Next Article in Journal
Multicomponent X-ray Shielding Using Sulfated Cerium Oxide and Bismuth Halide Composites
Next Article in Special Issue
Preanalytical Quality Evaluation of Citrate Evacuated Blood Collection Tubes—Ultraviolet Molecular Absorption Spectrometry Confronted with Ion Chromatography
Previous Article in Journal
Effects of the Coculture Initiation Method on the Production of Secondary Metabolites in Bioreactor Cocultures of Penicillium rubens and Streptomyces rimosus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Analytical and Clinical Validation of Assays for Volumetric Absorptive Microsampling (VAMS) of Drugs in Different Blood Matrices: A Literature Review

1
Doctoral Study Program, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
2
Division of Pharmacology and Therapy, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
3
Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran—Hasan Sadikin Hospital, Bandung 40161, Indonesia
4
Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
5
Department of Pharmacy, Radboud University Medical Center, Research Institute for Medical Innovation, 6255 HB Nijmegen, The Netherlands
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(16), 6046; https://doi.org/10.3390/molecules28166046
Submission received: 13 July 2023 / Revised: 10 August 2023 / Accepted: 11 August 2023 / Published: 14 August 2023
(This article belongs to the Special Issue Analytical Techniques in Pharmaceutical and Biomedical Analysis II)

Abstract

:
Volumetric absorptive microsampling (VAMS) is the newest and most promising sample-collection technique for quantitatively analyzing drugs, especially for routine therapeutic drug monitoring (TDM) and pharmacokinetic studies. This technique uses an absorbent white tip to absorb a fixed volume of a sample (10–50 µL) within a few seconds (2–4 s), is more flexible, practical, and more straightforward to be applied in the field, and is probably more cost-effective than conventional venous sampling (CVS). After optimization and validation of an analytical method of a drug taken by VAMS, a clinical validation study is needed to show that the results by VAMS can substitute what is gained from CVS and to justify implementation in routine practice. This narrative review aimed to assess and present studies about optimization and analytical validation of assays for drugs taken by VAMS, considering their physicochemical drug properties, extraction conditions, validation results, and studies on clinical validation of VAMS compared to CVS. The review revealed that the bio-analysis of many drugs taken with the VAMS technique was optimized and validated. However, only a few clinical validation studies have been performed so far. All drugs that underwent a clinical validation study demonstrated good agreement between the two techniques (VAMS and CVS), but only by Bland–Altman analysis. Only for tacrolimus and mycophenolic acid were three measurements of agreement evaluated. Therefore, VAMS can be considered an alternative to CVS in routine practice, especially for tacrolimus and mycophenolic acid. Still, more extensive clinical validation studies need to be performed for other drugs.

1. Introduction

The essential goal of clinical pharmacology is to understand the dose–concentration–effect relationship. The study of pharmacokinetics seeks to explain the time course of drug concentrations in the body (dose–concentration relationship). However, the time course of drug concentrations cannot predict the magnitude of the drug effect (concentration–effect relationship) [1,2]. This dose–concentration–effect relationship is now used as the central concept of therapeutic drug monitoring (TDM), which individualizes drug dosage by attaining specific target plasma concentrations (therapeutic ranges) guided by the measurement of plasma drug concentrations [3]. TDM is used for assessing efficacy, diagnosing undertreatment, preventing adverse effects, guidance to stop the treatment, monitoring and detecting drug interaction, and monitoring adherence [4]. It is recommended for drugs with a narrow therapeutic range, significant pharmacokinetic variability, and a clear relationship between drug concentrations and clinical response [5].
The sampling technique commonly used to routinely measure drug concentration, either for TDM or pharmacokinetic studies, is conventional venous sampling (CVS), which draws blood from the vein. CVS can lead to unexpected complications, such as hematoma and thrombophlebitis [6,7]. This technique requires much blood for each sample (up to 5 mL), is invasive and inconvenient, has a relatively high cost, sometimes needs special conditions (a cold chain) for transport, and samples must be centrifuged before storage as plasma or serum. If the sample is not immediately analyzed, it must be frozen at −20 up to −80 °C. These characteristics cause CVS to be less flexible for application in the field [8,9,10]. An advantage of CVS is that the sample volume is large, while analysis only needs a small volume; therefore, the leftover sample could be stored and re-analyzed [6,7].
Considering the many limitations of CVS, which can complicate TDM programs or pharmacokinetic studies, there have been many innovations in sampling techniques for drug concentration measurement in the past decade, called microsampling techniques. Microsampling is a sampling technique that only takes a small volume of samples (<100 µL for blood) from the body for analysis; it is less invasive, less painful, and more efficient. This technique is also more practical compared to CVS [11]. Therefore, this technique may improve routine clinical care in TDM and benefit pharmacokinetic studies, including those with children as subjects. Some microsampling techniques that have already been developed are capillary microsampling (CMS), dried blood spots (DBS), dried plasma spots (DPS), plasma preparation technologies, solid-phase microextraction (SPME), and volumetric absorptive microsampling (VAMS) [12]. VAMS and DBS have been the most researched to analyze drugs [12,13,14,15,16,17]. Previously, the most commonly used and straightforward microsampling design was DBS, which requires small volume sizes (30 µL per spot). But DBS has some limitations primarily related to the effect of hematocrit values that may affect measured concentrations. The VAMS technique was then developed to tide over the boundaries of DBS [12,18,19]. VAMS is a promising sample-collection technique for the quantitative analysis of drugs, which only takes a 10–50 µL volume of samples by an absorbent tip [20,21]. Compared to CVS, the VAMS technique has some superiority in practicality and flexibility of the sampling process because it only needs a small sample volume. The sampling process is relatively fast and less invasive and there is no need to freeze the sample before transport; thus, it may be more cost-effective, there is no need for centrifugation before storage, and it also reduces the amount of freezers needed to keep samples in the long term [18,19,22]. Therefore, this VAMS technique is more flexible for application in the field than CVS and can be innovative for sampling processes that are more practical and simpler [18,22].
Analytical methods for drugs need to be optimized and validated before the method is used [23]. Furthermore, VAMS can only be implemented in routine practice care, replacing CVS, after it has been successfully validated in a clinical validation study. In a clinical validation study, paired drug concentrations from samples taken by VAMS and CVS are obtained from actual patients and are statistically analyzed [24]. Until now, no review has described the clinical validation study of drugs that have undergone optimization and analytical validation. But there was a review describing the analytical validation of some drugs [25].
This narrative review aimed to assess and present studies about the optimization and validation of assays for drugs taken by VAMS, considering their physicochemical properties, extraction conditions, validation results, and the clinical validation of VAMS versus CVS.

2. Volumetric Absorptive Microsampling (VAMS)

VAMS is the newest innovation in microsampling techniques. VAMS was designed to absorb a minimal and fixed volume, such as 10, 20, and 50 µL. VAMS devices contain a white hydrophilic pore absorbent tip attached to a plastic handler [19]. The picture of the VAMS device is shown in Figure 1, which comes with two different types of devices, namely clamshells and cartridges. The sampling procedure with VAMS is simple: dipping the tip of VAMS in the location that has been punctured using a lancet at a 45° angle for 2–4 s until the tip is entirely red. The sample for this device is not only blood; VAMS has also been used to collect urine, saliva, or other liquid biological samples [18,19]. After obtaining the sample, the devices are dried at room temperature for at least 2 h. The samples can be transferred at room temperature for storage or analysis [19,26]. The drying of the devices could correlate with the limitation of VAMS itself. Care is also taken to ensure that the tips do not touch either other tips or their surroundings to prevent blood transfer during drying. If it touches others, it could be contaminated. Also, the variation in drying times could be an issue with reproducible recovery. Drug extraction is more inconvenient when samples become drier. Understanding the effect of drying on VAMS samples is essential for method development and validation. The total time for drying depends on the size of the absorbent tip on the VAMS devices. It has been known that 1 h of drying at room temperature is adequate when using 10 µL VAMS. Furthermore, the drying time also could generate the degradation of the sample; thus, the stability of analytics in drying time needs to be evaluated [18,27].
The dried sample from the VAMS technique has better stability than a liquid sample. Some studies reported that the desired analyte was stable for over a month in dried conditions at room temperature for an extended period (more than one month) [28,29,30,31,32,33,34,35,36,37]. Storage conditions need to be considered for the samples obtained from the VAMS technique, such as storing them in a closed and dark container and having a desiccant to prevent degradation [22]. The VAMS manufacturer provides clamshell storage with a hole for airflow that is important for drying. If the appropriate storage conditions are applied, it could extend the stability of the drugs [16,22,38]. However, stability still depends on many factors, such as analyte and storage conditions (temperature and how to save the sample). In the VAMS device, samples need to be stored in a tightly closed container that is dark and possibly with desiccant. Therefore, stability parameters must be evaluated during analytical method validation [22].
For the sample analysis, sample preparation must be conducted for the extraction process. Before the extraction, the tip from VAMS devices should be released from its handler [13,19,22,26,28], or the whole VAMS device could be used using automatic machines [19,22,39] to overcome the hematocrit issue of DBS [19]. The choice of type and solvent volume, extraction time, and extraction procedure are the most important in sample preparation for the VAMS technique. Ideally, a suitable extraction procedure is a procedure that can result in high reproducibility, maximal recovery of drugs, and minimal matrix effect [19,40]. In studies that used the VAMS technique, the type of solvent varied from 100% water [30,38,41,42,43] to 100% organic solvent [13,26,31,32,39,44,45,46,47,48]. Acid or base could be added to increase recovery [13,28,40,42,43,46,49], and the extraction time varied from minutes to one hour. A longer extraction time could increase the analyte’s recovery [19,39]. The VAMS technique, since it is a microsampling device, has a sensitivity issue based on its volume. It is known that the reduced sample volume impacts the sensitivity since the smaller volume also leads to small analyte/concentrations. Nevertheless, the small blood volume is sufficient for clinical assays because modern instruments are highly sensitive and can detect low concentrations of drugs and metabolites. Consequently, this highly sensitive instrument could overcome the issue of sensitivity of small-volume samples, especially in VAMS [27,50].
Assays for many drugs taken by VAMS have been optimized and analytically validated, such as cefepime, midazolam, tacrolimus, paracetamol, and voriconazole. Yet, only a few assays underwent clinical validations [26,51,52,53,54]. Some studies comparing VAMS and CVS demonstrated a strong correlation of drug concentrations from samples obtained by both sampling techniques, for example, lamotrigine, levetiracetam, phenytoin, valproic acid, and albendazole (r > 0.95) [51,52]. Furthermore, agreement analysis is also used for clinical validation. The clinical validation is discussed in another section [53,54].

3. Analytical Validation of Volumetric Absorptive Microsampling (VAMS) Assays of Drugs

3.1. Optimization and Validation

Before clinical validation, the analytical method of all drugs must be optimized and validated, which is different for every drug. Developing an analytical method consists of optimizing the method, including the extraction/detection of an analyte and subsequent validation of the analytical method [23]. Proper optimization and validation methods are essential for evaluating and interpreting bioavailability, bioequivalence, pharmacokinetic, and toxicokinetic research, which means it has a vital role in discovering, developing, and manufacturing drugs [55,56,57]. After optimization, the analytical method’s quality must be ensured to produce accurate and precise data [58,59]. Validation of the analytical method is a process to prove that the analytical method that has been optimized is appropriate for the analysis of the desired analyte [23]. Again, this process is vital to receive the quality and safety of the end product, especially in the pharmaceutical industry [58,59,60]. Selecting the extraction solvent and other conditions is essential for optimizing and validating. The extraction procedure should result in a reproducible, maximum recovery of the analytes of interest and low recovery of different compounds to reduce the matrix effect. The optimization of extraction conditions can be conducted by an experimental process [19,23].

3.1.1. Optimization and Validation of Assays for Acidic Drugs

As can be seen in Table 1 for the optimization and validation for acidic drugs, the extraction solvent in the conducted studies with VAMS varied from water:methanol [61,62], methanol 100% [14,39,46,63], acetonitrile 100% [16,49], and methanol:acetonitrile [13,51]. Some studies added an acid or base to increase the recovery [13,46,51,62]. Vortexing was used in some studies [13,16,63]. Shaking [39,62], sonication [14,51,61,64], and vortexing–sonication were also used to optimize acidic drugs [46,63,64].
In addition, the extraction time varies between studies, from a few minutes to more than one hour. Increasing the extraction time may result in a higher recovery of the analytes [19]. A difference in the recovery of 8.8% was observed for tamoxifen between vortexing for one hour and a combination of vortexing for 1 min and sonication for 25 min (100.7% and 91.9%, respectively). But the use of different extraction solvents may also influence the result of their recovery [13,63]. Details can be seen in Table 1.
Assays for acidic drugs, such as emixustat, paracetamol, cefepime, ethosuximide, felbamate, phenobarbital, phenytoin, primidone, topiramate, zonisamide, and tamoxifen were all validated and met the acceptance criteria by the FDA (Food and Drugs Administration) and/or EMA (European Medicines Agency) guidelines [16,46,62,63,64]. Other acidic drugs have also been validated, but some data were missing. Details of the validation results and the parameters missing from the analytical validation of acidic drugs are shown in Table 1, including the stability data. Most acidic drugs have a lower limit of quantification (LLOQ) below the concentration range in plasma or serum [65,66,67,68,69,70,71,72,73,74,75,76], except for atorvastatin with an LLOQ of 0.001 µg/mL [13], but clinically relevant concentrations are between 0.00033 and 0.01208 µg/mL [77]. Therefore, most acidic drugs could now undergo clinical validation, except atorvastatin, since its LLOQ was above the concentration range of the drug in plasma or serum.

3.1.2. Optimization and Validation of Assays for Basic Drugs

The extraction solvent in the performed studies with VAMS for basic drugs varied from water 100% [38], water:methanol [28], water:acetonitrile [37], methanol 100% [14,31,45,78,79], acetonitrile 100% [16], methanol:acetonitrile [13,48,80], methanol:ethyl acetate [81], and methanol:zinc sulfate [82]. Some studies added an acid or base to increase the analyte’s recovery [13,28]. Vortexing is used in most studies [13,16,38,45,48]. Shaking [79], sonication [14,78], thermomixing [28,37], vortexing–sonication [80], thermomixing–vortexing [31], and sonication–shaking were also used in the optimization of basic drugs [81,82]. Details can be seen in Table 2.
The use of extraction solvent, extraction time, and other conditions may influence the recovery of the analyte. However, there was no difference in the recovery of midazolam between using methanol 100% with a shaker for one hour and methanol–acetonitrile (1:1) with vortexing for one hour (99.95% vs. 101.2%) [13,79], which shows that the chosen combination of extraction solvent, extraction time, and other conditions may result in the same analyte’s recovery.
Most basic drugs, including miltefosine, hydroxychloroquine, voriconazole, voriconazole N-oxide, lamotrigine, and rufinamide, have been validated analytically. Assays met the acceptance criteria by the FDA and/or EMA guidelines [16,31,38,80]. Also, other basic drugs described in Table 2 that have been validated showed some missing parameter data. Details of the validation results from the analytical validation of basic drugs are shown in Table 2, incorporating stability data and the analytical instrument used for each drug’s analytical method. The LLOQ of all basic drugs were below the concentration ranges of their drugs [13,14,16,28,31,37,38,45,48,70,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96]. For example, mitotane has an LLOQ of 1 µg/mL [82], and its plasma concentration range is 11.3–23.3 µg/mL [92]. The assays for all other basic drugs could undergo clinical validation studies to compare with the assays after CVS.

3.1.3. Optimization and Validation of Assays for Neutral Drugs

As can be seen in Table 3 for the optimization and validation of assays for neutral drugs, the extraction solvent in the conducted studies with VAMS varied from water:methanol [61], acetonitrile 100% [16], methanol:zinc sulfate [97], and acetonitrile:zinc sulfate [53]. No studies used acid or base modification to enhance recovery since the analyte is a neutral drug. The most used procedure was vortexing. It was used for carbamazepine, lacosamide, oxcarbazepine, perampanel, levetiracetam, and tacrolimus [16,53]. A shaker [97] was used for sonication for tacrolimus, sirolimus, everolimus, and cyclosporin A [61]. Three different conditions were used to extract tacrolimus. Shaking for 6 min [97], vortexing for 1 min [53], and sonication for 15 min [61] were used to extract tacrolimus, resulting in different recoveries (98.5%, 80%, and 81.8%, respectively). The use of a shaker to extract tacrolimus gives the best recovery. Still, the extraction solvent may influence the recovery since the three used different extraction solvents (methanol:zinc sulfate, acetonitrile:zinc sulfate, and water:methanol, respectively) [53,61,97].
All assays for neutral drugs, such as tacrolimus, carbamazepine, lacosamide, oxcarbazepine, perampanel, and levetiracetam, were validated and met the acceptance criteria by the FDA and/or EMA guidelines [16,97]. Table 3 shows the detailed validation results and the missing parameter data of other neutral drugs. Most neutral drugs that underwent a validation study have an LLOQ concentration below the plasma/serum concentration range of their drugs, except for everolimus, perampanel, and sirolimus [16,53,61,97,98,99,100,101,102,103,104,105,106]. Everolimus has an LLOQ of 0.02 µg/mL [61], whereas its concentration range is 0.003–0.008 µg/mL [100]. The LLOQ of perampanel was 0.022 µg/mL [16], and its concentration range was 0.019–2.436 µg/mL [104]. Sirolimus has 0.02 µg/mL as the LLOQ concentration [61] and a plasma/serum concentration range within 0.004–0.012 µg/mL [105]. Hereafter, most neutral drugs could undergo a clinical validation study since their LLOQ concentration was below the concentration range of their drugs, except for everolimus, perampanel, and sirolimus.
In summary, many drugs taken by VAMS underwent optimization and validation of analytical methods. Assays for acid, basic, and neutral drugs could be optimized and validated with different preparation or extraction procedures. Assays for most acidic drugs [16,18,44,51,52,65,66,67], most basic drugs [14,16,28,31,38,45,79,80], and all neutral drugs have been validated and met the acceptance criteria by FDA and/or EMA guidelines [16,53,61,97].

4. Clinical Validation of Volumetric Absorptive Microsampling (VAMS) of Drugs

Generally, a new sampling technique can only be implemented in routine practice by replacing CVS after it has been successfully validated in clinical validation. This clinical validation aims to show that the results from the new sampling technique are exchangeable with what is gained from the conventional one. In this case, a clinical validation study compares CVS with VAMS, obtained by finger prick, if both samples are taken simultaneously in the same patient (paired samples). The sample was analyzed and statistically evaluated. Ideally, the whole concentration range is validated based on a large enough sample size of at least 40 patients/subjects. The sample can be collected at a single time point (trough or peak), or paired samples are taken at 2–3 time points covering the whole concentration range in blood, serum, or plasma with for CVS volume samples according to standardized volume; for VAMS, this is adjusted to the tip used (10–50 µL) [24,107].
Clinical validation studies can incorporate three different agreement measures: Passing–Bablock/Deming regression analysis, Bland–Altman analysis displaying agreement, and an assessment of predictive performance [24,107]. Passing–Bablock/Deming regression analysis is used by plotting the concentration of the new VAMS technique against results from the conventional CVS technique as a standard concentration to allow for measurement errors on both the x and y-axis, and this technique is not hampered by a few outliers. Bland–Altman analysis is another means to provide insight into the agreement between the two techniques. The ultimate goal of VAMS blood concentration measurement is to predict the corresponding CVS (plasma) concentration. Predictive performance can be quantified by calculating the median percentage prediction error (MPPE), given by the median [100% × (Predicted concentration − Observed concentration)/Observed concentration]. Then, the median absolute percentage prediction error (MAPE) can be calculated by [100% × │(Predicted concentration − Observed concentration)│/Observed concentration]. The MMPE is a measure of bias, while the MAPE measures precision. Acceptance criteria can vary, but MMPE and MAPE <15% values are often applied [107]. Until now, only eight studies have performed a clinical validation study to compare VAMS and CVS, such as in albendazole, tacrolimus, radiprodil, and mycophenolic acid (Table 4).
The analytical method for albendazole, a benzimidazole derivative and an anthelmintic for humans, from the sample taken by VAMS, was developed and validated by Schulz et al. [51] in 2019 and clinically validated by comparing the VAMS samples with plasma samples. In this study, 10 subjects receiving albendazole were sampled 10 times using CVS or VAMS for capillary blood until 24 h post-treatment. Bland–Altman analysis was used to determine the agreement between the two methods. The results showed a good agreement between the two matrices (VAMS blood and plasma). But in this study, measures of the agreement included only Bland–Altman analysis, and Passing–Bablock/Deming regression analysis and predictive performance were not assessed, and the number of subjects did not meet the minimum number of subjects [51]. Therefore, it would be better if the clinical validation of VAMS samples of albendazole were repeated using the three agreement measures with the minimum number of subjects in the future.
In 2020, Veenhof et al. [108] conducted a study about the clinical validation of VAMS and DBS versus CVS for tacrolimus. Tacrolimus is an immunosuppressant drug that has been part of routine transplant patient care for decades. This study included 88 matched samples from 72 patients, and VAMS and whole blood samples were taken during a regular visit. Because of insufficient sample quality, 62 duplicate VAMS samples were available for analysis. All three different measures of the agreement were performed. The Passing–Bablok fit was y = 0.88x + 0.01 (95% CI slope, 0.81–0.97; 95% CI intercept, −0.47–0.39), showing no significant constant difference, but a significant systematic difference of 12% lower tacrolimus concentration in VAMS to whole blood (WB) was established. This systematic difference was later used to obtain the following conversion formula: [tacrolimus WB concentration] = [tacrolimus VAMS concentration]/0.88. This conversion formula was used to recalculate all VAMS values, and these recalculated values were used in the Bland–Altman analysis. No significant bias was found in the Bland–Altman analysis, with a mean WB/VAMS ratio of 1.00 (95% CI 0.98–1.02), showing good agreement. Because of the correction factor, the bias estimation in the predictive performance was negligible. The MPPE and MAPE were within acceptable limits. This study suggested that VAMS (after correction) can be used in transplant patient care for tacrolimus monitoring [108].
Tacrolimus was also investigated by Vethe et al. [97] in 2019. In this study, two 12 h pharmacokinetic investigations were performed at the hospital in the early phase after transplantation (2–8 weeks after transplantation) when the tacrolimus dose was stable. Blood sampling, both VAMS and liquid venous blood, was performed at 13 time points after administration of individualized morning doses of tacrolimus. The agreement was measured using Passing–Bablock analysis, which was fit y = −0.5869 + 1.012x, and Bland–Altman analysis showed good agreement between the two matrices. It reported that less than 8% of the VAMS versus liquid venous sample pairs showed differences outside a ±20% range. Although the predictive performance was not assessed and the minimum number of subjects was not reached, the study suggested that the VAMS method is considered suitable for routine TDM in renal transplant recipients, either by trough or rich sampling strategies [97].
In 2020, an analytical method for immunosuppressant agents, such as mycophenolic acid, tacrolimus, sirolimus, everolimus, and cyclosporin, was developed and validated for samples taken by VAMS by Gonzalez et al. [61], and the two most used drugs (tacrolimus and mycophenolic acid) were clinically validated versus CVS/venous WB. In this study, 53 subjects (for tacrolimus) and 20 subjects (for mycophenolic acid) were obtained from adult patients on immunosuppressant treatment after liver transplantation. Passing–Bablock regression and Bland–Altman analysis were performed to evaluate the agreement between VAMS and CVS. Both tacrolimus and mycophenolic acid showed a strong linear relationship in the Passing–Bablock regression analysis. For tacrolimus, the association was not one of the equalities as the slope cannot be considered 1 since the estimated slope was 1.16, and its 95% CI (1.159–1.368) does not contain the value 1.0. Therefore, the estimated Passing–Bablok regression fit was used to transform VAMS concentration values into venous WB concentrations, and a new Passing–Bablok regression analysis was executed to assess the agreement between venous WB concentration and converted VAMS concentration. An agreement can be seen with an estimated slope of 0.997 (95% CI, 0.909–1.074). A robust linear relation and agreement can be seen for the mycophenolic acid. The estimated slope is similar to 1 (0.976 with 95% CI 0.936–1.053). Hence, no transformation or correction of the data is required. The MPPE and MAPE were within the acceptance criteria (<15%) for the predictive performance [61]. The VAMS method is suitable for routine TDM in liver transplant recipients for tacrolimus (after transformation/correction). For mycophenolic acid, more subjects should be added for another clinical validation before it can be used in routine practice.
Zwart et al. [109] developed and validated an LC-MS/MS assay capable of quantifying tacrolimus, everolimus, sirolimus, cyclosporin, mycophenolic acid, creatinine, and iohexol simultaneously in DBS and VAMS samples. Tacrolimus, mycophenolic acid, creatinine, and iohexol assays were clinically validated against plasma in 2022. This study sampled twenty-five stable kidney transplant recipients after more than one year of receiving immunosuppressive therapy. Passing–Bablock regression showed adequate linearity to CVS. The Passing–Bablok regression slope was 1.05 (95% CI, 0.98–1.14) for tacrolimus for the individual concentrations. No correction was used for the Bland–Altman analysis. For mycophenolic acid, the Passing–Bablok regression slope of 0.72 (95% CI, 0.66–0.77); therefore, this data requires correction with VAMS to plasma conversion factors of 1/0.73, and after correction, the corrected VAMS mycophenolic acid concentrations showed adequate linearity with the plasma concentrations demonstrating a Passing–Bablok slope of 1.07 (95% CI, 0.97–1.13). Bland–Altman analysis showed good agreement [109]. Future research should add more subjects for further clinical validation of tacrolimus and mycophenolic acid.
Radiprodil (UCB3491) is a selective negative allosteric modulator of NR2B-containing N-methyl-d-aspartate (NMDA) receptors and is currently under development for treating infantile spasms. An oral suspension was applied for pediatric use. In this study, radiprodil was involved in the clinical validation study in which 10 subjects were sampled at 15 time points post-dose, both with VAMS and CVS used for plasma samples. The agreement for this clinical validation was assessed using Bland–Altman analysis and showed good agreement with a mean bias of −11.4% (−39.1, 16.3). Passing–Bablock regression analysis and predictive performance assessment have yet to be performed [81]. We suggest that the other two agreement measures are assessed with a minimum of 40 subjects in future research.
The results of clinical validation studies of several drugs are summarized in Table 4.
According to Table 4, only some clinical validation studies met the requirements for measurement of agreement. Clinical validation studies ideally need three different agreement measurements: Passing–Bablock/Deming regression analysis; Bland–Altman analysis displaying agreement; and assessment of predictive performance [24,107]. Only tacrolimus and mycophenolic acid were assessed with the three measurements and showed good agreement [61,108]. Hence, clinical validation of the other drugs, namely albendazole and radiprodil, must be completed with other agreement measurements that still need to be conducted [51,81].

5. Future Prospective of Volumetric Absorptive Microsampling (VAMS)

VAMS may be an alternative to CVS to support clinical applications, such as TDM and pharmacokinetic studies, for research purposes. There has been much research on developing and validating analytical methods for drugs taken with VAMS. Yet, in the validation process, the LLOQ concentration needs to be below clinically relevant concentrations of the drugs to ensure that the method can be appropriately applied. More research is needed on the clinical validation of VAMS to make this technique applicable in routine practice as an alternative or replacement for conventional sampling. During clinical validation studies, sample acquisition by well-trained personnel is critical in obtaining high-quality samples. Also, re-analysis of VAMS samples must be incorporated in future studies to prove the reproducibility of results; thus, the samples must be taken more than once.

6. Conclusions

VAMS is a promising sampling technique for TDM and pharmacokinetic studies, and assays of many drugs have been developed and analytically validated. However, only a few studies have performed a clinical validation study. All studies used Bland–Altman analysis to assess agreement; only tacrolimus and mycophenolic acid evaluated the performance to predict plasma concentrations from VAMS concentrations. Most studies showed good agreement between concentration measurement and the two matrices. Therefore, VAMS can replace CVS in routine practice, especially for tacrolimus and mycophenolic acid. On the other hand, further research needs to be conducted for other drugs, as clinical validation is required before VAMS can replace CVS in routine care.

Author Contributions

Outline preparation, R.V.N.; writing—original draft preparation, R.V.N.; writing—review and editing, R.V.N., V.Y., P.S., A.N.H., R.E.A. and R.R.; supervision, V.Y., P.S., A.N.H., R.E.A. and R.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research and APC were funded by the Directorate of Research and Community Service, Deputy for Strengthening Research and Development, Ministry of Research, Technology/National Research and Innovation Agency of the Republic of Indonesia in the PMDSU Scholarship Program, grant number 2046/UN.6.3.1/PT.00/2022 and the Academic Leadership Grant (ALG), an internal Universitas Padjadjaran research fund, received by Rovina Ruslami, grant number 3855/UN.C/LT/2019.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Aronson, J.K. Concentration-effect and dose-response relations in clinical pharmacology. Br. J. Clin. Pharmacol. 2007, 63, 255–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Holford, N.H.G.; Sheiner, L.B. Understanding the Dose-Effect Relationship. Clin. Pharmacokinet. 1981, 6, 429–453. [Google Scholar] [CrossRef] [PubMed]
  3. Aarnoutse, R.E.; Schapiro, J.M.; Boucher, C.A.B.; Hekster, Y.A.; Burger, D.M. Therapeutic drug monitoring: An aid to optimising response to antiretroviral drugs? Drugs 2003, 63, 741–753. [Google Scholar] [CrossRef] [PubMed]
  4. Saleem, M.A.; Basharat, R.; Rana, N.A.; Akhtar Khan Khattak, S. Role of clinician in therapeutic drug monitoring practice. Clin. Pract. 2020, 17, 1429–1435. [Google Scholar]
  5. Ghiculesco, R. Abnormal laboratory results: Therapeutic drug monitoring: Which drugs, why, when and how to do it. Aust. Prescr. 2008, 31, 42–44. [Google Scholar] [CrossRef]
  6. Roberge, R.J. Venodilatation techniques to enhance venepuncture and intravenous cannulation. J. Emerg. Med. 2004, 27, 69–73. [Google Scholar] [CrossRef]
  7. Buowari, O.Y. Complications of venepuncture. Adv. Biosci. Biotechnol. 2013, 4, 126–128. [Google Scholar] [CrossRef] [Green Version]
  8. O’Connor, R.; O’Sullivan, J.F.; O’Kennedy, R. Determination of serum and tissue levels of phenazines including clofazimine. J. Chromatogr. B Biomed. Appl. 1996, 681, 307–315. [Google Scholar] [CrossRef]
  9. Schaad-Lanyi, Z.; Dieterle, W.; Dubois, J.P.; Theobald, W.; Vischer, W. Pharmacokinetics of clofazimine in healthy volunteers. Int. J. Lepr. 1987, 55, 9–15. [Google Scholar]
  10. Katzung, B.G. Basic & Clinical Pharmacology, 14th ed.; McGraw Hill Medical: New York, NY, USA, 2018; ISBN 9781259641152. [Google Scholar]
  11. Lei, B.U.W.; Prow, T.W. A review of microsampling techniques and their social impact. Biomed. Microdevices 2019, 21, 81. [Google Scholar] [CrossRef] [Green Version]
  12. Parker, S.L.; Dorofaeff, T.; Lipman, J.; Ballot, D.E.; Bandini, R.M.; Wallis, S.C.; Roberts, J.A. Is there a role for microsampling in antibiotic pharmacokinetic studies? Expert Opin. Drug Metab. Toxicol. 2016, 12, 601–614. [Google Scholar] [CrossRef] [PubMed]
  13. Ye, Z.; Gao, H. Evaluation of sample extraction methods for minimizing hematocrit effect on whole blood analysis with volumetric absorptive microsampling. Bioanalysis 2017, 9, 349–357. [Google Scholar] [CrossRef] [PubMed]
  14. Barco, S.; Castagnola, E.; Moscatelli, A.; Rudge, J.; Tripodi, G.; Cangemi, G. Volumetric adsorptive microsampling-liquid chromatography tandem mass spectrometry assay for the simultaneous quantification of four antibiotics in human blood: Method development, validation and comparison with dried blood spot. J. Pharm. Biomed. Anal. 2017, 145, 704–710. [Google Scholar] [CrossRef] [PubMed]
  15. Koponen, J.; Rudge, J.; Kushon, S.; Kiviranta, H. Novel volumetric adsorptive microsampling technique for determination of perfluorinated compounds in blood. Anal. Biochem. 2018, 545, 49–53. [Google Scholar] [CrossRef] [PubMed]
  16. D’Urso, A.; Rudge, J.; Patsalos, P.N.; De Grazia, U. Volumetric Absorptive Microsampling: A New Sampling Tool for Therapeutic Drug Monitoring of Antiepileptic Drugs. Ther. Drug Monit. 2019, 41, 681–692. [Google Scholar] [CrossRef]
  17. Lee, K.; Jun, S.H.; Choi, M.S.; Song, S.H.; Park, J.S.; Lee, J.H.; Park, K.U.; Song, J. Application of the isoniazid assay in dried blood spots using the ultra-performance liquid chromatography-tandem mass spectrometry. Clin. Biochem. 2017, 50, 882–885. [Google Scholar] [CrossRef]
  18. Denniff, P.; Spooner, N. Volumetric absorptive microsampling: A dried sample collection technique for quantitative bioanalysis. Anal. Chem. 2014, 86, 8489–8495. [Google Scholar] [CrossRef]
  19. Kok, M.G.M.M.; Fillet, M. Volumetric absorptive microsampling: Current advances and applications. J. Pharm. Biomed. Anal. 2018, 147, 288–296. [Google Scholar] [CrossRef]
  20. Marchand, A.; Roulland, I.; Semence, F.; Audran, M. Volumetric Absorptive Microsampling (VAMS) technology for IGF-1 quantification by automated chemiluminescent immunoassay in dried blood. Growth Horm. IGF Res. 2019, 50, 27–34. [Google Scholar] [CrossRef]
  21. Spooner, N.; Denniff, P.; Michielsen, L.; De Vries, R.; Ji, Q.C.; Arnold, M.E.; Woods, K.; Woolf, E.J.; Xu, Y.; Boutet, V.; et al. A device for dried blood microsampling in quantitative bioanalysis: Overcoming the issues associated blood hematocrit. Bioanalysis 2015, 7, 653–659. [Google Scholar] [CrossRef]
  22. Protti, M.; Mandrioli, R.; Mercolini, L. Tutorial: Volumetric absorptive microsampling (VAMS). Anal. Chim. Acta 2019, 1046, 32–47. [Google Scholar] [CrossRef]
  23. Imre, S.; Vlase, L.; Muntean, D.L. Bioanalytical method validation. Rev. Rom. Med. Lab. 2008, 10, 13–21. [Google Scholar] [CrossRef]
  24. Capiau, S.; Veenhof, H.; Koster, R.A.; Bergqvist, Y.; Boettcher, M.; Halmingh, O.; Keevil, B.G.; Koch, B.C.P.; Linden, R.; Pistos, C.; et al. Official International Association for Therapeutic Drug Monitoring and Clinical Toxicology Guideline: Development and Validation of Dried Blood Spot-Based Methods for Therapeutic Drug Monitoring. Ther. Drug Monit. 2019, 41, 409–430. [Google Scholar] [CrossRef] [PubMed]
  25. Kocur, A.; Pawiński, T. Volumetric Absorptive Microsampling in Therapeutic Drug Monitoring of Immunosuppressive Drugs—From Sampling and Analytical Issues to Clinical Application. Int. J. Mol. Sci. 2023, 24, 681. [Google Scholar] [CrossRef] [PubMed]
  26. Denniff, P.; Parry, S.; Dopson, W.; Spooner, N. Quantitative bioanalysis of paracetamol in rats using volumetric absorptive microsampling (VAMS). J. Pharm. Biomed. Anal. 2015, 108, 61–69. [Google Scholar] [CrossRef] [PubMed]
  27. Moorthy, G.S.; Vedar, C.; Downes, K.J.; Fitzgerald, J.C.; Scheetz, M.H.; Zuppa, A.F. Microsampling Assays for Pharmacokinetic Analysis and Therapeutic Drug Monitoring of Antimicrobial Drugs in Children: A Critical Review. Ther. Drug Monit. 2021, 43, 335–345. [Google Scholar] [CrossRef]
  28. De Kesel, P.M.M.; Lambert, W.E.; Stove, C.P. Does volumetric absorptive microsampling eliminate the hematocrit bias for caffeine and paraxanthine in dried blood samples? A comparative study. Anal. Chim. Acta 2015, 881, 65–73. [Google Scholar] [CrossRef] [Green Version]
  29. Marahatta, A.; Megaraj, V.; McGann, P.T.; Ware, R.E.; Setchell, K.D.R. Stable-isotope dilution HPLC-electrospray ionization tandem mass spectrometry method for quantifying hydroxyurea in dried blood samples. Clin. Chem. 2016, 62, 1593–1601. [Google Scholar] [CrossRef] [Green Version]
  30. Anoshkina, Y.; Costas-Rodríguez, M.; Vanhaecke, F. Iron isotopic analysis of finger-prick and venous blood by multi-collector inductively coupled plasma-mass spectrometry after volumetric absorptive microsampling. J. Anal. At. Spectrom. 2017, 32, 314–321. [Google Scholar] [CrossRef]
  31. Kip, A.E.; Kiers, K.C.; Rosing, H.; Schellens, J.H.M.; Beijnen, J.H.; Dorlo, T.P.C. Volumetric absorptive microsampling (VAMS) as an alternative to conventional dried blood spots in the quantification of miltefosine in dried blood samples. J. Pharm. Biomed. Anal. 2017, 135, 160–166. [Google Scholar] [CrossRef]
  32. Protti, M.; Rudge, J.; Sberna, A.E.; Gerra, G.; Mercolini, L. Dried haematic microsamples and LC–MS/MS for the analysis of natural and synthetic cannabinoids. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2017, 1044–1045, 77–86. [Google Scholar] [CrossRef] [PubMed]
  33. Nys, G.; Cobraiville, G.; Kok, M.G.M.; Wéra, O.; Servais, A.C.; Fillet, M. Comparison of nanofluidic and ultra-high performance liquid chromatography-tandem mass spectrometry for high sensitive pharmacokinetic studies of estrogens starting from whole blood microsampling. J. Chromatogr. A 2017, 1524, 160–168. [Google Scholar] [CrossRef]
  34. Kita, K.; Mano, Y. Application of volumetric absorptive microsampling device for quantification of tacrolimus in human blood as a model drug of high blood cell partition. J. Pharm. Biomed. Anal. 2017, 143, 168–175. [Google Scholar] [CrossRef]
  35. Youhnovski, N.; Mayrand-Provencher, L.; Bérubé, E.R.; Plomley, J.; Montpetit, H.; Furtado, M.; Keyhani, A. Volumetric absorptive microsampling combined with impact-assisted extraction for hematocrit effect free assays. Bioanalysis 2017, 9, 1761–1769. [Google Scholar] [CrossRef] [PubMed]
  36. Protti, M.; Catapano, M.C.; Samolsky Dekel, B.G.; Rudge, J.; Gerra, G.; Somaini, L.; Mandrioli, R.; Mercolini, L. Determination of oxycodone and its major metabolites in haematic and urinary matrices: Comparison of traditional and miniaturised sampling approaches. J. Pharm. Biomed. Anal. 2018, 152, 204–214. [Google Scholar] [CrossRef]
  37. Kovač, J.; Panic, G.; Neodo, A.; Meister, I.; Coulibaly, J.T.; Schulz, J.D.; Keiser, J. Evaluation of a novel micro-sampling device, MitraTM, in comparison to dried blood spots, for analysis of praziquantel in Schistosoma haematobium-infected children in rural Côte d’Ivoire. J. Pharm. Biomed. Anal. 2018, 151, 339–346. [Google Scholar] [CrossRef]
  38. Qu, Y.; Brady, K.; Apilado, R.; O’Malley, T.; Reddy, S.; Chitkara, P.; Ibarra, C.; Alexander, R.V.; Dervieux, T. Capillary blood collected on volumetric absorptive microsampling (VAMS) device for monitoring hydroxychloroquine in rheumatoid arthritis patients. J. Pharm. Biomed. Anal. 2017, 140, 334–341. [Google Scholar] [CrossRef]
  39. Parker, S.L.; Roberts, J.A.; Lipman, J.; Wallis, S.C. Quantitative bioanalytical validation of fosfomycin in human whole blood with volumetric absorptive microsampling. Bioanalysis 2015, 7, 2585–2595. [Google Scholar] [CrossRef]
  40. Houbart, V.; Cobraiville, G.; Servais, A.C.; Napp, A.; Merville, M.P.; Fillet, M. Hepcidin determination in dried blood by microfluidic LC-MS/MS: Comparison of DBS and volumetric absorptive microsampling for matrix effect and recovery. Bioanalysis 2015, 7, 2789–2799. [Google Scholar] [CrossRef] [PubMed]
  41. Bolea-Fernandez, E.; Phan, K.; Balcaen, L.; Resano, M.; Vanhaecke, F. Determination of ultra-trace amounts of prosthesis-related metals in whole blood using volumetric absorptive micro-sampling and tandem ICP-Mass spectrometry. Anal. Chim. Acta 2016, 941, 1–9. [Google Scholar] [CrossRef]
  42. Cañabate, Á.; García-Ruiz, E.; Resano, M.; Todolí, J.L. Analysis of whole blood by ICP-MS equipped with a high temperature total sample consumption system. J. Anal. At. Spectrom. 2017, 32, 78–87. [Google Scholar] [CrossRef] [Green Version]
  43. John, H.; Willoh, S.; Hörmann, P.; Siegert, M.; Vondran, A.; Thiermann, H. Procedures for Analysis of Dried Plasma Using Microsampling Devices to Detect Sulfur Mustard-Albumin Adducts for Verification of Poisoning. Anal. Chem. 2016, 88, 8787–8794. [Google Scholar] [CrossRef] [PubMed]
  44. Luo, Y.; Korfmacher, W.; Ho, S.; Shen, L.; Wang, J.; Wu, Z.; Guo, Y.; Snow, G.; O’shea, T. Evaluation of two blood microsampling approaches for drug discovery PK studies in rats. Bioanalysis 2015, 7, 2345–2359. [Google Scholar] [CrossRef] [PubMed]
  45. Mercolini, L.; Protti, M.; Catapano, M.C.; Rudge, J.; Sberna, A.E. LC-MS/MS and volumetric absorptive microsampling for quantitative bioanalysis of cathinone analogues in dried urine, plasma and oral fluid samples. J. Pharm. Biomed. Anal. 2016, 123, 186–194. [Google Scholar] [CrossRef] [PubMed]
  46. Miao, Z.; Farnham, J.G.; Hanson, G.; Podoll, T.; Reid, M.J. Bioanalysis of emixustat (ACU-4429) in whole blood collected with volumetric absorptive microsampling by LC-MS/MS. Bioanalysis 2015, 7, 2071–2083. [Google Scholar] [CrossRef]
  47. Nys, G.; Gallez, A.; Kok, M.G.M.; Cobraiville, G.; Servais, A.C.; Piel, G.; Pequeux, C.; Fillet, M. Whole blood microsampling for the quantitation of estetrol without derivatization by liquid chromatography-tandem mass spectrometry. J. Pharm. Biomed. Anal. 2017, 140, 258–265. [Google Scholar] [CrossRef]
  48. Thiry, J.; Evrard, B.; Nys, G.; Fillet, M.; Kok, M.G.M. Sampling only ten microliters of whole blood for the quantification of poorly soluble drugs: Itraconazole as case study. J. Chromatogr. A 2017, 1479, 161–168. [Google Scholar] [CrossRef]
  49. Parker, S.L.; Guerra Valero, Y.C.; Lipman, J.; Roberts, J.A.; Wallis, S.C. Effect of time on recovery of plasma microsamples for the quantitative determination of vancomycin. Bioanalysis 2016, 8, 2235–2242. [Google Scholar] [CrossRef]
  50. Londhe, V.; Rajadhyaksha, M. Opportunities and obstacles for microsampling techniques in bioanalysis: Special focus on DBS and VAMS. J. Pharm. Biomed. Anal. 2020, 182, 113102. [Google Scholar] [CrossRef]
  51. Schulz, J.D.; Neodo, A.; Coulibaly, J.T.; Keiser, J. Pharmacokinetics of Albendazole, Albendazole Sulfoxide, and Albendazole Sulfone Determined from Plasma, Blood, Dried Blood Spots, and Mitra Samples of Hookworm-Infected Adolescents. Antimicrob. Agents Chemother. 2019, 63, e02489-18. [Google Scholar] [CrossRef] [Green Version]
  52. Canisius, T.P.I.J.M.; Soons, J.W.P.H.; Verschuure, P.; Wammes-Van Der Heijden, E.A.; Rouhl, R.P.W.; Majoie, H.J.M. Therapeutic drug monitoring of anti-epileptic drugs—A clinical verification of volumetric absorptive micro sampling. Clin. Chem. Lab. Med. 2020, 58, 828–835. [Google Scholar] [CrossRef] [PubMed]
  53. Tron, C.; Ferrand-Sorre, M.J.; Querzerho-Raguideau, J.; Chemouny, J.M.; Houssel-Debry, P.; Verdier, M.C.; Bellissant, E.; Lemaitre, F. Volumetric absorptive microsampling for the quantification of tacrolimus in capillary blood by high performance liquid chromatography-tandem mass spectrometry. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2021, 1165, 122521. [Google Scholar] [CrossRef] [PubMed]
  54. Koster, R.A.; Niemeijer, P.; Veenhof, H.; van Hateren, K.; Alffenaar, J.-W.C.; Touw, D.J. A volumetric absorptive microsampling LC-MS/MS method for five immunosuppressants and their hematocrit effects. Bioanalysis 2019, 11, 495–508. [Google Scholar] [CrossRef] [PubMed]
  55. Gani, M.R.; Isnaeni; Prawita, A.; Hafid, A.F.; Widyawaruyanti, A. Bioanalytical method development and validation for quantification of morachalcone A in rabbit plasma using high performance liquid chromatography. Pak. J. Pharm. Sci. 2018, 31, 311–315. [Google Scholar]
  56. Arora, K.; Gangadharappa, H.V. An Approach to Bioanalytical Method Development and Validation: A Review. Int. J. Pharm. Sci. Res. 2016, 7, 2291–2301. [Google Scholar] [CrossRef]
  57. Moein, M.M.; El Beqqali, A.; Abdel-Rehim, M. Bioanalytical method development and validation: Critical concepts and strategies. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2017, 1043, 3–11. [Google Scholar] [CrossRef]
  58. Chanda Gupta, P. A View on Analytical Method Validation of Drugs. J. Cell Sci. Ther. 2016, 7, 1000235. [Google Scholar] [CrossRef] [Green Version]
  59. Ozkan, S.A. Analytical method validation: The importance for pharmaceutical analysis. Pharm. Sci. 2018, 24, 1–2. [Google Scholar] [CrossRef] [Green Version]
  60. Ravichandran, V.; Shalini, S.; Sundram, K.M.; Rajak, H. Validation of analytical methods—Strategies & importance International Journal of Pharmacy and Pharmaceutical Sciences. Int. J. Pharm. Pharm. Sci. 2010, 2, 18–22. [Google Scholar]
  61. Paniagua-González, L.; Díaz-Louzao, C.; Lendoiro, E.; Otero-Antón, E.; Cadarso-Suárez, C.; López-Rivadulla, M.; Cruz, A.; de-Castro-Ríos, A. Volumetric Absorptive Microsampling (VAMS) for assaying immunosuppressants from venous whole blood by LC–MS/MS using a novel atmospheric pressure ionization probe (UniSprayTM). J. Pharm. Biomed. Anal. 2020, 189, 113422. [Google Scholar] [CrossRef]
  62. Delahaye, L.; Dhont, E.; De Cock, P.; De Paepe, P.; Stove, C.P. Volumetric absorptive microsampling as an alternative sampling strategy for the determination of paracetamol in blood and cerebrospinal fluid. Anal. Bioanal. Chem. 2019, 411, 181–191. [Google Scholar] [CrossRef] [PubMed]
  63. Maggadani, B.P.; Harmita; Haryono, S.J.; Rinaldi, M.R.; Harahap, Y. Volumetric absorptive microsampling as a new biosampling tool for monitoring of tamoxifen, endoxifen, 4-oh tamoxifen and n-desmethyltamoxifen in breast cancer patients. Drug Des. Devel. Ther. 2021, 15, 2417–2430. [Google Scholar] [CrossRef] [PubMed]
  64. Moorthy, G.S.; Vedar, C.; Zane, N.R.; Downes, K.J.; Prodell, J.L.; DiLiberto, M.A.; Zuppa, A.F. Development and validation of a volumetric absorptive microsampling- liquid chromatography mass spectrometry method for the analysis of cefepime in human whole blood: Application to pediatric pharmacokinetic study. J. Pharm. Biomed. Anal. 2020, 179, 113002. [Google Scholar] [CrossRef] [PubMed]
  65. Arroyo, G.; Bustos, J.A.; Lescano, A.G.; Gonzales, I.; Saavedra, H.; Rodriguez, S.; Pretell, E.J.; Bonato, P.S.; Lanchote, V.L.; Takayanagui, O.M.; et al. Albendazole Sulfoxide Plasma Levels and Efficacy of Antiparasitic Treatment in Patients with Parenchymal Neurocysticercosis. Clin. Infect. Dis. 2019, 69, 1996–2002. [Google Scholar] [CrossRef]
  66. Wang, F.; Agnihotri, A.; Adabag, S.; Roukoz, H. Clinical Utility of Plasma Amiodarone Level Measurement: A Single Center Experience. Innov. Card. Rythm Manag. 2011, 2, 508–511. [Google Scholar]
  67. Lamoth, F.; Buclin, T.; Pascual, A.; Vora, S.; Bolay, S.; Decosterd, L.A.; Calandra, T.; Marchetti, O. High cefepime plasma concentrations and neurological toxicity in febrile neutropenic patients with mild impairment of renal function. Antimicrob. Agents Chemother. 2010, 54, 4360–4367. [Google Scholar] [CrossRef] [Green Version]
  68. Reid, M.J.; Eyre, R.; Podoll, T. Oxidative deamination of emixustat by human vascular adhesion protein-1/semicarbazide-sensitive amine oxidase. Drug Metab. Dispos. 2019, 47, 504–515. [Google Scholar] [CrossRef] [Green Version]
  69. Aubert, G.; Carricajo, A.; Coudrot, M.; Guyomarch, S.; Auboyer, C.; Zeni, F. Prospective determination of serum ceftazidime concentrations in intensive care units. Ther. Drug Monit. 2010, 32, 517–519. [Google Scholar] [CrossRef]
  70. Patsalos, P.N. Antiepileptic Drug Interactions; Springer: Berlin, Germany, 2014; ISBN 9781118456989. [Google Scholar]
  71. Wenzler, E.; Ellis-Grosse, E.J.; Rodvold, K.A. Pharmacokinetics, safety, and tolerability of single-dose intravenous (ZTI-01) and oral fosfomycin in healthy volunteers. Antimicrob. Agents Chemother. 2017, 61, e00775-17. [Google Scholar] [CrossRef] [Green Version]
  72. Thalhammer, F.; Schenk, P.; Burgmann, H.; El Menyawi, I.; Hollenstein, U.M.; Rosenkranz, A.R.; Sunder-Plassmann, G.; Breyer, S.; Ratheiser, K. Single-dose pharmacokinetics of meropenem during continuous venovenous hemofiltration. Antimicrob. Agents Chemother. 1998, 42, 2417–2420. [Google Scholar] [CrossRef] [Green Version]
  73. Lo Re, F.; Angelini, J.; Sponga, S.; Nalli, C.; Zucchetto, A.; Biasizzo, J.; Livi, U.; Baraldo, M. Therapeutic Drug Monitoring of Mycophenolic Acid as a Precision Medicine Tool for Heart Transplant Patients: Results of an Observational Pharmacokinetic Pilot Study. Pharmaceutics 2022, 14, 1304. [Google Scholar] [CrossRef]
  74. Brett, C.N.; Barnett, S.G.; Pearson, J. Postoperative plasma paracetamol levels following oral or intravenous paracetamol administration: A double-blind randomised controlled trial. Anaesth. Intensive Care 2012, 40, 166–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Cohen-Wolkowiez, M.; Watt, K.M.; Zhou, C.; Bloom, B.T.; Poindexter, B.; Castro, L.; Gao, J.; Capparelli, E.V.; Smitha, P.B.; Benjamin, D.K. Developmental pharmacokinetics of piperacillin and tazobactam using plasma and dried blood spots from infants. Antimicrob. Agents Chemother. 2014, 58, 2856–2865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ximenez, J.P.B.; De Andrade, J.M.; Marques, M.P.; Coelho, E.B.; Suarez-Kurtz, G.; Lanchote, V.L. Hormonal status affects plasma exposure of tamoxifen and its main metabolites in tamoxifen-treated breast cancer patients. BMC Pharmacol. Toxicol. 2019, 20, 81. [Google Scholar] [CrossRef]
  77. Valera, H.R.; Ganguly, B. Study of plasma level of atorvastatin and its effect on lipid profile. Indian J. Physiol. Pharmacol. 2009, 53, 73–82. [Google Scholar] [PubMed]
  78. Cordell, R.L.; Valkenburg, T.S.E.; Pandya, H.C.; Hawcutt, D.B.; Semple, M.G.; Monks, P.S. Quantitation of salbutamol using micro-volume blood sampling–applications to exacerbations of pediatric asthma. J. Asthma 2018, 55, 1205–1213. [Google Scholar] [CrossRef]
  79. Abu-Rabie, P.; Neupane, B.; Spooner, N.; Rudge, J.; Denniff, P.; Mulla, H.; Pandya, H. Validation of methods for determining pediatric midazolam using wet whole blood and volumetric absorptive microsampling. Bioanalysis 2019, 11, 1737–1754. [Google Scholar] [CrossRef]
  80. Moorthy, G.S.; Vedar, C.; Zane, N.; Prodell, J.L.; Zuppa, A.F. Development and validation of a volumetric absorptive microsampling assay for analysis of voriconazole and voriconazole N-oxide in human whole blood. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1105, 67–75. [Google Scholar] [CrossRef]
  81. Sciberras, D.; Otoul, C.; Lurquin, F.; Smeraglia, J.; Lappert, A.; De Bruyn, S.; Jaap van Lier, J. A pharmacokinetic study of radiprodil oral suspension in healthy adults comparing conventional venous blood sampling with two microsampling techniques. Pharmacol. Res. Perspect. 2019, 7, e00459. [Google Scholar] [CrossRef] [Green Version]
  82. Friedl, B.; Kurlbaum, M.; Kroiss, M.; Fassnacht, M.; Scherf-Clavel, O. A method for the minimally invasive drug monitoring of mitotane by means of volumetric absorptive microsampling for a home-based therapeutic drug monitoring. Anal. Bioanal. Chem. 2019, 411, 3951–3962. [Google Scholar] [CrossRef]
  83. Gal, P. Caffeine Therapeutic Drug Monitoring Is Necessary and Cost-effective. J. Pediatr. Pharmacol. Ther. 2007, 12, 212–215. [Google Scholar] [CrossRef]
  84. Karch, S. Cathinone Neurotoxicity (“The “3Ms”). Curr. Neuropharmacol. 2014, 13, 21–25. [Google Scholar] [CrossRef] [Green Version]
  85. Blanchet, B.; Jallouli, M.; Allard, M.; Ghillani-Dalbin, P.; Galicier, L.; Aumaître, O.; Chasset, F.; Le Guern, V.; Lioté, F.; Smail, A.; et al. Hydroxychloroquine levels in patients with systemic lupus erythematosus: Whole blood is preferable but serum levels also detect non-adherence. Arthritis Res. Ther. 2020, 22, 223. [Google Scholar] [CrossRef] [PubMed]
  86. Domínguez-Gil Hurlé, A.; Sánchez Navarro, A.; García Sánchez, M.J. Therapeutic drug monitoring of itraconazole and the relevance of pharmacokinetic interactions. Clin. Microbiol. Infect. 2006, 12, 97–106. [Google Scholar] [CrossRef] [Green Version]
  87. Douglas-Hall, P.; Dzahini, O.; Gaughran, F.; Bile, A.; Taylor, D. Variation in dose and plasma level of lamotrigine in patients discharged from a mental health trust. Ther. Adv. Psychopharmacol. 2017, 7, 17–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Pea, F.; Furlanut, M.; Cojutti, P.; Cristini, F.; Zamparini, E.; Franceschi, L.; Viale, P. Therapeutic drug monitoring of linezolid: A retrospective monocentric analysis. Antimicrob. Agents Chemother. 2010, 54, 4605–4610. [Google Scholar] [CrossRef] [Green Version]
  89. Jawad, A.M. Rational use of Metoprolol: The Relationship of Its Blood Concentration to Patient Compliance, Poor Quality Medicines and Side Effects. Univ. Thi-Qar J. Med. 2019, 16, 132–141. [Google Scholar] [CrossRef]
  90. Persson, P.; Nilsson, A.; Hartvig, P.; Tamsen, A. Pharmacokinetics of midazolam in total I. V. Anaesthesia. Br. J. Anaesth. 1987, 59, 548–556. [Google Scholar] [CrossRef]
  91. Mbui, J.; Olobo, J.; Omollo, R.; Solomos, A.; Kip, A.E.; Kirigi, G.; Sagaki, P.; Kimutai, R.; Were, L.; Omollo, T.; et al. Pharmacokinetics, safety, and efficacy of an allometric miltefosine regimen for the treatment of visceral leishmaniasis in eastern African children: An open-label, phase II clinical trial. Clin. Infect. Dis. 2019, 68, 1530–1538. [Google Scholar] [CrossRef]
  92. Kerkhofs, T.M.A.; Derijks, L.J.J.; Ettaieb, M.H.T.; Eekhoff, E.M.W.; Neef, C.; Gelderblom, H.; Den Hartigh, J.; Guchelaar, H.J.; Haak, H.R. Short-term variation in plasma mitotane levels confirms the importance of trough level monitoring. Eur. J. Endocrinol. 2014, 171, 677–683. [Google Scholar] [CrossRef] [Green Version]
  93. Meister, I.; Kovac, J.; Duthaler, U.; Odermatt, P.; Huwyler, J.; Vanobberghen, F.; Sayasone, S.; Keiser, J. Pharmacokinetic Study of Praziquantel Enantiomers and Its Main Metabolite R-trans-4-OH-PZQ in Plasma, Blood and Dried Blood Spots in Opisthorchis viverrini-Infected Patients. PLoS Negl. Trop. Dis. 2016, 10, e0004700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Auvin, S.; Dozières-Puyravel, B.; Avbersek, A.; Sciberras, D.; Collier, J.; Leclercq, K.; Mares, P.; Kaminski, R.M.; Muglia, P. Radiprodil, a NR2B negative allosteric modulator, from bench to bedside in infantile spasm syndrome. Ann. Clin. Transl. Neurol. 2020, 7, 343–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Vet, N.J.; de Winter, B.C.M.; Koninckx, M.; Boeschoten, S.A.; Boehmer, A.L.M.; Verhallen, J.T.; Plötz, F.B.; Vaessen-Verberne, A.A.; van der Nagel, B.C.H.; Knibbe, C.A.J.; et al. Population Pharmacokinetics of Intravenous Salbutamol in Children with Refractory Status Asthmaticus. Clin. Pharmacokinet. 2020, 59, 257–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Cojutti, P.; Candoni, A.; Forghieri, F.; Isola, M.; Zannier, M.E.; Bigliardi, S.; Luppi, M.; Fanin, R.; Pea, F. Variability of Voriconazole Trough Levels in Haematological Patients: Influence of Comedications with cytochrome P450(CYP) Inhibitors and/or with CYP Inhibitors plus CYP Inducers. Basic Clin. Pharmacol. Toxicol. 2016, 118, 474–479. [Google Scholar] [CrossRef] [Green Version]
  97. Vethe, N.T.; Gustavsen, M.T.; Midtvedt, K.; Lauritsen, M.E.; Andersen, A.M.; Asberg, A.; Bergan, S. Tacrolimus Can Be Reliably Measured with Volumetric Absorptive Capillary Microsampling Throughout the Dose Interval in Renal Transplant Recipients. Ther. Drug Monit. 2019, 41, 607–614. [Google Scholar] [CrossRef]
  98. Grześk, G.; Stolarek, W.; Kasprzak, M.; Grześk, E.; Rogowicz, D.; Wiciński, M.; Krzyżanowski, M. Therapeutic drug monitoring of carbamazepine: A 20-year observational study. J. Clin. Med. 2021, 10, 5396. [Google Scholar] [CrossRef]
  99. Narula, A.S.; Murthy, M.S.N.; Patrulu, K.S.K.; Saxena, V.K. Routine cyclosporine concentration—C2 level monitoring. Is it helpful during the early post transplant period? Med. J. Armed Forces India 2004, 60, 326–328. [Google Scholar] [CrossRef] [Green Version]
  100. Hirabatake, M.; Mizuno, T.; Kato, H.; Hashida, T. Everolimus pharmacokinetics and exposure-response relationship in Japanese patients with advanced breast cancer. Front. Pharmacol. 2022, 13, 984002. [Google Scholar] [CrossRef]
  101. May, T.W.; Brandt, C.; Helmer, R.; Bien, C.G.; Cawello, W. Comparison of lacosamide concentrations in cerebrospinal fluid and serum in patients with epilepsy. Epilepsia 2015, 56, 1134–1140. [Google Scholar] [CrossRef]
  102. Gupta, V.; Gupta, K.; Singh, G.; Kaushal, S. An analytical study to correlate serum levels of levetiracetam with clinical course in patients with epilepsy. J. Neurosci. Rural Pract. 2016, 7, S31–S36. [Google Scholar] [CrossRef]
  103. González-Esquivel, D.F.; Ortega-Gavilán, M.; Alcántara-López, G.; Jung-Cook, H. Plasma level monitoring of oxcarbazepine in epileptic patients. Arch. Med. Res. 2000, 31, 202–205. [Google Scholar] [CrossRef] [PubMed]
  104. Steinhoff, B.J.; Hübers, E.; Kurth, C.; Jürges (Kehl-Kork), U. Plasma concentration and clinical effects of perampanel—The Kork experience. Seizure 2019, 67, 18–22. [Google Scholar] [CrossRef] [Green Version]
  105. Stenton, S.B.; Partovi, N.; Ensom, M.H.H. Sirolimus: The evidence for clinical pharmacokinetic monitoring. Clin. Pharmacokinet. 2005, 44, 769–786. [Google Scholar] [CrossRef]
  106. Alghanem, S.S.; Soliman, M.M.; Alibrahim, A.A.; Gheith, O.; Kenawy, A.S.; Awad, A. Monitoring Tacrolimus Trough Concentrations During the First Year After Kidney Transplantation: A National Retrospective Cohort Study. Front. Pharmacol. 2020, 11, 566638. [Google Scholar] [CrossRef] [PubMed]
  107. Martial, L.C.; Aarnoutse, R.E.; Mulder, M.; Schellekens, A.; Brüggemann, R.J.M.; Burger, D.M.; Schene, A.H.; Batalla, A. Dried Blood Spot sampling in psychiatry: Perspectives for improving therapeutic drug monitoring. Eur. Neuropsychopharmacol. 2017, 27, 205–216. [Google Scholar] [CrossRef] [PubMed]
  108. Veenhof, H.; Koster, R.A.; Junier, L.A.T.; Berger, S.P.; Bakker, S.J.L.; Touw, D.J. Volumetric absorptive microsampling and dried blood spot microsampling vs. Conventional venous sampling for tacrolimus trough concentration monitoring. Clin. Chem. Lab. Med. 2020, 58, 1687–1695. [Google Scholar] [CrossRef]
  109. Zwart, T.C.; Metscher, E.; van der Boog, P.J.M.; Swen, J.J.; de Fijter, J.W.; Guchelaar, H.J.; de Vries, A.P.J.; Moes, D.J.A.R. Volumetric microsampling for simultaneous remote immunosuppressant and kidney function monitoring in outpatient kidney transplant recipients. Br. J. Clin. Pharmacol. 2022, 88, 4854–4869. [Google Scholar] [CrossRef]
Figure 1. Volumetric absorptive microsampling devices (left picture is VAMS in clamshell device; right picture is VAMS in cartridge device).
Figure 1. Volumetric absorptive microsampling devices (left picture is VAMS in clamshell device; right picture is VAMS in cartridge device).
Molecules 28 06046 g001
Table 1. Optimization and Validation of VAMS Assays for Acidic Drugs.
Table 1. Optimization and Validation of VAMS Assays for Acidic Drugs.
NoDrugExtraction SolventExtraction ConditionsAdditional Sample PreparationAnalytical InstrumentValidation ResultRef.
Available ParameterMissing Parameter
1FosfomycinMethanol + ISOn a lateral shaker for 30 min at 1200 rpm-LC-MS/MSCalibration range of 5–2000 µg/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stability was only assessed for 500 µg/mL concentrations and was stable at RT for 10 days in an autosampler at 4 °C for 48 h
Stability for QC concentrations and dilution integrity was not assessed[39]
2EmixustatMethanol containing 1% ammonium hydroxide + ISSonication for 15 min and vortex for 15 minEvaporation at 40 °C with nitrogen, reconstitution in methanol–water (3:7, v/v) containing 0.1% formic acidLC-MS/MSCalibration range of 0.05–10 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 1 week; −4 °C for 1 months; −20 °C for 1 months
-[46]
3AtorvastatinMethanol–acetonitrile (1:1, v/v) and 1% formic acid + ISVortex for 1 hCentrifugation at 3000 rpm for 15 minLC-MS/MSCalibration range of 1–2000 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Sensitivity, selectivity, matrix effect, dilution integrity, carry-over, and stability were not assessed[13]
4TamoxifenMethanol–acetonitrile (1:1, v/v) and 1% formic acid + ISVortex for 1 hCentrifugation at 3000 rpm for 15 minLC-MS/MSCalibration range of 1–2000 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Sensitivity, selectivity, matrix effect, dilution integrity, carry-over, and stability were not assessed[13]
5AmiodaroneMethanol–acetonitrile (1:1, v/v) and 1% formic acid + ISVortex for 1 hCentrifugation at 3000 rpm for 15 minLC-MS/MSCalibration range of 1–2000 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Sensitivity, selectivity, matrix effect, dilution integrity, carry-over, and stability were not assessed[13]
6PiperacillinMethanol Sonication for 10 minRehydration using water and incubation for 10 min at 37 °C in the thermos block heaterLC-MS/MSCalibration range of 3.125–200 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT and 4 °C for 72 h; −20 °C for 1 month
Dilution integrity was not assessed[14]
7TazobactamMethanol Sonication for 10 minRehydration using water and incubation for 10 min at 37 °C in the thermos block heaterLC-MS/MSCalibration range of 0.625–40 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT and 4 °C for 72 h; −20 °C for 1 month
Dilution integrity was not assessed[14]
8MeropenemMethanol Sonication for 10 minRehydration using water and incubation for 10 min at 37 °C in the thermos block heaterLC-MS/MSCalibration range of 0.625–40 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT and 4 °C for 72 h; −20 °C for 1 month
Dilution integrity was not assessed[14]
9CeftazidimeMethanol Sonication for 10 minRehydration using water and incubation for 10 min at 37 °C in the thermos block heaterLC-MS/MSCalibration range of 3.125–200 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at 4 °C for 72 h; −20 °C for 1 month
Dilution integrity was not assessed[14]
10ParacetamolMethanol–water–formic acid (80:20:0.01, v/v/v) + ISShaking for 15 min in a thermomixer comfort Centrifugation at RT for 10 min at 10,000× g, supernatant dilution with water with 0.01% formic acidLC-MS/MSCalibration range of 4.0–32.8 µg/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at 50 °C for 1 week; −80 °C, −20 °C, and 4 °C for 9 months; RT for at least 8 months
-[62]
11Albendazole Acetonitrile-methanol (1:1, v/v) with 0.1% formic acid + ISSonication in an ultrasonic bath for 1 h and agitation for 1 h in RT at 1200 rpm Centrifugation for 10 min at 3300× g, evaporation of supernatant, and reconstitution in methanol LC-MS/MSCalibration range of 1–200 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, and matrix effect were assessed and met the acceptance criteria
Stable at RT for 2 months; in autosampler at 4 °C for 72 h; −80 °C for 2 months
Dilution integrity and carry-over were not assessed[51]
12Cefepime Acetonitrile + ISSonication for 15 min, vortex at 700 rpm for 10 min, and centrifugation for 30 min at 3220× g at 4 °CRehydration using water, vortex for 2 min at 1000 rpm, and incubation for 10 min at 37 °CLC-MS/MSCalibration range of 0.100–100 µg/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at 4 °C for 1 week; −20 °C for 1 month (39 days); −78 °C for 3 months (91 days); in autosampler at 10 °C for 20 h
-[64]
13EthosuximideAcetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 3.48–234 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
14Felbamate Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 9.55–428 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
15Phenobarbital Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.89–60 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
16Phenytoin Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.58–39 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
17Primidone Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.76–51 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
18Topiramate Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.67–45 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
19Zonisamide Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 1.07–72 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
20Mycophenolic acidWater and methanol + ISSonication for 15 minCentrifugation for 5 min at 14,500 rpm, evaporation of supernatant, and reconstitution using ammonium formate and 0.1% formic acid in water-acetonitrile (6:4, v/v)LC-MS/MSCalibration range of 75–7500 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable in autosampler at 6 °C for 72 h; 20 °C, 4 °C, and RT for 15 days; −20 °C for 8 months
Dilution integrity was not assessed[61]
21TamoxifenMethanol + IS Vortex for 1 min, sonication for 25 minDry in a water bath at 55 °C under nitrogen steam, reconstitution using 0.1% formic acid and 0.1% formic acid in acetonitrile, vortex for 20 sLC-MS/MSCalibration range of 2.5–200 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 24 h; −20 °C for 1 month; in autosampler for 24 h
-[63]
IS: internal standard; LC-MS/MS: liquid chromatograph–tandem mass spectrometer; RT: room temperature; QC: quality control.
Table 2. Optimization and Validation of VAMS Assays for Basic Drugs.
Table 2. Optimization and Validation of VAMS Assays for Basic Drugs.
NoDrugExtraction SolventExtraction ConditionsAdditional Sample PreparationAnalytical InstrumentValidation ResultRef.
Available ParameterMissing Parameter
1ParaxanthineMethanol–water (8:2, v/v) and 0.01% formic acid + ISIn a thermomixer at 1000 rpm and 22 °CCentrifugation at RT (10 min, 10,000× g), supernatant diluted with 0.01% formic acidLC-MS/MSCalibration range of 0.025–5 µg/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT for 82 days; 60 °C for 4 days; −20 °C for 30 days; in autosampler at 4 °C for 4 days
Dilution integrity was not assessed[28]
2Caffeine Methanol–water (8:2, v/v) and 0.01% formic acid + ISIn a thermomixer at 1000 rpm and 22 °CCentrifugation at RT (10 min, 10,000× g), supernatant diluted with 0.01% formic acidLC-MS/MSCalibration range of 0.05–10 µg/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT for 82 days; 60 °C for 4 days; −20 °C for 30 days; in autosampler at 4 °C for 4 days
Dilution integrity was not assessed[28]
3MiltefosineMethanol Mixing for 15 min at 1250 rpm and vortex for 30 sA 20-fold dilution of highly concentrated samples with methanol and the addition of ISLC-MS/MSCalibration range of 10–5000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 1 month; 37 °C for 1 month; 2–8 °C for 5 days
-[31]
4Itraconazole Acetonitrile-methanol (6:4, v/v) + ISIncubation for 5 min, agitation for 5 min with a vortex mixerPhospholipid removal with Ostroplate, evaporation under vacuum at 50 °C for 1 h, reconstitution in methanol–water (1:1, v/v) LC-MS/MSCalibration range of 10–1000 ng/mL
Linearity, accuracy and precision, selectivity, recovery, and matrix effect were assessed and met the acceptance criteria
Stable at RT for 24 h; −80 °C for 2 weeks; in autosampler at 6 °C for 24 h
Sensitivity, dilution integrity, and carry-over were not assessed[48]
5Cathinone analogsMethanol Ultrasound agitation for 15 min and vortex for 1 minEvaporation under vacuum and reconstitution in acetonitrile-water (1:1, v/v) containing 0.1% formic acidLC-MS/MSCalibration range of 10–500 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, and matrix effect were assessed and met the acceptance criteria
Stable at RT for 7 days only for one concentration of 250 ng/mL
Dilution integrity, carry-over, and stability for QC concentrations were not assessed[45]
6MetoprololMethanol–acetonitrile (1:1, v/v) and 1% formic acid + ISVortex for 1 hCentrifugation at 3000 rpm for 15 minLC-MS/MSCalibration range of 1–2000 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Sensitivity, selectivity, matrix effect, dilution integrity, carry-over, and stability were not assessed[13]
7MidazolamMethanol–acetonitrile (1:1, v/v) and 1% formic acid + ISVortex for 1 hCentrifugation at 3000 rpm for 15 minLC-MS/MSCalibration range of 1–2000 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Sensitivity, selectivity, matrix effect, dilution integrity, carry-over, and stability were not assessed[13]
8Salbutamol Methanol + ISSonication for 15 min Evaporation for 60 min, resuspended in N, O-Bis(trimethylsilyl)trifluoroacetamide (BSTFA) with 1% trimethylchlorosilane (TMCS) by vortexing, and derivatization at 60 °C for 30 minGC-MSCalibration range of 3–100 ng/mL
Linearity, accuracy and precision, and recovery were assessed and met the acceptance criteria
Stable at −20 °C, RT, and 30 °C for 145 days; 4 °C for 75 days
Sensitivity, selectivity, matrix effect, dilution integrity, and carry-over were not assessed[78]
9Hydroxychloroquine Water + ISVortex for 30 minProtein precipitation with 70% perchloric acid, centrifugation for 5 min at 14,000 rpmLC-MS/MSCalibration range of 10–2000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 10 days; 22 °C and 50 °C for 24 h
-[38]
10LinezolidMethanol Sonication for 10 minRehydration using water and incubation for 10 min at 37 °C in the thermos block heaterLC-MS/MSCalibration range of 0.625–40 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT and 4 °C for 72 h; −20 °C for 1 month
Dilution integrity was not assessed[14]
11PraziquantelAcetonitrile-water (4:1, v/v) + ISThermos-mixing for 5 min at 1400 rpm at RT and ultrasonication for 40 min-LC-MS/MSCalibration range of 0.2–50 µg/mL
Linearity, accuracy and precision, sensitivity, recovery, and matrix effect were assessed and met the acceptance criteria
Stable at RT for 4 months; 4 °C for 72 h; −80 °C for 2 months
Carry-over, selectivity, and dilution integrity were not assessed[37]
12RadiprodilMethanol–ethyl acetate (1:1, v/v) + ISSonication for 15 min, shaking for 1 h at 1200 rpmDry using nitrogen and reconstitution LC-ESI-MS/MSCalibration range of 1–1000 ng/mL
Linearity, accuracy, and precision were assessed and met the acceptance criteria
Sensitivity, selectivity, recovery, matrix effect, dilution integrity, carry-over, and stability were not assessed[81]
13Voriconazole Acetonitrile-methanol (1:1, v/v) + ISSonication for 15 min, vortex for 30 min at 350 rpmCentrifugation for 30 min at 4000 rpmLC-MS/MSCalibration range of 10–10,000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 19 h; −80 °C for 3 days; 4 °C, −20 °C, −80 °C for 1 month; in autosampler for 24 h
-[80]
14Voriconazole N-oxideAcetonitrile-methanol (1:1, v/v) + ISSonication for 15 min, vortex for 30 min at 350 rpmCentrifugation for 30 min at 4000 rpmLC-MS/MSCalibration range of 10–10,000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 19 h; −80 °C for 3 days; in autosampler for 24 h
-[80]
15Midazolam Methanol + ISOn a lateral shaker for 1 h -LC-MS/MSCalibration range of 5–5000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and dilution integrity were assessed and met the acceptance criteria
Stable at RT for 131 days; 4 °C for 192 h; 37 °C for 4 h; 40 °C for 43 days; −20 °C for 43 days
Carry-over was not assessed[79]
16Lamotrigine Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.67–45 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
17RufinamideAcetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.94–63 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
18MitotaneMethanol–aqueous zinc sulfate heptahydrate solution 2% (4:1, v/v)Ultrasonic bath for 15 min, shaking for 1 h at 1400 rpmCentrifugation at 12,000× g for 5 min at 4 °C LC-UVCalibration range of 1–50 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, and carry-over were assessed and met the acceptance criteria
Stable at RT for 1 week; 2–8 °C for 1 week; in autosampler for 24 h
Matrix effect and dilution integrity were not assessed[82]
IS: internal standard; LC-MS/MS: liquid chromatograph–tandem mass spectrometer; GC-MS: gas chromatograph–mass spectrometer; LC-ESI-MS/MS: liquid chromatograph–electrospray ionization–tandem mass spectrometer; LC-UV, liquid chromatograph–ultraviolet; RT, room temperature; QC, quality control.
Table 3. Optimization and Validation of VAMS Assays for Neutral Drugs.
Table 3. Optimization and Validation of VAMS Assays for Neutral Drugs.
NoDrugExtraction SolventExtraction ConditionsAdditional Sample PreparationAnalytical InstrumentValidation ResultRef.
Available ParameterMissing Parameter
1Tacrolimus Methanol–zinc sulfate 0.10 mol/L (2:1) + ISShaking on the orbital shaker at 1400 rpm for 6 minCentrifugation at 2000× g for 10 min at 4 °CLC-MS/MSCalibration range of 0.7–6 µg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at RT for 2 months
-[97]
2Carbamazepine Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.45–30 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
3Lacosamide Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.45–30 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
4Oxcarbazepine Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.18–12 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
5Perampanel Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.03–1.86 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
6Levetiracetam Acetonitrile + IS Vortex mixing for 30 min at 600 rpmCentrifugation for 10 min at 17,000× g, reconstitution of supernatant using water and 0.1% formic acid, and vortex for 30 sLC-MS/MSCalibration range of 0.94–63 mg/L
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, dilution integrity, and carry-over were assessed and met the acceptance criteria
Stable at −20 °C, 4 °C, 37 °C, 10 °C (autosampler) for 10 days; −20 °C for 4 months
-[16]
7TacrolimusWater and methanol + ISSonication for 15 minCentrifugation for 5 min at 14,500 rpm, evaporation of supernatant, and reconstitution using ammonium formate and 0.1% formic acid in water-acetonitrile (6:4, v/v)LC-MS/MSCalibration range of 0.5–50 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable in autosampler at 6 °C for 72 h; 20 °C, 4 °C, and RT for 15 days; −20 °C for 8 months
Dilution integrity was not assessed[61]
8Sirolimus Water and methanol + ISSonication for 15 minCentrifugation for 5 min at 14,500 rpm, evaporation of supernatant, and reconstitution using ammonium formate and 0.1% formic acid in water-acetonitrile (6:4, v/v)LC-MS/MSCalibration range of 0.5–50 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable in autosampler at 6 °C for 72 h; 20 °C, 4 °C, and RT for 15 days; −20 °C for 8 months
Dilution integrity was not assessed[61]
9Everolimus Water and methanol + ISSonication for 15 minCentrifugation for 5 min at 14,500 rpm, evaporation of supernatant, and reconstitution using ⁰ammonium formate and 0.1% formic acid in water-acetonitrile (6:4, v/v)LC-MS/MSCalibration range of 0.5–50 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable in autosampler at 6 °C for 72 h; 20 °C, 4 °C, and RT for 15 days; −20 °C for 8 months
Dilution integrity was not assessed[61]
10Cyclosporin AWater and methanol + ISSonication for 15 minCentrifugation for 5 min at 14,500 rpm, evaporation of supernatant, and reconstitution using ammonium formate and 0.1% formic acid in water-acetonitrile (6:4, v/v)LC-MS/MSCalibration range of 20–2000 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable in autosampler at 6 °C for 72 h; 20 °C, 4 °C, and RT for 15 days; −20 °C for 8 months
Dilution integrity was not assessed[61]
11TacrolimusAcetonitrile-zinc sulfate 0.05 M in water (1:1, v/v) + ISVortex for 1 minAddition of ammonium sulfate 40% for salting out, vortex for 1 min, centrifugation at 16,260× g for 5 min at 8 °CLC-MS/MSCalibration range of 2.25–42.9 ng/mL
Linearity, accuracy and precision, sensitivity, selectivity, recovery, matrix effect, and carry-over were assessed and met the acceptance criteria
Stable at RT for 7 days; 4 °C and 60 °C for 48 h; −80 °C for 1 month
Dilution integrity was not assessed[53]
IS: internal standard; LC-MS/MS: liquid chromatograph–tandem mass spectrometer; RT: room temperature.
Table 4. Clinical Validation of VAMS Assays versus Assays for CVS for Several Drugs.
Table 4. Clinical Validation of VAMS Assays versus Assays for CVS for Several Drugs.
NoDrugSample (n)Patient (n)Measures of Clinical ValidationRef.
Passing–Bablock Analysis
(Slope [95% CI])
Bland–Altman AnalysisPredictive Performance
1Albendazole 10010NAGood AgreementNA[51]
2Tacrolimus8872Linear
(0.88 [0.81; 0.97]) a
(1.00 [0.98; 1.02]) b
Good agreement
(after correction)
Met the acceptance criteria[108]
67927Linear
(1.01 [NA])
Good agreementNA[97]
5353Linear
(1.26 [1.16; 1.4]) a
(0.99 [0.91; 1.07]) b
Good agreement
(after correction)
Met the acceptance criteria[61]
9725Linear
(1.05 [0.98; 1.14])
Good agreementNA[109]
3Radiprodil 15010NAGood agreementNA[81]
4Mycophenolic acid2020Linear
(0.98 [0.94; 1.05])
Good agreementMet the acceptance criteria[61]
6425Linear
(0.72 [0.66; 0.77]) a
(1.07 [0.97; 1.13]) b
Good agreement
(after correction)
NA[109]
NA, not available; a, uncorrected concentration; b, corrected concentration.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nugraha, R.V.; Yunivita, V.; Santoso, P.; Hasanah, A.N.; Aarnoutse, R.E.; Ruslami, R. Analytical and Clinical Validation of Assays for Volumetric Absorptive Microsampling (VAMS) of Drugs in Different Blood Matrices: A Literature Review. Molecules 2023, 28, 6046. https://doi.org/10.3390/molecules28166046

AMA Style

Nugraha RV, Yunivita V, Santoso P, Hasanah AN, Aarnoutse RE, Ruslami R. Analytical and Clinical Validation of Assays for Volumetric Absorptive Microsampling (VAMS) of Drugs in Different Blood Matrices: A Literature Review. Molecules. 2023; 28(16):6046. https://doi.org/10.3390/molecules28166046

Chicago/Turabian Style

Nugraha, Rhea Veda, Vycke Yunivita, Prayudi Santoso, Aliya Nur Hasanah, Rob E. Aarnoutse, and Rovina Ruslami. 2023. "Analytical and Clinical Validation of Assays for Volumetric Absorptive Microsampling (VAMS) of Drugs in Different Blood Matrices: A Literature Review" Molecules 28, no. 16: 6046. https://doi.org/10.3390/molecules28166046

Article Metrics

Back to TopTop