WO2015142130A1 - Method for detecting porphyrin in a biological sample by using lc-ms/ms - Google Patents

Method for detecting porphyrin in a biological sample by using lc-ms/ms Download PDF

Info

Publication number
WO2015142130A1
WO2015142130A1 PCT/KR2015/002779 KR2015002779W WO2015142130A1 WO 2015142130 A1 WO2015142130 A1 WO 2015142130A1 KR 2015002779 W KR2015002779 W KR 2015002779W WO 2015142130 A1 WO2015142130 A1 WO 2015142130A1
Authority
WO
WIPO (PCT)
Prior art keywords
porphyrin
biological sample
blood
samples
methanol
Prior art date
Application number
PCT/KR2015/002779
Other languages
French (fr)
Inventor
Young Shin Lee
Seung Woo Kang
Yeoun Hur
Sang Beom Han
Ju Hee Jung
Kwang Jin Kim
Original Assignee
Inernational Scientific Standards, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Inernational Scientific Standards, Inc. filed Critical Inernational Scientific Standards, Inc.
Publication of WO2015142130A1 publication Critical patent/WO2015142130A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/88Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/72Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving blood pigments, e.g. haemoglobin, bilirubin or other porphyrins; involving occult blood
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/88Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86
    • G01N2030/8809Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample
    • G01N2030/8813Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample biological materials
    • G01N2030/8822Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample biological materials involving blood
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2560/00Chemical aspects of mass spectrometric analysis of biological material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • G01N30/7233Mass spectrometers interfaced to liquid or supercritical fluid chromatograph

Definitions

  • the present invention relates to a method for detecting porphyrin by using LC-MS/MS (Liquid Chromatography coupled to Tandem Mass Spectrometry).
  • porphyrin makes up heme, which is a component of hemoglobin. Porphyrin is also a component of cytochrome, which plays an important role in oxidation and reduction in vivo. Further, porphyrin is a component of chlorophyll, which is involved in photosynthesis.
  • Porphyrin is related to various diseases.
  • a typical example of porphyrin-related disease is porphyria, which is caused by accumulation of porphyrin and iron in internal organs caused by the lack of enzymes involved in the synthesis of heme due to inherited or acquired genetic defects.
  • the common symptoms of Porphyria are neurovisceral or cutaneous symptoms. Neurovisceral symptoms appear after puberty and are more common in women, and include acute seizures, abdominal pain, and damaged sensorimotor nervous systems. Cutaneous symptoms, which appear after 30s in most cases, include excessive peeling, blisters and scars after being exposed to sunlight. Hepatopathy associated with the accumulation of porphyrin may appear together.
  • porphyrin is receiving attention as a biomarker of autism or autistic spectrum disorder.
  • concentration of porphyrin such as coproporphyrin, hexacarboxylporphyrin and pentacarboxylporphyrin is high in the urine of children with autism.
  • excessive amount of porphyrin causes heavy metal such as mercury to remain longer in vivo and triggers autism in result.
  • chelators such as DMSA or ALA improved the symptoms of autism.
  • Porphyrin is excreted in urine, feces and bile, and is particularly released in a large amount in the urine. Porphyrin is also present in blood, but its amount is too small to detect in tests. Tests for porphyrin are usually performed by using LC-MS/MS. However, it is hard to detect a small amount of porphyrin with conventional methods.
  • the purpose of the present invention is to provide a method for detecting porphyrin in a biological sample using LC-MS/MS, wherein the method includes pretreating the biological sample with a mixed solution of methanol and strong acid.
  • the present invention provides a method for detecting porphyrin in a biological sample, comprising: 1) obtaining a biological sample from a subject; 2) pretreating the biological sample with a mixed solution of methanol and strong acid; and 3) analyzing the pretreated biological sample by using LC-MS/MS.
  • the subject may be a human.
  • the pre-treatment of 2) may be performed by bringing the biological sample into contact with the mixed solution of methanol and strong acid or mixing the biological sample with the mixed solution of methanol and strong acid.
  • porphyrin commonly denotes compounds having the following basic structure and derivatives thereof.
  • porphyrin may vary. Protoporphyrin, harderoporphyrin, coproporphyrin, pentacarboxylporphyrin, hexacarboxylporphyrin, heptacarboxylporphyrin, and uroporphyrin, etc. are generally present in blood, and excreted in urine and feces. They are all referred to as porphyrin. Furthermore, there are type I, II, and III for each porphyrin, and they are all referred to as porphyrin.
  • porphyrin include one or more carboxyl groups.
  • protoporphyrin has two carboxyl groups
  • harderoporphyrin has three carboxyl groups
  • coproporphyrin has four carboxyl groups
  • pentacarboxylporphyrin has five carboxyl groups
  • hexacarboxylporphyrin has six carboxyl groups
  • heptacarboxylporphyrin has seven carboxyl groups
  • uroporphyrin has eight carboxyl groups.
  • the porphyrin is more soluble to water and more likely to be excreted in urine. Porphyrins with 4 carboxyl groups tend to be excreted in urine or feces, and harderoporphyrin or protoporphyrin tend to be excreted in feces.
  • LC-MS/MS Liquid Chromatography coupled to Tandem Mass Spectrometry
  • LC-MS Liquid Chromatography coupled to Tandem Mass Spectrometry
  • a sample is injected into the column by using HPLC and the components contained in the sample are separated. Then the components enter the Mass spectrometer and are ionized, and show signal peaks at different times, from which the corresponding materials may be identified.
  • LC-MS/MS which has more than one (typically two) mass spectrometers, enables more accurate analysis of materials. Since LC-MS/MS is capable of separating materials in samples and identifying the materials, it is widely used in analysis of a variety of materials.
  • the method of the present invention includes pretreating the obtained biological sample with the mixed solution of methanol and strong acid, and thereby porphyrin is esterified so that the carboxyl group is substituted with a methyl ester group and the produced water is removed, ultimately enabling precise detection of porphyrin.
  • the method for the present invention is able to detect a small amount of porphyrin, it is more useful when the amount of biological sample to be obtained is small.
  • One example where only a small amount of sample can be obtained is the case of newborn infants. Since the newborn infants are small in size and very sensitive to stimulation, the number of times obtaining biological samples and the amount of obtainable biological samples are limited.
  • the method of the present invention it is possible to detect porphyrin in a droplet of blood, which is about 3 ⁇ L.
  • the subjects include but not limited to a baby within one month of birth, more preferably seven days of birth, and much more preferably 72 hours of birth.
  • One example the method of the present invention can be utilized is the neonatal screening tests.
  • Some of congenital metabolic disorders may cause irreparable damage upon late diagnosis but such damage may be prevented by treating in early stage.
  • the Korean Government supports neonatal screening tests for six kinds of congenital metabolic disorders with high incidence rate, and most parents additionally perform tests for the other dozens of diseases. These tests are carried out in a manner that the blood is drawn from the heel of a 3 ⁇ 7 day-old baby, dropped on a blood filter paper, dried, and analyzed in lab.
  • the blood filter paper contains only about 3 ⁇ L of blood. Since the method of the present invention enables the detection of porphyrin using blood of the blood filter paper, there is no need to obtain more biological sample from a subject to detect porphyrin.
  • the porphyrins include but not limited to porphyrins with four or more carboxyl groups, such as coproporphyrin, pentacarboxylporphyrin, hexacarboxylporphyrin, heptacarboxylporphyrin, or uroporphyrin.
  • the method of the present invention is effective in detecting porphyrin from a biological sample containing a small amount of porphyrin.
  • the biological samples in 1) include but not limited to blood, for example, whole blood, plasma and serum.
  • the biological samples in 1) include but not limited to urine, feces, and bile.
  • the mixing ratio of methanol and strong acid in 2) is 9:1 ⁇ 8:2 (v/v), and preferably 9:1.
  • the mixing ratio is not limited thereto.
  • 1) may include:
  • eluting the biological sample in c) may be conducted by bringing formic acid into contact with the blood filter paper on which the biological sample was dropped.
  • formic acid enables effective elution of the blood contained in the porous blood filter paper.
  • 0.5 M formic acid in distilled water can be used.
  • concentration of the solution which can be used in the method herein is not limited thereto.
  • the present invention By using the present invention, a small amount of porphyrin can be detected effectively. Accordingly, it is possible to detect porphyrin from biological samples containing a small amount of porphyrin such as blood. Even in the case where the number of times obtaining biological samples and the amount of obtainable biological samples are very limited, such as newborn infants, a precise detection of porphyrin is possible. Therefore, the method of the present invention can be utilized for early diagnosis of porphyrin-related diseases.
  • FIG. 1 is a schematic illustration of a blood filter paper
  • FIG. 2A illustrates the structures of coproporphyrin (CP), pentacarboxylporphyrin (PP), hexacarboxylporphyrin (HexaP), heptacarboxylporphyrin (HeptaP), uroporphyrin (UP), and coproporphyrin I- 15 N 4
  • FIG. 2B illustrates the structures of their esterified forms
  • FIG. 3A illustrates the results of mass analysis of CP, PP, HexaP, HeptaP, and UP
  • FIG.3B illustrates the results of mass analysis of their esterified forms
  • FIG. 4A illustrates the results of analysis of LC-MS/MS of a blank
  • FIG. 4B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L CPTE calibration solution
  • FIG. 5A illustrates the results of analysis of LC-MS/MS of a blank
  • FIG. 5B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L PPPE calibration solution
  • FIG. 6A illustrates the results of analysis of LC-MS/MS of a blank
  • FIG. 6B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L HexaPHE calibration solution
  • FIG. 7A illustrates the results of analysis of LC-MS/MS of a blank
  • FIG. 7B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L HeptaPHE calibration solution
  • FIG. 8A illustrates the results of analysis of LC-MS/MS of a blank
  • FIG. 8B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L UPOE calibration solution.
  • Example 1 Testing instruments, testing devices, and materials
  • LC-MS/MS used in the Examples herein includes the mass spectrometers and columns listed in the following Table 1.
  • testing devices used in the Examples herein are as in the Table 2 below.
  • CP, PP, HexaP, HeptaP, and UP which were purchased from Frontier Scientific, were added to the blood prepared in (1), and solutions containing the above compounds at a concentration of 10 nmol/L respectively were prepared.
  • Each of the prepared solutions was dropped on a blood filter paper schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare control samples (dried blood spot samples). Then 200 ⁇ L of 0.5 M formic acid in DW was added to the control sample to elute the compounds from the blood filter paper.
  • CP, PP, HexaP, HeptaP, and UP which were purchased from Frontier Scientific, were added to the blood prepared in (1), and solutions containing the above compounds at a concentration of 10 nmol/L respectively were prepared.
  • Each of the prepared solutions was dropped on a blood filter paper schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare testing samples (dried blood spot samples).
  • CP, PP, HexaP, HeptaP, UP and coproporphyrin I- 15 N 4 were esterified via the reaction mechanism below to coproporphyrin tetramethyl ester (CPTE), pentacarboxylporphyrin pentamethyl ester (PPPE), hexacarboxylporphyrin hexamethyl ester (HexaPHE), heptacarboxylporphyrin heptamethyl ester (HeptaPHE), uroporphyrin octamethyl ester (UPOE), and coproporphyrin I- 15 N 4 tetramethyl ester, respectively.
  • CPTE coproporphyrin tetramethyl ester
  • PPPE pentacarboxylporphyrin pentamethyl ester
  • HexaPHE hexacarboxylporphyrin hexamethyl ester
  • HeptaPHE heptacar
  • the X axis represents time (min) and the Y axis represents the concentration (cps).
  • Each peak corresponds to CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE from the right. Since the mass of each compound is increased upon esterification, the peaks of FIG. 3B were shifted rightward compared to FIG. 3A.
  • Calibration standards and quality control (QC) samples were prepared as follows, and measured to check system suitability, specificity, carry-over, accuracy and precision, thereby evaluating whether the tests of Example 3 were appropriately performed.
  • CP 0.75 mg of CP, 0.86 mg of PP, 0.91 mg of HexaP, 0.96 mg of HeptaP, and 1.00 mg of UP were weighed, dissolved in 100 mL of 6 M formic acid in DW, to prepare 10,000 nmol/L stock solutions for CP, PP, HexaP, HeptaP, and UP, respectively. The stock solutions were then frozen and stored.
  • the working solutions for CP, PP, HexaP, HeptaP, and UP prepared in (1) were subjected to serial dilution with 6 M formic acid as shown in Table 7 below, to prepare QC solutions.
  • the kinds of solutions taken in Table 7 below ((A), (E), (F)) are as represented in Table 6.
  • QH denotes high-concentration QC solution (H: High)
  • QM denotes medium-concentration QC solution (M: Medium)
  • QL denotes low-concentration QC solution (L: Low)
  • LLOQ denotes a QC solution having a concentration corresponding to the lowest limit of quantification.
  • Each of the prepared calibration solutions was dropped on the blood filter paper as schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare calibration standards (dried blood spot samples).
  • the suitability of the present tester to ensure objectivity of the tests of Example 3 was evaluated as below. S1, among the calibration standards, and QL, among the QC samples, were injected into LC-MS/MS to ensure sensitivity. As an acceptance criterion for system suitability, the precision (CV, %) of the peak area ratio of each of CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE relative to the internal standard is 10% or less in principle, but was set to 15% in the present tests because five kinds of materials were analyzed simultaneously.
  • the sample was determined to be free of any material interfering the subject material.
  • FIGS. 4A to 8A show the results of mass analysis of the blanks
  • FIGS. 4B to 8B show the results of analysis of each compound.
  • the blank does not include any material showing a peak at 8.59 min, which corresponds to the peak of CPTE.
  • FIGS. 5A and 5B it can be understood that the blank does not include any material showing a peak at 7.64 min, which corresponds to the peak of PPPE.
  • FIGS. 4A to 8A show the results of mass analysis of the blanks
  • FIGS. 4B to 8B show the results of analysis of each compound.
  • the blank does not include any material showing a peak at 8.59 min, which corresponds to the peak of CPTE.
  • the blank does not include any material showing a peak at 7.64 min, which corresponds to the peak of PPPE.
  • the blank does not include any material showing a peak at 6.88 min, which corresponds to the peak of HexaPHE.
  • FIGS. 7A and 7B it can be understood that the blank does not include any material showing a peak at 6.24 min, which corresponds to the peak of HeptaPHE.
  • FIGS. 8A and 8B it can be understood that the blank does not include any material showing a peak at 5.77 min, which corresponds to the peak of UPOE. Accordingly, the porphyrin-free whole blood used in the tests did not include any material that interfere CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE.
  • a calibration curve was drawn, from which correlation coefficients were determined.
  • An acceptance criterion is as follows: a deviation from the nominal concentration at LOQ has to be within 20% and a deviation from the nominal concentration at a concentration other than LOQ has to be within 15%, where r (correlation coefficient) has to be 0.98 or more.
  • the correlation coefficient (r) in the concentration range of 0.2 ⁇ 50 nmol/L is 0.9982 ⁇ 0.9999 for CPTE, 0.9815 ⁇ 0.9999 for PPPE, 0.9958 ⁇ 0.9999 for HexaPHE, 0.9938 ⁇ 0.9986 for HeptaPHE, and 0.9980 ⁇ 0.9990 for UPOE, all of which were equal to or higher than the acceptance criterion, and the accuracy results satisfied the acceptance criterion.
  • the specific numerical values thereof are provided in Tables 12 to 16 below.
  • Accuracy was expressed as a percentage (%) of the value obtained in such a manner that the numerical values obtained from injecting QC samples into LC-MS/MS were substituted into the calibration curves to give the mean concentration that was then divided by the concentration of the actual QC samples as known. Five tests were performed per day using QH, QM, QL, and LLOQ samples to calculate intra-day accuracy, and the tests were repeated for five days to calculate inter-day accuracy.
  • the precision was expressed as a percentage (%) of the value obtained in such a manner that the numerical values resulting from injecting the QC samples in LC-MS/MS were divided by the mean value of the peak area ratios of CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE and coproporphyrin tetramethyl ester I- 15 N 4 as the internal standard. Five tests were performed per day using QH, QM, QL, and LLOQ samples to calculate intra-day precision, and the tests were repeated for five days to calculate inter-day precision.
  • CPTE precision of CPTE was 4.35%, 4.76%, 9.20% and 6.48% at LLOQ, QL, QM and QH, respectively.
  • accuracy of CPTE was 114.63%, 109.10%, 88.06% and 106.51% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 17 below.
  • the precision of PPPE was 4.14%, 7.58%, 5.93% and 5.39% at LLOQ, QL, QM and QH, respectively, and the accuracy of PPPE was 108.69%, 103.64%, 92.87% and 93.32% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 18 below.
  • HexaPHE The precision of HexaPHE was 11.53%, 4.88%, 2.63% and 2.28% at LLOQ, QL, QM and QH, respectively, and the accuracy of HexaPHE was 94.13%, 86.06%, 86.74% and 85.20% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 19 below.
  • HeptaPHE The precision of HeptaPHE was 17.79%, 7.57%, 14.27% and 13.15% at LLOQ, QL, QM and QH, respectively, and the accuracy of HeptaPHE was 104.07%, 101.50%, 93.47% and 103.24% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 20 below.
  • the precision of UPOE was 4.35%, 4.76%, 9.20% and 6.48% at LLOQ, QL, QM and QH, respectively, and the accuracy of UPOE was 114.63%, 109.10%, 88.06% and 106.51% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 21 below.
  • CPTE precision of CPTE was 4.80%, 12.11%, 12.71% and 7.14% at LLOQ, QL, QM and QH, respectively.
  • accuracy of CPTE was 114.72%, 103.08%, 97.49% and 109.05% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 22 below.
  • the precision of PPPE was 3.99%, 10.61%, 6.46% and 8.85% at LLOQ, QL, QM and QH, respectively.
  • the accuracy of PPPE was 111.49%, 102.53%, 101.14% and 102.60% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 23 below.
  • the precision of HexaPHE was 8.30%, 6.91%, 10.91% and 12.62% at LLOQ, QL, QM and QH, respectively.
  • the accuracy of HexaPHE was 99.32%, 94.33%, 100.69% and 103.69% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 24 below.
  • HeptaPHE The precision of HeptaPHE was 9.11%, 12.37%, 13.37% and 10.55% at LLOQ, QL, QM and QH, respectively.
  • the accuracy of HeptaPHE was 106.66%, 104.24%, 100.13% and 104.72% at LLOQ, QL, QM and QH, respectively.
  • the specific numerical values thereof are provided in Table 25 below.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Provided herein is a method for detecting porphyrin using LC-MS/MS by pretreating a biological sample. This method is very effective in detecting a small amount of porphyrin. Accordingly, it is possible to detect porphyrin from biological samples which contain a small amount of porphyrin such as blood. Furthermore, even in the case where the number of times obtaining biological samples and the amount of obtainable biological samples are very limited, such as newborn infants, a precise detection of porphyrin is possible. Hence, this method can be utilized for early diagnosis of porphyrin-related diseases.

Description

METHOD FOR DETECTING PORPHYRIN IN A BIOLOGICAL SAMPLE BY USING LC-MS/MS
The present invention relates to a method for detecting porphyrin by using LC-MS/MS (Liquid Chromatography coupled to Tandem Mass Spectrometry).
Together with iron, porphyrin makes up heme, which is a component of hemoglobin. Porphyrin is also a component of cytochrome, which plays an important role in oxidation and reduction in vivo. Further, porphyrin is a component of chlorophyll, which is involved in photosynthesis.
Porphyrin is related to various diseases. A typical example of porphyrin-related disease is porphyria, which is caused by accumulation of porphyrin and iron in internal organs caused by the lack of enzymes involved in the synthesis of heme due to inherited or acquired genetic defects. The common symptoms of Porphyria are neurovisceral or cutaneous symptoms. Neurovisceral symptoms appear after puberty and are more common in women, and include acute seizures, abdominal pain, and damaged sensorimotor nervous systems. Cutaneous symptoms, which appear after 30s in most cases, include excessive peeling, blisters and scars after being exposed to sunlight. Hepatopathy associated with the accumulation of porphyrin may appear together.
Methods to treat porphyria have not yet been developed. However, reducing exposure to sun and drugs, alcohol, female hormone, etc., or taking measures to prevent porphyrin from being accumulated in liver by periodic bloodletting early in the stage of porphyria is known to alleviate the symptoms. Mortality in patients who were not diagnosed early is reported to range from 10 ~ 40%.
Recently, porphyrin is receiving attention as a biomarker of autism or autistic spectrum disorder. There are reports that the concentration of porphyrin such as coproporphyrin, hexacarboxylporphyrin and pentacarboxylporphyrin is high in the urine of children with autism. Further, there are many papers reporting that excessive amount of porphyrin causes heavy metal such as mercury to remain longer in vivo and triggers autism in result. There are no methods available to treat autism now. However, Nafta et al.(2006) reported that chelators such as DMSA or ALA improved the symptoms of autism.
Although early diagnosis of porphyrin-related diseases by detecting and analyzing porphyrin is important, there is a limit to the tests for porphyrin to date. Porphyrin is excreted in urine, feces and bile, and is particularly released in a large amount in the urine. Porphyrin is also present in blood, but its amount is too small to detect in tests. Tests for porphyrin are usually performed by using LC-MS/MS. However, it is hard to detect a small amount of porphyrin with conventional methods.
The purpose of the present invention is to provide a method for detecting porphyrin in a biological sample using LC-MS/MS, wherein the method includes pretreating the biological sample with a mixed solution of methanol and strong acid.
The present invention provides a method for detecting porphyrin in a biological sample, comprising: 1) obtaining a biological sample from a subject; 2) pretreating the biological sample with a mixed solution of methanol and strong acid; and 3) analyzing the pretreated biological sample by using LC-MS/MS.
In the method for the present invention, the subject may be a human.
In the method for the present invention, the pre-treatment of 2) may be performed by bringing the biological sample into contact with the mixed solution of methanol and strong acid or mixing the biological sample with the mixed solution of methanol and strong acid.
As used herein, the term “porphyrin” commonly denotes compounds having the following basic structure and derivatives thereof.
[Structural Formula]
Figure PCTKR2015002779-appb-I000001
The form of porphyrin may vary. Protoporphyrin, harderoporphyrin, coproporphyrin, pentacarboxylporphyrin, hexacarboxylporphyrin, heptacarboxylporphyrin, and uroporphyrin, etc. are generally present in blood, and excreted in urine and feces. They are all referred to as porphyrin. Furthermore, there are type I, II, and III for each porphyrin, and they are all referred to as porphyrin.
Many kinds of porphyrin include one or more carboxyl groups. For example, protoporphyrin has two carboxyl groups, harderoporphyrin has three carboxyl groups, coproporphyrin has four carboxyl groups, pentacarboxylporphyrin has five carboxyl groups, hexacarboxylporphyrin has six carboxyl groups, heptacarboxylporphyrin has seven carboxyl groups, and uroporphyrin has eight carboxyl groups. As a porphyrin has more carboxyl groups, the porphyrin is more soluble to water and more likely to be excreted in urine. Porphyrins with 4 carboxyl groups tend to be excreted in urine or feces, and harderoporphyrin or protoporphyrin tend to be excreted in feces.
LC-MS/MS (Liquid Chromatography coupled to Tandem Mass Spectrometry) is composed of more than one mass spectrometers arranged in a row and connected to a liquid chromatography instrument consisting of columns and HPLC (High Performance Liquid Chromatography). In LC-MS, a sample is injected into the column by using HPLC and the components contained in the sample are separated. Then the components enter the Mass spectrometer and are ionized, and show signal peaks at different times, from which the corresponding materials may be identified. LC-MS/MS, which has more than one (typically two) mass spectrometers, enables more accurate analysis of materials. Since LC-MS/MS is capable of separating materials in samples and identifying the materials, it is widely used in analysis of a variety of materials.
However, it is very difficult to analyze compounds which have carboxyl groups, such as porphyrin, by using LC-MS/MS. Since carboxyl groups interfere the ionization of porphyrin in mass spectrometer, the sensitivity of the instrument is decreased and the precision of the analysis is impaired in result. The method of the present invention includes pretreating the obtained biological sample with the mixed solution of methanol and strong acid, and thereby porphyrin is esterified so that the carboxyl group is substituted with a methyl ester group and the produced water is removed, ultimately enabling precise detection of porphyrin.
Since the method for the present invention is able to detect a small amount of porphyrin, it is more useful when the amount of biological sample to be obtained is small. One example where only a small amount of sample can be obtained is the case of newborn infants. Since the newborn infants are small in size and very sensitive to stimulation, the number of times obtaining biological samples and the amount of obtainable biological samples are limited. When using the method of the present invention, it is possible to detect porphyrin in a droplet of blood, which is about 3 μL. Hence, in the present invention, the subjects include but not limited to a baby within one month of birth, more preferably seven days of birth, and much more preferably 72 hours of birth.
One example the method of the present invention can be utilized is the neonatal screening tests. Some of congenital metabolic disorders may cause irreparable damage upon late diagnosis but such damage may be prevented by treating in early stage. Hence, the Korean Government supports neonatal screening tests for six kinds of congenital metabolic disorders with high incidence rate, and most parents additionally perform tests for the other dozens of diseases. These tests are carried out in a manner that the blood is drawn from the heel of a 3 ~ 7 day-old baby, dropped on a blood filter paper, dried, and analyzed in lab. As such, the blood filter paper contains only about 3 μL of blood. Since the method of the present invention enables the detection of porphyrin using blood of the blood filter paper, there is no need to obtain more biological sample from a subject to detect porphyrin.
In the method of the present invention, the porphyrins include but not limited to porphyrins with four or more carboxyl groups, such as coproporphyrin, pentacarboxylporphyrin, hexacarboxylporphyrin, heptacarboxylporphyrin, or uroporphyrin.
Furthermore, the method of the present invention is effective in detecting porphyrin from a biological sample containing a small amount of porphyrin. Hence, the biological samples in 1) include but not limited to blood, for example, whole blood, plasma and serum. The biological samples in 1) include but not limited to urine, feces, and bile.
In the method of the present invention, the strong acids in 2) include but not limited to sulfuric acid, hydrochloric acid, and nitric acid, preferably sulfuric acid.
In the method of the present invention, the mixing ratio of methanol and strong acid in 2) is 9:1 ~ 8:2 (v/v), and preferably 9:1. However, the mixing ratio is not limited thereto.
In the method of the present invention, 1) may include:
a) obtaining a biological sample from a subject;
b) dropping the biological sample on a blood filter paper; and
c) eluting the biological sample from the blood filter paper.
In an embodiment of the present invention, eluting the biological sample in c) may be conducted by bringing formic acid into contact with the blood filter paper on which the biological sample was dropped. The use of formic acid enables effective elution of the blood contained in the porous blood filter paper. Preferably 0.5 M formic acid in distilled water can be used. However, the concentration of the solution which can be used in the method herein is not limited thereto.
By using the present invention, a small amount of porphyrin can be detected effectively. Accordingly, it is possible to detect porphyrin from biological samples containing a small amount of porphyrin such as blood. Even in the case where the number of times obtaining biological samples and the amount of obtainable biological samples are very limited, such as newborn infants, a precise detection of porphyrin is possible. Therefore, the method of the present invention can be utilized for early diagnosis of porphyrin-related diseases.
FIG. 1 is a schematic illustration of a blood filter paper;
FIG. 2A illustrates the structures of coproporphyrin (CP), pentacarboxylporphyrin (PP), hexacarboxylporphyrin (HexaP), heptacarboxylporphyrin (HeptaP), uroporphyrin (UP), and coproporphyrin I-15N4, and FIG. 2B illustrates the structures of their esterified forms;
FIG. 3A illustrates the results of mass analysis of CP, PP, HexaP, HeptaP, and UP, and FIG.3B illustrates the results of mass analysis of their esterified forms;
FIG. 4A illustrates the results of analysis of LC-MS/MS of a blank, and FIG. 4B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L CPTE calibration solution;
FIG. 5A illustrates the results of analysis of LC-MS/MS of a blank, and FIG. 5B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L PPPE calibration solution;
FIG. 6A illustrates the results of analysis of LC-MS/MS of a blank, and FIG. 6B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L HexaPHE calibration solution;
FIG. 7A illustrates the results of analysis of LC-MS/MS of a blank, and FIG. 7B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L HeptaPHE calibration solution; and
FIG. 8A illustrates the results of analysis of LC-MS/MS of a blank, and FIG. 8B illustrates the results of analysis of LC-MS/MS of a 50 nmol/L UPOE calibration solution.
The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.
[Examples]
Example 1. Testing instruments, testing devices, and materials
1-1. Testing instrument
LC-MS/MS used in the Examples herein includes the mass spectrometers and columns listed in the following Table 1.
[Table 1]
Figure PCTKR2015002779-appb-I000002
1-2. Testing devices
The testing devices used in the Examples herein are as in the Table 2 below.
[Table 2]
Figure PCTKR2015002779-appb-I000003
1-3. Reagents
The reagents used in the Examples herein are as in the Table 3 below.
[Table 3]
Figure PCTKR2015002779-appb-I000004
The structures of CP, PP, HexaP, HeptaP, UP, and coproporphyrin I-15N4 in the above Table 3 are illustrated in FIG. 2A.
1-4. Mobile phase
To 998 mL of distilled water (DW), 2 mL of formic acid was added to prepare a mobile phase A, and to 999 mL of acetonitrile, 1 mL of formic acid was added to prepare a mobile phase B.
1-5. Wash solvent
To 1000 mL of methanol, 1000 mL of distilled water was added to prepare 2000 mL of 50% methanol, which used as a wash solvent.
Example 2. LC-MS/MS analysis conditions and numerical treatment of values
2-1. LC-MS/MS analysis conditions
The specific conditions for LC-MS/MS used in the Examples herein are described in Table 4 below.
[Table 4]
Figure PCTKR2015002779-appb-I000005
Figure PCTKR2015002779-appb-I000006
2-2. The numerical treatment of values
The numerical values obtained from the result of LC-MS/MS were treated as described in Table 5 below. The numerical values are shown as N decimal points by rounding off the (N+1) decimal points.
[Table 5]
Figure PCTKR2015002779-appb-I000007
Example 3. The increase in sensitivity of esterified porphyrin in LC-MS/MS
3-1. Preparation of control and testing samples
(1) Preparation of intrinsic porphyrin-free blood
100 mL of whole blood was prepared. 10 g of activated carbon was added to the 100 mL of the whole blood, and mixed for at least 20 hours to remove porphyrin intrinsically contained in the blood. Then the activated carbon was removed from the blood by using a centrifuge.
(2) Preparation of control samples
CP, PP, HexaP, HeptaP, and UP, which were purchased from Frontier Scientific, were added to the blood prepared in (1), and solutions containing the above compounds at a concentration of 10 nmol/L respectively were prepared. Each of the prepared solutions was dropped on a blood filter paper schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare control samples (dried blood spot samples). Then 200 μL of 0.5 M formic acid in DW was added to the control sample to elute the compounds from the blood filter paper.
(3) Preparation of testing samples
Like control samples, CP, PP, HexaP, HeptaP, and UP, which were purchased from Frontier Scientific, were added to the blood prepared in (1), and solutions containing the above compounds at a concentration of 10 nmol/L respectively were prepared. Each of the prepared solutions was dropped on a blood filter paper schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare testing samples (dried blood spot samples).
Then 200 μL of 0.5 M formic acid in DW, and 500 μL of 9:1(v/v) solution of methanol (MeOH) and sulfuric acid (H2SO4) were added to the test samples, and then the samples were vortex-mixed for 50 min. Subsequently, to the resulting mixture, 1 mL of chloroform was added, and the mixture was vortex-mixed for 30 min, and allowed to stand for 30 min. After 30 min, layer separation was confirmed, centrifugation was performed at 13,000 rpm for 5 min, and 900 μL of the lower layer was completely dried at 40 ℃ using a nitrogen evaporator. To the dried samples, 100 μL of acetonitrile was added to re-dissolve the samples.
3-2. Esterification of porphyrin pretreated with methanol and sulfuric acid
When pre-treatment was carried out with a 9:1 (v/v) solution of methanol and sulfuric acid as in 3-1(3) through a separate assay, CP, PP, HexaP, HeptaP, UP and coproporphyrin I-15N4 were esterified via the reaction mechanism below to coproporphyrin tetramethyl ester (CPTE), pentacarboxylporphyrin pentamethyl ester (PPPE), hexacarboxylporphyrin hexamethyl ester (HexaPHE), heptacarboxylporphyrin heptamethyl ester (HeptaPHE), uroporphyrin octamethyl ester (UPOE), and coproporphyrin I-15N4 tetramethyl ester, respectively.
[Scheme]
Figure PCTKR2015002779-appb-I000008
The structures of CPTE, PPPE, HexaPHE, HeptaPHE, UPOE and coproporphyrin tetramethyl ester I-15N4 are illustrated in FIG. 2B.
3-3. Increased sensitivity of esterified porphyrin in LC-MS/MS
5 μL of each of the control samples prepared in 3-1 was injected into LC-MS/MS to be analyzed. The analysis results are shown in FIG. 3A.
Also, 5 μL of the upper layer of each of the testing samples re-dissolved in acetonitrile prepared in 3-1 was injected into LC-MS/MS to be analyzed. The analysis results are shown in FIG. 3B.
On the graphs of FIGS. 3A and 3B, the X axis represents time (min) and the Y axis represents the concentration (cps). Each peak corresponds to CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE from the right. Since the mass of each compound is increased upon esterification, the peaks of FIG. 3B were shifted rightward compared to FIG. 3A.
As illustrated in FIG. 3A, while CP showed an intensity of 9600 cps, the intensity of CPTE showed an intensity of 26,000 cps. As for PPPE, HexaPHE, HeptaPHE, and UPOE, such increases in signal intensities were also observed. Thus, it was understood that the pre-treatment of porphyrin with the mixed solution of methanol and sulfuric acid significantly increased the sensitivity in LC-MS/MS.
Example 4. Evaluation of the tests
Calibration standards and quality control (QC) samples were prepared as follows, and measured to check system suitability, specificity, carry-over, accuracy and precision, thereby evaluating whether the tests of Example 3 were appropriately performed.
4-1. Preparation of standard solution, QC solution, and internal standard solution
(1) Preparation of standard solution
0.75 mg of CP, 0.86 mg of PP, 0.91 mg of HexaP, 0.96 mg of HeptaP, and 1.00 mg of UP were weighed, dissolved in 100 mL of 6 M formic acid in DW, to prepare 10,000 nmol/L stock solutions for CP, PP, HexaP, HeptaP, and UP, respectively. The stock solutions were then frozen and stored.
Before the tests, each stock solution was subjected to serial dilution with 6 M formic acid as shown in Table 6 below, to prepare working solutions.
[Table 6]
Figure PCTKR2015002779-appb-I000009
(2) Preparation of QC solution
The working solutions for CP, PP, HexaP, HeptaP, and UP prepared in (1) were subjected to serial dilution with 6 M formic acid as shown in Table 7 below, to prepare QC solutions. The kinds of solutions taken in Table 7 below ((A), (E), (F)) are as represented in Table 6. In Table 7 below, QH denotes high-concentration QC solution (H: High), QM denotes medium-concentration QC solution (M: Medium), QL denotes low-concentration QC solution (L: Low), and LLOQ denotes a QC solution having a concentration corresponding to the lowest limit of quantification.
[Table 7]
Figure PCTKR2015002779-appb-I000010
(3) Preparation of internal standard solution
0.75 mg of coproporphyrin I-15N4 sodium bisulfate was weighed, and dissolved in 100 mL of 6 M formic acid, to prepare 10,000 nmol/L stock solution, which was then frozen and stored.
Before the tests, the stock solution was diluted as shown in Table 8 below, to prepare a working solution.
[Table 8]
Figure PCTKR2015002779-appb-I000011
4-2. Preparation of samples
(1) Preparation of a blank
100 mL of whole blood was prepared. 10 g of activated carbon was added to the 100 mL of the whole blood, and mixed for at least 20 hours to remove porphyrin intrinsically contained in the blood. Then the activated carbon was removed from the blood by using a centrifuge.
(2) Preparation of calibration standard
By adding the working solutions prepared in 4-1(1) as shown in Table 9 below to the blank prepared in (1), the calibration standards were prepared. The kinds of solutions taken in the following table are as represented in Table 6.
[Table 9]
Figure PCTKR2015002779-appb-I000012
Each of the prepared calibration solutions was dropped on the blood filter paper as schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight to prepare calibration standards (dried blood spot samples).
(3) Preparation of QC samples
QC solutions prepared in 4-1(2) were added to the blank prepared in (1) to prepare QC samples, as shown in Table 10 below. The kinds of solutions taken in the following Table 10 are as represented in Table 7.
[Table 10]
Figure PCTKR2015002779-appb-I000013
Each of the prepared QC solutions was dropped on the blood filter paper as schematically illustrated in FIG. 1, and dried at room temperature for at least 3 hours out of sunlight, to prepare QC samples (dried blood spot samples).
4-3. Pre-treatment of samples
To each of the calibration standards and the QC samples prepared in 4-2, 10 μL of coproporphyrin I-15N4 (50 nmol/L 6 M formic acid solution), the internal working solution prepared in 4-1(3), was added. Then, elution and pre-treatment were performed in the same manner as in 3-1(3) using 200 μL of a 0.5 M formic acid in DW and a 9:1 (v/v) solution of methanol (MeOH) and sulfuric acid (H2SO4), followed by drying and re-dissolution in acetonitrile.
4-4. Evaluation of system suitability, specificity, carry-over, linearity, and reproducibility
5 μL of the upper layer of the pretreated solution prepared in 4-3 was injected into LC-MS/MS to evaluate the following items.
(1) Evaluation of system suitability
The suitability of the present tester to ensure objectivity of the tests of Example 3 was evaluated as below. S1, among the calibration standards, and QL, among the QC samples, were injected into LC-MS/MS to ensure sensitivity. As an acceptance criterion for system suitability, the precision (CV, %) of the peak area ratio of each of CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE relative to the internal standard is 10% or less in principle, but was set to 15% in the present tests because five kinds of materials were analyzed simultaneously.
In all batches, the precision of CPTE was 2.23 ~ 9.30%, the precision of PPPE was 2.32 ~ 3.92%, the precision of HexaPHE was 3.17 ~ 8.94%, the precision of HeptaPHE was 3.14 ~ 8.67%, and the precision of UPOE was 2.92 ~ 8.97%, all of which satisfied the acceptance criterion. Specific numerical values for precision are shown in Table 11 below.
[Table 11]
Figure PCTKR2015002779-appb-I000014
(2) Evaluation of specificity
To determine whether materials which interfere the subject material to be analyzed and hinder the selective and accurate analysis of the subject material are present in the whole blood from which porphyrins are removed, the following evaluation was conducted.
Blanks prepared by using six kinds of whole blood from different origins, and the LLOQ sample, among the QC samples, were injected into LC-MS/MS, and whether materials that interferes CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE were present in the sample was determined. As an acceptance criterion for specificity, when there was no material that exceeds 20% of the peak area of the LLOQ sample for intra-day assays and exceeds 5% for the internal standard, the sample was determined to be free of any material interfering the subject material.
The results are shown in FIGS. 4 to 8 (for LC-MS/MS, there are slight differences in the positions of peaks from those in FIG. 3B. Such differences are within the range understood by one skilled in the art). FIGS. 4A to 8A show the results of mass analysis of the blanks, and FIGS. 4B to 8B show the results of analysis of each compound. In FIGS. 4A and 4B, it can be understood that the blank does not include any material showing a peak at 8.59 min, which corresponds to the peak of CPTE. In FIGS. 5A and 5B, it can be understood that the blank does not include any material showing a peak at 7.64 min, which corresponds to the peak of PPPE. In FIGS. 6A and 6B, it can be understood that the blank does not include any material showing a peak at 6.88 min, which corresponds to the peak of HexaPHE. In FIGS. 7A and 7B, it can be understood that the blank does not include any material showing a peak at 6.24 min, which corresponds to the peak of HeptaPHE. In FIGS. 8A and 8B, it can be understood that the blank does not include any material showing a peak at 5.77 min, which corresponds to the peak of UPOE. Accordingly, the porphyrin-free whole blood used in the tests did not include any material that interfere CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE.
(3) Evaluation of carry-over
When a series of samples were sequentially injected into LC-MS/MS, whether a sample was influenced by the previous sample was evaluated as below. At first, a blank was injected into LC-MS/MS, and 50 nmol/L ULOQ sample, which has the highest concentration among calibration standards, was injected into LC-MS/MS, and then the blank was injected into the instrument again. As such, whether there is a carry-over effect caused by the previously injected ULOQ sample was determined.
Consequently, no carry-over effect was observed.
(4) Evaluation of linearity
By using the accuracy and the correlation coefficient obtained from the calibration standards, whether the measured values could be linear with respect to the concentration of the analyte material within a predetermined range by the present testing method was evaluated.
Based on the results of analysis of the calibration standards for CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE using LC-MS/MS, a calibration curve was drawn, from which correlation coefficients were determined. An acceptance criterion is as follows: a deviation from the nominal concentration at LOQ has to be within 20% and a deviation from the nominal concentration at a concentration other than LOQ has to be within 15%, where r (correlation coefficient) has to be 0.98 or more.
The correlation coefficient (r) in the concentration range of 0.2 ~ 50 nmol/L is 0.9982 ~ 0.9999 for CPTE, 0.9815 ~ 0.9999 for PPPE, 0.9958 ~ 0.9999 for HexaPHE, 0.9938 ~ 0.9986 for HeptaPHE, and 0.9980 ~ 0.9990 for UPOE, all of which were equal to or higher than the acceptance criterion, and the accuracy results satisfied the acceptance criterion. The specific numerical values thereof are provided in Tables 12 to 16 below.
[Table 12]
Figure PCTKR2015002779-appb-I000015
[Table 13]
Figure PCTKR2015002779-appb-I000016
[Table 14]
Figure PCTKR2015002779-appb-I000017
[Table 15]
Figure PCTKR2015002779-appb-I000018
[Table 16]
Figure PCTKR2015002779-appb-I000019
(5) Evaluation of reproducibility (Accuracy and Precision)
Accuracy was expressed as a percentage (%) of the value obtained in such a manner that the numerical values obtained from injecting QC samples into LC-MS/MS were substituted into the calibration curves to give the mean concentration that was then divided by the concentration of the actual QC samples as known. Five tests were performed per day using QH, QM, QL, and LLOQ samples to calculate intra-day accuracy, and the tests were repeated for five days to calculate inter-day accuracy.
An acceptance criterion for accuracy is as follows: upon analysis of single component, the mean value has to be within ±15% of the nominal value except for LLOQ, and at LLOQ, the mean value does not exceed ±20% of the nominal value. However, since five kinds of materials were simultaneously analyzed in the present tests, the criterion was set within 20% of the nominal concentration in all concentrations.
The precision was expressed as a percentage (%) of the value obtained in such a manner that the numerical values resulting from injecting the QC samples in LC-MS/MS were divided by the mean value of the peak area ratios of CPTE, PPPE, HexaPHE, HeptaPHE, and UPOE and coproporphyrin tetramethyl ester I-15N4 as the internal standard. Five tests were performed per day using QH, QM, QL, and LLOQ samples to calculate intra-day precision, and the tests were repeated for five days to calculate inter-day precision.
An acceptance criterion for precision is as follows: upon analysis of single component, CV has to be within 15% at each concentration, and CV should not exceed 20% at LLOQ. However, since five kinds of materials were simultaneously analyzed in the present tests, CV was set within 20% in all concentrations.
The intra-day reproducibility results for accuracy and precision are as below.
The precision of CPTE was 4.35%, 4.76%, 9.20% and 6.48% at LLOQ, QL, QM and QH, respectively. The accuracy of CPTE was 114.63%, 109.10%, 88.06% and 106.51% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 17 below.
[Table 17]
Figure PCTKR2015002779-appb-I000020
The precision of PPPE was 4.14%, 7.58%, 5.93% and 5.39% at LLOQ, QL, QM and QH, respectively, and the accuracy of PPPE was 108.69%, 103.64%, 92.87% and 93.32% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 18 below.
[Table 18]
Figure PCTKR2015002779-appb-I000021
The precision of HexaPHE was 11.53%, 4.88%, 2.63% and 2.28% at LLOQ, QL, QM and QH, respectively, and the accuracy of HexaPHE was 94.13%, 86.06%, 86.74% and 85.20% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 19 below.
[Table 19]
Figure PCTKR2015002779-appb-I000022
The precision of HeptaPHE was 17.79%, 7.57%, 14.27% and 13.15% at LLOQ, QL, QM and QH, respectively, and the accuracy of HeptaPHE was 104.07%, 101.50%, 93.47% and 103.24% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 20 below.
[Table 20]
Figure PCTKR2015002779-appb-I000023
The precision of UPOE was 4.35%, 4.76%, 9.20% and 6.48% at LLOQ, QL, QM and QH, respectively, and the accuracy of UPOE was 114.63%, 109.10%, 88.06% and 106.51% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 21 below.
[Table 21]
Figure PCTKR2015002779-appb-I000024
The inter-day reproducibility results for accuracy and precision are shown below.
The precision of CPTE was 4.80%, 12.11%, 12.71% and 7.14% at LLOQ, QL, QM and QH, respectively. The accuracy of CPTE was 114.72%, 103.08%, 97.49% and 109.05% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 22 below.
[Table 22]
Figure PCTKR2015002779-appb-I000025
The precision of PPPE was 3.99%, 10.61%, 6.46% and 8.85% at LLOQ, QL, QM and QH, respectively. The accuracy of PPPE was 111.49%, 102.53%, 101.14% and 102.60% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 23 below.
[Table 23]
Figure PCTKR2015002779-appb-I000026
The precision of HexaPHE was 8.30%, 6.91%, 10.91% and 12.62% at LLOQ, QL, QM and QH, respectively. The accuracy of HexaPHE was 99.32%, 94.33%, 100.69% and 103.69% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 24 below.
[Table 24]
Figure PCTKR2015002779-appb-I000027
The precision of HeptaPHE was 9.11%, 12.37%, 13.37% and 10.55% at LLOQ, QL, QM and QH, respectively. The accuracy of HeptaPHE was 106.66%, 104.24%, 100.13% and 104.72% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 25 below.
[Table 25]
Figure PCTKR2015002779-appb-I000028
The precision of UPOE was 12.90%, 13.09%, 9.87% and 10.08% at LLOQ, QL, QM and QH, respectively. The accuracy of HeptaPHE was 105.35%, 105.42%, 101.94% and 101.99% at LLOQ, QL, QM and QH, respectively. The specific numerical values thereof are provided in Table 26 below.
[Table 26]
Figure PCTKR2015002779-appb-I000029

Claims (9)

  1. A method for detecting porphyrin in a biological sample, comprising:
    1) obtaining a biological sample from a subject;
    2) pretreating the biological sample with a mixed solution of methanol and strong acid; and
    3) analyzing the pretreated biological sample by using LC-MS/MS.
  2. The method of claim 1, wherein the pre-treatment in 2) is performed by bringing the biological sample into contact with a mixed solution of methanol and strong acid or mixing the biological sample with a mixed solution of methanol and strong acid.
  3. The method of claim 1, wherein the biological sample in 1) is blood.
  4. The method of claim 1, wherein the strong acid in 2) is sulfuric acid, hydrochloric acid, or nitric acid.
  5. The method of claim 1, wherein the mixed solution of methanol and strong acid in 2) comprises methanol and a strong acid mixed at a volume ratio of 9:1 ~ 8:2.
  6. The method of claim 1, wherein 1) comprises:
    a) obtaining a biological sample from a subject;
    b) dropping the biological sample on a blood filter paper; and
    c) eluting the biological sample from the blood filter paper.
  7. The method of claim 6, wherein eluting the biological sample in c) is performed by bringing formic acid into contact with the blood filter paper on which the biological sample was dropped.
  8. The method of claim 1, wherein the subject is a human.
  9. The method of claim 1, wherein the porphyrin is selected from the group consisting of coproporphyrin, pentacarboxylporphyrin, hexacarboxylporphyrin, heptacarboxylporphyrin, and uroporphyrin.
PCT/KR2015/002779 2014-03-21 2015-03-20 Method for detecting porphyrin in a biological sample by using lc-ms/ms WO2015142130A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020140033713A KR20150109995A (en) 2014-03-21 2014-03-21 A method for detecting porphyrin in a biospecimen using LC-MS/MS
KR10-2014-0033713 2014-03-21

Publications (1)

Publication Number Publication Date
WO2015142130A1 true WO2015142130A1 (en) 2015-09-24

Family

ID=54144989

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2015/002779 WO2015142130A1 (en) 2014-03-21 2015-03-20 Method for detecting porphyrin in a biological sample by using lc-ms/ms

Country Status (2)

Country Link
KR (1) KR20150109995A (en)
WO (1) WO2015142130A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017082915A1 (en) * 2015-11-12 2017-05-18 Halliburton Energy Services, Inc. Detection of porphyrins in subterranean formations

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102228072B1 (en) 2019-11-29 2021-03-15 대한민국 A method for detecting methanol in a biospecimen

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110207227A1 (en) * 2008-08-15 2011-08-25 Qiagen Gmbh Method for analysing a complex sample by mass spectrometry

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110207227A1 (en) * 2008-08-15 2011-08-25 Qiagen Gmbh Method for analysing a complex sample by mass spectrometry

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BENTON ET AL.: "Separation and fragmentation study of isocoproporphyrin derivatives by UHPLC-ESI-exact mass MS/MS and identification of a new isocoproporphyrin sulfonic acid metabolite", JOURNAL OF MASS SPECTROMETRY, vol. 49, no. Issue 1, January 2014 (2014-01-01), pages 80 - 85, XP055225442 *
DANTON ET AL.: "Porphyrin profiles in blood, urine and faeces by HPLC/electrospray ionization tandem mass spectrometry", BIOMEDICAL CHROMATOGRAPHY, vol. 20, no. 6-7, 2006, pages 612 - 621, XP055225439 *
HUR ET AL.: "Quantitative analysis of porphyrins in dried blood spots using liquid chromatography-tandem mass spectrometry with pre-column derivatization", BULLETIN OF THE KOREAN CHEMICAL SOCIETY, vol. 35, no. 10, 29 August 2014 (2014-08-29), pages 3103 - 3106, XP055225444 *
LI ET AL.: "Analysis of urinary porphyrins by high performance liquid chromatography-electrospray ionization mass spectometry", JOURNAL OF LIQUID CHROMATOGRAPHY & RELATED TECHNOLOGIES, vol. 34, no. 15, 2011, pages 1578 - 1593, XP055225440 *
WU ET AL.: "The prosthetic group of myeloperoxidase", FEBS LETTERS, vol. 60, no. 1, 1975, pages 141 - 144, XP025597805 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017082915A1 (en) * 2015-11-12 2017-05-18 Halliburton Energy Services, Inc. Detection of porphyrins in subterranean formations
GB2557801A (en) * 2015-11-12 2018-06-27 Haliburton Energy Services Inc Detection of porphyrins in subterranean formations

Also Published As

Publication number Publication date
KR20150109995A (en) 2015-10-02

Similar Documents

Publication Publication Date Title
HU180211B (en) Process for preparing a-guaiaconic acid and diagnostic composition containing thereof
WO2014104749A1 (en) Combustion pretreatment-isotope dilution mass spectrometry
WO2015142130A1 (en) Method for detecting porphyrin in a biological sample by using lc-ms/ms
WO2016144136A1 (en) Method for separating nucleic acids from fepe tissue
WO2019177345A1 (en) Method for ultrasensitive detection of multiple biomarkers
WO2019039888A1 (en) Real-time fluorescence imaging sensor for measuring glutathione in organelle and preparation method therefor
EA199800342A1 (en) A juvenile hormone and one of its agonists as a chemical ligand for the control of gene expression by means of transactivation mediated by a receptor
WO2017061818A1 (en) Mutant genes as diagnostic markers for amyotrophic lateral sclerosis and diagnostic method using same
KR20210133222A (en) Method for Quantification of Polysorbate in Samples Including LC-MS Using Internal Standards
WO2015020281A1 (en) Variable color two-photon fluorescent probe for detecting hydrogen sulfate, method for manufacturing same, and method for quantitatively imaging in vivo hydrogen sulfate using same
WO2017018735A1 (en) Biomarker for determining aging, determining obesity, and diagnosing cancer and diagnosing kit using same
Richards et al. Quantitative approach to the sequential degradation of proteins and peptides
Iwabuchi et al. Application of molecular-secondary-ion mass spectrometry for drug metabolism studies: I. Direct analysis of conjugates by thin-layer chromatography-secondary-ion mass spectrometry
WO2015167216A1 (en) Photocleavable mass tag material and use thereof
WO2020184905A2 (en) Method for quantitation of her2 in breast cancer sample by mass spectrometry and scoring of her2 state by using same
WO2023172102A1 (en) Bodipy derivative compound and fluorescent probe for detecting changes in ph and viscosity of oil comprising same
WO2020071868A1 (en) Dianhydride analysis method
WO2022270882A1 (en) Method for analyzing ages in blood of acute myocardial infarction patient and method for predicting prognosis of acute myocardial infarction by using same
WO2022075503A1 (en) Blocking agent, method for detecting analyte by using same, and kit for detecting analyte, comprising same
Podmore Routine determination of urinary pregnanediol using a gas chromatograph with automatic sample application
Matsuoka et al. Determination of three metabolites of a new angiotensin-converting enzyme inhibitor, imidapril, in plasma and urine by gas chromatography—mass spectrometry using multiple ion detection
WO2023200284A1 (en) Method for purifying and concentrating lipid-associated protein in biological sample to perform mass spectrometry of lipid-associated protein
WO2024136320A1 (en) Composition for single-cell proteomics analysis, sample treatment method using same, and method for unifying labeling of ultra-small amounts of peptides
WO2021137500A1 (en) Real-time fluorescence imaging sensor for measuring glutathione in endoplasmic reticulum and method using same
Rorem Ultra-violet fluorescence of quinine sulphate for detection of phosphate ester spots on paper

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15765495

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15765495

Country of ref document: EP

Kind code of ref document: A1