C19H14F2N6O
1207456-01-6
HRR gene-mutated (HRRm) metastatic castration-resistant prostate cancer (mCRPC), Metastatic Breast Cancer, Locally Advanced Breast Cancer (LABC)
Talazoparib, marketed under the brand name Talzenna®, is a highly potent, orally bioavailable small molecule inhibitor of the poly (ADP-ribose) polymerase (PARP) enzymes, PARP1 and PARP2. Its therapeutic effect is rooted in the principle of synthetic lethality, a mechanism particularly effective in tumors harboring deficiencies in the homologous recombination repair (HRR) pathway, most notably those with germline mutations in the BRCA1 or BRCA2 genes. Talazoparib is distinguished from other agents in its class by a dual mechanism of action that combines catalytic inhibition of the PARP enzyme with exceptionally potent "PARP trapping," a process that sequesters the enzyme on DNA, forming a cytotoxic complex that is profoundly disruptive to DNA replication. This high trapping efficiency is believed to be a key driver of its clinical activity.
Talazoparib has secured regulatory approval for two primary indications based on robust clinical evidence from pivotal Phase 3 trials. First, as a monotherapy for the treatment of adult patients with deleterious or suspected deleterious germline BRCA-mutated (gBRCAm), HER2-negative locally advanced or metastatic breast cancer, an approval supported by the EMBRACA trial. This study demonstrated a significant improvement in progression-free survival (PFS) for Talazoparib compared to standard chemotherapy. Second, it is approved in combination with the androgen receptor inhibitor enzalutamide for the treatment of adult patients with HRR gene-mutated metastatic castration-resistant prostate cancer (mCRPC). This indication was established by the TALAPRO-2 trial, which showed a marked improvement in radiographic progression-free survival (rPFS) and a clinically meaningful benefit in overall survival (OS).
The clinical use of Talazoparib is characterized by a well-defined and predictable safety profile. The primary dose-limiting toxicities are hematological, including high rates of anemia, neutropenia, and thrombocytopenia. These adverse events are considered on-target effects related to the drug's potent mechanism of action and are typically manageable through a structured regimen of monitoring, dose interruption, and dose reduction. This comprehensive monograph provides a detailed analysis of Talazoparib, covering its chemical and pharmaceutical properties, pharmacological profile, pivotal clinical trial data, safety and tolerability, and guidelines for its clinical application.
Talazoparib is a small molecule drug with a complex heterocyclic structure. Its unambiguous identification is established through a series of internationally recognized names and numbers. The active pharmaceutical ingredient possesses a specific stereochemistry at two chiral centers, designated as (8S,9R), which is critical for its potent biological activity.[1] Alternative names used during its development include BMN-673, LT-673, and MDV-3800.[1] A consolidation of its key identifiers and properties is provided in Table 1.
Table 1: Physicochemical Properties of Talazoparib
Property | Value | Source(s) |
---|---|---|
Drug Name | Talazoparib | 5 |
Brand Name | Talzenna® | 1 |
Alternate/Code Names | BMN-673, LT-673, MDV-3800 | 1 |
Systematic (IUPAC) Name | (8S,9R)-5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro-3H-pyrido[4,3,2-de]phthalazin-3-one | 1 |
CAS Number | 1207456-01-6 | 1 |
DrugBank ID | DB11760 | 1 |
Molecular Formula | C19H14F2N6O | 1 |
Molar Mass | 380.359 g·mol⁻¹ | 1 |
PubChem CID | 135565082 | 1 |
UNII | 9QHX048FRV | 1 |
The manufacturing of Talazoparib involves a multi-step chemical synthesis designed to efficiently produce the correct and biologically active (8S,9R) enantiomer. One patented method, suitable for industrial production, avoids the use of metal catalysts and cryogenic conditions, thereby simplifying the process.[6] This pathway can be summarized in four main stages [6]:
Talazoparib is formulated for oral administration as hard capsules, which allows for convenient once-daily dosing.[1] The drug is marketed under the brand name Talzenna® in a range of strengths to provide clinicians with the flexibility needed for dose adjustments in response to adverse events.[9] The availability of multiple, finely-graded capsule strengths is a direct and necessary consequence of the drug's potent, mechanism-based toxicities, particularly myelosuppression. This formulation strategy is inextricably linked to the clinical risk management plan, enabling precise dose titration to balance efficacy and tolerability for individual patients.
The available capsule strengths include 1 mg, 0.75 mg, 0.5 mg, 0.35 mg, 0.25 mg, and 0.1 mg.[10] To prevent medication errors, each strength has a distinct appearance, characterized by different colored caps and specific black ink imprints [13]:
Patients are instructed to swallow the capsules whole; they must not be opened, crushed, broken, or dissolved.[8]
Talazoparib is an inhibitor of the PARP family of enzymes, with high potency against PARP1 and PARP2.[3] These enzymes are central to the base excision repair (BER) pathway, a cellular mechanism responsible for repairing DNA single-strand breaks (SSBs).[1] The therapeutic strategy of Talazoparib is based on the concept of
synthetic lethality. In normal cells possessing a functional homologous recombination repair (HRR) pathway, the inhibition of PARP is generally tolerated because the HRR machinery can effectively repair the more complex double-strand breaks (DSBs) that arise from unrepaired SSBs during DNA replication.[16]
However, certain cancers, particularly those with germline mutations in BRCA1, BRCA2, or other HRR-related genes (ATM, PALB2, etc.), have a deficient HRR pathway.[16] These tumor cells become critically dependent on the PARP-mediated BER pathway for DNA repair and survival.[1] When Talazoparib inhibits PARP in these HRR-deficient cells, SSBs accumulate and are converted into DSBs during replication. Without a functional HRR pathway to repair these DSBs, the cell undergoes catastrophic genomic instability, cell cycle arrest, and ultimately, apoptosis (programmed cell death).[14]
A defining characteristic that distinguishes Talazoparib from other PARP inhibitors is its dual mechanism of action, which encompasses both catalytic inhibition and exceptionally potent PARP trapping. Beyond simply blocking the enzymatic activity of PARP, Talazoparib traps the PARP enzyme onto the DNA at the site of a break, forming a highly cytotoxic PARP-DNA complex.[1] This trapped complex acts as a physical barrier that stalls replication forks and obstructs access by other DNA repair proteins, leading to profound cellular toxicity.[2] Preclinical studies indicate that Talazoparib is the most potent PARP-trapper identified to date, with a trapping efficiency approximately 100-fold greater than that of olaparib.[1] Its half-maximal inhibitory concentration (
IC50) for PARP1 in cell-free assays is 0.57 nM, underscoring its high potency.[3]
This extreme potency in PARP trapping can be viewed as a double-edged sword. It is the molecular foundation for Talazoparib's robust anti-tumor efficacy in susceptible cancers. At the same time, this intense, on-target mechanism is also the primary driver of its most significant dose-limiting toxicities. Rapidly proliferating cells, such as hematopoietic stem cells in the bone marrow, are highly reliant on efficient DNA replication and repair. The profound replication stress induced by Talazoparib's trapping activity leads to significant collateral damage in these healthy tissues, manifesting clinically as the myelosuppression (anemia, neutropenia, and thrombocytopenia) that characterizes its safety profile. Thus, the drug's primary adverse effects are not off-target phenomena but are a direct and predictable consequence of its powerful on-target mechanism.
The pharmacokinetic profile of Talazoparib is characterized by rapid oral absorption, extensive tissue distribution, minimal metabolism, and primary elimination via the kidneys as an unchanged drug. This profile directly informs its clinical dosing schedule and management guidelines. A summary of key parameters is provided in Table 2.
Table 2: Summary of Key Pharmacokinetic Parameters of Talazoparib
Parameter | Value | Source(s) |
---|---|---|
Time to Max. Concentration (Tmax) | Median: 1-2 hours | 2 |
Effect of High-Fat Meal | Cmax increases by 46%, Tmax delayed to 4 hours, no effect on AUC | 15 |
Apparent Volume of Distribution (Vd/F) | 420 L | 15 |
Plasma Protein Binding | ~74% (concentration-independent) | 1 |
Metabolism | Minimal (<30%) | 1 |
Primary Elimination Route | Renal excretion of unchanged drug | 1 |
Excretion | 69% in urine (55% unchanged), 20% in feces (14% unchanged) | 1 |
Terminal Half-Life (t1/2) | 90 (±58) hours | 1 |
Apparent Oral Clearance (CL/F) | 6.45 L/h | 15 |
Time to Steady State | 2-3 weeks | 2 |
The pharmacokinetic properties of Talazoparib are the direct cause for its specific clinical management guidelines. The long terminal half-life of 90 hours supports a convenient once-daily dosing regimen.[1] The primary reliance on renal excretion for clearance means that patients with impaired kidney function are at risk of drug accumulation and increased toxicity. This directly explains why the prescribing information includes mandatory dose reduction guidelines for patients with moderate to severe renal impairment.[9] Conversely, the minimal hepatic metabolism explains the lack of specific dose adjustments for hepatic impairment.[25] Furthermore, this profile dictates that the most clinically relevant drug-drug interactions are not with metabolic enzyme modulators, but with inhibitors of efflux transporter proteins like P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP), which can affect the drug's absorption and distribution.[26]
The relationship between Talazoparib exposure and its clinical effects (both efficacy and safety) has not been fully characterized.[14] However, a clear exposure-toxicity relationship has been established for its primary adverse effect. Analysis from the EMBRACA trial demonstrated that higher plasma concentrations of Talazoparib were associated with a greater incidence of Grade ≥3 anemia, confirming that this on-target toxicity is dose- and exposure-dependent.[22]
In terms of cardiac safety, dedicated studies have shown that Talazoparib, at the recommended therapeutic dose of 1 mg daily, does not cause large prolongations of the QT interval on an electrocardiogram (ECG) (i.e., >20 ms).[2] This indicates a low risk of inducing potentially fatal cardiac arrhythmias such as Torsades de Pointes, a significant safety advantage.
The clinical development program for Talazoparib has established its efficacy in specific, biomarker-defined cancer populations. Early-phase (Phase 1) studies evaluated the safety, tolerability, and preliminary activity of Talazoparib in patients with a variety of advanced solid tumors, including breast, ovarian, pancreatic, prostate, colorectal, and non-small cell lung cancer.[29] These studies identified a promising signal in tumors with known DNA damage repair defects.
Subsequent Phase 2 trials, such as the ABRAZO study (NCT02034916), provided more robust evidence of clinical benefit in heavily pretreated patients with gBRCA-mutated advanced breast cancer.[21] The promising activity observed in these trials provided the rationale for launching two large, pivotal Phase 3 studies—EMBRACA and TALAPRO-2—which ultimately formed the basis for its regulatory approvals. Concurrently, Phase 2 trials continue to explore Talazoparib's potential in other settings, such as in combination with agents like abemaciclib and capivasertib for various breast cancer subtypes.[31]
The EMBRACA trial (NCT01945775) was the cornerstone study that established Talazoparib as a standard of care for patients with gBRCA-mutated advanced breast cancer.[30]
EMBRACA was a global, multicenter, open-label, randomized Phase 3 trial that enrolled 431 patients.[21] Eligible patients had deleterious or suspected deleterious germline mutations in either the
BRCA1 or BRCA2 gene and had HER2-negative locally advanced or metastatic breast cancer.[32] The patient population was representative of this disease setting, including individuals with hormone receptor-positive (HR+) disease (54%) and triple-negative breast cancer (TNBC) (46%).[35] Patients were required to have received no more than three prior cytotoxic chemotherapy regimens for advanced disease and must have been treated with an anthracycline and/or a taxane in a prior setting, unless contraindicated.[8]
Patients were randomized in a 2:1 ratio to receive either Talazoparib (1 mg orally once daily) or the physician's choice of standard single-agent chemotherapy (PCT), which included capecitabine, eribulin, gemcitabine, or vinorelbine.[21]
The EMBRACA trial successfully met its primary endpoint, demonstrating the superiority of Talazoparib over standard chemotherapy. However, the results for the key secondary endpoint of overall survival presented a more complex picture. A summary of the key efficacy outcomes is presented in Table 3.
Table 3: Summary of Efficacy Results from the EMBRACA Trial
Efficacy Endpoint | Talazoparib (n=287) | Chemotherapy (n=144) | Hazard Ratio (HR) / Odds Ratio (OR) (95% CI) | p-Value | Source(s) |
---|---|---|---|---|---|
Median PFS (Primary) | 8.6 months | 5.6 months | HR: 0.54 (0.41, 0.71) | <0.0001 | 30 |
Objective Response Rate (ORR) | 62.6% | 27.2% | OR: 4.99 (2.9, 8.8) | <0.0001 | 30 |
Median OS (Final) | 19.3 months | 19.5 months | HR: 0.848 (0.670, 1.073) | 0.17 | 21 |
The primary outcome, progression-free survival (PFS) as assessed by blinded independent central review, was significantly longer in the Talazoparib arm. The median PFS was 8.6 months for patients treated with Talazoparib compared to 5.6 months for those receiving chemotherapy.[30] This corresponded to a hazard ratio (HR) of 0.54, representing a 46% reduction in the risk of disease progression or death (p<0.0001).[32] This robust PFS benefit was consistent across all key patient subgroups, including by HR status (HR+ or TNBC), type of BRCA mutation (
BRCA1 or BRCA2), and number of prior chemotherapy lines.[30] Furthermore, the objective response rate (ORR) was more than doubled with Talazoparib (62.6%) compared to chemotherapy (27.2%).[30]
In contrast to the strong PFS data, the final analysis of the key secondary endpoint, overall survival (OS), did not demonstrate a statistically significant benefit for Talazoparib. After a median follow-up of approximately 45 months in the Talazoparib arm, the median OS was 19.3 months, nearly identical to the 19.5 months observed in the chemotherapy arm (HR 0.848; p=0.17).[36] This apparent discrepancy between a large PFS benefit and no OS benefit is a classic example of how post-progression therapies can confound survival outcomes in modern oncology trials. The trial data revealed that a substantial proportion of patients in the control arm received effective subsequent treatments after their disease progressed on chemotherapy. Specifically, 32.6% of patients in the chemotherapy arm crossed over to receive a subsequent PARP inhibitor, the very class of drug being tested.[21] Additionally, high rates of subsequent platinum-based chemotherapy, which is known to be particularly effective in BRCA-mutated cancers, were used in both arms.[21] The use of these effective subsequent therapies in the control group likely "rescued" those patients, extending their survival and masking or diluting the true survival benefit of receiving Talazoparib as an earlier line of treatment. This confounding effect was acknowledged in the trial's final publications.[21] Therefore, the OS result should not be interpreted as a failure of the drug's activity but rather as an artifact of the trial's design and the evolving treatment landscape. The significant improvements in PFS and quality of life remain the primary indicators of Talazoparib's clinical value in this patient population.
Despite the lack of an OS benefit, Talazoparib provided a significant and meaningful improvement in patients' quality of life (QoL). Patient-reported outcome (PRO) analyses showed a statistically significant delay in the time to definitive clinically meaningful deterioration of global health status.[21] The median time to deterioration was 26.3 months for patients on Talazoparib, a stark contrast to just 6.7 months for those on chemotherapy.[35] This demonstrates that, in addition to controlling the disease for longer (PFS), Talazoparib allowed patients to maintain their overall well-being and functionality for a significantly longer period compared to standard cytotoxic treatments.
The TALAPRO-2 trial (NCT03395197) was a landmark study that established a new combination therapy standard for first-line metastatic castration-resistant prostate cancer (mCRPC) and expanded Talazoparib's indications.[17]
TALAPRO-2 was a large, Phase 3, randomized, double-blind, placebo-controlled trial that evaluated Talazoparib in combination with the standard-of-care androgen receptor pathway inhibitor (ARPI), enzalutamide.[17] The study enrolled over 1,000 men with mCRPC who had not yet received systemic treatment for their castration-resistant disease.[41] The trial was designed with two primary cohorts: an "all-comer" population (unselected for biomarker status) and a cohort of patients prospectively confirmed to have mutations in one of 12 pre-specified HRR genes (including
BRCA1/2, ATM, PALB2, and CHEK2).[17] Patients were randomized 1:1 to receive either Talazoparib (at a reduced dose of 0.5 mg daily) plus enzalutamide or placebo plus enzalutamide.[17]
The rationale for this combination is based on preclinical evidence of synergy. Androgen receptor signaling can regulate DNA repair pathways, and its inhibition with drugs like enzalutamide may induce a state of "BRCAness," impairing HRR function and thereby sensitizing tumor cells to PARP inhibition.
The TALAPRO-2 trial met its primary and key secondary endpoints, demonstrating significant benefits for the combination therapy. The FDA's initial approval was based on the rPFS results in the HRR-mutated population, while subsequent final OS data have supported its broader clinical utility. A summary of the key efficacy outcomes is presented in Table 4.
Table 4: Summary of Efficacy Results from the TALAPRO-2 Trial
Efficacy Endpoint | Population | Talazoparib + Enzalutamide | Placebo + Enzalutamide | Hazard Ratio (HR) (95% CI) | p-Value | Source(s) |
---|---|---|---|---|---|---|
Median rPFS (Primary) | HRR-Mutated | Not Reached | 13.8 months | HR: 0.45 (0.33, 0.61) | <0.0001 | 17 |
Median rPFS (Final) | All-Comers | 33.1 months | 19.5 months | HR: 0.667 (0.551, 0.807) | <0.0001 | 42 |
Median OS (Final) | HRR-Deficient | 45.1 months | 31.1 months | HR: 0.622 (0.475, 0.814) | 0.0005 | 39 |
Median OS (Final) | All-Comers | 45.8 months | 37.0 months | HR: 0.796 (0.661, 0.958) | 0.0155 | 42 |
The primary endpoint of radiographic progression-free survival (rPFS) was met with high statistical significance. In the HRR-mutated population, the combination of Talazoparib and enzalutamide resulted in a 55% reduction in the risk of radiographic progression or death compared to enzalutamide alone (HR 0.45; p<0.0001).[17] This profound benefit in the biomarker-selected group was the basis for the FDA's approval.[20] A significant rPFS benefit was also observed in the all-comer population (HR 0.63).[40]
Most importantly, the final analysis of overall survival demonstrated a statistically significant and clinically meaningful benefit. In the all-comer population, the combination therapy extended median OS by 8.8 months (45.8 months vs. 37.0 months; HR 0.796; p=0.0155).[39] As expected, the benefit was even greater in the HRR-deficient subgroup, where the combination extended median OS by approximately 14 months and reduced the risk of death by 38% (HR ~0.62).[39]
Subgroup analyses confirmed that the greatest magnitude of benefit was observed in patients with BRCA mutations (rPFS HR 0.20).[17] However, the demonstration of a significant OS benefit in the unselected "all-comer" population is a critical finding with broad clinical implications. It suggests that the synergistic mechanism between Talazoparib and enzalutamide may extend beyond the classic model of synthetic lethality in tumors with pre-existing HRR defects. The data suggest that ARPI therapy may induce a vulnerable state in a wider range of prostate cancers, sensitizing them to PARP inhibition. While the FDA approval was appropriately restricted to the HRR-mutated population where the benefit-risk ratio is most favorable [20], the final OS data provide a strong rationale for considering this combination as a new standard-of-care option for a much broader population of men with first-line mCRPC.[42]
The safety profile of Talazoparib is well-characterized and is dominated by on-target hematological toxicities. Management of these adverse events is a central aspect of its clinical use.
The prescribing information for Talazoparib carries several important warnings and precautions that require diligent clinical monitoring.
A rare but very serious risk associated with PARP inhibitors, including Talazoparib, is the development of therapy-related myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). These conditions are often fatal. In clinical studies, MDS/AML was reported in approximately 0.4% of solid tumor patients treated with Talazoparib.[45] Many of these patients had confounding factors, such as prior exposure to platinum-based chemotherapy or other DNA-damaging agents.[47] Clinicians must monitor complete blood counts monthly. If a patient develops prolonged hematological toxicity (lasting more than 4 weeks), Talazoparib should be interrupted, and the patient should be referred to a hematologist for further investigation, including bone marrow analysis and cytogenetics. If MDS/AML is confirmed, Talazoparib must be permanently discontinued.[45]
Myelosuppression is the most common and clinically significant toxicity of Talazoparib.[32] It manifests as anemia (low red blood cells), neutropenia (low neutrophils), and/or thrombocytopenia (low platelets). Grade 3 or higher events are common and require active management.
Baseline and monthly monitoring of complete blood counts is mandatory throughout treatment. Treatment should not be initiated until patients have recovered from hematological toxicity caused by prior therapies.[13]
Based on its mechanism of action (interfering with DNA repair) and findings in animal reproduction studies, Talazoparib can cause harm to a developing fetus and may lead to miscarriage.[13] Therefore, its use is contraindicated during pregnancy.
Beyond the major warnings, a range of other adverse reactions are commonly associated with Talazoparib. The most frequent events are hematologic and gastrointestinal in nature. Table 5 provides a summary of the most common adverse reactions observed in the pivotal EMBRACA trial.
Table 5: Incidence of Common (≥20%) and Grade ≥3 Adverse Reactions with Talazoparib (EMBRACA Trial)
Adverse Reaction | Talazoparib (Any Grade) | Chemotherapy (Any Grade) | Talazoparib (Grade ≥3) | Chemotherapy (Grade ≥3) | Source(s) |
---|---|---|---|---|---|
Hematologic | |||||
Anemia | 53% | 18% | 39% | 5% | 1 |
Neutropenia | 35% | 43% | 21% | 35% | 1 |
Thrombocytopenia | 27% | 7% | 15% | 2% | 1 |
Non-Hematologic | |||||
Fatigue | 62% | 50% | - | - | 32 |
Nausea | 49% | 47% | - | - | 32 |
Headache | 33% | 22% | - | - | 32 |
Vomiting | 25% | 23% | - | - | 32 |
Alopecia (Hair Loss) | 25% | 28% | - | - | 32 |
Diarrhea | 22% | 26% | - | - | 32 |
Decreased Appetite | ≥20% | - | - | - | 32 |
The most common laboratory abnormalities (≥25%) reflect the myelosuppressive effects, with high rates of decreases in hemoglobin, leukocytes, lymphocytes, and platelets.[46]
The cornerstone of managing Talazoparib-related toxicity is dose modification, which may involve treatment interruption, dose reduction, or, in severe cases, permanent discontinuation.[9] The availability of multiple capsule strengths facilitates this management strategy. Specific, protocol-defined thresholds are used to guide these decisions, particularly for hematological toxicities. Table 6 outlines the recommended dose reduction schedules and the criteria for managing myelosuppression.
Table 6: Recommended Dose Modifications for Adverse Reactions
Parameter | Action | Source(s) |
---|---|---|
Dose Reduction Levels (Breast Cancer) | ||
Starting Dose | 1 mg once daily | 10 |
First Reduction | 0.75 mg once daily | 10 |
Second Reduction | 0.5 mg once daily | 10 |
Third Reduction | 0.25 mg once daily | 10 |
Dose Reduction Levels (Prostate Cancer) | ||
Starting Dose | 0.5 mg once daily | 10 |
First Reduction | 0.35 mg once daily | 10 |
Second Reduction | 0.25 mg once daily | 10 |
Third Reduction | 0.1 mg once daily | 10 |
Management of Hematologic Toxicity | ||
Hemoglobin <8 g/dL | Withhold TALZENNA. Resume at a reduced dose when level resolves to ≥9 g/dL. | 10 |
Platelet count <50,000/μL | Withhold TALZENNA. Resume at a reduced dose when count resolves to ≥75,000/μL. | 10 |
Neutrophil count <1,000/μL | Withhold TALZENNA. Resume at a reduced dose when count resolves to ≥1500/μL. | 10 |
Management of Non-Hematologic Toxicity | ||
Grade 3 or 4 | Withhold TALZENNA. Consider resuming at a reduced dose or discontinuing when toxicity resolves to ≤Grade 1. | 10 |
Treatment with Talazoparib should be permanently discontinued if a patient requires more than three dose reductions.[9]
The use of Talazoparib is approved for specific, biomarker-defined patient populations, requiring genetic testing for patient selection.
For both indications, patient eligibility must be confirmed by an FDA-approved companion diagnostic test to detect the presence of the qualifying genetic mutations.[18]
The recommended dosing for Talazoparib differs by indication.
For both indications, treatment should continue until disease progression or the development of unacceptable toxicity. The capsules can be taken with or without food and must be swallowed whole. If a patient vomits or misses a dose, they should not take an additional dose but should resume with the next scheduled dose at the usual time.[9]
Specific dose adjustments are required for patients with renal impairment and for those taking certain interacting medications. These adjustments are critical for maintaining a safe exposure level. Table 7 summarizes these required modifications.
Table 7: Recommended Dosing Adjustments for Renal Impairment and Drug Interactions
Condition | Indication | Recommended TALZENNA Dose | Source(s) |
---|---|---|---|
Moderate Renal Impairment (CLcr 30-59 mL/min) | Breast Cancer | 0.75 mg once daily | 9 |
Prostate Cancer | 0.35 mg once daily | 10 | |
Severe Renal Impairment (CLcr 15-29 mL/min) | Breast Cancer | 0.5 mg once daily | 9 |
Prostate Cancer | 0.25 mg once daily | 10 | |
Coadministration with Strong P-gp Inhibitors* | Breast Cancer | 0.75 mg once daily | 9 |
*Strong P-gp inhibitors include amiodarone, carvedilol, clarithromycin, itraconazole, and verapamil. For other P-gp or BCRP inhibitors, dose reduction is not mandated, but increased monitoring for adverse reactions is recommended.[27]
Talazoparib is a substrate of the P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) efflux transporters.[26] Coadministration with drugs that inhibit these transporters can increase plasma concentrations of Talazoparib, thereby increasing the risk of toxicity.[15] If concurrent use with a potent P-gp inhibitor cannot be avoided, the dose of Talazoparib must be reduced as specified. When the inhibitor is discontinued, the Talazoparib dose should be increased back to the original dose after 3-5 half-lives of the interacting drug have passed.[9]
The development and commercialization of Talazoparib involved several pharmaceutical companies. It was initially developed by BioMarin Pharmaceutical Inc. In August 2015, Medivation Inc. acquired the worldwide rights to the compound. Subsequently, in 2016, Pfizer acquired Medivation and now holds the global marketing rights for Talazoparib under the brand name Talzenna®.[1]
Talazoparib has received approvals from major regulatory agencies based on the strength of its pivotal clinical trial data.
Across all major global markets, including the United States, European Union, Canada, United Kingdom, and Australia, Talazoparib is classified as a prescription-only medicine (℞-only).[1]
Talazoparib (Talzenna®) represents a significant advancement in the field of precision oncology, solidifying the role of PARP inhibitors as a cornerstone of therapy for cancers with underlying DNA damage repair deficiencies. Its defining feature is an exceptionally potent dual mechanism of action, combining catalytic inhibition with profound PARP trapping, which drives its robust clinical efficacy. This potency, however, is directly linked to its primary and dose-limiting toxicity of myelosuppression, creating a clear and predictable relationship between its on-target mechanism, therapeutic benefit, and safety profile.
The clinical development program, highlighted by the pivotal EMBRACA and TALAPRO-2 trials, has unequivocally established its value. In gBRCA-mutated advanced breast cancer, it offers a significant progression-free survival and quality-of-life advantage over standard chemotherapy. In metastatic castration-resistant prostate cancer, its combination with enzalutamide has set a new standard of care for HRR-mutated disease by delivering a clinically meaningful overall survival benefit.
The successful clinical application of Talazoparib hinges on a comprehensive understanding of its properties. This includes mandatory biomarker testing for patient selection, diligent monitoring for hematological toxicities, and adherence to established guidelines for dose modification in response to adverse events or in specific populations, such as those with renal impairment. As research continues, the full potential of Talazoparib, both as a monotherapy and in novel combinations, will likely expand, further refining its role in the treatment of cancers vulnerable to the principle of synthetic lethality.
Published at: August 4, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.