MedPath

Darolutamide Advanced Drug Monograph

Published:Aug 2, 2025

Generic Name

Darolutamide

Brand Names

Nubeqa

Drug Type

Small Molecule

Chemical Formula

C19H19ClN6O2

CAS Number

1297538-32-9

Associated Conditions

Metastatic Hormone-Sensitive Prostate Cancer (mHSPC), Non-Metastatic Castration-Resistant Prostate Cancer

Darolutamide (Nubeqa®): A Comprehensive Monograph on Pharmacology, Clinical Efficacy, and Safety

I. Introduction and Executive Synopsis

Darolutamide, marketed under the brand name Nubeqa®, represents a significant therapeutic advancement in the management of advanced prostate cancer, establishing itself as a cornerstone therapy across multiple disease states.[1] As a third-generation, nonsteroidal androgen receptor inhibitor (ARi), also referred to as a nonsteroidal antiandrogen (NSAA), darolutamide has redefined treatment paradigms by offering a unique combination of potent, broad-spectrum antineoplastic activity and a distinctly favorable safety and tolerability profile.[2] This advantageous clinical profile is not a fortuitous outcome but is directly rooted in the drug's unique molecular structure, which was rationally designed to limit central nervous system (CNS) penetration and minimize the potential for clinically significant drug-drug interactions.[3]

The clinical and commercial success of darolutamide is built upon this compelling synergy. Its robust efficacy has been validated in a series of large, well-designed, pivotal Phase III trials, leading to global regulatory approvals for a broad spectrum of advanced prostate cancer indications. These approvals demonstrate its versatility and underscore its importance in the oncologist's armamentarium.[2] The currently approved indications for darolutamide include:

  • The treatment of adult men with non-metastatic castration-resistant prostate cancer (nmCRPC) who are at high risk of developing metastatic disease.[2]
  • The treatment of adult men with metastatic hormone-sensitive prostate cancer (mHSPC) in combination with androgen deprivation therapy (ADT).[2]
  • The treatment of adult men with mHSPC in combination with both ADT and the chemotherapeutic agent docetaxel.[2]

This comprehensive range of indications allows physicians to tailor treatment intensification strategies for patients across different stages of advanced disease. The drug was jointly developed through a collaboration between Bayer HealthCare Pharmaceuticals Inc. and Orion Corporation, a globally operating Finnish pharmaceutical company.[1] Reflecting its significant clinical impact and rapid adoption into practice, darolutamide achieved blockbuster commercial status in September 2024, with a projected peak sales potential exceeding €3 billion.[5] This monograph provides an exhaustive review of darolutamide's physicochemical properties, its preclinical and clinical pharmacology, the pivotal clinical trial data supporting its efficacy, its comprehensive safety profile, and its global regulatory and commercial trajectory.

II. Physicochemical Properties and Drug Identification

The foundational chemical and identification data for darolutamide are essential for understanding its pharmacological behavior. These properties, consolidated from numerous chemical and drug databases, provide a precise molecular fingerprint for the compound.

Chemical Identity and Physical Properties

Darolutamide is a specific, optically active stereoisomer, a critical feature that contributes to its targeted biological activity. Its International Union of Pure and Applied Chemistry (IUPAC) name is N-{(2S)-1-[3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl]propan-2-yl}-5-(1-hydroxyethyl)-1H-pyrazole-3-carboxamide.[3] The

(2S) designation confirms that it is a single enantiomer, not a racemic mixture of isomers. This stereospecificity is a deliberate aspect of modern rational drug design, intended to optimize binding affinity to the target receptor—in this case, the androgen receptor—while minimizing off-target interactions that could lead to undesirable side effects. This "safety-by-design" approach is likely a key contributor to darolutamide's high potency and its favorable clinical tolerability, particularly its low affinity for other receptors such as GABA-A, which are implicated in the CNS side effects seen with other ARis.[3]

The compound has the molecular formula C19​H19​ClN6​O2​ and a molecular weight of 398.85 g/mol.[3] Physically, it presents as a white to off-white solid powder.[12] Its solubility characteristics are also noteworthy; it is soluble in organic solvents like dimethyl sulfoxide (DMSO) but is not soluble in water.[12] This low aqueous solubility necessitates its formulation into oral tablets and underscores the clinical importance of administering the drug with food to enhance its absorption and bioavailability.

Drug Identifiers

A comprehensive list of identifiers is crucial for cross-referencing the compound across various scientific and regulatory databases. The following table consolidates these key identifiers.

Table 1: Darolutamide Identification and Physicochemical Properties

PropertyValueSource(s)
Drug NameDarolutamide1
Brand NameNubeqa®2
Drug TypeSmall Molecule15
Synonyms / CodesODM-201, BAY-18417881
IUPAC NameN-{(2S)-1-[3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl]propan-2-yl}-5-(1-hydroxyethyl)-1H-pyrazole-3-carboxamide3
Molecular FormulaC19​H19​ClN6​O2​3
Molecular Weight398.85 g/mol1
CAS Number1297538-32-91
DrugBank IDDB129411
PubChem CID671718673
UNIIX05U0N2RCO1
KEGG IDD110451
ChEMBL IDCHEMBL42971851
SMILESC[C@@H](Cn1ccc(n1)c2ccc(c(c2)Cl)C#N)NC(=O)c3cc([nH]n3)C(C)O3
AppearanceWhite to off-white solid powder12
SolubilitySoluble in DMSO; not soluble in water12

III. Preclinical and Clinical Pharmacology

The clinical utility of darolutamide is a direct result of its distinct pharmacological profile. This section dissects the drug's mechanism of action, its unique structural properties that confer a favorable safety profile, and its pharmacokinetic journey through the body, linking its molecular behavior to its observed clinical outcomes.

A. Mechanism of Action and Pharmacodynamics

Darolutamide functions as a selective, competitive, and potent "silent" antagonist of the androgen receptor (AR).[3] The term "silent" antagonist implies that it effectively blocks the receptor without exhibiting any partial agonist activity. It binds with high affinity to the ligand-binding domain of the AR, thereby preventing the natural androgens—testosterone and its more potent metabolite, dihydrotestosterone (DHT)—from activating the receptor.[17]

The antineoplastic activity of darolutamide is derived from a comprehensive, three-pronged inhibition of the AR signaling pathway, a critical driver of prostate cancer cell growth and survival [1]:

  1. Inhibition of Androgen Binding: It competitively blocks testosterone and DHT from binding to the AR.
  2. Prevention of Nuclear Translocation: It prevents the AR, once bound, from translocating from the cell's cytoplasm into the nucleus. This is a crucial step, as the AR must enter the nucleus to exert its effects on gene expression.
  3. Inhibition of AR-Mediated Transcription: By preventing nuclear translocation, it ultimately inhibits AR-mediated transcription, blocking the activation of a suite of androgen-responsive genes that regulate prostate cancer cell proliferation, survival, and PSA production.

In vitro assays have quantitatively demonstrated the high potency of darolutamide. It exhibits an inhibition constant (Ki​) of 11 nM and a half-maximal inhibitory concentration (IC50​) of 26 nM for AR antagonism.[12] This high potency translates into efficacious suppression of AR signaling at clinically achievable concentrations.

A particularly critical feature of darolutamide is its demonstrated activity against known AR mutations that can arise in response to other antiandrogen therapies and drive treatment resistance. Notably, it effectively blocks the activity of the clinically relevant F876L mutation, which is known to confer resistance to the second-generation ARis enzalutamide and apalutamide.[3] This suggests that darolutamide may retain activity in patients whose tumors have developed this specific resistance mechanism, potentially offering an advantage in later lines of therapy or in patients progressing on other ARis. Furthermore, the drug's antiproliferative effects are highly specific to AR-dependent prostate cancer cells. Studies have shown it has no effect on the viability of AR-negative cell lines, such as the DU-145 prostate cancer cell line, confirming that its anticancer activity is mediated specifically through the AR pathway.[12]

B. The Structural Basis for a Favorable CNS Profile

A key differentiator for darolutamide is its unique chemical structure, which is the foundation of its superior CNS safety profile. It possesses a flexible, polar-substituted pyrazole structure that is chemically distinct from the structures of other ARis like enzalutamide and apalutamide.[6]

This unique molecular architecture results in a very low potential to cross the blood-brain barrier (BBB). This property is not merely a theoretical claim; it is substantiated by a body of evidence from both preclinical animal models and human neuroimaging studies.[5] The clinical consequence of this low CNS penetration is profound: it is the primary reason for the markedly low incidence of CNS-related side effects, most notably seizures, which are a known class effect and a significant concern with other ARis. The mechanism is believed to involve a reduced risk of off-target inhibition of the GABAA receptor in the brain, a pathway that has been linked to the proconvulsant effects of other ARis.[3] This favorable CNS profile is a cornerstone of darolutamide's overall tolerability and a major advantage in clinical practice.

C. Pharmacokinetics (PK): Absorption, Distribution, Metabolism, and Excretion (ADME)

The journey of darolutamide through the body is characterized by several key pharmacokinetic properties that influence its dosing and clinical effects.

Absorption: Darolutamide is administered orally. In a fasted state, peak plasma concentrations (Tmax​) are reached within 3 to 5 hours. However, its absorption is significantly influenced by food. When administered with a meal, its bioavailability increases by a factor of 2.0 to 2.5.[15] This is a critical clinical consideration, as the absolute bioavailability is only approximately 30% after a single 300 mg dose taken while fasting.[15] This substantial food effect is the basis for the label instruction to always take Nubeqa with food to ensure adequate and consistent drug exposure.[8] Steady-state concentrations are typically achieved within 2 to 5 days of repeated twice-daily administration with food.[15]

Distribution: After absorption, darolutamide distributes throughout the body, with an apparent volume of distribution of approximately 119 L, indicating that it distributes into tissues beyond the plasma volume.[15] It is highly bound to plasma proteins, primarily albumin. The plasma protein binding is 92% for darolutamide and an even higher 99.8% for its major active metabolite, keto-darolutamide.[15]

Metabolism and the Role of Keto-Darolutamide: Darolutamide is primarily metabolized in the liver by the cytochrome P450 enzyme CYP3A4, with additional contributions from the UGT (uridine 5'-diphospho-glucuronosyltransferase) enzymes UGT1A9 and UGT1A1.[15] This metabolism leads to the formation of several metabolites, the most important of which is

keto-darolutamide.

The role of keto-darolutamide is a crucial aspect of the drug's overall pharmacology. This is not a minor or inactive byproduct; it is a major, pharmacologically active metabolite. Keto-darolutamide exhibits antiandrogenic activity that is very similar to the parent drug, with a reported Ki​ of 8 nM and an IC50​ of 38 nM.[3] Critically, at steady state, it is found in the plasma at approximately twice the concentration of darolutamide itself.[15] Therefore, the clinical efficacy observed with Nubeqa is not derived from a single agent but from the combined, sustained action of two potent ARis: the parent drug and its more abundant active metabolite. This "dual-agent" action likely contributes to the profound and durable AR blockade observed in clinical trials, providing a more robust and persistent suppression of the AR pathway than the parent drug could achieve alone.

Excretion: Darolutamide and its metabolites are eliminated from the body through both renal and fecal routes. Following a radiolabeled dose, approximately 63.4% of the material was recovered in the urine (with 7% as unchanged drug) and 32.4% was recovered in the feces.[15] The elimination half-life for both darolutamide and keto-darolutamide is approximately 20 hours, a pharmacokinetic property that supports the convenience of a twice-daily dosing regimen.[15]

The pharmacokinetic properties of darolutamide and its active metabolite are summarized in the table below.

Table 2: Summary of Darolutamide and Keto-Darolutamide Pharmacokinetic Parameters

ParameterDarolutamideKeto-DarolutamideSource(s)
Time to Peak (Tmax)3-8 hours (fed)~3-6 hours15
Bioavailability~30% (fasting); increases 2-2.5x with foodN/A (metabolite)15
Plasma Protein Binding92% (to albumin)99.8% (to albumin)15
Elimination Half-life (t1/2​)~20 hours~20 hours15
Primary Metabolizing EnzymeCYP3A4, UGT1A9, UGT1A1N/A15
Relative Exposure (AUC)Lower~2x higher than Darolutamide15
Relative Potency (Ki​)11 nM8 nM3

The pharmacokinetic profile also reveals a favorable drug-drug interaction (DDI) profile, which appears to be another outcome of its rational design. At clinically relevant concentrations, darolutamide does not significantly inhibit or induce most major CYP enzymes, with only a mild induction of CYP3A4 noted.[3] This contrasts with other ARis that have more complex DDI profiles. The primary DDI risks for darolutamide are as a

victim of strong CYP3A4 inducers (e.g., rifampicin) or as a perpetrator via inhibition of the BCRP transporter (e.g., with rosuvastatin).[3] However, its overall DDI liability is considered low. This is a major clinical advantage in the target population of elderly men with prostate cancer, who frequently present with multiple comorbidities and are on numerous concomitant medications (polypharmacy).[6] A physician can prescribe darolutamide with greater confidence that it will not disrupt a patient's existing medication regimen, thereby simplifying management and enhancing overall safety.

IV. Clinical Efficacy in Non-Metastatic Castration-Resistant Prostate Cancer (nmCRPC): The ARAMIS Trial

The ARAMIS trial was the first pivotal study to establish the clinical efficacy and safety of darolutamide, leading to its initial FDA approval and setting a new standard of care in high-risk non-metastatic castration-resistant prostate cancer (nmCRPC).

A. Trial Design and Patient Population

The ARAMIS (Androgen Receptor Antagonizing Agent for Metastasis-free Survival) trial was a large, multinational, randomized, double-blind, placebo-controlled, Phase III study.[23] The trial enrolled 1,509 men with nmCRPC who were deemed to be at high risk of developing metastases. This high-risk status was defined by a prostate-specific antigen (PSA) doubling time (PSADT) of 10 months or less, coupled with a baseline PSA level of 2 ng/mL or higher.[10] All patients enrolled were required to continue on a gonadotropin-releasing hormone (GnRH) analog or have had a prior bilateral orchiectomy to maintain castrate levels of testosterone.[10] Notably, the trial design permitted the enrollment of patients with a prior history of seizures, a reflection of the confidence in darolutamide's favorable CNS safety profile.[11]

Patients were randomized in a 2:1 ratio to receive either darolutamide at a dose of 600 mg administered orally twice daily (n=955) or a matching placebo (n=554).[4]

B. Efficacy Outcomes: Meeting Primary and Key Secondary Endpoints

The ARAMIS trial successfully met both its primary and key secondary endpoints with a high degree of statistical significance, demonstrating a profound clinical benefit for darolutamide.

Primary Endpoint: Metastasis-Free Survival (MFS): The primary endpoint of the trial was MFS, defined as the time from randomization to the first evidence of distant metastasis or death from any cause.[26] Darolutamide demonstrated a dramatic improvement in MFS, with a median MFS of

40.4 months for the darolutamide group compared to just 18.4 months for the placebo group.[10] This represents a clinically meaningful delay of 22 months in the onset of metastatic disease. This outcome corresponded to a

59% reduction in the risk of metastasis or death, with a Hazard Ratio (HR) of 0.41 (95% Confidence Interval [CI]: 0.34-0.50; p<0.0001).[27]

Key Secondary Endpoint: Overall Survival (OS): The final analysis of overall survival, a critical secondary endpoint, confirmed that the benefit in MFS translated into a significant advantage in longevity. Treatment with darolutamide resulted in a 31% reduction in the risk of death compared to placebo (HR 0.69; 95% CI: 0.53-0.88; p=0.003).[23] At the 3-year mark, the survival rate was 83% for patients in the darolutamide arm versus 77% for those in the placebo arm.[29] This survival benefit is particularly impressive given that a substantial number of patients in the placebo group (170 patients, or 31%) were allowed to cross over and receive open-label darolutamide after the primary analysis unblinding, which would tend to dilute the observed survival difference.[27]

Other Clinically Relevant Secondary Endpoints: The benefits of darolutamide extended to other important measures of disease progression and patient well-being. Treatment with darolutamide significantly delayed the time to:

  • Pain Progression: Median of 40.3 months versus 25.4 months for placebo (HR 0.65; p<0.001).[27]
  • First Use of Cytotoxic Chemotherapy: Significantly delayed (HR 0.58; p<0.001).[27]
  • First Symptomatic Skeletal Event (SSE): Significantly delayed (HR 0.48; p=0.005).[27]

C. Quality of Life and Subgroup Analyses

A crucial aspect of treating an often asymptomatic patient population is the ability to control the disease without negatively impacting quality of life. Darolutamide excelled in this regard. Analyses of health-related quality of life (HRQoL) data from ARAMIS showed that treatment with darolutamide maintained HRQoL by significantly delaying the time to deterioration of prostate cancer-specific symptoms, including urinary and bowel issues, as well as preserving overall well-being compared to placebo.[26]

The tolerability profile was exceptionally favorable and was a standout feature of the trial. Discontinuation rates due to treatment-emergent adverse events (TEAEs) were nearly identical between the darolutamide and placebo groups (8.9% vs. 8.7%, respectively), indicating that the addition of darolutamide to ADT did not add a significant toxicity burden.[4]

Furthermore, the profound efficacy benefits in both MFS and OS were consistent across all predefined patient subgroups. This included patients with a high burden of comorbidities (more than six) and those taking numerous concomitant medications (more than ten), as well as patients regardless of the type of prior local therapy they had received (e.g., radiotherapy or radical prostatectomy).[23] This consistency reinforces the robustness of the findings and their broad applicability to the real-world nmCRPC population.

The results of the ARAMIS trial provided landmark evidence that a substantial improvement in the surrogate endpoint of MFS could directly translate into a significant and clinically meaningful overall survival benefit. The initial FDA approval for darolutamide in this setting was based on the MFS endpoint, a pragmatic regulatory decision that was debated at the time.[28] The final OS analysis from ARAMIS definitively answered the critical question of whether delaying the radiological detection of metastases would actually help patients live longer.[27] The 31% reduction in the risk of death, achieved despite significant crossover, cemented the clinical value of early intervention in high-risk nmCRPC and validated the FDA's use of MFS as an approvable endpoint in this disease space. This result solidified a paradigm shift in the management of nmCRPC, moving the field away from "watchful waiting" and toward active, early treatment in high-risk patients to delay progression and prolong life.

V. Clinical Efficacy in Metastatic Hormone-Sensitive Prostate Cancer (mHSPC): The ARASENS and ARANOTE Trials

Following its success in nmCRPC, the clinical development program for darolutamide strategically expanded into the metastatic hormone-sensitive prostate cancer (mHSPC) setting. This was accomplished through two major, complementary Phase III trials—ARASENS and ARANOTE—which were designed to establish darolutamide's role across the full spectrum of mHSPC treatment intensities. The following table provides a high-level comparison of these pivotal trials.

Table 3: Comparative Overview of Pivotal Phase III Trials (ARAMIS, ARASENS, ARANOTE)

FeatureARAMISARASENSARANOTE
Trial NameAndrogen Receptor Antagonizing Agent for Metastasis-free SurvivalAndrogen Receptor Antagonist for Metastatic Hormone-Sensitive Prostate CancerN/A
Patient PopulationHigh-Risk nmCRPCmHSPC (Chemo-eligible)mHSPC
N1,5091,306669
Treatment ArmsDarolutamide + ADT vs. Placebo + ADTDarolutamide + ADT + Docetaxel vs. Placebo + ADT + DocetaxelDarolutamide + ADT vs. Placebo + ADT
Primary EndpointMetastasis-Free Survival (MFS)Overall Survival (OS)Radiographic Progression-Free Survival (rPFS)
Primary Endpoint ResultMedian: 40.4 vs. 18.4 mos HR: 0.41 (95% CI: 0.34-0.50) p<0.0001Median: Not Reached vs. 48.9 mos HR: 0.68 (95% CI: 0.57-0.80) p<0.001Median: Not Reached vs. 25 mos HR: 0.54 (95% CI: 0.41-0.71) p<0.0001
Source(s)24335

A. The ARASENS Trial: Establishing Triplet Therapy (Darolutamide + ADT + Docetaxel)

The ARASENS trial was an international, randomized (1:1), double-blind, placebo-controlled, Phase III study that was prospectively designed to test the efficacy and safety of a "triplet" therapy regimen.[33] The trial enrolled 1,306 men with mHSPC who were considered candidates for both ADT and docetaxel chemotherapy. A high proportion of these patients (86.1%) had de novo metastatic disease, meaning their cancer had already spread at the time of initial diagnosis.[38]

Patients were randomized to receive either darolutamide (600 mg twice daily) in addition to standard of care ADT and docetaxel (for 6 cycles), or placebo plus ADT and docetaxel.[40]

The primary endpoint of the ARASENS trial was Overall Survival (OS). The trial met this high bar, demonstrating a statistically significant and clinically meaningful survival advantage for the triplet regimen. The addition of darolutamide to ADT and docetaxel resulted in a 32.5% reduction in the risk of death compared to the control arm (HR 0.68; 95% CI: 0.57-0.80; p<0.001).[20] This landmark result established the darolutamide-based triplet therapy as a new standard of care for eligible patients with mHSPC, providing a more effective option for treatment intensification.[35]

The robustness of this survival benefit was underscored by its consistency across all prespecified patient subgroups, including those with high-volume or low-volume disease.[39] Furthermore, the benefit was achieved despite a high rate (76%) of patients in the placebo arm receiving subsequent life-prolonging therapies, including other ARis, upon disease progression.[35]

B. The ARANOTE Trial: Validating Doublet Therapy (Darolutamide + ADT)

The ARANOTE trial was a global, randomized (2:1), double-blind, placebo-controlled, Phase III study designed to evaluate darolutamide in a less intensive, chemo-free regimen.[5] The trial enrolled 669 men with mHSPC, who were randomized to receive either darolutamide (600 mg twice daily) plus ADT or placebo plus ADT.[5] This trial was designed to address the large population of mHSPC patients who are not candidates for, or who wish to avoid the toxicities of, upfront chemotherapy.

The primary endpoint of the ARANOTE trial was Radiographic Progression-Free Survival (rPFS). The trial successfully met this endpoint, demonstrating that the darolutamide doublet significantly delayed disease progression. Treatment with darolutamide plus ADT resulted in a 46% reduction in the risk of radiographic progression or death compared to ADT alone (HR 0.54; 95% CI: 0.41-0.71; p<0.0001).[5]

While the OS data from ARANOTE showed a positive trend favoring darolutamide, the difference was not statistically significant at the time of the final analysis (HR ~0.8).[6] However, all other secondary endpoints, such as time to progression to castration-resistant prostate cancer (mCRPC) (HR 0.40) and time to pain progression, showed clear and significant clinical benefits for the darolutamide arm.[6]

The design and execution of the ARASENS and ARANOTE trials represent a highly effective and complementary clinical development strategy. This dual-pronged approach was designed to establish darolutamide as a versatile agent across the entire mHSPC landscape. ARASENS targeted the most intensive treatment setting, proving superiority as a triplet therapy for fit patients where a survival benefit is the highest clinical bar.[34] Success in this trial cemented its role as a premier agent for treatment intensification. Concurrently, ARANOTE addressed the needs of the broader mHSPC population, including those for whom chemotherapy is not suitable.[6] By demonstrating a strong rPFS benefit as a doublet, it secured a critical role as a potent, well-tolerated, chemotherapy-free option. This comprehensive evidence base provides physicians with maximum flexibility, allowing them to use the ARASENS data to justify triplet therapy for fit patients and the ARANOTE data to justify doublet therapy for others. This strategic coverage gives Nubeqa a significant competitive advantage and solidifies its position as a go-to ARi in mHSPC.

VI. Comprehensive Safety and Tolerability Profile

A central component of darolutamide's value proposition is its exceptional safety and tolerability profile, which has been consistently demonstrated across its pivotal clinical trials. This section synthesizes safety data from the ARAMIS, ARASENS, and ARANOTE trials to provide a holistic view of the drug's risk profile.

A. Adverse Reactions and Laboratory Abnormalities

The overall safety profile of darolutamide is characterized by excellent tolerability, with an adverse event (AE) profile that is often comparable to placebo, particularly with respect to CNS effects and fatigue.[4]

Profile with ADT (from ARAMIS & ARANOTE): When used as a doublet with ADT, darolutamide adds minimal toxicity. The most common AEs occurring at a rate of at least 2% over placebo were:

  • Fatigue or asthenic conditions (16% vs. 11% for placebo).[2]
  • Pain in extremity (6% vs. 3% for placebo).[2]
  • Rash (3-4% vs. 1% for placebo).[2]
  • Common laboratory abnormalities included increased AST, decreased neutrophil count, and increased bilirubin.[2]

Profile with ADT + Docetaxel (from ARASENS): When added to the more toxic backbone of ADT plus docetaxel, the most common AEs (≥10% with a ≥2% increase over the control arm) included:

  • Constipation (23%).[2]
  • Rash (20%).[2]
  • Decreased appetite (19%).[2]
  • Hemorrhage (18%).[2]
  • Increased weight (18%).[2]
  • Hypertension (14%).[2]
  • Common laboratory abnormalities (≥30%) in this setting included anemia, hyperglycemia, decreased lymphocyte and neutrophil counts, increased AST/ALT, and hypocalcemia.[2]

The results from ARASENS demonstrated a "tolerability dividend": the favorable intrinsic safety profile of darolutamide allowed it to be added to an intensive chemotherapy regimen to unlock a significant survival benefit without a corresponding and prohibitive penalty in safety and tolerability. The overall incidence of TEAEs and serious AEs was broadly similar between the triplet and doublet-plus-placebo arms.[35]

Warnings and Precautions in Detail:

  • Ischemic Heart Disease: An increased incidence of ischemic heart disease has been observed. Across trials, it occurred in approximately 4% of patients receiving darolutamide compared to approximately 2-3% in control arms.[9]
  • Seizure: This is a key differentiating safety feature. The incidence of seizures with darolutamide is very low, a direct result of its limited BBB penetration. While early trials observed no seizures [3], a pooled analysis of the larger trials shows an incidence of just 0.2% with darolutamide monotherapy (plus ADT) and 0.8% when combined with docetaxel.[18] This represents a significant safety advantage over other ARis.
  • Embryo-Fetal Toxicity: As an antiandrogen, darolutamide can cause fetal harm and is an expected teratogen. It is contraindicated for use in women who are or may become pregnant. Male patients with female partners of reproductive potential must be advised to use effective contraception during treatment and for one week after the final dose.[2]

The following table compares the incidence of key adverse events across the pivotal trials.

Table 4: Incidence (%) of Key Adverse Events in Pivotal Phase III Trials

Adverse EventARAMIS (nmCRPC)ARANOTE (mHSPC)ARASENS (mHSPC)
Daro + ADT (N=955)Placebo + ADT (N=554)Daro + ADT (N=446)
Fatigue16%11%5.6%
Rash3%1%N/A
HypertensionN/AN/AN/A
Ischemic Heart Disease4.0%3.4%N/A
Heart Failure2.1%0.9%N/A
Seizure0.2% (pooled)N/A0.2% (pooled)
Neutropenia (Any Grade)20%9%N/A
AST Increased (Any Grade)23%N/A (vs placebo)N/A
ALT Increased (Any Grade)N/AN/AN/A
Note: Data compiled from multiple sources.2 "N/A" indicates data not specifically reported in the available sources in a comparable format.

B. Drug-Drug Interactions (DDIs)

The DDI profile of darolutamide is generally favorable but requires attention to specific interactions.

Darolutamide as a Victim:

  • Darolutamide is a substrate of the enzyme CYP3A4 and the transporter P-glycoprotein (P-gp).[9]
  • Concomitant use with strong inducers of CYP3A4 and P-gp (e.g., rifampicin, carbamazepine, phenobarbital, St. John's Wort) is not recommended. These agents can significantly decrease darolutamide plasma concentrations by over 70%, which could compromise its efficacy.[9]
  • Concomitant use with drugs that are combined strong inhibitors of both CYP3A4 and P-gp (e.g., itraconazole) can increase darolutamide exposure by approximately 1.7-fold. In such cases, patients should be monitored more frequently for adverse reactions.[9]

Darolutamide as a Perpetrator:

  • Darolutamide is a clinically relevant inhibitor of the transporter proteins BCRP (Breast Cancer Resistance Protein) and OATP1B1/OATP1B3 (Organic Anion Transporting Polypeptides).[3]
  • This inhibition can significantly increase the exposure of drugs that are substrates of these transporters. A key example is rosuvastatin (a BCRP substrate), whose exposure can be increased by approximately 5-fold when co-administered with darolutamide. This combination should be avoided due to the increased risk of myopathy.[3]
  • Darolutamide is only a mild inducer of CYP3A4, and this effect is not expected to be clinically relevant for most co-administered CYP3A4 substrates.[9]

C. Contraindications and Use in Special Populations

Contraindications:

  • Known hypersensitivity to darolutamide or any of its excipients.[9]
  • Use in women who are or may become pregnant.[9]

Use in Special Populations: A dose reduction to 300 mg twice daily is recommended for patients with:

  • Severe renal impairment (eGFR < 30 mL/min/1.73 m², not on hemodialysis).[3]
  • Moderate hepatic impairment (Child-Pugh Class B).[3]
  • Darolutamide has not been studied in patients with severe hepatic impairment (Child-Pugh Class C).[9]

VII. Regulatory and Commercial Trajectory

Darolutamide's journey from a promising clinical candidate to a global blockbuster therapy has been marked by a rapid and successful regulatory and commercial trajectory, reflecting the strength of its clinical data and its significant unmet need.

A. Global Regulatory Approvals Timeline

Developed jointly by Bayer and Orion Corporation, darolutamide received expedited reviews from major regulatory agencies, facilitating its swift entry into the market.

United States (Food and Drug Administration - FDA):

The FDA recognized the potential of darolutamide by granting it Fast Track and Priority Review designations.11

  • July 30, 2019: The first FDA approval was granted for the treatment of patients with high-risk nmCRPC, based on the pivotal results of the ARAMIS trial.[1]
  • August 5, 2022: The indication was expanded to include the treatment of mHSPC in combination with docetaxel, following the positive OS data from the ARASENS trial.[2]
  • June 3, 2025: A third indication was approved for the treatment of mHSPC as a doublet therapy with ADT, based on the rPFS benefit shown in the ARANOTE trial. This review was conducted under the FDA's Project Orbis, an initiative for concurrent submission and review of oncology drugs among international partners, including agencies in Australia, Canada, Switzerland, and the UK.[2]

European Union (European Medicines Agency - EMA):

  • March 27, 2020: The first marketing authorization was granted for the treatment of high-risk nmCRPC.[22]
  • March 1, 2023: The approval was expanded to include mHSPC in combination with ADT and docetaxel.[7]
  • July 21, 2025: The EMA granted approval for the third indication, for use in mHSPC in combination with ADT alone.[21]

Other Regions: Beyond the US and EU, darolutamide is approved in over 85 countries worldwide, including other major markets such as Japan and China, for its various indications.[7]

B. Dosing, Administration, and Commercialization

  • Brand Name: The drug is marketed globally under the brand name Nubeqa®.[1]
  • Dosage Form: It is supplied as 300 mg film-coated tablets.[9]
  • Recommended Dosage: The standard recommended dose is 600 mg (administered as two 300 mg tablets) taken orally twice daily, for a total daily dose of 1200 mg. It is crucial that the tablets are taken with food to ensure optimal absorption.[3]
  • Commercial Performance: Nubeqa® has seen rapid uptake in clinical practice. It officially achieved blockbuster status in September 2024. Its developer, Bayer, forecasts that the drug's peak sales potential will exceed €3 billion, reflecting its strong clinical profile and broad applicability in the treatment of advanced prostate cancer.[5]

VIII. Synthesis and Concluding Analysis

Darolutamide's development and clinical success serve as a compelling case study in modern, rational drug design and strategic clinical development. Its structurally distinct nature, deliberately engineered for high potency and low CNS penetration, translates directly into a unique and highly advantageous pharmacological profile. This profile is characterized by a potent, dual-agent (parent drug plus active metabolite) blockade of the androgen receptor pathway, which drives its profound antineoplastic efficacy.

This pharmacological foundation underpins its exceptional performance in the clinic. Across the three pivotal Phase III trials—ARAMIS, ARASENS, and ARANOTE—darolutamide has consistently demonstrated statistically significant and clinically meaningful efficacy. It has been proven to improve metastasis-free survival, radiographic progression-free survival, and, most importantly, overall survival across a wide spectrum of advanced prostate cancer settings, from high-risk non-metastatic disease to metastatic, hormone-sensitive disease.

Crucially, this profound efficacy is delivered with what can be described as a "tolerability dividend." Darolutamide possesses a safety profile so favorable that it is often comparable to placebo, particularly with regard to burdensome side effects like fatigue and CNS events. This allows for the full realization of its efficacy by promoting better treatment adherence and, critically, preserving patient quality of life—a primary goal of therapy, especially in asymptomatic or minimally symptomatic populations. This tolerability also enables treatment intensification, as shown in the ARASENS trial, where it could be added to a toxic chemotherapy regimen to unlock a major survival benefit without a prohibitive cost in safety.

In the competitive landscape of androgen receptor inhibitors, where efficacy is often similar, darolutamide's safety profile is its primary strategic differentiator. This makes it an especially attractive option for the typical prostate cancer patient: an elderly man, often with multiple comorbidities and on numerous concomitant medications. The comprehensive evidence base from its three major trials provides physicians with the flexibility and confidence to use it as a doublet or triplet therapy in mHSPC, or as a highly effective and well-tolerated monotherapy (with ADT) in nmCRPC.

In conclusion, through a combination of intelligent drug design and a robust, strategically executed clinical development program, darolutamide (Nubeqa®) has firmly established itself as a foundational agent and a global standard of care in the management of advanced prostate cancer. It offers a best-in-class combination of efficacy and safety that directly addresses the core clinical goals of prolonging survival while protecting and maintaining the patient's quality of life.

Works cited

  1. Darolutamide | C19H19ClN6O2 | CID 67171867 - PubChem, accessed August 2, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/67171867
  2. Nubeqa (darolutamide) FDA Approval History - Drugs.com, accessed August 2, 2025, https://www.drugs.com/history/nubeqa.html
  3. Darolutamide - Wikipedia, accessed August 2, 2025, https://en.wikipedia.org/wiki/Darolutamide
  4. Extended Safety and Tolerability of Darolutamide for Nonmetastatic ..., accessed August 2, 2025, https://academic.oup.com/oncolo/article/29/7/581/7613339
  5. U.S. FDA approves third indication of Bayer blockbuster Nubeqa™ (darolutamide) for patients with advanced prostate cancer, accessed August 2, 2025, https://www.bayer.com/media/en-us/us-fda-approves-third-indication-of-bayer-blockbuster-nubeqa-darolutamide-for-patients-with-advanced-prostate-cancer/
  6. Darolutamide in Combination With Androgen-Deprivation Therapy in Patients With Metastatic Hormone-Sensitive Prostate Cancer From the Phase III ARANOTE Trial | Journal of Clinical Oncology - ASCO Publications, accessed August 2, 2025, https://ascopubs.org/doi/10.1200/JCO-24-01798
  7. Nubeqa™ (darolutamide) receives EU approval for additional indication in prostate cancer, accessed August 2, 2025, https://www.bayer.com/media/en-us/nubeqa-darolutamide-receives-eu-approval-for-additional-indication-in-prostate-cancer/
  8. Reference ID: 5602092 - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2025/212099s008lbl.pdf
  9. Nubeqa, INN-darolutamide - EMA, accessed August 2, 2025, https://www.ema.europa.eu/en/documents/product-information/nubeqa-epar-product-information_en.pdf
  10. FDA Oncology Update - PMC - PubMed Central, accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6996617/
  11. Darolutamide Receives FDA Approval for the Treatment of Non-metastatic Castration-resistant Prostate Cancer - UroToday, accessed August 2, 2025, https://www.urotoday.com/recent-abstracts/urologic-oncology/prostate-cancer/114081-darolutamide-receives-fda-approval-for-the-treatment-of-non-metastatic-castration-resistant-prostate-cancer.html
  12. Darolutamide | ODM-201 | CAS#1297538-32-9 | Androgen Receptor (AR) Inhibitor, accessed August 2, 2025, https://www.medkoo.com/products/7589
  13. NUBEQA (darolutamide) tablets, for oral use - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212099Orig1s000lbl.pdf
  14. Darolutamide, ODM-201, BAY-1841788 supplier | CAS 1297538-32-9 - AOBIOUS, accessed August 2, 2025, https://aobious.com/aobious/pathway-inhibitors/13340-darolutamide-odm-201.html
  15. Darolutamide: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 2, 2025, https://go.drugbank.com/drugs/DB12941
  16. Darolutamide | CAS 1297538-32-9 | SCBT - Santa Cruz Biotechnology, accessed August 2, 2025, https://www.scbt.com/p/darolutamide-1297538-32-9
  17. darolutamide 300mg film-coated tablets (Nubeqa®) - Scottish Medicines Consortium, accessed August 2, 2025, https://scottishmedicines.org.uk/media/5563/darolutamide-nubeqa-final-october-2020-for-website.pdf
  18. U.S. FDA Approves NUBEQA® (darolutamide) to Treat Patients with Metastatic Castration-Sensitive Prostate Cancer | Bayer United States, accessed August 2, 2025, https://www.bayer.com/en/us/news-stories/metastatic-castration-sensitive-prostate-cancer
  19. Darolutamide (ODM-201) | Androgen Receptor Antagonist | MedChemExpress, accessed August 2, 2025, https://www.medchemexpress.com/ODM-201.html
  20. ASCO GU: ARANOTE and ARASENS Trials Reinforce Darolutamide in Metastatic Hormone-Sensitive Prostate Cancer - Pharmacy Times, accessed August 2, 2025, https://www.pharmacytimes.com/view/asco-gu-aranote-and-arasens-trials-reinforce-darolutamide-in-metastatic-hormone-sensitive-prostate-cancer
  21. Bayer's Nubeqa™ (darolutamide) receives EU approval in third indication for patients with advanced prostate cancer, accessed August 2, 2025, https://www.bayer.com/media/en-us/bayers-nubeqa-darolutamide-receives-eu-approval-in-third-indication-for-patients-with-advanced-prostate-cancer/
  22. Nubeqa | European Medicines Agency (EMA), accessed August 2, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/nubeqa
  23. Efficacy and safety outcomes of darolutamide in patients with non-metastatic castration-resistant prostate cancer with comorbidities and concomitant medications from the randomised phase 3 ARAMIS trial - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/37660438/
  24. Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/30763142/
  25. Efficacy and Safety Study of Darolutamide (ODM-201) in Men With High-risk Non-metastatic Castration-resistant Prostate Cancer (ARAMIS) - ClinicalTrials.gov, accessed August 2, 2025, https://clinicaltrials.gov/study/NCT02200614
  26. Darolutamide and health-related quality of life in patients with non-metastatic castration-resistant prostate cancer: An analysis of the phase III ARAMIS trial - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/34273811/
  27. Overall survival (OS) results of phase III ARAMIS study of darolutamide (DARO) added to androgen deprivation therapy (ADT) for nonmetastatic castration-resistant prostate cancer (nmCRPC). | Journal of Clinical Oncology - ASCO Publications, accessed August 2, 2025, https://ascopubs.org/doi/10.1200/JCO.2020.38.15_suppl.5514
  28. FDA approves darolutamide for non-metastatic castration-resistant prostate cancer, accessed August 2, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-darolutamide-non-metastatic-castration-resistant-prostate-cancer
  29. Darolutamide Continues to Show Strong Efficacy in nmCRPC After 3 Years, Regardless of Prior Therapy - Targeted Oncology, accessed August 2, 2025, https://www.targetedonc.com/view/darolutamide-continues-to-show-strong-efficacy-in-nmcrpc-after-3-years-regardless-of-prior-therapy
  30. Darolutamide Combo Significantly Increased Overall Survival in Metastatic Hormone-Sensitive Prostate Cancer - CancerNetwork, accessed August 2, 2025, https://www.cancernetwork.com/view/darolutamide-combo-significantly-increased-overall-survival-in-metastatic-hormone-sensitive-prostate-cancer
  31. The ARAMIS Trial: Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer, accessed August 2, 2025, https://www.urotoday.com/transformative-evidence/aramis-trial.html
  32. The Relevance of MFS as a Primary End Point in the ARAMIS Trial - Targeted Oncology, accessed August 2, 2025, https://www.targetedonc.com/view/the-relevance-of-mfs-as-a-primary-end-point-in-the-aramis-trial
  33. Efficacy and Safety of Darolutamide in Combination With Androgen-Deprivation Therapy and Docetaxel in Black Patients From the Randomized ARASENS Trial - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/37812679/
  34. Darolutamide in combination with docetaxel and ADT meets primary endpoint of significantly increasing overall survival in patients with metastatic hormone-sensitive prostate cancer - Bayer Global, accessed August 2, 2025, https://www.bayer.com/media/en-us/darolutamide-in-combination-with-docetaxel-and-adt-meets-primary-endpoint-of-significantly-increasing-overall-survival-in-patients-with-metastatic-hormone-sensitive-prostate-cancer/
  35. ASCO GU 2022: Overall Survival With Darolutamide Versus Placebo in Combination With Docetaxel and Androgen-Deprivation Therapy for Metastatic Hormone-Sensitive Prostate Cancer in the Phase 3 ARASENS Trial - UroToday, accessed August 2, 2025, https://www.urotoday.com/conference-highlights/asco-gu-2022/asco-gu-2022-prostate-cancer/135400-asco-gu-2022-overall-survival-with-darolutamide-versus-placebo-in-combination-with-docetaxel-and-androgen-deprivation-therapy-for-metastatic-hormone-sensitive-prostate-cancer-in-the-phase-3-arasens-trial.html
  36. FDA approves darolutamide for metastatic castration-sensitive prostate cancer, accessed August 2, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-darolutamide-metastatic-castration-sensitive-prostate-cancer
  37. ARASENS trial with darolutamide in combination with docetaxel and androgen deprivation therapy meets primary endpoint of significantly increasing overall survival in patients with metastatic hormone-sensitive prostate cancer - Orion Pharma, accessed August 2, 2025, https://www.orionpharma.com/newsroom/all-news/releases/stock-releases/2021/arasens-trial-with-darolutamide-in-combination-with-docetaxel-and-androgen-deprivation-therapy-meets-primary-endpoint-of-significantly-increasing-overall-survival-in-patients-with-metastatic-hormone-sensitive-prostate-cancer/
  38. Darolutamide in Addition to Standard Androgen Deprivation Therapy and Docetaxel in Metastatic Hormone-Sensitive Prostate Cancer (ARASENS) - ClinicalTrials.gov, accessed August 2, 2025, https://clinicaltrials.gov/study/NCT02799602
  39. SES AUA 2025: Overall Survival with Darolutamide Versus Placebo ..., accessed August 2, 2025, https://www.urotoday.com/conference-highlights/ses-aua-2025/ses-aua-2025-prostate-cancer/158987-ses-aua-2025-overall-survival-with-darolutamide-versus-placebo-in-combination-with-androgen-deprivation-therapy-and-docetaxel-accounting-for-subsequent-therapy-a-sensitivity-analysis-from-arasens.html
  40. Darolutamide and Survival in Metastatic, Hormone-Sensitive Prostate Cancer - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/35179323/
  41. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer: a plain language summary of the ARASENS trial - Video Journal of Biomedicine, accessed August 2, 2025, https://www.biomedicine.video/animated-videos/plain-language-summary-arasens-study-darolutamide-metastatic-hormone-sensitive-prostate-cancer
  42. Efficacy and Safety of Darolutamide in Combination With Androgen-Deprivation Therapy and Docetaxel in Black Patients From the Randomized ARASENS Trial - Oxford Academic, accessed August 2, 2025, https://academic.oup.com/oncolo/article/29/3/235/7302202
  43. Darolutamide receives EU approval in third indication for patients with advanced prostate cancer - Orion Pharma, accessed August 2, 2025, https://www.orionpharma.com/newsroom/all-news/releases/press-releases/2025/darolutamide-receives-eu-approval-in-third-indication-for-patients-with-advanced-prostate-cancer/

Published at: August 2, 2025

This report is continuously updated as new research emerges.

MedPath

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

© 2025 MedPath, Inc. All rights reserved.