1.[1] The primary and explicitly stated reason for the discontinuation of ASP-8731's development was the premature termination of the Phase 1 trial NCT05167526. This termination occurred because "protocol defined stopping criteria" were met during the study in healthy volunteers.[1] As discussed previously, this strongly indicates that unacceptable safety or tolerability issues emerged when ASP-8731 was administered to humans, preventing further investigation and development for SCD.
The development and subsequent discontinuation of ASP-8731 offer valuable learning points regarding the translation of preclinical science to clinical reality, particularly for novel targets like BACH1 in complex diseases such as SCD.
ASP-8731 was supported by a robust preclinical data package that demonstrated its intended mechanism of action – BACH1 inhibition leading to NRF2 activation – and showed promising efficacy across multiple relevant endpoints in SCD models. The dual benefit of enhancing antioxidant defenses and inducing fetal hemoglobin was a particularly compelling aspect of its therapeutic rationale.[1]
However, the failure in Phase 1, attributed to meeting protocol-defined stopping criteria in healthy volunteers, underscores the critical translational gap that often exists between preclinical animal models and human studies.[1] Animal models, even humanized ones, may not fully replicate human physiology or predict all potential toxicities. The specific safety or tolerability issues that led to the termination of NCT05167526 remain undisclosed in the available information. This lack of transparency limits a full understanding of ASP-8731's shortcomings and the precise nature of the adverse human response. Without these details, it is challenging to ascertain whether the issues were related to the drug's specific chemistry (e.g., off-target effects, metabolic profile) or potentially a class effect related to systemic BACH1 inhibition.
Despite the discontinuation of ASP-8731, the BACH1 pathway remains a target of considerable interest for SCD and potentially other diseases characterized by oxidative stress, inflammation, or conditions where HbF induction would be beneficial.[1] The underlying scientific rationale for targeting BACH1 in SCD is still considered valid by some in the field.
Evidence for this continued interest includes the activities of other companies. For instance, IMMvention Therapeutix is developing oral BACH1 inhibitors for SCD and has entered into a co-development deal with Novo Nordisk.[1] IMMvention has claimed that the "druglike properties" of its candidate differentiate it from other BACH1 inhibitors.[1] This statement might subtly allude to potential challenges related to pharmacokinetics, toxicity, or specificity that may have affected earlier BACH1 inhibitors like ASP-8731. If ASP-8731 suffered from suboptimal pharmaceutical properties (e.g., poor solubility, metabolic instability, off-target activity), a new chemical entity with improved characteristics might indeed fare better.
Additionally, vTv Therapeutics has also been involved in the development of BACH1 inhibitors. One of their candidates was licensed to Anteris Bio for renal diseases, while vTv retained rights in other indications, including brain-penetrant BACH1 inhibitors for potential applications in neurodegenerative conditions like Parkinson's and Alzheimer's diseases.[1] This diverse approach suggests that the therapeutic utility and safety profile of BACH1 inhibition might be highly dependent on the specific chemical entity, its tissue distribution, and the particular disease context.
The broad physiological roles of BACH1, which extend to the regulation of heme homeostasis, cell cycle, immune responses, and its implication in conditions such as cancer, cardiovascular diseases, and neurodegenerative disorders, highlight both the therapeutic potential and the challenges of targeting this protein.[9] Achieving a desirable therapeutic window with high selectivity and an acceptable safety profile will be paramount for any BACH1 inhibitor to succeed. The critical question arising from ASP-8731's failure is whether the observed issues were compound-specific (attributable to the unique properties of ASP-8731) or target-related (suggesting that systemic BACH1 inhibition via an oral drug inherently leads to unacceptable toxicity in humans). The progression and clinical data from other BACH1 inhibitors currently in development will be crucial in clarifying this distinction.
The therapeutic landscape for SCD is dynamic, with numerous investigational agents and diverse mechanisms of action being explored. The discontinuation of ASP-8731 provides a data point for competitors in this space. However, the absence of specific details regarding its clinical trial termination limits the direct lessons that can be drawn by other developers concerning specific toxicities to avoid or biomarkers to monitor.
Future BACH1 inhibitors aiming for success in SCD or other indications will need to unequivocally demonstrate a favorable safety profile in human clinical trials, particularly addressing any liabilities that may have led to the cessation of ASP-8731's development. Their success will depend on careful molecular design, thorough preclinical safety assessment, and meticulous clinical trial execution.
ASP-8731 (HRS-6719/ML-0207) embarked on its development journey as a promising oral BACH1 inhibitor with a strong preclinical rationale for the treatment of sickle cell disease. Its proposed dual mechanism of enhancing antioxidant defenses and inducing fetal hemoglobin offered a comprehensive approach to addressing the multifaceted pathophysiology of SCD. Preclinical studies in relevant cellular and animal models consistently supported its potential efficacy.
However, the trajectory of ASP-8731 was decisively altered during its first-in-human Phase 1 clinical trial (NCT05167526). The trial's premature termination in healthy adult volunteers due to the meeting of "protocol defined stopping criteria" was the pivotal event that led to Astellas's decision to discontinue the drug's development for SCD. This outcome, occurring despite prior U.S. FDA Orphan Drug Designation, highlights the significant hurdles in translating promising preclinical science into viable human therapeutics.
A critical limitation in analyzing the failure of ASP-8731 is the lack of publicly available information regarding the specific safety or tolerability issues that triggered the trial's termination. Without these details, it is difficult to definitively conclude whether the problems were unique to the ASP-8731 molecule or indicative of broader challenges associated with systemic BACH1 inhibition.
The experience with ASP-8731 serves as a reminder of the complexities and high attrition rates in drug development. While this particular candidate did not succeed, the BACH1 pathway continues to be explored as a therapeutic target. The fate of other BACH1 inhibitors currently in development will be instrumental in determining the ultimate viability of this approach for sickle cell disease and potentially other conditions. Future research in this area will need to focus intently on optimizing drug properties to ensure a favorable safety and tolerability profile in humans.
Published at: May 16, 2025
This report is continuously updated as new research emerges.