Bioanalytical Methods for Poly(ADP-Ribose) Polymerase Inhibitor Quantification: A Review for Therapeutic Drug Monitoring : Therapeutic Drug Monitoring

Secondary Logo

Journal Logo

Review Article

Bioanalytical Methods for Poly(ADP-Ribose) Polymerase Inhibitor Quantification: A Review for Therapeutic Drug Monitoring

Orleni, Marco MSc*,†; Canil, Giovanni PhD*; Posocco, Bianca PhD*; Gagno, Sara PhD*; Toffoli, Giuseppe MD*

Author Information
Therapeutic Drug Monitoring ():10.1097/FTD.0000000000001081, January 12, 2023. | DOI: 10.1097/FTD.0000000000001081
  • Open
  • PAP

Abstract

INTRODUCTION

Poly(ADP-ribose) polymerase inhibitors (PARPis) are a rapidly growing class of targeted anticancer drugs that have been developed over the past 2 decades. PARP belongs to a family of enzymes that play a key role in DNA damage repair.1,2 In normal cells, the repair of DNA double-strand breaks is regulated by proteins encoded by breast cancer (BRCA) 1 and BRCA2 genes through the homologous recombination mechanism of repair (HRR). Homologous recombination mechanism of repair is an essentially error-free mechanism; however, when altered, as in BRCA-mutated tumors, other repair mechanisms take over to minimize the damage. One of these alternate repair mechanisms requires proper functioning of the PARP enzyme. However, when an inhibitor impairs PARP function, permanent damage accumulates in DNA, leading to a cytotoxic effect.2–4 The process outlined is based on the synthetic lethality theory that initiated the development of PARPis.

Since their first clinical trial in 2003,2,4 the following 4 PARPis have been approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) for cancer therapy: olaparib (Lynparza), rucaparib (Rubraca), niraparib (Zejula), and talazoparib (Talzenna). These drugs are primarily used as monotherapies for the maintenance treatment of platinum-sensitive advanced or recurrent ovarian cancer and human epidermal growth factor receptor (HER) 2–negative locally advanced or metastatic breast cancer, which are most commonly associated with BRCA mutations.5–8 In addition, olaparib has been approved for the treatment of prostate and pancreatic cancer, further stimulating the interest and investigation of PARPis for their therapeutic potential in other tumor types, regardless of the BRCA mutation status of the patient.3,4

Poly(ADP-ribose) polymerase inhibitors are orally administered at a fixed dose over a prolonged period and are substrates of various drug-metabolizing enzymes and transporters.5–8 These factors, along with the pharmacogenetic background, varying concomitant medications, and dietary regimens of the patient, often result in wide interindividual variability in drug exposure. Consequently, some patients may experience an insufficient response because of suboptimal drug concentrations, while others may experience severe side effects when the drug exceeds optimal levels.9 To optimize response and personalize treatment, therapeutic drug monitoring (TDM) may be a useful strategy. Specifically, this practice measures drug concentrations in biological fluids of the patient and helps support clinical decisions with the goal of maximizing treatment efficacy and minimizing the occurrence of toxicity.

The aim of this review was to highlight the relevant pharmacological features of PARPis that suggest their use in TDM practice and to discuss the available analytical methods for PARPis quantification in human plasma using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS).

METHODS

This review considered published methods for quantifying PARPis in human plasma, which is the standard method of quantifying pharmaceutical levels in clinical trials and practice. In addition, the search was refined to include only those methods that use LC-MS/MS with a triple quadrupole because this is the most accurate technique for drug quantification in human plasma.10 Finally, only methods that quantify FDA-approved and EMA-approved drugs and provide sufficient information to be reproduced by analysts were considered. The search was performed using SciFinder, Google Scholar, PubMed, and Web of Science with the keywords “PARP inhibitors,” “bioanalytical methods,” “LC-MS/MS,” “human plasma,” “TDM,” and a combination thereof. Medical subject heading terms such as “Poly(ADP-ribose) Polymerase Inhibitors,” “Chromatography, High Pressure Liquid,” “Tandem Mass Spectrometry,” “Drug Monitoring,” and “Analytic Sample Preparation Methods” were considered during the PubMed search. Thirteen manuscripts,11–23 published before August 2022, met the inclusion criteria described above. Moreover, the investigation of PARPis pharmacology and the clinical utility of TDM was based on the official documents of the regulatory agencies, namely the FDA multidisciplinary reviews,5–8,24 EMA public assessment reports,25–29 and other relevant papers found in the literature.30–40 In the following discussion, information on the pharmacology, physicochemical features of the drugs, methods used for biological sample preparation, hardware used for the analysis (LC and MS), and validation issues are provided.

TDM AND PARP INHIBITORS

Therapeutic drug monitoring is a branch of clinical chemistry and pharmacology that is applied to several classes of drugs, such as antibiotics, immunosuppressants, antipsychotics, and anticonvulsants. Clinical studies have also demonstrated a correlation between drug exposure [eg, area under the curve (AUC) and trough concentration (Ctrough)] and treatment response (efficacy and/or toxicity) in some cancers.34 However, a strong correlation has not been found for many anticancer drugs, and their potential for TDM is still being investigated. In the case of PARPis, several authors point out that they should be further explored as candidates for TDM.30,31,35 Recently, a review by Groenland et al30 gathered together 7 criteria known to be valid indications of the suitability of a drug for TDM, and PARPis satisfy most of these criteria. Specifically, they are prescribed as long-term therapy, taken daily for at least 2 years or until disease progression or unacceptable toxicity.41–44 Furthermore, their dose can be easily adapted to meet individual patient needs because PARPis are formulated as fixed dose capsules or tablets.25–29 Moreover, no measurable biomarkers to assess drug effects are available,5–8,24 and validated bioanalytical methods for their quantification have been published.11–23 Finally, PARPis display significant variability in pharmacokinetic (PK) exposure, as highlighted by relevant studies on olaparib,35,45 niraparib,46 rucaparib,6,47 and talazoparib.48 By contrast, the therapeutic window of PARPis is wide,37 and not all PARPis have clear exposure–response relationships.35 In the subsequent sections, the controversial exposure–response relationships, drug–drug interactions, and renal and hepatic impairments have been addressed thoroughly. The last 2 circumstances are included in the current discussion because a change in dosage might be involved, leading to nonoptimal plasma concentrations, which could be monitored by TDM.

Pharmacology of PARP Inhibitors

Olaparib was first formulated as capsule with a standard dose of 400 mg twice daily (BID) and then as tablet with a standard dose of 300 mg BID. Both are addressed in the subsequent discussion.

Exposure Safety

For talazoparib8 and olaparib,5 an exposure–toxicity relationship for anemia has been reported, that is, a higher risk of anemia at higher steady-state concentrations. For olaparib specifically, the likelihood of anemia has been correlated with its maximum concentration (Cmax). In addition, olaparib tablets are associated with a higher risk of anemia than the capsules because the drug exposure of the tablets exceeds that of the capsules.5 Moreover, a Ctrough of 2500 ng/mL for olaparib was identified in a recent retrospective study36 as the threshold for early adverse events. In addition, higher exposure to talazoparib was associated with a higher risk of thrombocytopenia.39 For niraparib, a correlation between AUC and thrombocytopenia has been observed7; however, its tolerability could be managed through laboratory monitoring. For rucaparib, a correlation between exposure and some hematological parameters,40 including grade 2+ creatinine, has been reported.6

Exposure-Efficacy

The regulatory paper by FDA reports that higher exposure to talazoparib leads to longer progression-free survival (PFS).8 For rucaparib, the situation is unclear and requires further investigation. Based on previous studies on olaparib and niraparib, the FDA and EMA report a desired target Ctrough of 650 ng/mL for rucaparib6 and that all patients treated with the recommended dose (600 mg BID) had a plasma rucaparib concentration well above this threshold.6,27 By contrast, a recent review35 reported the absence of a correlation between exposure and efficacy end points. Notably, the exposure–efficacy relationship for olaparib and niraparib was not significant. However, this relationship should be further studied because frequent dose modification and/or treatment discontinuation occurred in past studies.7,24

Drug–Drug Interactions

Olaparib is primarily metabolized by cytochrome P450 (CYP) 3A4; therefore, the concomitant administration of potent CYP3A4 inhibitors or inducers should be avoided.32 When strong and moderate CYP3A4 inhibitors are present, a dose reduction to 100 and 150 mg BID, respectively, is recommended for tablet administration.24 When talazoparib is coadministered with potent P-glycoprotein (P-gp) inhibitors, dose reduction from the standard dose of 1 mg–0.75 mg is suggested and patients should be monitored for adverse effects so the dose can be adjusted according to tolerability.33 Similarly, breast cancer resistance protein (BCRP) inhibitors may increase talazoparib exposure.8 Niraparib has a low risk of drug–drug interactions because it is primarily metabolized by carboxylesterases, rather than cytochromes.7 Finally, rucaparib has been reported to interact with many CYPs and transporters, resulting in a potential effect on other drugs.35

Renal and Hepatic Impairment

According to the latest guidelines,49 renal impairment is considered either mild or moderate when creatinine clearance is between 60 and 89 mL/min or 30 and 59 mL/min, respectively. For olaparib, the creatinine clearance values used to assess renal impairment refer to older guidelines.50 Hepatic impairment is considered mild and moderate when the Child–Pugh score is classified as A and B, respectively.51

For olaparib, no dose adjustment is required for patients with mild renal impairment24; however, patients should be monitored for toxicity.5 By contrast, a tablet dose reduction to 200 mg BID is suggested for patients with moderate renal impairment.24 Furthermore, no dose adjustment is required in cases of mild or moderate hepatic impairment.38 Similarly, for talazoparib, dose reduction is required if the renal impairment experienced by the patient is moderate (0.75 mg),8,33 whereas no adjustments are required if mild renal or hepatic impairment is present.28 No dose adjustments are required for niraparib if the renal impairment experienced by the patient is mild or moderate7; however, a recent postmarketing study52 suggested a dose reduction to 200 mg for cases with moderate hepatic impairment. In the case of rucaparib, the situation requires further investigation because mild or moderate renal impairment is known to increase the AUC of rucaparib, although no dose adjustment is recommended.6 However, in cases of severe renal impairment, some authors35 indicate that TDM of rucaparib might be beneficial.

BIOANALYTICAL METHODS FOR PARP INHIBITOR QUANTIFICATION

General Considerations

The compounds analyzed in this literature review and their most relevant physicochemical features are listed in Table 1. The information presented in Table 1 is useful for the development of a robust bioanalytical method and is directed to the analyst required to prepare the analyte solutions and set up the sample preparation protocol, as well as the LC and MS methods and conditions. In addition, a discussion of other analytical aspects (ie, single versus multianalyte methods and analyte calibration range) relevant to TDM is provided.

TABLE 1. - Main Physicochemical Features of EMA-Approved and FDA-Approved Poly (ADP-Ribose) Polymerase Inhibitors (PARPis)
Olaparib Rucaparib Niraparib Talazoparib
MW (g/mol) 434.46 555.67 510.61 552.56
Free base MW (g/mol) N/A 323.36 320.4 380.35
Counterion N/A Camsylate Tosylate Tosylate
pKa N/A ∼9.5 9.95 N/A
Solubility in water (mg/mL) 0.1 at 20°C 1.7 at 37°C 0.7–1.1 at 37°C Low
Solubility in organic solvents (mg/mL) ACN = 3.1; EtOH = 5.5; MeOH = 10.6 N/A N/A N/A
ACN, acetonitrile; EtOH, ethanol; MeOH, methanol; N/A, not applicable or reported.

Physicochemical Properties of the Analytes

Olaparib is a neutral compound with a molecular weight (MW) of 434.46 g/mol that is sold as a free base species. It has a basic pKa of −1.25 and an acidic pKa of 12.07, which means that it is unionized at physiological pH.5 It is only slightly soluble in aqueous media (0.1 mg/mL at 20°C), but it is soluble in organic solvents (Table 1).5

Niraparib, rucaparib, and talazoparib are weak bases that are commercialized as salts with their specific counterions in their pharmacological formulations (Table 1). Niraparib is found in capsules as a tosylate salt (monohydrate). It has a MW of 510.61 g/mol and a free base MW of 320.40 g/mol. It has a pKa of 9.95, and its aqueous solubility at 37°C ranges between 0.7 and 1.1 mg/mL below its pKa (ionized form).7 Rucaparib is found as tablets in the form of a camsylate salt with a MW of 555.67 g/mol and a free base MW of 323.36 g/mol. Based on a report by EMA,27 its solubility decreases above pH 9.5, likely because of deprotonation, and its structure is that of a secondary amine, suggesting its pKa to be approximately 9.5. It has an aqueous solubility of 1.4 mg/mL at 25°C and 1.7 mg/mL at 37°C, which is pH independent between pH 3 and 7 (under its pKa value).6 Talazoparib is commercialized in capsules in the form of a tosylate salt with a MW of 552.56 g/mol and a free base of 380.35 g/mol. Information on its solubility and pKa was not clearly indicated in the official EMA28 and FDA8 documents, which state that the solubility of talazoparib is “low across the physiological pH range” (at 37°C), but they report that 1 mg of talazoparib is soluble in less than 250 mL of aqueous medium (pH not specified).

Other Analytical Considerations

For TDM purposes, either single-analyte or multianalyte methods can be found in the literature. Of the 13 methods considered in this review, more than half are single-analyte methods. Specifically, 4 methods for the quantification of olaparib,12,13,15,16 3 for that of rucaparib,14,18,22 and one for that of niraparib17 were considered. The major advantage of single-analyte methods is the simplicity of their development as both sample preparation and detection are optimized for this analyte, and chromatography does not require the separation of multiple species. However, multianalyte methods are more convenient because they save time and money by analyzing different molecules simultaneously. Indeed, several authors use multianalyte methods for PARPi quantification. For example, Krens et al21 investigated olaparib and niraparib together with other anticancer drugs, and similarly, Jolibois et al,20 Pressiat et al,19 and Sparidans et al11 investigated olaparib together with other anticancer drugs. These methods could be useful if the analyst is interested in quantifying all the drugs included in the assay; otherwise, a single-analyte method might be more convenient and easier to transfer from one laboratory to another. Finally, the method proposed by Bruin et al23 allows the use of a single assay to simultaneously quantify all the FDA-approved and EMA-approved PARPis, along with veliparib, which is yet to be approved by the regulatory agencies. Reportedly, this is the first published method to quantify talazoparib in human plasma.23Table 2 summarizes the details of all bioanalytical methods.

TABLE 2. - Summary of General Features, Sample Preparation, and LC Conditions of Bioanalytical Methods for PARPi Quantification
Author (yr) Analytes IS Calibration Range Matrix Volume Extraction Procedure Addition of IS Injection Solvent Mobile Phases Instrument Specification and Set Up Column Elution Type Runtime Other Chromatographic Parameters (TF, Oven T, AS T, Vinj) Reference Number
Bruin et al (2020) Olaparib, rucaparib, niraparib, talazoparib, veliparib 2H8-olaparib,13C,2H3-rucaparib, 13C6-niraparib as hydrochloride salt, 13C,2H4-Talazoparib, 13C,2H3,15N-Veliparib as dihydrochloride salt 100–10000 for olaparib, 30–3000 for niraparib, 50–5000 for rucaparib, and veliparib; 0.5–50 for talazoparib (ng/mL for all) 50 µL PP (1 + 2 ACN) Before precipitation Dilution 1 + 1 with MPA 0.1% HCOOH in water (MP A) and 0.1% HCOOH in MeOH (MP B) Shimadzu nexera 2 with binary pump, degasser, autosampler, valco diverter valve and oven Waters acquity UPLC BEH C18 (100 × 2.1 mm, 1.7 µm) + its vanguard pre-column Gradient 6.2 min TF: 0.3; oven T: 40; AS T: 2–8 23
Krens et al (2020) Olaparib, niraparib, regorafenib, regorafenib M2, vemurafenib, cobimetinib, cabozantinib and dabrafenib 2H8-olaparib,13C6-niraparib 200–20000 for olaparib and 200–2000 for niraparib (ng/mL for all) 50 µL PP (1 + 4 ACN) With precipitant Dilution 1 + 9 with water 0.1% HCOOH in water (MP A) and 0.1% HCOOH in ACN (MP B) Waters acquity H-class UPLC Waters Cortex UPLC C18 (50 × 2.1 mm, 1.6 µm) Gradient 7 min TF: 0.8; oven T: 50; AS T: RT; Vinj: 10 21
Shapiro et al (2019) Rucaparib d7-rucaparib 5–10000 ng/mL 20 or 50 µL PP with 96-well plate, solvent not reported N/A N/A 20% ACN and 0.1% HCOOH in H2O N/A Agilent Polaris C18-A (50 × 2.1 mm, 3 µm) Isocratic N/A TF: 0.5 22
Jolibois et al (2019) Olaparib, cabozantinib, palbociclib HCl, pazopanib HCl, sorafenib, Sunitinib l-malate, N-desethyl-sunitinib HCl 2H8-olaparib 170–20000 ng/mL 150 µL LLE with EtOAc Before extraction Reconstitution with 55% MeOH/45% water-based solvent 92% ACN (vol/vol) and 8% of H2O in 0.1% HCOOH Dionex HPLC Agilent zorbax Bonus-RP (150 × 2.1 mm, 1.8 µm) Isocratic 2.5 min TF: 0.5, then 0.3; oven T: 50; AS T: 4; Vinj: 2 20
Pressiat et al (2018) Olaparib, ruxolitinib, vismodegib, pazopanib 2H8-olaparib 100–100000 ng/mL 50 µL PP (1 + 2,5 ACN) With precipitant Organic supernatant injected 10 mM ammonium formate buffer containing 0.1% HCOOH (MP A) and ACN with 0.1% HCOOH (MP B) Waters acquity I-class UPLC Waters acquity UPLC BEH C18 (50 × 2.1 mm, 1.7 µm) Gradient 5 min TF: 0.3; oven T: 40; AS T: RT; Vinj: 0.15 19
Gorijavolu et al (2018) Rucaparib 13C,2H3-rucaparib 5–5000 pg/mL 400 µL LLE with EtOAc and DCM Before extraction Reconstitution with mobile phase 10 mM ammonium formate and MeOH 20:80 vol/vol Agilent 1200 series HPLC Waters Xbridge C18 (50 × 4.6 mm, 5 µm) Isocratic 2.5 min TF: 0.7; oven T: 40; Vinj: 5 18
van Andel et al (2017) Niraparib, Niraparib-M1 M002151 (niraparib deuterated IS), D5-M1 1–500 ng/mL for niraparib and Niraparib–M1 100 µL PP (1 + 2 ACN/MeOH) then dryness Before precipitation Reconstitution with initial mobile phase 20 mM ammonium acetate in water (MP A) and 0.1% HCOOH in ACN/MeOH 50:50 (vol/vol) (MP B) Waters acquity I-class with diverter valve Waters SunFire C18 (50 × 2.1 mm, 5 µm) Gradient 7 min TF: 0.7; oven T: 40; AS T: 8; Vinj: 3 17
Rolfo et al (2015) Olaparib 2H8-olaparib 0.5–500 and 0.02–20 µg/mL 100 µL SPE elution with ACN Before extraction Reconstitution with mobile phase 1 mM pH 3 ammonium formate buffer/ACN (73/27 vol/vol) N/A Waters xterra phenyl (50 × 2.1 mm, 3.5 µm) Isocratic N/A TF: 0.2 16
Roth et al (2014) Olaparib 2H8-olaparib 0.5–5000 ng/mL 100 µL LLE with EtOAc containing IS With extraction solvent Reconstitution with 50% ACN in water 0.1% HCOOH in H2O (MP A) and 0.1% HCOOH in ACN (MP B) Waters acquity UPLC Waters acquity UPLC BEH C18 (50 × 2.1 mm, 1.7 µm) Gradient 5 min TF: 0.25; oven T: 30; AS T: 4; Vinj: 10 15
Sparidans et al (2014) Rucaparib Gefitinib 1.25–2000 ng/mL 50 µL PP (1 + 1,5 ACN) With precipitant Dilution 1 + 1 with water 0.02% HCOOH in H2O (MP A) and MeOH (MP B) Shimadzu LC10 with diverter valve Varian polaris 3 C18-A (50 × 2 mm, 3 µm) Gradient 3 min TF: 0.5; oven T: 40; AS T: 4; Vinj: 2 14
Nijenhuis et al (2013) Olaparib 2H8-olaparib 10–5000 ng/mL 100 µL LLE with MTBE Before extraction Reconstitution with MeOH 10 mM ammonium acetate in water (MP A) and MeOH (MP B) Shimadzu LC20AD prominence with diverter valve Phenomenex gemini C18 (50 × 2.0 mm, 5 µm) + its guard column Gradient 6 min TF: 0.25; oven T: 40; AS T: 4; Vinj: 1 13
Rajan et al (2012) Olaparib 2H8-olaparib 2–2000 ng/mL 100 µL LLE with EtOAc N/A Reconstitution with mobile phase 0.1% HCOOH in water (MP A) and ACN (MP B) N/A Phenomenex luna C18(2) (50 × 2 mm, 5 µm) Gradient 7.5 min Vinj: 10 12
Sparidans et al (2011) Olaparib, melphalan Erlotinib 10–5000 ng/mL 100 µL PP (1 + 1,5 ACN) With precipitant Dilution 1 + 1 with water 0.1% HCOOH in water (10% vol/vol) + water (47% vol/vol) + MeOH (43% vol/vol) Accela system with diverter valve Waters acquity UPLC BEH C18 (30 × 2.1 mm, 1.7 µm) + its VanGuard pre-column Isocratic 1.2 min TF: 0.6; oven T: 40; AS T: 4; Vinj: 1 11
ACN, acetonitrile; AS T, autosampler temperature (°C); DCM, dichloromethane; EtOAc, ethyl acetate; HCOOH, formic acid; MeOH, methanol; MP A, mobile phase A; MP B, mobile phase B; MTBE, methyl tert-butyl ether; N/A, not applicable or not reported; oven T, oven temperature (°C); RT, room temperature; TF, total flow (mL/min); Vinj, volume of injection (µL).

One of the most important aspects of a bioanalytical method is the calibration range of the analytes that can be reliably and reproducibly quantified. This range is established by considering several factors, including whether the purpose of the analysis is TDM or PK, the expected concentration of the analyte in the matrix, and the need for dilution.53,54 In TDM, the range of analytes is generally limited because the main objective is to determine whether the analyte reaches a target drug value, often referred to as Ctrough.23,31 By contrast, the purpose of PK studies was to examine the entire concentration range from intake to elimination. Only 4 recent methods claim that their goal is TDM19–21,23 and they use the amount of data for Ctrough accumulated by earlier studies, whose goal was a more comprehensive PK analysis. It is also possible to use other methods for TDM, but some of them12,14,17,18 may require a sample dilution step because they do not properly include Ctrough values in their quantification range (Table 2).

Sample Preparation: Extraction from Human Plasma

Among the many extraction techniques available,55 the reviewed papers used protein precipitation (PP),11,14,17,19,21–23 liquid–liquid extraction (LLE),12,13,15,18,20 and solid-phase extraction (SPE) (Table 2).16 Sample preparation is necessary to avoid any ion suppression or enhancement phenomena that may occur because of matrix effects (MEs).56 As reported by Nijenhuis et al13 and Gorijavolu et al,18 when problems similar to these occur, the method is flawed and the sample preparation must be revised. In both studies, the preliminary PP had to be changed to LLE13,18 and the analytes were extracted using ethyl acetate/dichloromethane and tert-butyl methyl ether, respectively. Although LLE proved to be the superior extraction technique and was necessary in the aforementioned cases, PP was used in most of the methods11,14,17,19,21–23 because it is faster and more simple. Most of the studies that used PP extracted the analytes using various amounts of acetonitrile, starting from 1.5 volumes,11,14 all the way up to 4 volumes.19,21,23 Alternatively, one of these studies used a mixture of acetonitrile/methanol as the extraction solvent.17 In a clinical TDM environment where high throughput is essential, a fast and easy PP might be the best choice for extraction, especially because it is easily amenable to automation with 96-well plates, as reported by Shapiro et al.22 However, special care should be taken to not contaminate the electrospray ionization (ESI) source. Some authors managed to avoid this using a divert valve11,13,14,17,23 that directed the flow to the mass spectrometer only when analytes were eluting from the column.

Another aspect to consider is the matrix volume needed for the analysis as almost all authors using PP kept the volume of human plasma down to 50 µL.14,19,21–23 This is small compared with the average amount required by LLE and SPE, which ranges between 100 and 400 µL.12,13,15,16,18,20 Therefore, when the analysis requires a limited matrix volume, the use of PP is a good solution.

All methods reviewed here added an internal standard (IS) during sample preparation to compensate for losses that might occur during sample handling and injection. Specifically, almost every study presented in Table 2 used the following stable isotope-labelled analogs of the analytes as the IS (SIL IS): 2H8-olaparib, 13C,2H3-rucaparib, 13C6-niraparib, 13C,2H4-talazoparib, and 13C,2H3,15N-veliparib. These molecules have the same chemical behavior as the analytes; therefore, they elute together from the column, minimizing the potential MEs57 at their retention time. This aspect was highlighted in the work of Nijenhuis et al,13 where the use of dasatinib, erlotinib, or imatinib as an IS for olaparib led to an unacceptable lack of accuracy and precision. However, when 2H8-olaparib became commercially available and could be used as the IS, these problems were resolved. Importantly, van Andel et al17 demonstrated the use of one SIL IS for niraparib and another SIL IS for its metabolite niraparib-M1, highlighting this method as the best choice for reliable metabolite quantification. However, when an SIL IS is neither commercially available nor accessible because of its high cost, a generic organic compound could be used as the IS. This was reported by Sparidans et al,11,14 who used erlotinib and gefitinib, respectively. Regardless of the type of IS used, IS addition during the sample preparation was performed in 2 main ways in the papers reviewed. In 6 papers,13,16–18,20,23 the addition was performed by dispensing a small amount of the IS before adding the extraction solvent. In another 5 papers,11,14,15,19,21 the IS was dissolved in the extraction solvent and added to the matrix in one step. The necessity of either approach depends on the solubility of the IS; however, the dissolution of the IS in the extraction solvent can shorten sample preparation and reduce sources of error.

Some studies, such as those by Nijenhuis et al13 and Pressiat et al,19 ended the sample preparation after the extraction of the analyte, whereas others required further steps. Although direct injection of the organic extract seems to be the simplest and the fastest route, it can lead to solvent effects that interfere with chromatography in the form of fronting peaks, as reported by Bruin et al.23 In this case, the organic extract was diluted with an equivalent amount of an aqueous mobile phase to improve the peak shape of the analytes. Similarly, Sparidans et al11,14 diluted the organic extract with an equivalent amount of water, whereas Krens et al21 chose to inject a solution with an even higher (>90%) percentage of water. Other studies included an evaporation step with a subsequent reconstitution. Four of them used the mobile phase as the reconstituting agent,12,16–18 while 2 of them used a 1/1 aqueous/organic solution.15,20 Ideally, the composition of the final extract should match the initial composition of the mobile phase, which can be easily obtained through evaporation and reconstitution steps. Moreover, this procedure is often advantageous when low limits of quantification must be reached but is subject to more errors because of extensive manipulation of the sample.

Liquid Chromatography Separation

In previously published methods, chromatographic separation of PARPis was performed exclusively on reverse phase columns. Almost every author used columns functionalized with C18 chains, except Rolfo et al, who took advantage of the phenyl functionalization to retain olaparib. Regarding C18 columns, most studies selected columns based on hybrid particles built with resistance to high pH,11,13,15,16,18,19,23 although they were used at low to mid pH. In 2 studies, the researchers selected ordinary silica-based C18 columns suitable for work at low to mid pH,12,17 and one study used a core–shell column,21 which should improve both the resolution and efficiency. Finally, only a few studies used silica columns functionalized with embedded polar groups that provide good peak shapes for mixtures of polar and nonpolar compounds, especially for bases at low pH.14,20,22 This last column choice might be the most appropriate for the group of analytes and conditions used in the methods reviewed, where the elution of rucaparib, niraparib, talazoparib (weak bases), and/or olaparib (neutral) was performed under acidic conditions.11,12,14–17,19–23 In fact, protonated bases (at a pH below their pKa) are known to produce tailing, which should be minimized to avoid inaccuracies during peak integration. An FDA guideline58 recommends the tailing factor (TF) to be less than 2. However, although the TF was clearly visible in some papers, it was neither calculated nor reported by any of them.

In addition to the chemistry of the columns, their physical properties varied substantially among the methods reviewed. In about half of them, the columns were packed with a stationary phase consisting of sub 2 μm particles,11,15,19–21,23 whereas the others used columns with particle sizes in the range of 3–5 μm.12–14,16–18,22 Smaller particles exhibit superior performance than larger ones for resolution but are associated with higher back pressures and typically require specific equipment. This is referred to as ultra-high-performance liquid chromatography (UHPLC) or ultra-performance liquid chromatography (UPLC). Although the use of such a system allows for faster methods with lower solvent consumption and better resolution, it requires cleaner samples and more frequent maintenance. For laboratories that are not equipped with this instrumentation or cannot afford its cost, a few of the methods presented in Table 2 explicitly use the conventional high-performance liquid chromatography (HPLC) system.13,14,18 However, it should be noted that a similar run time13,15,17 and solvent flow13,14,18,19,21 has been reported in comparisons among the HPLC with the UHPLC methods. Therefore, the use of UHPLC in the papers reviewed here might be appropriate only when many species are to be separated and/or when a higher resolution is needed.

Most of the methods listed in Table 2 used a gradient elution program,12–15,17,19,21,23 but isocratic separation was still widely used.11,16,18,20,22 With a duration of less than 3 minutes, a major advantage of isocratic methods is their rapidity,11,18,20 although some papers in the literature debate whether isocratic separation is always faster than a gradient elution program.59 Indeed, the method developed by Sparidans et al14 used a gradient with a total run time of only 3 minutes, which is very close to the mean isocratic performance. Such a short run time translates into higher throughput and lower solvent consumption, which are necessary features in contexts where many samples are analyzed each day, such as in a hospital. Another advantage of isocratic methods is their ease of transferability from one system to another, a property of paramount importance when running an existing method from the literature in one's laboratory. Although methods using gradients are transferable, some fundamental features of the original equipment should be known (eg, the dwell volume) to avoid gradient delays and distortions after the transfer.60–62 However, none of the methods reviewed reported these features, and in addition, some did not report any hardware specifications.11,12,16,18,20,22

Mass Spectrometry Conditions

The ESI source was used in all the reviewed methods. Because it generally allows for high sensitivity with polar analytes, it is the most common ionization source chosen in clinical laboratories. Only Gorijavolu et al18 examined the atmospheric pressure chemical ionization (APCI) source as a viable alternative to ESI because APCI is generally less susceptible to MEs. However, they reported better results with ESI for the detection of rucaparib.57

Almost every reviewed paper reported the type of mass spectrometer used for this study. The most common instrument brand was AB Sciex, followed by ThermoFisher Scientific and Waters. Moreover, all the authors accurately listed the fundamental parameters of the mass spectrometer source for temperature, voltage, and gases. This information is useful when transferring a method from the literature to a laboratory using the same type of mass spectrometer.

All authors ionized PARPis in positive mode and detected them using selected reaction monitoring (SRM) or multiple reaction monitoring (MRM) MS programs. Detailed information on the fragmentation of each analyte was provided in each paper, except for that by Rajan et al.12 For olaparib, a precursor ion of 435 m/z was reported in all methods,11,13,15,16,19–21 except in a study by Bruin et al23 wherein the [M + H]+ + 1 isotopologue at 436 m/z was used instead. For rucaparib, a precursor ion of 324 m/z was described in all methods,14,22,23 except in the case of Gorijavolu et al18 wherein a molecule (the nature of which was not described) at 323 m/z was used. Furthermore, the niraparib precursor ion at 321 m/z was used by all authors,17,21,23 and the study by van Andel et al17 additionally analyzed the niraparib M1 metabolite at 322 m/z. Finally, talazoparib was reported by Bruin et al,23 with a precursor ion at 381 m/z. Regardless of the analyte involved, only a few studies11,14,19,20 have used 2 different product ions for the analyte quantification and confirmation, although the importance of this is emphasized in official guidelines.63,64 Moreover, most studies used a product ion of the analyte which had the same m/z as the product ion of its relative SIL IS.15–18,20,21,23 This choice could be problematic for quantification, particularly when the 2 product ions are collected closely—one immediately after the other—in the MS acquisition method. Instead, the product ion of the SIL IS should still contain stable isotopes for more robust quantification.

Finally, 5 studies explored the fragmentation pattern of the analytes in detail and reported the representative figures. Three of the studies examined olaparib,11,13,21 2 of them examined niraparib,17,21 and 1 of them examined rucaparib.14 No information was found for the fragmentation pattern of talazoparib. Moreover, the fragmentation pattern was not reported for any IS. This information is superfluous in studies where the SIL IS has the same product ion as the relative analyte; however, it might be useful where other compounds were used as the IS in place of the SIL IS.11,14

Validation Issues

Regulatory agencies, such as the FDA53 and EMA,54 define the key parameters required to validate a bioanalytical method. Among the methods reviewed (Table 3), 3 of them12,16,22 did not specify how validation was performed, 4 of them13,17,20,21 followed the EMA guidelines, and 7 of them11,13–15,17–19 followed the older version of the FDA guidelines from 2001.65 In addition, Jolibois et al20 also used the French Accreditation Committee (COFRAC) guidelines for biological methods, and Bruin et al23 used another validation protocol.66 Most of the studies evaluated linearity, accuracy, precision, sensitivity, and stability using the rationale proposed by the respective regulatory agencies with minimal deviations; therefore, they do not require further comments. Some studies also evaluated carryover,13,17,20,23 recovery,11,14,15,17–21 and dilution,12,17,21 and Pressiat et al19 did a thorough investigation of the possible interference of other drugs (selectivity). However, the evaluation of MEs was performed using several approaches, some of which were more exhaustive than other experimental protocols. The reason for this heterogeneity might be the complexity of ME evaluation, as already pointed out in a recent perspective article.67 Most of the reviewed studies13–15,17,19–21 used the postextraction method,68 which requires evaluation from 6 different matrices at high and low concentrations. Some of these studies used more levels of concentration19 or less than 6 matrices15 or did not specify these details,14,19 but their rationales remained the same. One study18 used an alternative method that was unclear to us. Finally, Sparidans et al11 evaluated MEs with the postcolumn infusion method,69 a qualitative procedure often used during method development to evaluate ion enhancement/suppression phenomena at the retention time of analytes.56 This method is not currently sufficient for the regulatory agencies, which require a quantitative determination of MEs. As demonstrated, the evaluation of MEs is one of the most challenging investigations required by regulatory agencies and is susceptible to misinterpretation.70

TABLE 3. - Summary of Mass Spectrometry Conditions and Validation Issues of Bioanalytical Methods for PARPi Quantification
Author (yr) Mass Spectrometer Set up Detection Mode MS Transitions (m/z) Type of Validation Validation Experiments Evaluation of ME Reference Number
Bruin et al (2020) Triple quadrupole 6500+ (AB Sciex) ESI, positive ion mode, MRM Olaparib 436 > 253, niraparib 321 > 205, rucaparib 324 > 236, talazoparib 381 > 298, veliparib 245 > 90, olaparib IS 443 > 253, niraparib IS 327 > 211, rucaparib IS 328 > 236, talazoparib IS 386 > 303, veliparib IS 250 > 90. Simplified validation procedure for TDM purposes Linearity, A&P, selectivity, sensitivity, carryover, stability Not evaluated 23
Krens et al (2020) Xevo RQ-S micro tandem MS (Waters) ESI, positive ion mode, MRM Olaparib 435 > 281, niraparib 321 > 205, olaparib IS 433 > 281, niraparib IS, 327 > 211 EMA 2011 Linearity, A&P, selectivity, sensitivity, recovery (not done according to guidelines), matrix effect, dilution, stability Determined with postextraction spike from 6 plasma batches 21
Shapiro et al (2019) API4000 system (AB Sciex) ESI, positive ion mode, SRM Rucaparib 324.1 > 293.1, rucaparib IS 331.2 > 300.2 N/A N/A N/A 22
Jolibois et al (2019) Endura (Thermo Fisher Scientific) triple quadrupole MS ESI, positive ion mode, MRM Olaparib 435.03 > 367.11 and 435.03 > 281.00; olaparib IS 443.26 > 375.16 and 443.26 > 281.07 EMA 2011 + French committee of accreditation's guidelines for biological method (2018), when appropriate. Linearity, A&P (not clear), selectivity, sensitivity, carryover, recovery, matrix effect, stability Determined with postextraction spike from 6 plasma batches 20
Pressiat et al (2018) Triple quadrupole (TQ-S) Xevo MS (Waters) ESI, positive ion mode, MRM Olaparib 435.21 > 367.18 and 435.21 > 133.12; olaparib IS 443.29 > 375.25 and 443.29 > 280.03 FDA 2001 + others Linearity, A&P, selectivity, sensitivity, recovery, matrix effect, stability Determined with postextraction spike at 3 concentration levels (not specified how many different plasma batches) 19
Gorijavolu et al (2018) API4000 triple quadrupole instrument (AB Sciex) ESI, positive ion mode, MRM Rucaparib 323.4 > 170.1, rucaparib IS 328.4 > 170.1 FDA 2001 Linearity, A&P, selectivity, sensitivity, recovery, matrix effect, stability Not evaluated with standard procedure according to EMA guidelines 18
van andel et al (2017) API5500 tandem mass spectrometer (AB Sciex) ESI, positive ion mode, MRM Niraparib 321 > 304, niraparib-M1 322 > 304, niraparib IS 328 > 304, niraparib-M1 IS 329 > 304 FDA 2001 and EMA 2011 Linearity, A&P, selectivity, sensitivity, carryover, recovery, matrix effect, dilution, stability Determined with postextraction spike from 6 plasma batches, plus one hemolyzed sample in triplicate 17
Rolfo et al (2015) N/A N/A Olaparib 435.2 > 281.1, olaparib IS 443.0 > 281.1 N/A N/A N/A 16
Roth et al (2014) Quattro micro triple quadrupole MS (Waters) ESI, positive ion mode, MRM Olaparib 435.4 > 281.1, olaparib IS 443.2 > 281.1 FDA 2001 Linearity, A&P, selectivity, recovery, matrix effect, stability Determined with postextraction spike from 5 plasma batches 15
Sparidans et al (2014) Finnigan TSQ Quantum Discovery Max triple quadrupole (Thermo Electron) ESI, positive ion mode, SRM Rucaparib 324.1 > 293 and 324.1 > 236; gefitinib 447.1 > 128 FDA 2001 Linearity, A&P, selectivity, sensitivity, recovery, matrix effect, stability Determined with postextraction spike (not specified how many plasma batches) 14
Nijenhuis et al (2013) Finnigan TSQ quantum ultra triple quadrupole (Thermo Fisher Scientific) ESI, positive ion mode, MRM Olaparib 435.3 > 367.1; olaparib IS 443.1 > 375.2 FDA 2001 and EMA 2011 in GLP environment Linearity, A&P, selectivity, sensitivity, carryover, matrix effect, stability Determined with postextraction spike at high and low levels of QC from 6 plasma batches 13
Rajan et al (2012) 5500 Qtrap (AB Sciex) MRM N/A N/A Linearity, A&P, dilution N/A 12
Sparidans et al (2011) Finnigan TSQ quantum ultra triple quadrupole (Thermo Fisher Scientific) ESI, positive ion mode, SRM Olaparib 435.2 > 281.1 and 435.2 > 367.2; erlotinib 394.1 > 278.1 FDA 2001 plus others Linearity, A&P, selectivity, recovery, stability Determined with postcolumn infusion-no matrix effect at the retention time of the analytes 11
A&P, accuracy and precision; GLP, good laboratory practice; N/A, not applicable or not reported; QC, quality control.

DISCUSSION, GAP ANALYSIS, AND OUTLOOK

Our literature search identified 13 validated bioanalytical methods for the quantification of PARPis, suggesting a high level of interest in this subject. This could be attributed to the fact that PARPis are good candidates for TDM because they satisfy most of the criteria reported in the literature.30 Moreover, interactions with other drugs, as well as renal and hepatic impairments, are situations in which clinical monitoring may be especially useful. In addition, the relationship between exposure and response to PARPis needs to be better defined. Although some clear relationships were found for both safety and efficacy, PK thresholds were not established for all the reviewed drugs. This ambiguity arises from various reasons, including frequent dose modification and treatment discontinuation during clinical trials. Accordingly, further studies should be conducted to mitigate this. TDM might be a useful tool to accomplish this task with the additional aims of reducing toxicity and increasing treatment efficacy.

This comparison of PARPi quantification methods highlighted the most relevant aspects to consider when choosing the appropriate method from the literature to reproduce. The experimental procedures of a method and its ease of transfer from one laboratory to another are crucial in the choice. Detailed information regarding all these aspects is necessary if the method has to be reproduced and used by the scientific community. In addition, the validation of these bioanalytical methods is fundamental, and several guidelines exist for different purposes. The lack of information provided by some authors and the heterogeneity of validation protocols are significant issues for the analyst that adopts a published method. In the future, newer and standardized guidelines focusing specifically on the clinical practice of TDM are required to help the scientific community develop robust and reliable bioanalytical methods.

ACKNOWLEDGMENTS

The authors thank the Italian Ministry of Health—Ricerca Corrente.

REFERENCES

1. Lindahl T, Satoh MS, Poirier GG, et al. Post-translational modification of poly(ADP-ribose) polymerase induced by DNA strand breaks. Trends Biochem Sci. 1995;20:405–411.
2. Curtin NJ, Szabo C. Poly(ADP-ribose) polymerase inhibition: past, present and future. Nat Rev Drug Discov. 2020;19:711–736.
3. Schettini F, Giudici F, Bernocchi O, et al. Poly (ADP-ribose) polymerase inhibitors in solid tumours: systematic review and meta-analysis. Eur J Cancer. 2021;149:134–152.
4. Chen Z, Wang X, Li X, et al. Deep exploration of PARP inhibitors in breast cancer: monotherapy and combination therapy. J Int Med Res. 2021;49:300060521991019.
5. US Food and Drug Administration (FDA). Center for Drug Evaluation and Research Multi Disciplinary Review, Olaparib (Capsules) 2014 (Application Number 206162Orig1s000); 2014. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206162Orig1s000ClinPharmR.pdf. Accessed May 4, 2022.
6. US Food and Drug Administration (FDA). Center for Drug Evaluation and Research Multi Disciplinary Review, Rucaparib 2016 (Application Number 209115Orig1s000); 2016. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/209115Orig1s000MultiDisciplineR.pdf. Accessed May 4, 2022.
7. US Food and Drug Administration (FDA). Center for Drug Evaluation and Research Multi Disciplinary Review, Niraparib 2016 (Application Number 208447Orig1s000); 2016. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/208447Orig1s000MultidisciplineR.pdf. Accessed May 4, 2022.
8. US Food and Drug Administration (FDA). Center for Drug Evaluation and Research Multi Disciplinary Review, Talazoparib 2018 (Application Number 211651Orig1s000); 2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/211651Orig1s000MultidisciplineR.pdf. Accessed May 4, 2022.
9. Widmer N, Bardin C, Chatelut E, et al. Review of therapeutic drug monitoring of anticancer drugs part two—targeted therapies. Eur J Cancer. 2014;50:2020–2036.
10. Hopfgartner G, Bourgogne E. Quantitative high-throughput analysis of drugs in biological matrices by mass spectrometry. Mass Spectrom Rev. 2003;22:195–214.
11. Sparidans RW, Martens I, Valkenburg-van Iersel LBJ, et al. Liquid chromatography–tandem mass spectrometric assay for the PARP-1 inhibitor olaparib in combination with the nitrogen mustard melphalan in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879:1851–1856.
12. Rajan A, Carter CA, Kelly RJ, et al. A phase I combination study of olaparib with cisplatin and gemcitabine in adults with solid tumors. Clin Cancer Res. 2012;18:2344–2351.
13. Nijenhuis CM, Lucas L, Rosing H, et al. Development and validation of a high-performance liquid chromatography–tandem mass spectrometry assay quantifying olaparib in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;940:121–125.
14. Sparidans RW, Durmus S, Schinkel AH, et al. Liquid chromatography–tandem mass spectrometric assay for the PARP inhibitor rucaparib in plasma. J Pharm Biomed Anal. 2014;88:626–629.
15. Roth J, Peer C, Mannargudi B, et al. A sensitive and robust ultra HPLC assay with tandem mass spectrometric detection for the quantitation of the PARP inhibitor olaparib (AZD2281) in human plasma for pharmacokinetic application. Chromatography. 2014;1:82–95.
16. Rolfo C, Swaisland H, Leunen K, et al. Effect of Food on the pharmacokinetics of olaparib after oral dosing of the capsule formulation in patients with advanced solid tumors. Adv Ther. 2015;32:510–522.
17. van Andel L, Zhang Z, Lu S, et al. Liquid chromatography-tandem mass spectrometry assay for the quantification of niraparib and its metabolite M1 in human plasma and urine. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1040:14–21.
18. Gorijavolu V, Gupta AK, Chowdary YA. A Sensitive bioanalytical method development and validation of Rucaparib in human plasma by LC-ESI-MS/MS. Int J Adv Res. 2018;6:836–843.
19. Pressiat C, Huynh HH, Plé A, et al. Development and validation of a simultaneous quantification method of ruxolitinib, vismodegib, olaparib, and pazopanib in human plasma using liquid chromatography coupled with tandem mass spectrometry. Ther Drug Monit. 2018;40:337–343.
20. Jolibois J, Schmitt A, Royer B. A simple and fast LC-MS/MS method for the routine measurement of cabozantinib, olaparib, palbociclib, pazopanib, sorafenib, sunitinib and its main active metabolite in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2019;1132:121844–121852.
21. Krens SD, van der Meulen E, Jansman FGA, et al. Quantification of cobimetinib, cabozantinib, dabrafenib, niraparib, olaparib, vemurafenib, regorafenib and its metabolite regorafenib M2 in human plasma by UPLC–MS/MS. Biomed Chromatogr. 2020;34:e4758.
22. Shapiro GI, Kristeleit RS, Burris HA, et al. Pharmacokinetic study of rucaparib in patients with advanced solid tumors. Clin Pharmacol Drug Develop. 2019;8:107–118.
23. Bruin MAC, de Vries N, Lucas L, et al. Development and validation of an integrated LC-MS/MS assay for therapeutic drug monitoring of five PARP-inhibitors. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1138:121925–121933.
24. US Food and Drug Administration (FDA). Center for Drug Evaluation and Research Multi Disciplinary Review, Olaparib (Tablets) 2016 (Application Number 208558Orig1s000); 2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/208558Orig1s000MultidisciplineR.pdf. Accessed May 4, 2022.
25. European Medicines Agency (EMA). Committee for Medicinal Products for Human Use (CHMP), Lynparza Capsules, Public Assessment Report (2014), Procedure Number EMEA/H/C/003726/0000. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/documents/assessment-report/lynparza-epar-public-assessment-report_en.pdf. Accessed August 31, 2022.
26. European Medicines Agency (EMA), Committee for Medicinal Products for Human Use (CHMP). Lynparza Tablets, Public Assessment Report (2018), procedure number EMEA/H/C/003726/X/0016/G. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/medicines/human/EPAR/lynparza. Accessed August 31, 2022.
27. European Medicines Agency (EMA). Committee for Medicinal Products for Human Use (CHMP), Rubraca Public Assessment Report (2018). Procedure Number EMEA/H/C/004272/0000. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/medicines/human/EPAR/rubraca. Accessed June 29, 2022.
28. European Medicines Agency (EMA). Committee for Medicinal Products for Human Use (CHMP), Talzenna Public Assessment Report (2019), Procedure Number EMEA/H/C/004674/0000. Available at: https://www.ema.europa.eu/en/documents/assessment-report/talzenna-epar-public-assessment-report_en.pdf. Accessed May 4, 2022.
29. European Medicines Agency (EMA), Committee for Medicinal Products for Human Use (CHMP). Zejula Public Assessment Report (2017), procedure number EMEA/H/C/004249/0000. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/documents/assessment-report/zejula-epar-public-assessment-report_en.pdf. Accessed August 26, 2022.
30. Groenland SL, Mathijssen RHJ, Beijnen JH, et al. Individualized dosing of oral targeted therapies in oncology is crucial in the era of precision medicine. Eur J Clin Pharmacol. 2019;75:1309–1318.
31. Mueller-Schoell A, Groenland SL, Scherf-Clavel O, et al. Therapeutic drug monitoring of oral targeted antineoplastic drugs. Eur J Clin Pharmacol. 2021;77:441–464.
32. Dirix L, Swaisland H, Verheul HMW, et al. Effect of itraconazole and rifampin on the pharmacokinetics of olaparib in patients with advanced solid tumors: results of two phase I open-label studies. Clin Ther. 2016;38:2286–2299.
33. Yu Y, Durairaj C, Shi H, et al. Population pharmacokinetics of talazoparib in patients with advanced cancer. J Clin Pharmacol. 2020;60:218–228.
34. Yu H, Steeghs N, Nijenhuis CM, et al. Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: focus on the pharmacokinetic targets. Clin Pharmacokinet. 2014;53:305–325.
35. Bruin MAC, Sonke GS, Beijnen JH, et al. Pharmacokinetics and pharmacodynamics of PARP inhibitors in oncology. Clin Pharmacokinet. 2022;61:1649–1675.
36. Velev M, Puszkiel A, Blanchet B, et al. Association between olaparib exposure and early toxicity in BRCA-mutated ovarian cancer patients: results from a retrospective multicenter study. Pharmaceuticals. 2021;14:804.
37. LaFargue CJ, Dal Molin GZ, Sood AK, et al. Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol. 2019;20:e15–e28.
38. Rolfo C, Isambert N, Italiano A, et al. Pharmacokinetics and safety of olaparib in patients with advanced solid tumours and mild or moderate hepatic impairment. Br J Clin Pharmacol. 2020;86:1807–1818.
39. Elmeliegy M, Yu Y, Litton JK, et al. Exposure-safety analyses of talazoparib in patients with advanced breast cancer and germline BRCA1/2 mutations in the EMBRACA and ABRAZO trials. J Clin Pharmacol. 2020;60:1334–1343.
40. Konecny GE, Oza AM, Tinker AV, et al. Population exposure-efficacy and exposure-safety analyses for rucaparib in patients with recurrent ovarian carcinoma from Study 10 and ARIEL2. Gynecol Oncol. 2021;161:668–675.
41. Olaparib FDA Prescribing Information. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208558s014lbl.pdf. Accessed November 23, 2022.
42. Niraparib FDA Prescribing Information. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/208447s015s017lbledt.pdf. Accessed November 23, 2022.
43. Rucaparib FDA Prescribing Information. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/209115s004lbl.pdf. Accessed November 23, 2022.
44. Talazoparib FDA Prescribing Information. 2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211651s000lbl.pdf. Accessed November 23, 2022.
45. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123–134.
46. Sandhu SK, Schelman WR, Wilding G, et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2013;14:882–892.
47. Swisher EM, Lin KK, Oza AM, et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017;18:75–87.
48. Hoffman J, Chakrabarti J, Plotka A, et al. Talazoparib has no clinically relevant effect on QTc interval in patients with advanced solid tumors. Anticancer Drugs. 2019;30:523–532.
49. European Medicines Agency (EMA). Guideline on the Evaluation of the Pharmacokinetics of Medicinal Products in Patients With Decreased Renal Function, 2015. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-evaluation-pharmacokinetics-medicinal-products-patients-decreased-renal-function_en.pdf. Accessed October 27, 2022.
50. European Medicines Agency (EMA). Guideline on the Evaluation of the Pharmacokinetics of Medicinal Products in Patients With Decreased Renal Function, 2004. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/documents/scientific-guideline/note-guidance-evaluation-pharmacokinetics-medical-products-patients-impaired-renal-function_en.pdf. Accessed October 27, 2022.
51. European Medicines Agency (EMA). Guideline on the Evaluation of the Pharmacokinetics of Medicinal Products in Patients With Impaired Hepatic Function, 2005. Amsterdam, The Netherlands: EMA. Available at: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-evaluation-pharmacokinetics-medicinal-products-patients-impaired-hepatic-function_en.pdf. Accessed October 27, 2022.
52. Akce M, El-Khoueiry A, Piha-Paul SA, et al. Pharmacokinetics and safety of niraparib in patients with moderate hepatic impairment. Cancer Chemother Pharmacol. 2021;88:825–836.
53. US Food and Drug Administration (FDA), Center for Drug Evaluation and Research. Bioanalytical Method Validation: Guidance for Industry; 2018. Silver Spring, MD: FDA. Available at: https://www.fda.gov/files/drugs/published/Bioanalytical-Method-Validation-Guidance-for-Industry.pdf.
54. European Medicines Agency (EMA). Committee for Medicinal Products for Human Use (CHMP). Guideline on Bioanalytical Method Validation 2011, 2011. Available at: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-bioanalytical-method-validation_en.pdf. Accessed June 20, 2022.
55. Chang MS, Ji Q, Zhang J, El-Shourbagy TA. Historical review of sample preparation for chromatographic bioanalysis: pros and cons. Drug Dev Res. 2007;68:107–133.
56. Taylor PJ. Matrix effects: the Achilles heel of quantitative high-performance liquid chromatography–electrospray–tandem mass spectrometry. Clin Biochem. 2005;38:328–334.
57. Clinical and Laboratory Standard Institute. Liquid Chromatography-Mass Spectrometry Methods; Approved Guideline. CLSI Document C62A. 2014;34. Wayne, PA: CLSI. Available at: https://clsi.org/media/1346/c62a_sample.pdf.
58. US Food and Drug Administration (FDA), Center for Drug Evaluation and Research. Reviewer Guidance, Validation of Chromatographic Methods, 1994. Silver Spring, MD: FDA. Available at: https://www.fda.gov/media/75643/download. Accessed July 6, 2022.
59. Schellinger AP, Carr PW. Isocratic and gradient elution chromatography: a comparison in terms of speed, retention reproducibility and quantitation. J Chromatogr A. 2006;1109:253–266.
60. Snyder LR, Kirkland JJ, Dolan JW. Introduction to Modern Liquid Chromatography. Hoboken, NJ: John Wiley & Sons, Inc.; 2009.
61. Dolan J. How fast can a gradient Be run? LCGC N Am. 2011;29:652–657.
62. Hendriks G, Franke JP, Uges DRA. New practical algorithm for modelling retention times in gradient reversed-phase high-performance liquid chromatography. J Chromatogr A. 2005;1089:193–202.
63. WADA Laboratory Committee. WADA Technical Document—TD2010IDCR. Identification Criteria for Qualitative Assays: Incorporating Column Chromatography and Mass Spectrometry, 2010. Montreal, Canada: WADA. Available at: https://www.wada-ama.org/sites/default/files/resources/files/WADA_TD2010IDCRv1.0_Identification%20Criteria%20for%20Qualitative%20Assays_May%2008%202010_EN.doc.pdf. Accessed June 22, 2022.
64. U.S. Department of Health and Human Services, Food and Drug Administration (FDA), Center for Veterinary Medicine, Guidance for Industry. Mass Spectrometry for Confirmation of the Identity of Animal Drug Resides. 2003. Available at: http://www.fda.gov/AnimalVeterinary/GuidanceComplianceEnforcement/GuidanceforIndustry/default.htm. Accessed August 12, 2022.
65. US Food and Drug Administration (FDA), Center for Drug Evaluation and Research. Bioanalytical Method Validation: Guidance for Industry; 2001. Silver Spring, MD: FDA.
66. Nuland M, Rosing H, Schellens JHM, Beijnen JH. Bioanalytical LC–MS/MS validation of therapeutic drug monitoring assays in oncology. Biomed Chromatogr. 2020;34:1–22.
67. King K, Li P, Pietrasiewicz A, et al. Perspectives on updates, clarifications and controversies in chromatographic assay guidance for bioanalytical method validation from major regulatory agencies and organizations. Biomed Chromatogr. 2021;35:e5030–e5038.
68. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Strategies for the assessment of matrix effect in quantitative bioanalytical methods based on HPLC−MS/MS. Anal Chem. 2003;75:3019–3030.
69. Bonfiglio R, King RC, Olah TV, et al. The effects of sample preparation methods on the variability of the electrospray ionization response for model drug compounds. Rapid Commun Mass Spectrom. 1999;13:1175–1185.
70. Saar E, Beyer J, Gerostamoulos D, et al. The analysis of antipsychotic drugs in human matrices using LC-MS(/MS): review: the analysis of antipsychotic drugs in human matrices. Drug Test Anal. 2012;4:376–394.
Keywords:

PARPis; TDM; LC-MS/MS; bioanalytical methods; validation

Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology.