C24H28FN3O
313368-91-1
Depressive Episodes, Schizophrenia
Lumateperone, marketed under the brand name Caplyta®, is a novel second-generation (atypical) antipsychotic agent developed for the treatment of serious central nervous system (CNS) disorders.[1] It is specifically approved for schizophrenia and bipolar depression in adults.[1] Lumateperone is distinguished by a unique receptor binding profile, which contributes to its mechanism of action and a generally favorable tolerability profile, particularly concerning metabolic disturbances and extrapyramidal symptoms (EPS), when compared to some other antipsychotic medications.[1]
Schizophrenia, a complex mental illness affecting approximately 1% of the population, presents a significant treatment challenge [User Query]. While several antipsychotics, including aripiprazole, paliperidone, and clozapine, are available, their use is often accompanied by significant metabolic and/or neurological adverse effects [User Query]. Lumateperone's development appears to be a direct response to these limitations. The emphasis on its "novel" mechanism involving the modulation of glutamate, serotonin, and dopamine, coupled with "minimal off-target activity" and a "more favourable adverse effect profile," suggests a targeted drug design strategy aimed at improving upon existing therapies [User Query]. This approach seeks to provide efficacy in managing both positive and negative symptoms of schizophrenia with enhanced safety and tolerability [User Query].
The evolution of lumateperone's approved indications, from schizophrenia (December 2019) to subsequently include bipolar depression (December 2021, as monotherapy and adjunctive therapy) [1], reflects a broadening understanding of its therapeutic utility. This expansion is likely driven by its complex pharmacology that extends beyond simple dopamine D<sub>2</sub> receptor antagonism, engaging multiple neurotransmitter systems relevant to the pathophysiology of both psychotic and mood disorders.[8]
The fundamental identifying details for lumateperone are consolidated in Table 1. This information is crucial for unambiguous identification by healthcare professionals, researchers, and regulatory bodies, facilitating database searches, literature reviews, and precise communication regarding the compound.
Table 1: Lumateperone - Key Identifying Information
Attribute | Details | Reference(s) |
---|---|---|
Generic Name | Lumateperone | User Query |
Brand Name | Caplyta® | 1 |
DrugBank ID | DB06077 | User Query |
CAS Number | 313368-91-1 | User Query, 13 |
Type | Small Molecule | User Query |
Developer | Originally: Intra-Cellular Therapies, Inc. <br> Current: Johnson & Johnson (following acquisition of Intra-Cellular Therapies, Inc., completed April 2, 2025; operates as a unit within Johnson & Johnson Innovative Medicine) | 1 |
Chemical Name (IUPAC) | 1-(4-Fluorophenyl)-4-hexadeca-5,7,9(16)-trien-12-yl]butan-1-one | 13 |
Molecular Formula | C<sub>24</sub>H<sub>28</sub>FN<sub>3</sub>O | 13 |
Lumateperone's therapeutic efficacy is derived from its sophisticated multi-target engagement strategy, which involves the simultaneous and nuanced modulation of dopaminergic, serotonergic, and glutamatergic neurotransmitter systems.[3] This complex pharmacology distinguishes it from many conventional antipsychotics that primarily target dopamine D<sub>2</sub> receptors.
Serotonin System Modulation:
A cornerstone of lumateperone's action is its potent antagonism at serotonin 5-HT<sub>2A</sub> receptors, evidenced by a high binding affinity (K<sub>i</sub> = 0.54 nM).10 This affinity is approximately 60 times greater than its affinity for dopamine D<sub>2</sub> receptors.10 Such potent 5-HT<sub>2A</sub> antagonism is a hallmark of atypical antipsychotics and is believed to contribute significantly to antipsychotic efficacy, particularly in addressing negative symptoms, while concurrently reducing the risk of EPS and hyperprolactinemia commonly associated with D<sub>2</sub> blockade.10
Furthermore, lumateperone exhibits moderate binding affinity for the serotonin transporter (SERT), with reported K<sub>i</sub> values of 33 nM 11 or 62 nM.10 This inhibition of serotonin reuptake, especially when combined with its 5-HT<sub>2A</sub> antagonism, is thought to play a crucial role in its efficacy for depressive symptoms, making it suitable for its indication in bipolar depression.10 The combined actions on 5-HT<sub>2A</sub> receptors and SERT may offer synergistic antidepressant effects, potentially addressing the affective components of both schizophrenia and bipolar disorder more comprehensively than agents with more restricted serotonergic actions.
Dopamine System Modulation:
Lumateperone interacts with the dopamine system in a multifaceted manner. It functions as a dopamine D<sub>2</sub> receptor presynaptic partial agonist and postsynaptic antagonist (K<sub>i</sub> = 32 nM).10 This dual action is theorized to stabilize dopamine neurotransmission. As a presynaptic partial agonist, it can modulate dopamine release, thereby preventing excessive dopaminergic hyperactivity (often linked to the positive symptoms of schizophrenia) without inducing profound dopaminergic hypoactivity (which can exacerbate negative symptoms and cognitive impairment).10
A critical aspect of its dopaminergic action is its regional selectivity. Lumateperone preferentially targets D<sub>2</sub> receptors in the mesolimbic and mesocortical brain pathways, which are central to psychosis and mood regulation. Conversely, it demonstrates lower affinity for D<sub>2</sub> receptors in the nigrostriatal pathway, the region primarily involved in motor control. This selectivity is a key factor contributing to its observed lower liability for EPS.10 The significantly higher affinity of lumateperone for 5-HT<sub>2A</sub> receptors over D<sub>2</sub> receptors (K<sub>i</sub> ratio ~1:60) likely underpins this favorable EPS profile, as potent 5-HT<sub>2A</sub> blockade can mitigate some effects of D<sub>2</sub> blockade in the nigrostriatal pathway, a principle of atypicality that lumateperone appears to leverage effectively.10
Lumateperone also modulates dopamine D<sub>1</sub> receptor-mediated pathways, with a K<sub>i</sub> of 52 nM for D<sub>1</sub> receptors.10 This interaction is instrumental in its indirect effects on the glutamate system.
Glutamate System Modulation:
Uniquely among currently available antipsychotics, lumateperone has been shown to enhance neurotransmission mediated by both N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors.8 This effect is not achieved through direct binding to glutamate receptors but is believed to be an indirect consequence of its D<sub>1</sub> receptor agonism and SERT inhibition. These actions synergistically lead to increased levels of dopamine and glutamate in the medial prefrontal cortex (mPFC), a brain region crucial for cognitive functions and executive control.10
Deficiencies in NMDA receptor-mediated glutamatergic neurotransmission are strongly implicated in the pathophysiology of cognitive impairments and negative symptoms in schizophrenia. Lumateperone's capacity to enhance glutamate release via D<sub>1</sub> agonism offers a novel and potentially more nuanced pharmacological strategy to address these often treatment-resistant symptom domains, distinguishing it from traditional antipsychotics.10 This indirect modulation may provide a safer approach than direct glutamate receptor agonism, which has faced developmental challenges.
Minimal Affinity for Other Receptors and Clinical Implications:
Lumateperone exhibits negligible binding affinity for H<sub>1</sub> histaminergic, 5-HT<sub>2C</sub> serotonergic, and muscarinic cholinergic receptors.10 This "cleaner" receptor profile is clinically significant because it predicts a lower incidence of common and burdensome antipsychotic-related side effects. Specifically, the lack of affinity for H<sub>1</sub> and 5-HT<sub>2C</sub> receptors reduces the likelihood of sedation and weight gain, while minimal interaction with muscarinic receptors lessens the risk of anticholinergic side effects such as dry mouth, constipation, blurred vision, and cognitive impairment. This contributes substantially to lumateperone's overall favorable tolerability.10
The specific binding affinities of lumateperone for key neurotransmitter receptors and transporters are summarized in Table 2. These quantitative data provide the pharmacological basis for its mechanism of action and help to explain its clinical profile, including its efficacy and side-effect liability.
Table 2: Lumateperone Receptor Binding Affinities (K<sub>i</sub> values)
Receptor/Transporter | K<sub>i</sub> Value (nM) | Reference(s) |
---|---|---|
Serotonin 5-HT<sub>2A</sub> Receptor | 0.54 | 10 |
Dopamine D<sub>2</sub> Receptor | 32 | 10 |
Serotonin Transporter (SERT) | 33 / 62 | 11 / 10 |
Dopamine D<sub>1</sub> Receptor | 52 | 10 |
H<sub>1</sub> Histaminergic Receptor | Negligible affinity | 10 |
5-HT<sub>2C</sub> Serotonergic Receptor | Negligible affinity | 10 |
Muscarinic Cholinergic Receptors | Negligible affinity | 10 |
Note: For SERT, K<sub>i</sub> values of 33 nM [11] and 62 nM [10] have been reported, both indicating moderate affinity.
The pharmacokinetic profile of lumateperone has been characterized in adults.
Absorption:
Following oral administration, lumateperone is rapidly absorbed, with median T<sub>max</sub> (time to peak plasma concentration) achieved within 1 to 2 hours.6 With once-daily dosing, pharmacokinetic steady-state concentrations are typically reached in approximately 5 days.10 The absolute oral bioavailability of lumateperone is relatively low, reported at 4.4% 12, suggesting significant first-pass metabolism.
Lumateperone can be administered with or without food.13 However, the presence of food, particularly high-fat meals, increases the extent and slows the rate of absorption. Compared to fasted conditions, low-, moderate-, and high-fat meals increased lumateperone AUC<sub>(0-∞)</sub> by 33%, 41%, and 66%, respectively. Correspondingly, C<sub>max</sub> increased by 46%, 52%, and 67%, and T<sub>max</sub> was prolonged from 2 hours (fasted) to 3 hours (low-fat), 4 hours (moderate-fat), and 5 hours (high-fat).13 This food effect, while notable, is accommodated by the flexible dosing recommendations.
Distribution:
Lumateperone is highly bound to human plasma proteins (≥98.9%), primarily to albumin and α1-acid glycoprotein, and this binding is independent of its plasma concentration. The apparent volume of distribution (Vd/F) following a 50 mg once-daily dose is estimated to be 17.4 L, based on population pharmacokinetic analysis.
Metabolism:
Lumateperone undergoes extensive metabolism through multiple pathways.7 The primary route of metabolism is glucuronidation via UGT1A1, with a substantial contribution from oxidative metabolism mediated by CYP3A4. Other enzymes involved to a lesser extent include CYP2C8, CYP1A2, other UGT isoenzymes (such as UGT1A3 and UGT1A9), and aldo-keto reductase isoenzymes 13, S_S19, 27. The significant role of CYP3A4 and UGT1A1 in its clearance makes lumateperone susceptible to drug-drug interactions with modulators of these enzymes.
Excretion:
The terminal elimination half-life (t<sub>1/2</sub>) of lumateperone is approximately 18 hours 15, with other reports indicating a range of 13 to 29 hours. This half-life supports a once-daily dosing regimen. Following a single oral dose of radiolabeled lumateperone, approximately 53% of the total radioactivity was recovered in feces (primarily as unchanged drug) and 31% in urine (predominantly as metabolites: an ether glucuronide accounted for 18.9% of the total dose, a metabolite formed by oxidation at the benzylic carbon for 3.0%, and its hydrolytic N-dealkylation product for 3.6%). Renal elimination of unchanged lumateperone is minimal (<1% of the dose). The apparent clearance (CL/F) of lumateperone is approximately 1.0 L/h.
The pharmacokinetic characteristics of lumateperone are summarized in Table 3.
Table 3: Summary of Lumateperone Pharmacokinetic Parameters
Parameter | Value | Reference(s) |
---|---|---|
T<sub>max</sub> (Median) | 1-2 hours (fasted) | 6 |
3-5 hours (with low to high-fat meal) | 13 | |
Oral Bioavailability | 4.4% | 12 |
Effect of Food (vs. Fasted) | AUC<sub>(0-∞)</sub> ↑ by 33% (low-fat), 41% (mod-fat), 66% (high-fat) <br> C<sub>max</sub> ↑ by 46% (low-fat), 52% (mod-fat), 67% (high-fat) | 13 |
Protein Binding | ≥98.9% (primarily albumin, α1-acid glycoprotein) | S_S260 |
Apparent Volume of Distribution (Vd/F) | 17.4 L | S_S260 |
Terminal Half-life (t<sub>1/2</sub>) | Approx. 18 hours (range 13-29 hours) | S_S31, S_S27, S_S260 |
Apparent Clearance (CL/F) | Approx. 1.0 L/h | S_S260 |
Primary Metabolic Pathways | Glucuronidation (UGT1A1 primarily, also UGT1A3, UGT1A9), Oxidation (CYP3A4 primarily, also CYP2C8, CYP1A2), Aldo-keto reduction | S_S3, 13, S_S19, 27, S_S260 |
Primary Routes of Excretion (of total dose) | Feces: 53% (mostly unchanged drug) <br> Urine: 31% (as metabolites; <1% unchanged) | S_S260 |
The pharmacokinetic profile, particularly the ~18-hour half-life and achievement of steady state within 5 days, strongly supports the convenient once-daily dosing regimen. This simplicity is a notable clinical advantage, enhancing the potential for patient adherence in chronic conditions like schizophrenia and bipolar disorder, where complex medication schedules can be a barrier to effective long-term management.
Lumateperone, administered at a dose of 42 mg once daily, has demonstrated statistically significant and clinically meaningful efficacy in the treatment of adults experiencing an acute exacerbation of schizophrenia.[1] Its efficacy has been established in pivotal, randomized, double-blind, placebo-controlled, multicenter inpatient trials.[12]
Pivotal Clinical Trials (Study 1/NCT01499563 & Study 2/NCT02282761):
The primary efficacy endpoint in these studies was the change from baseline in the Positive and Negative Syndrome Scale (PANSS) total score at Week 4 compared to placebo. The PANSS is a 30-item clinician-rated scale used to measure the severity of schizophrenia symptoms, with scores ranging from 30 to 210; higher scores indicate greater overall symptom severity.16
The consistent efficacy of the 42 mg dose across these two pivotal trials, while other doses (28 mg and 84 mg) did not consistently outperform placebo, strongly supports 42 mg as the optimal therapeutic dose for acute schizophrenia, suggesting a well-defined therapeutic window.[16] The inclusion of risperidone in Study 1, showing a similar magnitude of PANSS reduction, validates the trial's ability to detect an antipsychotic effect and, when considered with lumateperone's distinct safety profile, highlights its potential as a valuable alternative.[16]
PANSS Subscales:
Lumateperone treatment was associated with improvements across the various domains of schizophrenia symptoms, including the PANSS positive subscale (e.g., delusions, hallucinations, conceptual disorganization), negative subscale (e.g., blunted affect, emotional withdrawal, poor rapport, lack of spontaneity), and general psychopathology subscale (e.g., anxiety, tension, depression, poor attention).17
Secondary Efficacy Endpoints:
In Study 2, the Clinical Global Impression-Severity (CGI-S) score was a key secondary endpoint. Lumateperone 42 mg resulted in a statistically significant improvement in the CGI-S score compared to placebo at Week 4. Patients receiving lumateperone showed a mean improvement of -0.8 points from a baseline mean of 4.8, compared to -0.5 points for placebo (LSMD -0.3; p<0.05).16 The CGI-S is a 7-point scale where clinicians rate the patient's overall illness severity.16
Long-term Efficacy/Relapse Prevention:
Further supporting its utility in schizophrenia, positive topline results evaluating the efficacy and safety of lumateperone for the prevention of relapse in adult patients with schizophrenia were announced in November 2024.18
Table 4: Summary of Efficacy in Pivotal Schizophrenia Trials (Lumateperone 42 mg vs. Placebo)
Trial Identifier | Primary Endpoint (PANSS Total Score Change from Baseline at Week 4) | Baseline PANSS Total Score (Mean) | Change for Lumateperone 42 mg (Mean) | Change for Placebo (Mean) | LSMD vs. Placebo (Points) | p-value | Key Secondary Endpoint (CGI-S Change from Baseline at Week 4) | Baseline CGI-S (Mean) | CGI-S Change Lumateperone 42 mg (Mean) | CGI-S Change Placebo (Mean) | LSMD vs. Placebo (Points) | p-value |
---|---|---|---|---|---|---|---|---|---|---|---|---|
Study 1 (NCT01499563) | Change in PANSS Total Score | LMT: 88.1 <br> PLB: 86.3 | -13.2 | -7.4 | -5.8 | Significant* | Not specified as key secondary in snippet | - | - | - | - | - |
Study 2 (NCT02282761) | Change in PANSS Total Score | LMT: 90.0 <br> PLB: 89.0 | Not directly stated, but LSMD given | Not directly stated | -4.2 | p<0.05 | Change in CGI-S Score | LMT: 4.8 <br> PLB: 4.8 | -0.8 | -0.5 | -0.3 | p<0.05 |
*LMT: Lumateperone; PLB: Placebo. p-value for Study 1 primary endpoint implied by superiority but not explicitly stated in.[16]
Lumateperone, at a dose of 42 mg once daily, is FDA-approved for the treatment of depressive episodes associated with bipolar I or bipolar II disorder in adults. It can be used as monotherapy or as adjunctive therapy with lithium or valproate.[1] This dual approval for both bipolar I and II depression is a notable feature, as lumateperone is one of only four treatments with this broad indication from the FDA.[2]
Monotherapy Trial (Study 404 / NCT03249376):
This was a Phase 3, randomized, double-blind, placebo-controlled outpatient trial conducted over 6 weeks in patients with a major depressive episode (MDE) associated with bipolar I or bipolar II disorder.19
Adjunctive Therapy Trial (with lithium or valproate):
This randomized, placebo-controlled clinical trial evaluated lumateperone over 6 weeks as adjunctive therapy in patients with bipolar I or bipolar II disorder experiencing an MDE who had an inadequate therapeutic response to lithium or valproate alone.20
The consistent efficacy of the 42 mg dose in both monotherapy and adjunctive settings for bipolar depression, similar to its efficacy in schizophrenia, reinforces this as the established therapeutic dose across its approved indications.[19]
Table 5: Summary of Efficacy in Pivotal Bipolar Depression Trials (Lumateperone 42 mg vs. Placebo)
Trial Setting / Identifier | Primary Endpoint (MADRS Total Score Change from Baseline at Week 6) | LSMD vs. Placebo (Points) | p-value | Key Secondary Endpoint (CGI-BP-S Change from Baseline at Week 6) | LSMD vs. Placebo (Points) | p-value |
---|---|---|---|---|---|---|
Monotherapy (Study 404 / NCT03249376) | (CGI-BP-S Total Score) | |||||
- With Mixed Features | Change in MADRS Total Score | -4.4 | <.01 | Change in CGI-BP-S Total Score | -0.7 | <.05 |
- Without Mixed Features | Change in MADRS Total Score | -4.2 | <.001 | Change in CGI-BP-S Total Score | -1.0 | <.001 |
Adjunctive Therapy (to Lithium/Valproate) | Change in MADRS Total Score | -2.4 | 0.02 | (CGI-BP-S Depression Subscore) | -0.3 | 0.01 |
MADRS: Montgomery-Åsberg Depression Rating Scale; CGI-BP-S: Clinical Global Impression Scale-Bipolar Version-Severity; LSMD: Least Squares Mean Difference.
Lumateperone is generally well-tolerated across its approved indications for schizophrenia and bipolar depression.[1] A key feature of its safety profile is the low incidence of extrapyramidal symptoms (EPS), including akathisia, and minimal effects on metabolic parameters (such as weight, glucose, and lipids) and prolactin levels, with rates often similar to those observed with placebo.[3] In clinical studies, no single adverse reaction leading to treatment discontinuation occurred at a rate greater than 2% in patients treated with Caplyta.[4]
The most frequently reported adverse events associated with lumateperone treatment are generally mild to moderate in severity.
Table 6: Common Treatment-Emergent Adverse Events (TEAEs) for Lumateperone 42 mg (Incidence ≥5% in any trial and >Placebo)
Adverse Event | Indication(s) | Lumateperone 42 mg Incidence (%) | Placebo Incidence (%) | Reference(s) |
---|---|---|---|---|
Somnolence/Sedation | Schizophrenia | 24 | 10 | 4 |
Bipolar Depression (Monotherapy) | 13 | 3 | 4 | |
Bipolar Depression (Adjunctive) | 13 | 3 | 4 | |
Dry Mouth | Schizophrenia | 6 | 2 | 4 |
Bipolar Depression (Monotherapy) | 5 | 1 | 4 | |
Bipolar Depression (Adjunctive) | 5 | 1 | 4 | |
Dizziness | Bipolar Depression (Monotherapy) | 8 | 4 | 4 |
Bipolar Depression (Adjunctive) | 11 | 2 | 4 | |
Nausea | Bipolar Depression (Monotherapy) | 8 | 3 | 4 |
Bipolar Depression (Adjunctive) | 9 | 4 | 4 | |
Headache | Schizophrenia (Open-label study) | 5.3 | N/A (Open-label) | 5 |
Lumateperone, like other antipsychotic medications, carries several warnings and precautions that clinicians should be aware of.[4]
While these are standard class warnings for antipsychotics, the collective clinical trial data for lumateperone generally indicate a lower actual incidence of several of these concerns, such as TD, significant metabolic changes, and EPS, when compared to what might be anticipated from older antipsychotics or even some other atypical agents. This distinction reinforces the "favorable adverse effect profile" often attributed to lumateperone and is a critical consideration in clinical decision-making, especially for long-term treatment.
Atypical antipsychotic agents have been associated with metabolic changes, including hyperglycemia, diabetes mellitus, dyslipidemia, and weight gain.[4] However, lumateperone has demonstrated a generally minimal impact on these metabolic parameters, with effects often similar to placebo in clinical trials.[3]
Lumateperone's relatively neutral metabolic profile represents a significant clinical advantage. Metabolic syndrome is a major concern with many other atypical antipsychotics, profoundly impacting patients' long-term physical health and adherence to treatment. The differentiation of lumateperone in this regard, as highlighted in multiple sources [3], makes it a more attractive option for individuals at risk for, or concerned about, these metabolic complications.
Table 7: Metabolic Parameters - Changes from Baseline (Lumateperone 42 mg vs. Placebo, representative data)
(Specific pooled mean change data for all parameters across all indications were not consistently available in the provided snippets for a comprehensive comparative table. Clinical trial publications should be consulted for detailed breakdowns. The general finding is similarity to placebo for most metabolic parameters.4)
Parameter | Indication(s) | Lumateperone 42 mg Change from Baseline | Placebo Change from Baseline | Note | Reference(s) |
---|---|---|---|---|---|
Weight | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Generally low incidence of clinically significant weight gain. | 4 |
Fasting Glucose | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Monitoring recommended. | 4 |
Total Cholesterol | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Monitoring recommended. Increase noted post-discontinuation in one study.5 | 4 |
LDL Cholesterol | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Monitoring recommended. Increase noted post-discontinuation in one study.5 | 4 |
HDL Cholesterol | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Monitoring recommended. | 4 |
Triglycerides | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Monitoring recommended. | 4 |
Prolactin | Schiz, Bipolar Dep | Minimal / Similar to Placebo | Minimal | Low risk of hyperprolactinemia. Increase noted post-discontinuation in one study.5 | 4 |
Schiz: Schizophrenia; Bipolar Dep: Bipolar Depression.
Lumateperone is associated with a low incidence of EPS-related TEAEs, with rates generally similar to placebo and notably lower than some active comparators like risperidone in short-term studies.[3] This favorable EPS profile is a cornerstone of its clinical advantage.
The consistently low rates of EPS, comparable to placebo and significantly lower than an active comparator like risperidone, strongly support the pharmacological rationale behind lumateperone's design, particularly its D<sub>2</sub> receptor selectivity for mesolimbic/mesocortical pathways and its potent 5-HT<sub>2A</sub> antagonism.[10] This profile is a key differentiator that can improve patient experience and adherence to long-term treatment.
Data from clinical trials in both bipolar depression and schizophrenia indicate that lumateperone has a low incidence of sexual side effects. Rates of adverse reactions such as erectile dysfunction, decreased libido, and abnormal orgasm were generally similar to or lower than those observed with placebo.[4] For example, in schizophrenia trials, erectile dysfunction was reported in 0.7% of lumateperone-treated patients versus 0.5% for placebo, with no reports of decreased libido for either group.[4] In bipolar depression monotherapy trials, erectile dysfunction was 0.0% for lumateperone vs 0.7% for placebo, and decreased libido was 1.1% vs 0.3%.[4] These low rates of sexual dysfunction are an important aspect of overall tolerability, positively impacting quality of life and potentially improving adherence to long-term antipsychotic therapy.
The recommended dosage for lumateperone (Caplyta) is straightforward for its approved indications in adults:
Lumateperone is extensively metabolized, primarily by CYP3A4 and UGT enzymes, making it susceptible to interactions with drugs that modulate these pathways.[7] Careful medication review is essential.
The significant impact of CYP3A4 modulators on lumateperone exposure underscores the critical importance of thorough medication reconciliation and appropriate dose adjustments to prevent toxicity or loss of efficacy. While the "titration-free" nature of the 42 mg starting dose offers convenience, this must be balanced against the need for vigilance regarding potential interactions and patient-specific factors like hepatic function.
Table 8: Lumateperone Dosage Adjustments for Drug Interactions and Special Populations
Condition / Interacting Drug Class | Specific Examples (if applicable) | Recommended Lumateperone Dosage | Reference(s) |
---|---|---|---|
Drug Interactions | |||
Strong CYP3A4 Inhibitors | Itraconazole, ketoconazole, clarithromycin | 10.5 mg once daily | 13 |
Moderate CYP3A4 Inhibitors | Diltiazem, erythromycin, fluconazole, verapamil | 21 mg once daily | 13 |
Strong/Moderate CYP3A4 Inducers | Rifampin, carbamazepine, phenytoin, St. John’s wort, efavirenz | Avoid concomitant use | 5 |
UGT Inhibitors | Avoid concomitant use | 5 | |
Special Populations | |||
Mild Hepatic Impairment | Child-Pugh Class A | No adjustment (42 mg once daily) | 13 |
Moderate Hepatic Impairment | Child-Pugh Class B | 21 mg once daily | 13 |
Severe Hepatic Impairment | Child-Pugh Class C | 21 mg once daily | 13 |
Renal Impairment | All severities | No adjustment (42 mg once daily) | 13 |
Lumateperone, marketed as Caplyta®, was developed by Intra-Cellular Therapies, Inc..[1] It has received the following approvals from the U.S. Food and Drug Administration (FDA):
Potential Future Indications:
Intra-Cellular Therapies submitted a supplemental New Drug Application (sNDA) to the FDA for Caplyta® as an adjunctive treatment for adults with Major Depressive Disorder (MDD). This submission was announced in December 2024 18, and the FDA accepted the sNDA for review in February 2025.28
As of the latest available information from the provided sources, lumateperone's formal marketing authorization is primarily established in the United States.[1] There is no conclusive evidence from these documents indicating that lumateperone has received marketing authorization from the European Medicines Agency (EMA), or in other major jurisdictions such as Japan or Canada.[1] However, clinical trials, including those for bipolar depression, have been conducted at international sites.[1] This international trial presence suggests a potential strategy for future regulatory submissions in these regions, although a staggered approach, prioritizing the U.S. market first, is common.
Lumateperone (Caplyta®) was originally developed by Intra-Cellular Therapies, Inc..1
A significant development in its commercialization occurred with the acquisition of Intra-Cellular Therapies, Inc. by Johnson & Johnson. This transaction was completed on April 2, 2025, with Intra-Cellular Therapies subsequently operating as a business unit within Johnson & Johnson Innovative Medicine.18
This acquisition is viewed as a strategic move for Johnson & Johnson, expected to be a near- and long-term growth catalyst by strengthening its neuroscience portfolio.25 The transaction is anticipated to accelerate Johnson & Johnson's sales growth in 2025.28 The backing by a major pharmaceutical company like Johnson & Johnson signifies strong confidence in lumateperone's clinical value and market potential. This could lead to expanded research programs, development into further indications, and broader global commercialization efforts, leveraging Johnson & Johnson's extensive resources and global reach.
Lumateperone (Caplyta®) has emerged as a significant advancement in the pharmacotherapy of schizophrenia and bipolar depression. Its key distinguishing features include:
Lumateperone represents a valuable treatment option within the current psychopharmacological armamentarium:
The favorable side effect profile of lumateperone aligns with the broader goals of patient-centered care in psychiatry, aiming not only for symptom control but also for improved quality of life and long-term treatment adherence by minimizing the burden of adverse effects.
The development trajectory of lumateperone suggests several promising future directions:
In conclusion, lumateperone represents a significant evolution in antipsychotic therapy, offering a unique pharmacological profile that translates into effective symptom management for schizophrenia and bipolar depression with an improved tolerability profile. Its ongoing development and the backing of a major pharmaceutical entity suggest a promising future for this agent in addressing the complex needs of individuals with serious mental illness.
Published at: June 9, 2025
This report is continuously updated as new research emerges.