Isha
Taneja†
ab,
Kanumuri Siva Rama
Raju†
ab,
Monika
Mittal
c,
Kapil
Dev
ad,
Mohammad Faheem
Khan
ad,
Rakesh
Maurya
ad and
Muhammad
Wahajuddin
*ab
aAcademy of Scientific and Innovative Research, New Delhi, India
bPharmacokinetics and Metabolism Division, CSIR- Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India. E-mail: wahajuddin@cdri.res.in; wahajuddin@gmail.com; Fax: +91-522-2771941; Tel: +91-522-2772450 ext. 4849 Tel: +91-522-2772550 ext. 4850
cDivision of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, India
dMedicinal and Process Chemistry Division, CSIR- Central Drug Research Institute, Lucknow, India
First published on 22nd May 2015
Alda-1 is an activator of the enzyme ALDH2. It has been suggested as a novel therapeutic for cardiovascular implications such as myocardial infarction, coronary bypass surgery, heart transplantation, peripheral artery disease, ischemia reperfusion injury, angina and alcoholic cardiomyopathy. Despite its widespread experimental use, no reports are available on its pharmacokinetics or bioanalytical quantification. In the present study, a simple, precise and reliable LC-ESI-MS/MS method has been developed and validated for the first time for quantification of alda-1 in plasma. Alda-1 was analyzed on a C18 column using methanol and 0.1% formic acid (60
:
40, v/v) as the mobile phase at a flow rate of 0.7 mL min−1. The method was found to be linear within the concentration range of 1–500 ng mL−1. The intra- and inter-day precision and accuracy were within acceptable limits. For the first time, the preclinical oral and intravenous pharmacokinetics of alda-1 were conducted. Alda-1 was found to be a rapidly absorbed, high clearance and poorly bioavailable compound in rats. Its plasma protein binding was found to be 82–86%. In view of the new regulatory guidelines, incurred sample reanalysis was also performed and all the samples were found within 15% of the mean value. From the in vitro microsomal incubation studies, it was found to be a high extraction compound. The data presented here provide important information to support the in vivo efficacy of alda-1 and would be helpful in its further development as a therapeutic agent and synthesis of its analogs with better systemic exposure and disposition properties.
In approximately 40% of the East Asians, ALDH2 occurs as mutant ALDH2*2 due to a single nucleotide substitution of G to A in exon 12, resulting in glutamate at position 487 instead of lysine. This single amino acid polymorphism causes disruption of the co-enzyme NAD binding thereby leading to reduced catalytic activity. The mutant ALDH2*2 has 200 fold higher Km for NAD+ and 10 fold lower kcat as compared to the wild-type enzyme.3 As a result, the homozygous ALDH2*2 have 1–5% activity of the wild-type ALDH2*1 while the heterozygous ALDH2*1/*2 have 10–45% activity. In individuals having mutant ALDH2, acetaldehyde accumulates upon consumption of alcohol causing Asian flushing syndrome/alcohol flush reaction, characterized by facial flushing, nausea, drowsiness and increased heartbeat. The mutant ALDH2 also leads to decreased vasodilatory effect of nitroglycerin used in the treatment of angina, hypertension and myocardial infarction. An increased susceptibility to upper digestive tract cancers and neurodegenerative diseases has also been associated with ALDH2*2.
Alda-1 (Fig. 1), chemically N-(1,3-benzodioxol-5-ylmethyl)-2,6-dichlorobenzamide, is an activator of the enzyme ALDH2, acting as a structural chaperone and restoring the activity of ALDH2*2.1 It has been proposed that alda-1 promotes the apparent binding affinity for NAD+, the essential cofactor for the functioning of ALDH2. Thus, it reinstates the structural and functional conformation of the active site of ALDH2 resulting in its increased dehydrogenase, esterase and nitroglycerin denitration activity.4 The binding of alda-1 decreases the apparent Km for NAD+ by 6.7-fold and increases the Vmax by 2-fold.3 Due to the protective role of ALDH2 in regulation of free radical generation, metabolism of 4-HNE and mitochondrial dysfunction, activators of ALDH2 such as alda-1 that restore its activity have been advocated as novel therapeutics for cardiovascular implications such as myocardial infarction, coronary bypass surgery, heart transplantation, peripheral artery disease, ischemia reperfusion injury, angina and alcoholic cardiomyopathy.5,6 The in vivo cardio-protective role of alda-1 has been confirmed in myocardial infarction rat model. Administration of alda-1 at 8 mg kg−1 prior to ischemia reduced the infarct size by 60%.2
Despite the progressive research on its clinical utility and its popular use as an experimental tool, no literature is reported describing its pharmacokinetic and disposition aspects. Also, to the best of our knowledge, no method is reported for its quantification in biomatrix. Thus, in the present study, we have developed and validated an LC-ESI-MS/MS method for the quantification of alda-1 in rat plasma for the first time. We have also determined its in vivo preclinical pharmacokinetics, protein binding and in vitro metabolic profile for the first time. Understanding the pharmacokinetic and metabolic aspects of any therapeutic agent is important since its in vitro and in vivo efficacy is correlated to the systemic concentration and the exposure time. In vivo pharmacokinetic studies give an insight into the dose-exposure relationship while the in vitro metabolic profile provides useful information regarding elimination mechanism of the compound and rate of biotransformation, both of which decide the residence time of the compound in biological system. Since, it is only the free fraction of the drug that is available for exerting the pharmacodynamics response and that gets distributed and excreted from the body, the plasma protein binding studies becomes important. Thus, the data presented here provide important information to support the in vivo efficacy of alda-1 and would be helpful in its further development as a therapeutic agent and synthesis of its analogs with better systemic exposure and disposition properties.
:
6
:
1 approximately, which is the typical mass spectra peak pattern for chlorine atom. Its sodium adduct was also observed in the similar isotopic pattern in the ratio of 9
:
6
:
1 for [M + Na]+, [M + Na + 2]+ and [M + Na + 4]+. For medicarpin (internal standard, IS), the protonated molecular ion [M + H]+ appeared at m/z 271.6. During the MRM scan, m/z 325.9 was found to have better signal-to-noise ratio than 323.9 or 328.0 and was chosen as the precursor ion for quantifying alda-1. Following detailed optimization of mass spectrometry conditions, the transitions m/z 325.9 precursor ion [M + H + 2]+ to the m/z 135.1 product ion and m/z 271.6 precursor ion [M + H]+ to the 137.1 m/z product ion were selected as the quantifier transition for alda-1 and IS, respectively. The transitions m/z 325.9 precursor ion [M + H + 2]+ to the m/z 77.2 product ion and m/z 325.9 precursor ion [M + H + 2]+ to the m/z 51 were selected as the qualifier transitions for alda-1. The ratio of the qualifier peak to the quantifier peak for m/z 77.2 product ion was found to be 0.175 and 0.170 for m/z product ion 55 and this ratio deviated within ±5.6% and ±5.5% for the respective transitions. The Q1 and product ion spectra of alda-1 and IS are shown in Fig. 2. The fragmentation pattern of alda-1 is shown in Fig. 3.
The LC method was developed by examining the feasibility of various combinations of solvents such as acetonitrile and methanol as organic modifiers; and ammonium acetate, ammonium formate, formic acid and acetic acid as aqueous phase. Different C18 columns were tried along with altered mobile phase compositions and flow rates to achieve good peak shape, sensitivity and selectivity for alda-1 and IS. The LC method was optimized with mobile phase comprising of methanol and 0.1% formic acid in ratio of 60
:
40 (v/v) which was delivered at a flow rate of 0.7 mL min−1 for chromatographic separation on a Waters XBridge® C18 column (4.6 mm × 50 mm, 5.0 μm).
| Alda-1 (ng mL−1) | ||||
|---|---|---|---|---|
| 1 (LLOQ) | 3 (LQC) | 150 (MQC) | 420 (HQC) | |
| a Calculated as (mean determined concentration/nominal concentration) × 100. b Expressed as %R.S.D. (S.D./mean) × 100. | ||||
| Intra-day | ||||
| Day 1 | ||||
| Mean ± SD | 0.91 ± 0.05 | 2.77 ± 0.11 | 156 ± 2.97 | 415.83 ± 20.36 |
| Accuracya (%) | 91.12 | 92.44 | 104.00 | 99.01 |
| Precisionb (%) | 5.14 | 4.06 | 1.90 | 4.90 |
![]() |
||||
| Day 2 | ||||
| Mean ± SD | 1.06 ± 0.04 | 2.83 ± 0.25 | 142.83 ± 6.05 | 398.33 ± 9.44 |
| Accuracya (%) | 106.33 | 94.33 | 95.22 | 94.84 |
| Precisionb (%) | 3.29 | 8.67 | 4.23 | 2.37 |
![]() |
||||
| Day 3 | ||||
| Mean ± SD | 1.01 ± 0.12 | 3.07 ± 0.36 | 140.17 ± 6.88 | 415.83 ± 20.36 |
| Accuracya (%) | 100.83 | 102.33 | 93.44 | 99.01 |
| Precisionb (%) | 11.78 | 11.63 | 4.91 | 4.90 |
![]() |
||||
| Inter-day | ||||
| Mean ± SD | 0.95 ± 0.08 | 2.92 ± 0.29 | 152.67 ± 7.51 | 401.61 ± 22.60 |
| Accuracya (%) | 94.85 | 97.41 | 101.78 | 95.62 |
| Precisionb (%) | 8.00 | 9.82 | 4.92 | 5.63 |
| Alda-1 (ng mL−1) | ||||
|---|---|---|---|---|
| 1 (LLOQ) | 3 (LQC) | 150 (MQC) | 420 (HQC) | |
| Bench top stability (6 h) | ||||
| Mean ± SD | 1.00 ± 0.10 | 2.97 ± 0.19 | 150.33 ± 3.20 | 394.33 ± 19.99 |
| Accuracy | 99.16 | 96.85 | 96.37 | 94.83 |
| Precision | 10.27 | 6.52 | 2.13 | 5.07 |
![]() |
||||
| Auto sampler stability (24 h) | ||||
| Mean ± SD | 0.99 ± 0.13 | 2.86 ± 0.18 | 155.50 ± 5.09 | 444.67 ± 22.92 |
| Accuracy | 98.20 | 93.00 | 99.68 | 106.93 |
| Precision | 13.51 | 6.17 | 3.27 | 5.16 |
![]() |
||||
| Long term stability (30 days) | ||||
| Mean ± SD | 0.92 ± 0.06 | 2.92 ± 0.30 | 157.33 ± 6.57 | 381.17 ± 12.59 |
| Accuracy | 91.45 | 95.22 | 100.85 | 91.66 |
| Precision | 6.35 | 10.10 | 4.18 | 3.30 |
![]() |
||||
| Freeze thaw stability (3 cycles) | ||||
| Mean ± SD | 1.02 ± 0.07 | 3.14 ± 0.25 | 138.50 ± 7.26 | 371.50 ± 8.69 |
| Accuracy | 101.47 | 102.28 | 88.78 | 89.34 |
| Precision | 7.07 | 8.06 | 5.24 | 2.34 |
| Parameter | Alda-1 |
|---|---|
| Plasma protein binding (%) | 82.83 ± 1.81 (at 100 ng mL−1) |
| 85.94 ± 2.15 (at 1000 ng mL−1) | |
| In vitro t 1/2 (min) | 11.01 ± 0.93 |
| Intrinsic clearance (CLint) (μL min−1 kg−1) | 126.3 ± 10.63 |
| Whole liver clearance (mL min−1 kg−1) | 227.34 ± 19.14 |
| Hepatic clearance (mL min−1 kg−1) | 53.49 ± 1.06 |
| Hepatic extraction ratio | 0.76 ± 0.01 |
![]() | ||
| Fig. 7 Mean plasma concentration–time plots of alda-1 following intravenous (10 mg kg−1) and oral (40 mg kg−1) administration (mean ± S.D). | ||
| Parameter | Intravenous (10 mg kg−1) | Oral (40 mg kg−1) |
|---|---|---|
| t 1/2 (h) | 1.67 ± 0.54 | 7.33 ± 1.37 |
| C o (ng mL−1) | 8774.71 ± 955.72 | — |
| C max (ng mL−1) | — | 69.88 ± 4.60 |
| T max (h) | — | 0.63 ± 0.25 |
| V d (L kg−1) | 9.39 ± 1.80 | 41.50 ± 2.95 |
| CL (L h−1 kg−1) | 3.97 ± 0.54 | 4.01 ± 0.72 |
| AUC0–t (h × ng mL−1) | 2536.07 ± 347.25 | 415.83 ± 57.04 |
| AUC0–∞ (h × ng mL−1) | 2540.80 ± 246.19 | 469.12 ± 81.24 |
| Bioavailability (%) | — | 4.62 |
Upon oral administration, the peak concentration was reached at 0.63 h and was found to be 69.88 ng mL−1. This suggests that alda-1 has a high absorption rate. It was found to have a low oral bioavailability of 4.62%. Upon intravenous administration, alda-1 was detectable up to 9 h only. The high clearance, 3.97 ± 0.54 L h−1 kg−1, of alda-1 suggest it to be a high extraction compound. However, the in vivo clearance was found to be significantly higher than the hepatic clearance (1.28 L h−1 kg−1) obtained from microsomal studies. This implies that apart from CYP-mediated phase I biotransformation, other elimination mechanisms such as phase II biotransformation or renal elimination might also be involved that are responsible for the high in vivo clearance of this compound. The high hepatic extraction of alda-1 and its poor bioavailability could prove to be a limiting factor in clinical development of this compound. During the reanalysis of the incurred samples, all the samples were found to be within ±15% of the mean value, thus, re-establishing the validity and robustness of the developed method.
Animals and prerequisites: blank, drug free plasma samples were collected from adult, healthy female Sprague–Dawley (SD) rats at the Division of Laboratory Animals (DOLA) of Central Drug Research Institute (Lucknow, India). Prior approval from the Institutional Animal Ethics Committee (IAEC) was sought for maintenance, experimental studies, euthanasia and disposal of carcass of animals. Plasma was obtained by centrifuging the heparinised blood (25 IU per mL) at 2000× g for 10 min at 20 °C.
:
0.1% formic acid in the ratio of 60
:
40 (v/v) at a flow rate of 0.7 mL min−1. Mobile phase was duly filtered through 0.22 μm Millipore filter (Billerica, USA) and degassed ultrasonically for 15 min prior to use. Separations were performed at room temperature. The injection volume was kept at 20 μL.
Mass spectrometric detection was performed in multiple reaction monitoring (MRM) mode on an API 4000 QTRAP mass spectrometer (Applied Biosystems, MDS Sciex Toronto, Canada) equipped with an API electrospray ionization (ESI) source and triple quadrupole mass analyser. Quadrupoles Q1 and Q3 were set on unit resolution in positive ion mode. The ion spray voltage was set at 5500 V. The instrument parameters viz., nebulizer gas, curtain gas, auxillary gas and collision gas were set at 50, 20, 50 and 8 psi, respectively. Compound related parameters viz., declustering potential (DP), collision energy (CE), entrance potential (EP) and collision cell exit potential (CXP) were 66, 29, 10, 26 V; and 50, 25, 10, 10 V for alda-1 and IS, respectively. Zero air was used as source gas while nitrogen was used as both curtain and collision gas. The transition of m/z 325.9 precursor ion [M + H + 2]+ to the m/z 135.1 product ion was monitored for alda-1 and m/z 271.6 precursor ion [M + H]+ to the 137.1 m/z product ion for IS. Data acquisition and quantitation were performed using Analyst® software version 1.6 (Applied Biosystems, MDS Sciex Toronto, Canada).
000 g on Sigma 3-16K (Frankfurt, Germany). The supernatant was separated and 20 μL was injected for LC-MS/MS analysis.
Specificity was tested by analyzing plasma samples spiked with alda-1 and IS in comparison to six randomly selected blank plasma samples to investigate potential interferences at the LC peak region of both the analytes. Recovery was assessed by comparing the peak responses of the pre-extracted plasma samples to those of post-extracted blank plasma samples spiked with equivalent concentration of the analytes. The recovery of IS was determined at 50 ng mL−1. The matrix effect was evaluated by comparing the peak response of the post extracted spiked plasma samples and the unextracted analytical neat samples at equivalent concentrations.8,9 The intraday accuracy and precision were determined by six repeat analyses (intrabatch variability) of the four QC concentrations. The interday accuracy and precision were determined by analyzing four QC concentrations in three different runs (interbatch variability) on 3 days. Accuracy should lie within ±15% deviation of the nominal value and precision within ±15% relative standard deviation (RSD) within except for LLOQ where both should lie within ±20%. The stability of plasma samples was assessed under different storage conditions: bench top (BT) stability at room temperature for 6 h; long term (LT) stability at −80 °C for 30 days, autosampler (AS) stability at 4 °C for 24 h and; freeze/thaw (FT) stability after 3 freeze/thaw cycles. Autosampler carry over was determined in triplicate by injecting the highest calibration standard followed by a blank sample and was considered insignificant if the measured peak area was less than 20% of the lowest calibrator area. Dilution integrity was performed by 20 times dilution of the plasma samples containing 8000 ng mL−1 of alda-1 with blank plasma to obtain plasma samples containing 400 ng mL−1 of alda-1. The response after dilution should not vary more than 15% of the true value. An incurred sample reanalysis (ISR) was performed by selecting the in vivo pharmacokinetic study samples, to show reproducibility of method. Four samples were selected from each rat including the time points at Cmax and elimination phase to cover the entire profile of the individual subject. Thus, the ISR was performed on 24 samples out of total 88 samples. The difference between the original and reanalyzed concentrations of the analyte should be within 20% for at least 67% of the total samples reanalyzed.7
000 rpm. 20 μL of supernatant was directly analyzed by LC-MS/MS. The data was analysed using GraphPad Prism 5.0 software. Non-linear regression of percent parent remaining against time was performed as per the equation:| %Parent drug remaining = span × e−kt + plateau |
The in vitro parameters were estimated using the following formulae:
000 rpm for 10 min on Sigma 1-15K (Frankfurt, Germany). Samples were stored at −80 ± 10 °C until bioanalysis. All the four QC concentrations (n = 5 each) were analysed with each batch of study samples. The observed maximum serum concentration (Cmax) and the time to reach the maximum serum concentration (Tmax) were obtained by visual inspection of the experimental data. The data was subjected to non-compartmental pharmacokinetic analysis using WinNonlin (version 5.1, Pharsight Corporation, Mountain View, USA). Area under curve (AUC) from 0 to 24 h (AUC0–24) was calculated using linear trapezoidal rule. AUC from 0 to infinity (AUC0–∞) was calculated as the sum of AUC0–t and Clast/kel, where, Clast represents the last quantifiable concentration and kel represents the elimination rate constant.
Footnote |
| † Equally contributing authors. |
| This journal is © The Royal Society of Chemistry 2015 |