Bitopertin (DB12426): A Comprehensive Analysis of a GlyT1 Inhibitor's Journey from Neuropsychiatry to Rare Hematologic Disease
Executive Summary
Bitopertin (DB12426) is an investigational, orally administered small molecule that functions as a potent and selective inhibitor of the glycine transporter 1 (GlyT1). Its development history represents a compelling and instructive case study in translational science, characterized by a significant strategic pivot from a large-scale neuropsychiatry program to a focused rare disease application. Initially developed by Roche, Bitopertin was designed to treat the negative and cognitive symptoms of schizophrenia. The therapeutic rationale was based on the well-supported N-methyl-D-aspartate (NMDA) receptor hypofunction hypothesis of the disease. By inhibiting GlyT1 in the central nervous system, Bitopertin was intended to increase synaptic glycine concentrations, thereby enhancing NMDA receptor function.
The early clinical data were promising, with a Phase II proof-of-concept study demonstrating a statistically significant improvement in negative symptoms. This success prompted the initiation of an extensive six-study Phase III program, "SearchLyte," which ultimately failed to confirm the efficacy of Bitopertin in either negative or sub-optimally controlled positive symptoms of schizophrenia, leading to the program's discontinuation in 2014. However, a consistent pharmacological effect observed during these trials—a dose-dependent, reversible reduction in hemoglobin—provided the scientific basis for a remarkable second act. This hematological signal was correctly identified as a result of GlyT1 inhibition on erythroid precursors, thereby limiting the glycine supply necessary for heme biosynthesis.
In 2021, Disc Medicine licensed Bitopertin to repurpose it as a first-in-class, disease-modifying therapy for erythropoietic porphyrias (EPP), a group of rare genetic disorders caused by the toxic accumulation of the heme precursor protoporphyrin IX (PPIX). In this new context, the drug's mechanism is leveraged as a substrate reduction therapy to decrease PPIX production. Subsequent Phase II clinical trials in EPP have been highly successful, demonstrating robust, dose-dependent reductions in PPIX that correlate with significant clinical improvements in photosensitivity and quality of life. With a well-characterized safety profile from over 4,000 trial participants and a clear regulatory path forward, including an agreement with the U.S. FDA for a potential accelerated approval pathway, Bitopertin is now a promising late-stage asset for a significant unmet need in a rare disease population.
Section 1: Molecular Profile and Physicochemical Characteristics
The foundation of any pharmaceutical agent's biological activity and developmental trajectory lies in its molecular structure and physicochemical properties. This section details the fundamental identity of Bitopertin, outlining its chemical structure, nomenclature, and the key physical characteristics that govern its behavior as a drug.
1.1 Chemical Identity and Structure
Bitopertin is a synthetic organic small molecule belonging to the pyridinylpiperazine class of compounds.[1] Its structure is defined by a central piperazine ring linking a substituted pyridine moiety to a substituted benzoyl moiety.
- Generic Name: Bitopertin [1]
- Synonyms and Developmental Codes: The compound has been known by several identifiers throughout its development, including RG1678, RO-4917838, RO-4917939, R-1678, and Paliflutine.[1]
- Systematic (IUPAC) Name: The formal chemical name is [3-fluoro-5-(trifluoromethyl)pyridin-2-yl]piperazin-1-yl]-oxyphenyl]methanone.[4] An alternative name is 1-[3-fluoro-5-(trifluoromethyl)pyridin-2-yl]-4-(5-methanesulfonyl-2-{oxy}benzoyl)piperazine.[1]
- Chemical Formula: The molecular formula for Bitopertin is C21H20F7N3O4S.[2]
- Molecular Weight: The average molecular weight is 543.46 g/mol, with a monoisotopic mass of 543.1063 g/mol.[1]
The molecule's identity is further specified by a series of unique identifiers used across various chemical and drug databases, which are consolidated in Table 1 below.
1.2 Physicochemical Properties and Formulation Considerations
The physicochemical properties of Bitopertin are critical for understanding its absorption, distribution, and formulation requirements.
- Solubility: Bitopertin is characterized by low aqueous solubility, with a reported value of 0.0508 mg/mL.[1] This property is a key consideration for oral drug delivery. Its solubility is significantly higher in certain organic solvents, such as dimethylformamide (DMF) at 30 mg/mL and dimethyl sulfoxide (DMSO) at 25 mg/mL.[5] Importantly for its oral formulation, it exhibits satisfactory solubility in simulated intestinal fluids, measured at 20 μg/mL in the fasted state and 60 μg/mL in the fed state, suggesting that absorption can be facilitated in the gastrointestinal environment.[11]
- Lipophilicity: The molecule is lipophilic, a characteristic necessary for crossing biological membranes, including the blood-brain barrier. This is quantified by its partition coefficient (logP), with values reported as 4.09 (ALOGPS) and 3.46 (Chemaxon).[1]
- Other Properties: Bitopertin has a polar surface area (PSA) of 79.81 Ų, 6 hydrogen bond acceptors, and 0 hydrogen bond donors.[1] According to Lipinski's Rule of Five, a set of guidelines for predicting drug-likeness, Bitopertin has one violation, which is not uncommon for centrally acting therapeutic agents.[1]
Table 1: Key Chemical and Physicochemical Properties of Bitopertin
Property | Value | Source(s) |
---|
Identifiers | | |
CAS Number | 845614-11-1 | 1 |
DrugBank ID | DB12426 | 1 |
ChEMBL ID | CHEMBL1171829 | 4 |
PubChem CID | 24946690 | 1 |
UNII | Q8L6AN59YY | 1 |
InChIKey | YUUGYIUSCYNSQR-LBPRGKRZSA-N | 1 |
Structural & Chemical | | |
Chemical Formula | C21H20F7N3O4S | 2 |
Average Molecular Weight | 543.46 g/mol | 2 |
IUPAC Name | [3-fluoro-5-(trifluoromethyl)pyridin-2-yl]piperazin-1-yl]-oxyphenyl]methanone | 4 |
Physicochemical | | |
Water Solubility | 0.0508 mg/mL | 1 |
logP | 3.46 - 4.09 | 1 |
Polar Surface Area | 79.81 Ų | 1 |
Hydrogen Bond Acceptors | 6 | 1 |
Hydrogen Bond Donors | 0 | 1 |
Rotatable Bonds | 5 - 7 | 1 |
1.3 Implications of Physicochemical Profile on Development
The physicochemical profile of Bitopertin, particularly its low aqueous solubility, was not merely a data point but a central challenge that profoundly shaped its clinical development. This property created a direct causal link to a significant hurdle: formulation-dependent bioavailability. Clinical pharmacokinetic studies revealed that the drug's absorption and resulting plasma concentrations were highly sensitive to changes in the particle size of the active pharmaceutical ingredient and the specific dosage form used (e.g., capsules vs. tablets).[13]
This challenge prompted the development team at Roche to employ a modern, model-informed drug development strategy. Rather than relying solely on empirical clinical studies, they constructed a physiologically based pharmacokinetic (PBPK) absorption model.[13] This computational model allowed them to simulate the impact of different particle sizes on key pharmacokinetic parameters. The simulations predicted that the maximum plasma concentration (
Cmax) would be reduced by over 20% if the median particle diameter exceeded 15 μm, while the total drug exposure (AUCinf) would remain relatively stable until the diameter surpassed 30 μm.[13] These simulations, verified against measured plasma concentrations in both preclinical and clinical studies, provided a quantitative understanding of the structure-property-absorption relationship. Consequently, this modeling effort was instrumental in de-risking the program by allowing the team to establish strict manufacturing specifications for particle size in the final tablet formulation, thereby ensuring consistent and predictable drug exposure in patients across large-scale clinical trials. This proactive management of a key biopharmaceutical risk highlights a sophisticated approach to overcoming the inherent challenges posed by the molecule's physical properties.
Section 2: Dual Pharmacological Mechanisms of a Singular Target
The therapeutic story of Bitopertin is unique in that its entire development, including its dramatic pivot from one disease area to another, is rooted in the inhibition of a single molecular target: the glycine transporter 1 (GlyT1). The drug's two distinct therapeutic rationales emerge from the different physiological roles GlyT1 plays in two separate biological systems: the central nervous system and the hematopoietic system.
2.1 GlyT1 Inhibition and NMDA Receptor Modulation in the Central Nervous System (CNS)
The initial development of Bitopertin was centered on its activity within the brain.
- Primary Target and Potency: Bitopertin is a potent, selective, and non-competitive inhibitor of GlyT1, a protein encoded by the SLC6A9 gene that is responsible for regulating glycine concentrations at synapses.[1] In vitro assays have demonstrated its high affinity and inhibitory activity, with a binding affinity (
Ki) of 8.1 nM for the human GlyT1b isoform and a half-maximal inhibitory concentration (IC50) of 22-25 nM in cell-based glycine uptake assays.[3] This potency is coupled with excellent selectivity; Bitopertin shows minimal activity against the related glycine transporter 2 (GlyT2), with an
IC50 greater than 30 µM, and was found to be inactive against a broad panel of 86 to 107 other receptors, transporters, and enzymes at concentrations up to 10 µM.[5]
- CNS Mechanism of Action: In the CNS, particularly in forebrain regions such as the neocortex and hippocampus, GlyT1 is predominantly expressed on astrocytes that ensheath glutamatergic synapses.[14] Its primary function here is to clear glycine from the synaptic cleft via reuptake.[2] By blocking this transporter, Bitopertin effectively increases the ambient extracellular concentration of glycine at these synapses.[2]
- Link to NMDA Receptors: This increase in synaptic glycine is therapeutically relevant because glycine serves as an obligatory co-agonist for the NMDA receptor, a key ion channel involved in synaptic plasticity, learning, and memory.[2] For the NMDA receptor channel to open, it must bind not only the primary neurotransmitter, glutamate, but also a co-agonist—either glycine or D-serine—at a distinct site on its NR1 subunit.[16] Under normal physiological conditions, this co-agonist site is thought to be sub-saturated.[17] Therefore, by elevating local glycine levels, Bitopertin enhances the probability of NMDA receptor activation in the presence of glutamate, effectively potentiating glutamatergic neurotransmission.[6]
- Therapeutic Hypothesis for Schizophrenia: This mechanism was directly aimed at addressing the "glutamate hypofunction hypothesis" of schizophrenia. This hypothesis posits that a core deficit in the disease, particularly underlying the persistent negative symptoms (e.g., social withdrawal, apathy) and cognitive impairments, is reduced signaling through NMDA receptors.[11] The strategy behind Bitopertin was to counteract this hypofunction by boosting NMDA receptor activity, thereby offering a novel treatment for symptoms that are poorly addressed by conventional antipsychotic medications.[2]
2.2 GlyT1 Inhibition and Heme Biosynthesis Regulation in Erythropoiesis
The second, later-discovered mechanism of Bitopertin operates in a completely different physiological context: the development of red blood cells.
- Hematological Mechanism of Action: GlyT1 is also expressed as a key membrane transporter on the surface of developing red blood cells, known as erythroid precursors.[20] In this setting, its function is not to modulate neurotransmission but to supply the cell with sufficient glycine to support erythropoiesis—the process of red blood cell production.[20]
- Role of Glycine in Heme Synthesis: The synthesis of heme, the iron-containing compound that enables hemoglobin to carry oxygen, is a fundamental multi-step enzymatic process. The very first and rate-limiting step of this pathway is the condensation of glycine with succinyl-CoA, a reaction catalyzed by the enzyme 5'-aminolevulinate synthase 2 (ALAS2).[20] Glycine is therefore an essential, foundational substrate for all downstream heme production.[2]
- Therapeutic Hypothesis for Porphyrias: This hematological role of GlyT1 forms the basis for Bitopertin's repurposing. In erythropoietic porphyrias, specifically Erythropoietic Protoporphyria (EPP) and X-linked Protoporphyria (XLPP), genetic mutations in the heme synthesis pathway (affecting the enzymes FECH or ALAS2, respectively) cause a metabolic bottleneck. This leads to the massive and toxic accumulation of an upstream intermediate, protoporphyrin IX (PPIX).[20] The therapeutic strategy with Bitopertin is one of "substrate reduction." By inhibiting GlyT1 on erythroid precursors, the drug limits the cellular uptake of glycine. This reduction in the initial substrate effectively throttles the entire downstream heme synthesis pathway, thereby decreasing the pathological production and accumulation of the toxic PPIX metabolite.[20]
- Therapeutic Hypothesis for β-Thalassemia: A similar rationale was explored for β-thalassemia. In this disease, the pathophysiology is driven by an imbalance between α-globin and β-globin chains, which leads to an excess of free heme and severe oxidative stress, culminating in ineffective erythropoiesis and anemia.[21] The hypothesis was that by using Bitopertin to restrict glycine supply, a "heme-restricted" environment could be created. This was thought to potentially rebalance heme and globin synthesis, reduce the oxidative damage caused by excess heme, and consequently improve red cell survival and anemia.[21]
2.3 Repurposing Driven by Context-Dependent Target Biology
The developmental arc of Bitopertin is a powerful demonstration of how a profound understanding of a target's tissue-specific biology can unlock entirely new therapeutic paradigms. The drug's pivot from neuropsychiatry to rare hematologic disease hinges on the dual, context-dependent roles of its single molecular target, GlyT1. The initial development program was predicated exclusively on the role of GlyT1 in the CNS, where the therapeutic goal was to enhance NMDA receptor signaling by increasing the availability of its co-agonist, glycine.[6] During the extensive clinical trials for this indication, a systemic pharmacological effect was consistently observed: a dose-dependent reduction in hemoglobin.[24] While initially categorized as an undesirable side effect, a crucial intellectual connection was made between this observation and the fundamental role of glycine as the primary substrate for heme synthesis in red blood cells.[21]
This led to the realization that the observed hemoglobin reduction was, in fact, evidence of on-target activity in the hematopoietic system. A new hypothesis was formulated: if systemic GlyT1 inhibition could modulate normal heme synthesis, it could be therapeutically applied to reduce the pathological overproduction of heme precursors in diseases like EPP.[20] In this new context, the therapeutic goal became the
restriction of a metabolic pathway's output. Thus, the exact same molecular action—the blockade of glycine transport by GlyT1—is leveraged for two diametrically opposed therapeutic aims simply by virtue of the different physiological functions of the target protein in two distinct tissues: brain astrocytes versus erythroid precursors. This represents a sophisticated application of pharmacological principles and a strategically astute pivot from a failed high-risk, high-reward program to a targeted, de-risked rare disease application.
Section 3: Pharmacokinetic and Pharmacodynamic Profile
A thorough understanding of a drug's absorption, distribution, metabolism, and excretion (ADME) is fundamental to its clinical development. This section details the pharmacokinetic profile of Bitopertin, highlighting how its properties were characterized and how advanced computational modeling was employed to predict its behavior, manage formulation challenges, and inform clinical trial design.
3.1 Absorption, Distribution, Metabolism, and Excretion (ADME)
Bitopertin's journey through the body has been well-characterized through numerous preclinical and clinical studies.
- Absorption: Bitopertin is designed for oral administration.[20] Its absorption is generally good, with validated PBPK models simulating a bioavailability of over 90% at doses below 80 mg.[26] However, its low aqueous solubility becomes a limiting factor at higher doses; above 50 mg, absorption becomes slightly less than dose-proportional as the system's ability to dissolve and absorb the drug is saturated.[26] As previously discussed, the rate and extent of absorption are highly dependent on the particle size of the drug substance.[13]
- Distribution: As a lipophilic molecule, Bitopertin undergoes rapid and extensive distribution into body tissues following absorption.[26] The steady-state volume of distribution (
Vss) has been estimated to be approximately 2.85 L/kg, indicating that the drug does not remain confined to the bloodstream but partitions widely into peripheral tissues.[26]
- Metabolism: The primary mechanism of Bitopertin clearance from the body is hepatic metabolism. In vitro and in vivo studies have established that approximately 90% of the drug is eliminated via oxidative metabolism, with the cytochrome P450 enzyme CYP3A4 identified as the major contributing enzyme.[27] This reliance on a single major metabolic pathway makes Bitopertin susceptible to clinically significant drug-drug interactions. Consequently, the concurrent use of strong inhibitors or inducers of CYP3A4 is a key exclusion criterion in its recent clinical trials to avoid unpredictable alterations in drug exposure.[29]
- Excretion: Direct renal excretion of the parent drug is a negligible pathway for elimination, accounting for less than 0.1% of total clearance.[27] Preclinical studies in rats indicated a very long terminal half-life (
T1/2) of 35 to 110 hours and low systemic clearance, suggesting that the drug is eliminated slowly from the body following its extensive distribution and metabolism.[27]
3.2 Insights from Physiologically Based Pharmacokinetic (PBPK) Modeling
The development of Bitopertin serves as an exemplar of a modern, model-informed approach, where computational simulations were used to anticipate and solve clinical challenges.
- Predictive Power and Model Refinement: PBPK models were constructed early in the drug discovery process, integrating preclinical data on physicochemical properties and in vitro metabolism.[28] These initial models successfully predicted human pharmacokinetics in the first-in-human studies, with predicted total exposure (AUC) values falling within a twofold margin of the observed mean values.[28] As more clinical data became available, the model was refined, further improving the accuracy of its predictions for both single- and multiple-dose regimens.[28]
- Formulation De-risking: The PBPK model proved to be a critical tool for managing the biopharmaceutical risks associated with Bitopertin's low solubility. By simulating the precise relationship between particle size and oral absorption, the model enabled the development team to establish data-driven specifications for the drug substance, ensuring consistent bioavailability without the need for extensive and costly clinical relative bioavailability studies.[13]
- Ethnic Sensitivity Prediction: The PBPK platform was also leveraged to predict potential pharmacokinetic differences between ethnic populations. Simulations incorporating known physiological differences (e.g., average liver weight and CYP3A4 abundance) predicted that the geometric mean clearance of Bitopertin in Caucasians would be approximately 1.3-fold higher than in Chinese and Japanese populations.[26] This foresight was valuable for planning and interpreting data from global clinical trials.
3.3 Dose-Exposure and Dose-Response Relationships
The relationship between the administered dose, the resulting drug concentration in the body, and the ultimate pharmacological effect is a cornerstone of clinical pharmacology.
- Dose-Exposure: The relationship between the dose of Bitopertin and the resulting systemic exposure (AUC) is generally linear and predictable across the therapeutic dose range.[27] The slight deviation from dose-proportionality at doses above 50 mg, caused by solubility-limited absorption, was well-characterized and understood through modeling.[27]
- Dose-Response (Pharmacodynamics): A particularly crucial finding emerged from the Phase II schizophrenia trial (NCT00616798), which revealed a complex, non-linear dose-response relationship. While the 10 mg and 30 mg daily doses demonstrated efficacy in reducing negative symptoms, the higher 60 mg dose was ineffective.[30] This suggested a "bell-shaped" or "inverted-U" dose-response curve, a phenomenon where increasing the dose beyond a certain point leads to a diminished or absent therapeutic effect. This observation led to the hypothesis that an optimal, moderate level of GlyT1 receptor occupancy (estimated to be around 50%) was necessary to achieve a clinical benefit, and that higher levels of occupancy were counterproductive.[17] This type of complex dose-response has also been observed with other GlyT1 inhibitors, suggesting it may be a class effect related to the intricate regulation of the glutamatergic system.[16]
3.4 A Model-Informed Development Paradigm
The clinical development program for Bitopertin showcases the value of integrating computational modeling into the strategic decision-making process. The initial data revealed a significant liability in the form of low aqueous solubility and high sensitivity of its pharmacokinetics to particle size.[1] In a more traditional development paradigm, this would necessitate a series of resource-intensive clinical studies to empirically define an acceptable formulation. Instead, the development team proactively built and validated a PBPK model using preclinical data.[28] This model was not merely an academic exercise; it was applied to solve concrete problems. It quantitatively predicted the impact of specific particle size distributions on Cmax and AUC, allowing the team to set precise manufacturing controls to ensure bioequivalence.[13] It was also used to prospectively estimate pharmacokinetic differences between Caucasian and Asian populations, providing a rational basis for dose selection and data interpretation in global trials.[26] This represents a best-practice example of a model-informed drug development strategy, where in vitro and preclinical data are leveraged through sophisticated modeling to make faster, more efficient, and less risky decisions for the clinical program.
Section 4: The Schizophrenia Campaign: A Case Study in Clinical Development
The initial and most extensive chapter in Bitopertin's history was its development by Roche as a potential treatment for schizophrenia. This campaign, from its scientifically compelling rationale to its definitive late-stage failure, serves as a significant case study in the challenges of translating neuroscience hypotheses into clinical reality.
4.1 Rationale and Preclinical Evidence for Targeting NMDA Hypofunction
The entire schizophrenia program was built upon the robust scientific foundation of the NMDA receptor hypofunction hypothesis.[11] This hypothesis proposes that a core pathophysiological element of schizophrenia is diminished signaling through glutamatergic pathways mediated by the NMDA receptor. This deficit is thought to be particularly relevant to the negative symptoms (e.g., apathy, social withdrawal) and cognitive deficits that are poorly addressed by existing dopamine-blocking antipsychotics.[2] The therapeutic strategy was to indirectly enhance NMDA receptor signaling by inhibiting GlyT1, thereby increasing the synaptic availability of the co-agonist glycine.[2]
This hypothesis was supported by preclinical evidence. In vitro, Bitopertin was shown to increase long-term potentiation, a cellular correlate of learning and memory, in rat hippocampal slices.[5] In vivo, it demonstrated target engagement by reversing the hyperlocomotion induced in mice by an NMDA receptor partial agonist, with a half-maximal effective dose (
ID50) of 0.5 mg/kg.[5] These results provided the necessary proof-of-concept to proceed with clinical investigation.
4.2 Promising Signals: Analysis of Phase II Proof-of-Concept Data (NCT00616798)
The initial human efficacy data for Bitopertin in schizophrenia were highly encouraging, generating considerable excitement within the field.
- Study Design: The Phase II trial (NCT00616798) was a well-designed, randomized, double-blind, placebo-controlled study conducted at 66 sites worldwide. It enrolled 323 patients diagnosed with schizophrenia who had stable but predominant negative symptoms. For a duration of 8 weeks, patients received either placebo or one of three doses of Bitopertin (10, 30, or 60 mg per day) as an add-on to their existing standard antipsychotic therapy.[30]
- Key Findings: The study's primary outcome was the change from baseline in the PANSS negative symptom factor score. In the per-protocol population, treatment with both the 10 mg/d and 30 mg/d doses of Bitopertin resulted in a statistically significant reduction in negative symptoms compared to placebo. The mean reduction in the negative symptom score was approximately -25% for both effective doses, versus -19% for the placebo group (p=0.049 for 10 mg; p=0.03 for 30 mg).[2] Furthermore, the 10 mg group showed a significantly higher clinical response rate and a positive trend toward improved overall functioning.[30]
- Dose-Response Curve: A critical finding was that the highest dose tested, 60 mg/d, was not superior to placebo. This strongly suggested a non-linear, bell-shaped dose-response relationship.[30] Pharmacokinetic-pharmacodynamic modeling indicated that the optimal therapeutic effect was achieved at low to medium levels of GlyT1 target occupancy.[30] These positive results, published in the high-impact journal
JAMA Psychiatry, provided strong proof-of-concept and were the primary impetus for advancing Bitopertin into a large-scale Phase III program.[30]
4.3 The "SearchLyte" Program: Deconstruction of the Phase III Failures
Buoyed by the Phase II success, Roche launched an ambitious and comprehensive Phase III clinical development program named "SearchLyte." This program was designed to definitively establish the efficacy and safety of Bitopertin across the spectrum of difficult-to-treat schizophrenia symptoms.
- Program Overview: The SearchLyte program was a massive undertaking, comprising six large, multicenter, placebo-controlled Phase III studies that collectively enrolled over 3,600 patients in 32 countries.[19] The program was divided into two parallel tracks:
- Negative Symptoms: Three studies—FlashLyte (NN25310), DayLyte (WN25309), and SunLyte (WN25308)—were designed to confirm the Phase II findings in patients with persistent, predominant negative symptoms. The primary endpoint was the change in the PANSS negative symptom factor score after 24 weeks of adjunctive therapy.[19]
- Sub-optimally Controlled Symptoms: Three other studies—TwiLyte (NN25307), MoonLyte (WN25306), and NightLyte (WN25305)—evaluated Bitopertin as an adjunctive therapy for patients with sub-optimally controlled positive symptoms (e.g., hallucinations, delusions) that persisted despite standard antipsychotic treatment. The primary endpoint for these trials was the change in the PANSS positive symptom factor score at 12 weeks.[19]
- Failure to Confirm Efficacy: Despite the promising Phase II data, the SearchLyte program resulted in a near-complete failure.
- In January 2014, Roche announced that the first two negative symptom trials, FlashLyte and DayLyte, had failed to meet their primary endpoints. While patients in all treatment arms (placebo and Bitopertin doses of 5, 10, and 20 mg) showed some improvement, there was no statistically significant separation between any Bitopertin dose and placebo.[19] Subsequently, the third negative symptom trial, SunLyte, also failed, leading Roche to terminate development for this indication.[32]
- The results for sub-optimally controlled symptoms were equally disappointing. Across the three studies and six active treatment arms (doses ranging from 5 mg to 20 mg), only a single arm—the 10 mg dose in the NightLyte study—showed a statistically significant, albeit modest, improvement over placebo.[35] All other active arms in the TwiLyte, MoonLyte, and NightLyte studies failed to demonstrate superiority over placebo.[35]
- Program Termination: Faced with this overwhelming lack of efficacy across multiple large, well-conducted trials, Roche performed futility analyses on the remaining studies and made the decision to discontinue the entire Bitopertin for schizophrenia program in 2014.[6]
Table 2: Summary of the Phase III "SearchLyte" Schizophrenia Clinical Trial Program
Trial Name (NCT ID) | Target Population | Doses Tested | Primary Endpoint | Outcome | Source(s) |
---|
FlashLyte (NN25310) | Negative Symptoms | 10 mg, 20 mg | Change in PANSS Negative Score @ 24 wks | Failed | 19 |
DayLyte (WN25309) | Negative Symptoms | 5 mg, 10 mg | Change in PANSS Negative Score @ 24 wks | Failed | 19 |
SunLyte (WN25308) | Negative Symptoms | Not Specified | Change in PANSS Negative Score @ 24 wks | Failed | 19 |
TwiLyte (NN25307) | Sub-optimal Positive | 10 mg, 20 mg | Change in PANSS Positive Score @ 12 wks | Failed | 19 |
MoonLyte (WN25306) | Sub-optimal Positive | 5 mg, 10 mg | Change in PANSS Positive Score @ 12 wks | Failed | 19 |
NightLyte (WN25305) | Sub-optimal Positive | 10 mg, 20 mg | Change in PANSS Positive Score @ 12 wks | 10 mg Met, 20 mg Failed | 19 |
4.4 Post-Hoc Analysis: Factors Contributing to the Translational Failure
The stark contrast between the positive Phase II results and the definitive Phase III failures warrants a critical examination of the potential contributing factors. The translational failure of Bitopertin was likely not due to a single cause but rather a confluence of challenges related to the drug's complex pharmacology, the nature of the disease, and the difficulties of clinical trial execution in this population.
First, the complex dose-response relationship identified in Phase II may have been a critical factor. The "bell-shaped" curve suggested a narrow therapeutic window, where only a moderate level of GlyT1 occupancy was beneficial.[30] It is highly plausible that the doses selected for Phase III (5, 10, and 20 mg) did not consistently achieve this optimal occupancy across a large and heterogeneous patient population, with the 20 mg dose potentially being supra-optimal and thus less effective.[34] The developers themselves acknowledged the need for "careful dose titration" after the Phase II study, a nuance that is difficult to implement in large, fixed-dose registration trials.[17]
Second, the underlying biology of the target system is profoundly complex. Systemic inhibition of GlyT1 affects its function on multiple cell types (neurons and astrocytes) and in various brain regions, which may have opposing or nullifying effects on overall network function.[17] Furthermore, GlyT1 inhibition can have intricate downstream consequences on the metabolism of other NMDA co-agonists, such as D-serine, which may not have been fully accounted for in the therapeutic model.[17] This suggests that broad, non-specific enhancement of the glutamatergic system may be an overly simplistic approach.
Finally, the clinical trial endpoints themselves present a challenge. Negative symptoms are notoriously difficult to measure objectively and are known to have a high placebo response rate, which was observed in the Phase III trials where all arms, including placebo, showed improvement.[32] This high placebo effect can obscure a modest drug effect, making it statistically difficult to demonstrate superiority. The failure of Bitopertin, along with several other GlyT1 inhibitors, suggests a broader, class-wide difficulty in translating this mechanism into a robust clinical benefit for schizophrenia, underscoring the immense challenge of developing novel treatments for this devastating disorder.[17]
Section 5: Exploratory CNS Indications
In parallel with the primary focus on schizophrenia, the developers of Bitopertin explored its potential utility in other CNS disorders where glutamatergic dysfunction is implicated. These investigations, though less extensive, provide additional context on the perceived scope and ultimate limitations of the drug's CNS-directed mechanism.
5.1 Investigation in Obsessive-Compulsive Disorder (OCD)
Given the growing body of evidence implicating the glutamate system in the pathophysiology of OCD, Roche initiated a clinical trial to evaluate Bitopertin for this indication.
- Rationale and Trial Design: A Phase II clinical trial, identified as NCT01674361, was launched to assess the efficacy and safety of Bitopertin as an adjunctive therapy for patients with OCD who had an inadequate response to selective serotonin reuptake inhibitors (SSRIs).[1] The study was a multicenter, randomized, double-blind, placebo-controlled trial that evaluated two doses of Bitopertin, 10 mg and 30 mg, added to a stable background SSRI regimen.[37]
- Trial Status and Outcome: The trial enrolled 99 participants and was officially completed in April 2015.[37] Despite its completion, the sponsor has not published or publicly presented any efficacy data from this study. While several databases list the trial, they provide no information on its results.[6] However, one review source explicitly states that the investigation in OCD, along with those in schizophrenia and β-thalassemia, demonstrated "negative results".[39]
5.2 Preclinical Evidence in Neuropathic Pain
The role of glycine in the spinal cord, where it acts as a primary inhibitory neurotransmitter, provided a rationale for exploring Bitopertin in pain models.
- Rationale: In the spinal cord dorsal horn, glycinergic inhibition is a key mechanism for modulating pain signals. Pharmacological facilitation of this system is considered a promising strategy for treating chronic pain. Inhibition of GlyT1 can potentiate this inhibitory neurotransmission via glycine receptors while also potentially modulating glutamatergic signaling through NMDA receptors.[40]
- Preclinical Findings: A study in animal models of chronic pain demonstrated that Bitopertin effectively ameliorates allodynia (pain from a non-painful stimulus) and hyperalgesia (exaggerated pain response) in both neuropathic and inflammatory pain models. Notably, this analgesic effect was achieved without altering the perception of acute pain and without producing severe side effects, suggesting a favorable therapeutic window for this indication in a preclinical setting.[40]
5.3 Implications of Exploratory Program Outcomes
The outcomes of these exploratory efforts, particularly in OCD, further reinforce the conclusion that the clinical application of Bitopertin's CNS mechanism is fraught with challenges. In the pharmaceutical industry, the absence of publicly reported data from a completed clinical trial is often a strong indicator of a negative outcome. Companies are highly motivated to disseminate positive or even mixed results to support their scientific platforms and development assets. The fact that the Phase II OCD trial (NCT01674361) was completed in 2015, yet years later no efficacy results have been formally presented, strongly implies that the study failed to meet its primary endpoints.[37]
This presumed failure in OCD, when viewed alongside the definitive failures in the much larger schizophrenia program, paints a consistent picture: despite a compelling preclinical rationale and a clear mechanism of action, systemic GlyT1 inhibition with Bitopertin did not translate into a demonstrable clinical benefit for complex neuropsychiatric disorders. This collective evidence provides the crucial context for understanding why Roche ultimately ceased all CNS development for the compound and made it available for out-licensing, paving the way for its strategic repurposing.
Section 6: A Strategic Pivot: Repurposing for Hematologic Disorders
The story of Bitopertin's development takes a remarkable turn following the discontinuation of its CNS program. In a prime example of strategic drug repurposing, a pharmacological effect initially classified as an adverse event was re-examined and astutely identified as a potential therapeutic mechanism for an entirely different class of diseases.
6.1 From Adverse Effect to Therapeutic Hypothesis: The Hematological Signal
The foundation for Bitopertin's second act was laid by a consistent observation made during its first.
- The Key Observation: Across the numerous schizophrenia clinical trials, a consistent, dose-dependent, and reversible reduction in blood hemoglobin levels was noted in participants receiving Bitopertin.[24] This was initially monitored as a potential safety liability.
- The Scientific Pivot: Crucially, this hematological effect was not dismissed but was instead interpreted as clear evidence of on-target pharmacological activity outside the CNS. It demonstrated that systemic administration of Bitopertin was sufficient to inhibit GlyT1 on erythroid precursors, thereby limiting the transport of glycine required for heme synthesis and creating what was termed a "heme-restricted" environment.[21] This insight transformed the perception of the hemoglobin effect from an unwanted side effect into a valuable pharmacodynamic marker.
- New Company, New Strategy: Recognizing the potential of this mechanism, Disc Medicine executed a licensing agreement with Roche in May 2021, acquiring the global rights to Bitopertin. The company's explicit strategy was to pivot away from the CNS and develop the drug specifically for the treatment of serious hematologic diseases.[20]
6.2 Clinical Investigation in Erythropoietic Porphyrias (EPP/XLPP)
The primary focus of the new development program became the erythropoietic porphyrias, a group of rare diseases for which Bitopertin's mechanism appeared ideally suited.
- Disease Background: EPP and the related X-linked Protoporphyria (XLPP) are rare, inherited metabolic disorders caused by defects in the heme biosynthesis pathway. This leads to the accumulation of protoporphyrin IX (PPIX), a photoactive and toxic intermediate. When patients are exposed to sunlight, PPIX generates reactive oxygen species, causing excruciating pain, swelling, and burning sensations in the skin. Chronic accumulation can also lead to severe liver damage.[20]
- Preclinical Proof of Concept: The therapeutic hypothesis was tested in relevant preclinical models. In mouse models of both EPP and XLPP, oral administration of Bitopertin resulted in a significant reduction in blood and liver PPIX levels. Importantly, this was accompanied by a reduction in histological evidence of liver cholestasis and fibrosis, indicating a potential to modify the course of the disease.[22]
- Clinical Program: Based on this strong rationale, Disc Medicine initiated a focused clinical program in adult EPP patients, which included two key Phase 2 studies: BEACON (ACTRN12622000799752), an open-label study, and AURORA (NCT05308472), a randomized, double-blind, placebo-controlled study.[44]
6.3 Analysis of Phase II Efficacy and Safety in EPP (BEACON & AURORA Studies)
The clinical data from the EPP program have been robust and consistent, providing strong evidence of both biological activity and clinical benefit.
- Primary Endpoint Success: Both the BEACON and AURORA studies successfully met their primary endpoint, demonstrating that Bitopertin produces a statistically significant, dose-dependent, and sustained reduction in whole blood PPIX concentrations.
- In the placebo-controlled AURORA study, after 17 weeks of treatment, the 60 mg dose of Bitopertin reduced mean PPIX levels by -40.7%, compared to an +8.0% increase in the placebo group (p<0.001).[46]
- In the open-label BEACON study, after 24 weeks, the 60 mg dose achieved a mean PPIX reduction of -61.1% from baseline (p<0.001).[47]
- Evidence of Clinical Benefit: Critically, this robust reduction in the toxic metabolite translated directly into meaningful clinical improvements for patients.
- In AURORA, patients in the 60 mg group experienced a 75% reduction in the incidence rate of phototoxic reactions with pain compared to placebo (p=0.011).[46]
- On the Patient Global Impression of Change (PGIC) scale, 86% of patients in the 60 mg group reported that their EPP was "much better," a significantly higher proportion than the 50% in the placebo group (p=0.022).[46]
- The BEACON study showed a dose-dependent increase in the cumulative time patients could spend in the sun without pain and a remarkable 92% reduction in the number of patient-reported phototoxic reactions compared to the screening period.[47]
- Favorable Safety Profile in EPP: In both studies, Bitopertin was generally well-tolerated. The most common adverse event was dizziness, consistent with previous studies. A crucial finding was the absence of clinically meaningful changes in hemoglobin levels in this patient population, mitigating a key theoretical safety concern.[46]
Table 3: Summary of Key Phase II Hematology Clinical Trials in EPP
Trial Name (NCT/ACTRN ID) | Design | Patient Population | Doses | Primary Endpoint Outcome (PPIX Reduction) | Key Clinical Outcomes | Source(s) |
---|
AURORA (NCT05308472) | Randomized, Double-Blind, Placebo-Controlled | 75 Adults with EPP | 20 mg, 60 mg | -40.7% for 60 mg vs. +8.0% for placebo (p<0.001) | 75% reduction in rate of phototoxic reactions (60 mg, p=0.011); 86% of patients "much better" on PGIC (60 mg, p=0.022) | 46 |
BEACON (ACTRN12622000799752) | Randomized, Open-Label | 22 Adults with EPP/XLP | 20 mg, 60 mg | -61.1% from baseline for 60 mg (p<0.001) | 92% reduction in phototoxic reactions; Dose-dependent increase in cumulative pain-free time in sun | 47 |
6.4 Parallel Investigations: β-Thalassemia and Diamond-Blackfan Anemia (DBA)
The exploration of Bitopertin's hematological mechanism extended to other disorders of erythropoiesis.
- β-Thalassemia: The rationale for β-thalassemia was to reduce heme-induced oxidative stress.[21] Despite promising results in a mouse model where Bitopertin improved anemia and reduced hemolysis [21], a Phase 2 clinical trial in patients with non-transfusion-dependent β-thalassemia (NCT03271541) yielded negative results and was terminated early. In patients, the treatment led to an expected inhibition of heme synthesis but also caused a clinically undesirable
decrease in hemoglobin levels, failing to improve the overall disease phenotype.[39]
- Diamond-Blackfan Anemia (DBA): In DBA, a rare inherited bone marrow failure syndrome, the rationale is to rebalance heme and globin synthesis. Preclinical studies have been encouraging, showing that Bitopertin can improve the expansion of erythroid precursor cells from DBA patients in vitro and can improve anemia in a mouse model of the disease.[49] An investigator-sponsored study in DBA is currently underway.[20]
6.5 Contrasting Outcomes and Key Learnings
The divergent clinical outcomes in EPP and β-thalassemia provide a critical lesson in precision medicine. The success in EPP stems from a direct and elegant match between the drug's mechanism and the disease's core pathophysiology. In EPP, the problem is the accumulation of a toxic precursor due to a downstream blockage; the solution is to reduce the input to the pathway.[20] Bitopertin's substrate reduction mechanism is perfectly suited for this task. In contrast, the pathophysiology of β-thalassemia is far more complex, involving globin chain imbalance and severe ineffective erythropoiesis.[21] In this context, the primary effect of restricting heme synthesis was simply to further impair hemoglobin production, which was detrimental to already anemic patients.[39] This highlights that a "one size fits all" approach to heme restriction is not viable and that success depends on precisely targeting the specific molecular defect.
Furthermore, a crucial and highly favorable finding from the EPP program is that Bitopertin did not cause clinically significant anemia in these patients, despite its known effect on hemoglobin in healthy individuals.[46] This observation significantly de-risks the therapy. It is likely that in the pathological state of EPP, where the heme pathway is already compromised, reducing the initial glycine substrate serves to normalize the metabolic flux rather than causing a complete shutdown of hemoglobin production. This allows the system to find a new, lower, but non-anemic homeostatic set point, thereby widening the therapeutic index and making Bitopertin a much more attractive clinical candidate for this indication.
Section 7: Comprehensive Safety and Tolerability Profile
A key asset for the current development of Bitopertin is the extensive body of safety data accumulated over its entire clinical history. This large database, spanning multiple patient populations and long-term exposure, provides a robust understanding of the drug's risk profile.
7.1 Synthesis of Safety Data from over 4,000 Clinical Trial Participants
Bitopertin has been administered to over 4,000 human subjects, including healthy volunteers and patients in both the large-scale schizophrenia program and the more recent hematology trials.[20] This extensive clinical experience provides a high degree of confidence in its overall safety profile. Across this broad range of studies, Bitopertin has been consistently described as generally safe and well-tolerated.[19] The vast majority of treatment-emergent adverse events (TEAEs) reported have been mild to moderate in severity.[25]
7.2 Characterization of Common and Dose-Dependent Adverse Events
The safety profile of Bitopertin is characterized by a few common, dose-dependent adverse events.
- CNS Effects: The most frequently reported adverse events are related to the central nervous system, which is consistent with the drug's ability to cross the blood-brain barrier and modulate neurotransmission. Dizziness and somnolence (drowsiness) are the most common TEAEs and their incidence increases with the dose.[25] For example, in the AURORA trial for EPP, dizziness was reported in 44% of patients in the 60 mg group, compared to 15% in the 20 mg group and 17% in the placebo group.[48] Headache has also been reported as a prominent event in some trials.[39]
- Hematological Effects: As discussed previously, a dose-dependent, gradual, and reversible decrease in blood hemoglobin was a hallmark finding in studies involving healthy volunteers and schizophrenia patients.[25] In a 52-week study, these decreased levels remained stable over long-term treatment and tended to return toward baseline after drug discontinuation.[25] While withdrawal criteria were met in a small number of schizophrenia patients due to significant hemoglobin drops, no associated bleeding was reported, and levels recovered after stopping the drug.[25] Importantly, in the target EPP patient population, this effect was not clinically meaningful, with no significant changes in hemoglobin observed.[46]
- Other Adverse Events: In a long-term (52-week) study in Japanese patients with schizophrenia, other commonly reported AEs included nasopharyngitis (common cold) and worsening of the underlying schizophrenia, the latter of which is difficult to distinguish from the natural course of the illness.[25]
- Serious Adverse Events (SAEs): The overall incidence of SAEs across all clinical programs has been low.[35] In the EPP program, no SAEs were reported in the BEACON study, and the single SAE in the AURORA study occurred in a patient receiving placebo.[47]
Table 4: Consolidated Safety Profile: Common Adverse Events Across Major Clinical Programs
Adverse Event | Incidence in Schizophrenia Program (52-wk study) 25 | Incidence in EPP Program (AURORA study) 48 | Comments |
---|
| 5 / 10 / 20 mg Doses | Placebo / 20 / 60 mg Doses | |
Dizziness | Not specified as common | 17% / 15% / 44% | Most common AE in EPP trials; dose-dependent. |
Somnolence | 0% / 12.3% / 24.7% | Not specified as common | Dose-dependent; common in CNS trials. |
Headache | 7.5% (overall) | Not specified as common | Reported as prominent in some hematology trials. |
Hemoglobin Decrease | Dose-dependent, gradual decrease from baseline | No meaningful changes observed | Key pharmacodynamic effect; clinically significant only in non-EPP populations. |
Nasopharyngitis | 39.1% (overall) | Not specified as common | Common in long-term schizophrenia study. |
Worsening Schizophrenia | 10.6% (overall) | N/A | Confounded by underlying disease. |
7.3 Strategic Value of the Pre-existing Safety Database
The extensive safety database amassed during the large-scale schizophrenia program represents a major strategic asset that significantly de-risks and accelerates the current development path for Bitopertin in EPP. Typically, developing a new drug for a rare disease is a slow process, often constrained by the difficulty of enrolling a sufficient number of patients to build a comprehensive safety database from scratch. Bitopertin, however, benefits from a pre-existing and well-characterized safety and tolerability profile derived from over 4,000 individuals, including data from long-term (52-week) exposure.[20]
This means that the nature, frequency, dose-dependency, and time course of its common adverse events are already well understood. This wealth of prior human data provides a high degree of confidence for patients, investigators, and regulatory agencies like the FDA. It reduces uncertainty about long-term safety, streamlines the design of pivotal trials, and strengthens the overall regulatory submission package. This pre-existing database is a significant competitive advantage for Disc Medicine that would not have existed had the drug's development originated in the rare disease space.
Section 8: Regulatory Trajectory and Future Outlook
Following the successful demonstration of efficacy and safety in Phase II trials for erythropoietic protoporphyria, Bitopertin has entered a new phase focused on securing regulatory approval and market access. This final section outlines the current regulatory strategy, the design of the confirmatory Phase III trial, and provides an expert synthesis of the drug's future prospects.
8.1 The Path to Market: FDA Engagement and the Accelerated Approval Pathway
Disc Medicine has established a clear and collaborative regulatory strategy with the U.S. Food and Drug Administration (FDA) aimed at bringing Bitopertin to patients as efficiently as possible.
- Regulatory Strategy: The company is actively pursuing an accelerated approval pathway for Bitopertin for the treatment of EPP.[43] This pathway is reserved for drugs that treat serious conditions and fill an unmet medical need, based on a surrogate endpoint that is reasonably likely to predict clinical benefit.
- FDA Alignment: Through a series of successful interactions, including an End-of-Phase-2 meeting and a Type C meeting, Disc Medicine has achieved alignment with the FDA on key aspects of its development plan. Most critically, the FDA has agreed with the potential for the reduction of whole blood PPIX to serve as an acceptable surrogate endpoint to support an accelerated approval.[43] This agreement is foundational to the entire regulatory strategy.
- New Drug Application (NDA) Submission: Based on this regulatory alignment and the strength of the existing clinical data package from the completed Phase II BEACON and AURORA trials, Disc Medicine has announced its plan to submit an NDA to the FDA in the second half of 2025.[43]
8.2 The APOLLO Confirmatory Trial: Design, Endpoints, and Implications
A condition of the accelerated approval pathway is the requirement to conduct a post-marketing study to verify and describe the drug's clinical benefit. This confirmatory trial for Bitopertin is named APOLLO.
- Purpose and Design: The APOLLO trial is a Phase III, randomized, double-blind, placebo-controlled study designed to provide definitive evidence of Bitopertin's clinical efficacy.[43] The trial will enroll adults and, importantly, adolescents (ages 12 and older) with EPP and XLPP. It will evaluate a single dose level of 60 mg of Bitopertin administered once daily over a 6-month treatment period.[44]
- Endpoints: The trial has been designed with robust co-primary endpoints that have been agreed upon with the FDA:
- Clinical Benefit Endpoint: The average monthly total time in sunlight (between 10:00 and 18:00) on days without pain during the last month of treatment. This is a direct, patient-centric measure of improved light tolerance and quality of life.[43]
- Surrogate Endpoint: The percent change from baseline in whole blood metal-free PPIX after 6 months of treatment. Including this endpoint allows for direct confirmation of the biological effect seen in Phase II.[43]
- Timeline and Strategic Importance: Disc Medicine plans to initiate the APOLLO trial by mid-2025.[43] This timeline is strategically important, as it ensures that enrollment in the confirmatory study will be well underway, or potentially complete, by the time the FDA makes a decision on the accelerated approval. This proactive approach helps to minimize the time between a potential conditional approval and full approval.
8.3 Synthesis and Expert Conclusion: Bitopertin's Evolving Therapeutic Identity and Future Potential
The development history of Bitopertin is a powerful narrative of scientific resilience and strategic acumen. The molecule began its life as a high-profile candidate for schizophrenia, designed to address the complex challenge of NMDA receptor hypofunction. After failing definitively in one of the largest Phase III programs of its time, it was effectively consigned to the "graveyard" of failed neuropsychiatric drugs. However, through astute scientific observation and a deep understanding of the drug's on-target pharmacology, a seemingly minor side effect was reconceptualized as a primary therapeutic mechanism. This led to the drug's rescue and repurposing as a highly promising, first-in-class, disease-modifying therapy for the debilitating rare disease, erythropoietic protoporphyria.
Today, Bitopertin stands as a mature, late-stage clinical asset. It is supported by robust Phase II data demonstrating a powerful and consistent effect on a key surrogate biomarker (PPIX) that is directly and logically linked to multiple, clinically meaningful patient-reported outcomes. It possesses a well-characterized safety profile established in an exceptionally large patient population, and it benefits from a clear, de-risked regulatory path toward accelerated approval.
The probability of technical and regulatory success for Bitopertin in EPP appears high. If approved, it would represent a significant therapeutic advance, addressing a substantial unmet medical need by offering patients an oral, convenient, and disease-modifying treatment option.[43] Beyond its clinical and commercial potential, the story of Bitopertin will endure as a premier case study in modern drug development. It exemplifies the value of rigorously understanding a drug's mechanism of action, the potential for serendipity in clinical observation, and the strategic intelligence required to successfully navigate the complexities of pharmaceutical R&D and turn failure into success.
Works cited
- Bitopertin: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 4, 2025, https://go.drugbank.com/drugs/DB12426
- Bitopertin - Wikipedia, accessed September 4, 2025, https://en.wikipedia.org/wiki/Bitopertin
- Bitopertin | 99.80%(HPLC) | In Stock | GlyT inhibitor - Selleck Chemicals, accessed September 4, 2025, https://www.selleckchem.com/products/bitopertin-rg1678-ro-4917838.html
- Bitopertin | C21H20F7N3O4S | CID 24946690 - PubChem, accessed September 4, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Bitopertin
- Bitopertin (CAS 845614-11-1) - Cayman Chemical, accessed September 4, 2025, https://www.caymanchem.com/product/19896/bitopertin
- bitopertin | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 4, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=7546
- bitopertin | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 4, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7546
- Compound: BITOPERTIN (CHEMBL1171829) - ChEMBL - EMBL-EBI, accessed September 4, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL1171829
- BITOPERTIN - precisionFDA, accessed September 4, 2025, https://precision.fda.gov/ginas/app/ui/substances/Q8L6AN59YY
- Bitopertin | CAS 845614-11-1 | SCBT - Santa Cruz Biotechnology, accessed September 4, 2025, https://www.scbt.com/p/bitopertin-845614-11-1
- Bitopertin | CAS NO.:845614-11-1 - GlpBio, accessed September 4, 2025, https://www.glpbio.com/bitopertin.html
- BITOPERTIN, accessed September 4, 2025, https://gsrs-dev-public.ncats.io/ginas/app/ui/substances/e66450ca-ee06-41c0-888c-5b2faf966c7b
- Physiologically Based Absorption Modelling to Predict the Impact of Drug Properties on Pharmacokinetics of Bitopertin - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/263516021_Physiologically_Based_Absorption_Modelling_to_Predict_the_Impact_of_Drug_Properties_on_Pharmacokinetics_of_Bitopertin
- GlyT | Inhibitors | MedChemExpress, accessed September 4, 2025, https://www.medchemexpress.com/Targets/GlyT.html
- What is Bitopertin used for? - Patsnap Synapse, accessed September 4, 2025, https://synapse.patsnap.com/article/what-is-bitopertin-used-for
- Development of the novel GlyT1 inhibitor, iclepertin (BI 425809), for the treatment of cognitive impairment associated with schizophrenia - PMC - PubMed Central, accessed September 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10465677/
- Inhibition of astrocytic glycine transporter-1: friend or foe for ameliorating NMDA receptor hypofunction? - Frontiers, accessed September 4, 2025, https://www.frontiersin.org/journals/cellular-neuroscience/articles/10.3389/fncel.2024.1389718/full
- Inhibition of glycine transporter 1: The yellow brick road to new schizophrenia therapy?, accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/26205290/
- Roche provides update on the first two of six phase III studies of bitopertin in schizophrenia, accessed September 4, 2025, https://assets.roche.com/imported/en/med-cor-2014-01-21-e.pdf
- Bitopertin (GlyT1 Inhibitor) for Erythropoietic Porphyrias - Disc Medicine, accessed September 4, 2025, https://www.discmedicine.com/our-pipeline/bitopertin/
- Bitopertin, a selective oral GLYT1 inhibitor, improves anemia in a mouse model of β-thalassemia - PMC, accessed September 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6948868/
- Proof of Mechanism Studies with Bitopertin, a Selective Glycine Transporter 1 Inhibitor Under Development - Disc Medicine, accessed September 4, 2025, http://www.discmedicine.com/wp-content/uploads/2022/05/Porphyria-Symposium-2021-Bitopertin-poster_final_w-siganature-page_FE-pages-deleted.pdf
- Erythropoietic Protoporphyria and X-Linked Porphyria, accessed September 4, 2025, https://www.porphyria.org/epp-xlp
- Bitopertin, a selective oral GLYT1 inhibitor, improves anemia in a mouse model of β-thalassemia - JCI Insight, accessed September 4, 2025, https://insight.jci.org/articles/view/130111
- A double-blind randomized study assessing safety and efficacy ..., accessed September 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4791769/
- Combining 'Bottom-Up'and 'Top-Down'Methods to Assess Ethnic ..., accessed September 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4916198/
- Physiologically Based Pharmacokinetic Modelling to Predict Single- and Multiple-Dose Human Pharmacokinetics of Bitopertin | Request PDF - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/236206224_Physiologically_Based_Pharmacokinetic_Modelling_to_Predict_Single-_and_Multiple-Dose_Human_Pharmacokinetics_of_Bitopertin
- Physiologically based pharmacokinetic modelling to predict single- and multiple-dose human pharmacokinetics of bitopertin - PubMed, accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/23591780/
- Study of Bitopertin to Evaluate the Safety, Tolerability, Efficacy, and PPIX Concentrations in Participants With EPP | Mount Sinai, accessed September 4, 2025, https://www.mountsinai.org/clinical-trials/study-of-bitopertin-to-evaluate-safety-tolerability-efficacy-ppix-concentrations-in-participants-with-epp
- (PDF) Effect of Bitopertin, a Glycine Reuptake Inhibitor, on Negative Symptoms of Schizophrenia A Randomized, Double-Blind, Proof-of-Concept Study - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/261328109_Effect_of_Bitopertin_a_Glycine_Reuptake_Inhibitor_on_Negative_Symptoms_of_Schizophrenia_A_Randomized_Double-Blind_Proof-of-Concept_Study
- Effect of bitopertin, a glycine reuptake inhibitor, on negative symptoms of schizophrenia: a randomized, double-blind, proof-of-concept study - PubMed, accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/24696094/
- Bitopertin Disappoints as Schizophrenia Treatment - Medscape, accessed September 4, 2025, https://www.medscape.com/viewarticle/826805
- Summary of the six phase III clinical trials of bitopertin initiated by... - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/figure/Summary-of-the-six-phase-III-clinical-trials-of-bitopertin-initiated-by-Hoffman-La-Roche_tbl1_280327796
- Bitopertin in Negative Symptoms of Schizophrenia-Results From the ..., accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/28117049/
- Efficacy and safety of adjunctive bitopertin versus placebo in patients with suboptimally controlled symptoms of schizophrenia treated with antipsychotics: results from three phase 3, randomised, double-blind, parallel-group, placebo-controlled, multicentre studies in the SearchLyte clinical trial programme - PubMed, accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/27816567/
- Roche adds a slate of PhIII schizophrenia trial failures to bitopertin's ..., accessed September 4, 2025, https://www.fiercebiotech.com/r-d/roche-adds-a-slate-of-phiii-schizophrenia-trial-failures-to-bitopertin-s-obituary
- Study Details | A Study of Bitopertin (RO4917838) in Combination ..., accessed September 4, 2025, https://clinicaltrials.gov/study/NCT01674361
- Bitopertin Completed Phase 2 Trials for Obsessive Compulsive Disorder (OCD) Treatment, accessed September 4, 2025, https://go.drugbank.com/drugs/DB12426/clinical_trials?conditions=DBCOND0033099&phase=2&purpose=treatment&status=completed
- Phase 2 Clinical Trial Results for Bitopertin, an Oral Glycine Transporter 1 Inhibitor, in Patients with Non-Transfusion Dependent Beta-Thalassemia | Request PDF - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/336542125_Phase_2_Clinical_Trial_Results_for_Bitopertin_an_Oral_Glycine_Transporter_1_Inhibitor_in_Patients_with_Non-Transfusion_Dependent_Beta-Thalassemia
- The GlyT1 Inhibitor Bitopertin Ameliorates Allodynia and Hyperalgesia in Animal Models of Neuropathic and Inflammatory Pain - Frontiers, accessed September 4, 2025, https://www.frontiersin.org/journals/molecular-neuroscience/articles/10.3389/fnmol.2017.00438/full
- A phase II/III trial of bitopertin monotherapy compared with placebo in patients with an acute exacerbation of schizophrenia – Results from the CandleLyte study | Request PDF - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/261184646_A_phase_IIIII_trial_of_bitopertin_monotherapy_compared_with_placebo_in_patients_with_an_acute_exacerbation_of_schizophrenia_-_Results_from_the_CandleLyte_study
- A double-blind randomized study assessing safety and efficacy following one-year adjunctive treatment with bitopertin, a glycine reuptake inhibitor, in Japanese patients with schizophrenia - ResearchGate, accessed September 4, 2025, https://www.researchgate.net/publication/298808931_A_double-blind_randomized_study_assessing_safety_and_efficacy_following_one-year_adjunctive_treatment_with_bitopertin_a_glycine_reuptake_inhibitor_in_Japanese_patients_with_schizophrenia
- Disc Medicine Announces Successful Type C Meeting with FDA for Bitopertin in Erythropoietic Protoporphyria (EPP) and Shares Plans for NDA Submission - FirstWord Pharma, accessed September 4, 2025, https://firstwordpharma.com/story/5929433
- Disc Medicine Announces Successful End of Phase 2 Meeting with FDA for Bitopertin in Erythropoietic Protoporphyria (EPP), Including Potential for Accelerated Approval, accessed September 4, 2025, https://ir.discmedicine.com/news-releases/news-release-details/disc-medicine-announces-successful-end-phase-2-meeting-fda/
- PB2562: STUDY DESIGN OF THE AURORA TRIAL: A PHASE 2, RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED TRIAL OF BITOPERTIN IN ERYTHROPOIETIC PROTOPORPHYRIA - PMC, accessed September 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10429793/
- Disc Reports Topline Results from Phase 2 AURORA Study of Bitopertin in Patients with Erythropoietic Protoporphyria (EPP), accessed September 4, 2025, https://ir.discmedicine.com/news-releases/news-release-details/disc-reports-topline-results-phase-2-aurora-study-bitopertin/
- Results from the Beacon Trial: A Phase 2, Randomized, Open-Label Study of Bitopertin in Erythropoietic Protoporphyria | Blood | American Society of Hematology, accessed September 4, 2025, https://ashpublications.org/blood/article/144/Supplement%201/1093/529163/Results-from-the-Beacon-Trial-A-Phase-2-Randomized
- Paper: Results from the Aurora Study: A Phase 2, Randomized, Double-Blind, Placebo-Controlled Study of Bitopertin in Erythropoietic Protoporphyria, accessed September 4, 2025, https://ash.confex.com/ash/2024/webprogram/Paper203503.html
- Preclinical Studies of the GlyT1 Inhibitor Bitopertin in Diamond-Blackfan Anemia | Blood, accessed September 4, 2025, https://ashpublications.org/blood/article/142/Supplement%201/1355/499264/Preclinical-Studies-of-the-GlyT1-Inhibitor
- A phase II/III trial of bitopertin monotherapy compared with placebo in patients with an acute exacerbation of schizophrenia - results from the CandleLyte study - PubMed, accessed September 4, 2025, https://pubmed.ncbi.nlm.nih.gov/24735806/
- Disc raises $225.5m as it eyes approval of rare skin disorder drug, accessed September 4, 2025, https://www.pharmaceutical-technology.com/news/disc-raises-225-5m-as-it-eyes-approval-of-rare-skin-disorder-drug/
- Disc Medicine Advances Bitopertin NDA for Erythropoietic Protoporphyria as Pipeline Progress Continues - MedPath, accessed September 4, 2025, https://trial.medpath.com/news/6420024f2b9d87ec/disc-medicine-advances-bitopertin-nda-for-erythropoietic-protoporphyria-as-pipeline-progress-continues
- Bitopertin for Erythropoietic Protoporphyria - Clinical Trials, accessed September 4, 2025, https://www.withpower.com/trial/phase-3-erythropoietic-protoporphyria-epp-3-2025-6f265