リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Safety comparisons among monoamine oxidase inhibitors against Parkinson’s disease using FDA adverse event reporting system」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Safety comparisons among monoamine oxidase inhibitors against Parkinson’s disease using FDA adverse event reporting system

Asano, Hiroto 大阪大学

2023.11.06

概要

Title

Safety comparisons among monoamine oxidase
inhibitors against Parkinson’s disease using
FDA adverse event reporting system

Author(s)

Asano, Hiroto; Tian, Yu Shi; Hatabu, Asuka et
al.

Citation

Scientific Reports. 2023, 13(1), p. 19272

Version Type VoR
URL
rights

https://hdl.handle.net/11094/93232
This article is licensed under a Creative
Commons Attribution 4.0 International License.

Note

Osaka University Knowledge Archive : OUKA
https://ir.library.osaka-u.ac.jp/
Osaka University

www.nature.com/scientificreports

OPEN

Safety comparisons
among monoamine oxidase
inhibitors against Parkinson’s
disease using FDA adverse event
reporting system
Hiroto Asano  , Yu‑Shi Tian 
Kenji Ikeda 

*

, Asuka Hatabu  , Tatsuya Takagi  , Mikiko Ueda  &

Monoamine oxidase B (MAO-B) inhibitors are used to control Parkinson’s disease (PD). Selegiline,
rasagiline, and safinamide are widely used as MAO-B inhibitors worldwide. Although these drugs
inhibit MAO-B, there are pharmacological and chemical differences, such as the inhibitory activity,
the non-dopaminergic properties in safinamide, and the amphetamine-like structure in selegiline.
MAO-B inhibitors may differ in adverse events (AEs). However, differences in actual practical
clinics are not fully investigated. A retrospective study was conducted using FAERS, the largest
database of spontaneous adverse events. AE signals for MAO-B inhibitors, including selegiline,
rasagiline, and safinamide, were detected using the reporting odds ratio method and compared.
Hypocomplementemia, hepatic cyst, hepatic function abnormal, liver disorder and cholangitis were
detected for selegiline as drug-specific signals. The amphetamine effect was not confirmed for any of
the three MAO-B inhibitors. The tyramine reaction was detected as an AE signal only for rasagiline.
Moreover, the REM sleep behavior disorder was not detected as an AE signal for safinamide,
suggesting that non-dopaminergic effects might be beneficial. Considering the differences in AEs for
MAO-B inhibitors will assist with the appropriate PD medication.
Parkinson’s disease (PD) is a highlighted neurodegenerative disorder and has become one of the most serious
health problems. Based on the Global Burden of Disease 2019, a significant 155.50% global increase in the
occurrence of PD has been estimated since 1990, and the prevalence reached over 8.5 million individuals. This
upward prevalence trend demonstrated consistency from 1990 to 2­ 0191. During the clinical progression of PD,
motor symptoms, including tremors, rigidity, akinesia, imbalance, and numerous non-motor complications
occur. These symptoms predominantly result from altered and abnormal neurotransmission related to deficits
of predominant biogenic amines. Dopamine is the most responsible neurotransmission amine, and its relative
amount in the midbrain nigrostriatal area of patients with PD decreases due to reduced or dropped out dopaminergic neurons. So far, the principal treatment of PD is to directly supplement dopamine, such as using the
combination of carbidopa and levodopa or dopamine agonists, or to achieve therapeutic maintenance of dopamine by reducing the key metabolism, such as targeting catechol-O-methyltransferase or monoamine oxidase
B (MAO-B) using i­ nhibitors2.
MAO is an enzyme with two isoforms of MAO-A and -B, which are high-expressing in the brain and gut.
Selective inhibition of MAO-B in the striatum can increase dopamine levels and show anti-PD action. However,
the non-selective inhibition in the brain and gut may cause serious hypertension when taking cheese together,
termed tyramine reaction. Therefore, inhibitory selectivity is important for MAO-B inhibitor development. To
date, three MAO-B inhibitors, selegiline, rasagiline, and safinamide, have been approved for clinical use. These
inhibitors can be used as monotherapy or add-on drugs to levodopa, improve the wear-off, delay the onset of
levodopa, and prolong the mean levodopa action ­duration3.
Selegiline, an irreversible inhibitor, was approved for PD by the FDA in 1989 and has been widely used as a
first-generation MAO-B inhibitor. The selectivity of MAO-B over MAO-A in the brain at low doses of selegiline
Graduate School of Pharmaceutical Sciences, Osaka University, 1‑6 Yamadaoka, Suita, Osaka 565‑0871, Japan.
*
email: yushi-tian@phs.osaka-u.ac.jp
Scientific Reports |

(2023) 13:19272

| https://doi.org/10.1038/s41598-023-44142-2

1
Vol.:(0123456789)

www.nature.com/scientificreports/
has been confirmed. However, it was also reported that when administered at high doses, the selegiline level
increased in plasma, indicating lower s­ electivities4. The chemical structure of selegiline contains an amphetamine
skeleton, and by first-pass metabolism, selegiline can be transformed into L-amphetamine and L-methamphetamine. Despite these aspects of selegiline, which raise the possibility of adverse effects, thus far, selegiline
has generally been considered a well-tolerated d
­ rug5, and its major adverse events are reported as headaches,
dizziness, insomnia, nausea, xerostomia, and c­ onstipation6. Furthermore, according to meta-analysis, there is
no difference in adverse event frequency between selegiline and placebo ­groups7. However, whether selegiline
metabolites can cause amphetamine-like adverse events, including cardiovascular and central neural system
adverse events, remains conversational. Some studies reported that cardiovascular adverse events in selegiline
users might be related to a­ mphetamine8–13, whereas other studies have suggested no association under ordinary
­use14, 15. Moreover, in high-dose use, whether tyramine reaction occurs remains unclear.
Rasagiline is another irreversible, selective MAO-B i­ nhibitor16, which FDA approved in 2006. As rasagiline
does not have an amphetamine skeleton and higher selectivities of MAO-B, rasagiline has been considered to have
fewer adverse events than selegiline. Sleep disturbances have been reported to be improved by switching from
selegiline to r­ asagiline17. Tyramine dietary restriction was removed based on the results from clinical s­ tudies18.
Conversely, the package insert of selegiline still warns of the tyramine ­response19. It is important to clarify the
frequency and risk of tyramine reactions using pharmacovigilance data.
Safinamide, an alpha-aminoamide derivative, has recently been on the market as the newest MAO-B inhibitor.
Unlike selegiline and rasagiline, safinamide is a reversible inhibitor, and its MAO-B selectivity is much higher
than the other two drugs, which may link to higher safety profiles. Furthermore, in addition to dopaminergic
properties, safinamide has non-dopaminergic properties, such as inhibiting voltage-gated sodium and calcium
channels and glutamate ­release20, 21. These characteristics may also distinguish safinamide from selegiline and
rasagiline from the view of drug safety.
These MAO-B inhibitors play important roles in the current PD treatments. However, these drugs differ in
selectivity, chemical structures, and dopaminergic properties, which may affect drug safety. Although clinical
studies and meta-analyses have focused on their safety, further pharmacovigilance analyses from real-world
patient data can provide deeper comprehension and address rare but important safety concerns. Therefore,
this study applied AE signal d
­ etection22 to the data from the FDA Adverse Event Reporting System (FAERS)23,
compared the AE signals associated with selegiline, rasagiline, and safinamide, and provided information on
drug safety, particularly on the considered differences.

Results

Baseline information

This study involved 3784, 10,584, and 1007 records reported in FAERS (2004Q1-2022Q2) for selegiline, rasagiline,
and safinamide, respectively (Table 1). The mean ages of these reports were 68, 70, and 71, and the female rates
Selegiline

Rasagiline

Safinamide

Age in years (mean ± SD (n))

67.8 ± 12.7 (2667)

70.0 ± 10.7 (7209)

71.4 ± 9.8 (713)

Number of combination ­drugsa (mean ± SD)

8.5 ± 6.2

7.5 ± 5.6

7.8 ± 4.9

Monotherapyb (n, %)

34 (0.8%)

883 (8.3%)

57 (5.6%)

Female (n, %)

1517 (42.6%)

3998 (40.5%)

415 (44.3%)

Area (n, %)
 Northern America

1523 (45.2%)

6082 (59.6%)

377 (40.4%)

 Western Europe

201 (6.0%)

1497 (14.7%)

251 (26.9%)

 Eastern Asia

991 (29.4%)

708 (6.9%)

29 (3.1%)

 Northern Europe

310 (9.2%)

799 (7.8%)

62 (6.6%)

 Southern Europe

231 (6.9%)

439 (4.3%)

197 (21.1%)

 Eastern Europe

23 (0.7%)

169 (1.7%)

0 (0.0%)

 South America

42 (1.2%)

94 (0.9%)

16 (1.7%)

 Western Asia

4 (0.1%)

130 (1.3%)

0 (0.0%)

 Central America

8 (0.2%)

110 (1.1%)

0 (0.0%)

 Australia and New Zealand

22 (0.7%)

90 (0.9%)

1 (0.1%)

 Southern Asia

5 (0.1%)

40 (0.4%)

1 (0.1%)

 South-eastern Asia

11 (0.3%)

19 (0.2%)

0 (0.0%)

 Southern Africa

0 (0.0%)

19 (0.2%)

0 (0.0%)

 Caribbean

0 (0.0%)

5 (0.0%)

0 (0.0%)

 Northern Africa

0 (0.0%)

1 (0.0%)

0 (0.0%)

 Micronesia

0 (0.0%)

1 (0.0%)

0 (0.0%)

Table 1.  Baseline information. In this table, unreported data were excluded from the calculation of both the
count (n) and the percentage (%). a The number of combination drugs was counted when other drugs were
reported for the same primary ID. b Monotherapy is counted when no other drugs are reported for one primary
ID.

Scientific Reports |
Vol:.(1234567890)

(2023) 13:19272 |

https://doi.org/10.1038/s41598-023-44142-2

2

www.nature.com/scientificreports/
were 40–44%, suggesting no obvious differences among MAO-B inhibitors. For each drug, males were more
frequently reported than females. The highest reporting proportion was confirmed in Northern America for all
three MAO-B inhibitors. However, differences were observed in some reported areas. For example, selegiline
has a higher reporting proportion than rasagiline and safinamide in Eastern Asia, especially in Japan (950 cases
of 991 cases). ...

この論文で使われている画像

参考文献

1. Ou, Z. et al. Global trends in the incidence, prevalence, and years lived with disability of Parkinson’s disease in 204 countries/

territories from 1990 to 2019. Front. Public Health 9, 776847 (2021).

2. Armstrong, M. J. & Okun, M. S. Diagnosis and treatment of Parkinson disease: A review. JAMA 323, 548–560 (2020).

Scientific Reports |

Vol:.(1234567890)

(2023) 13:19272 |

https://doi.org/10.1038/s41598-023-44142-2

www.nature.com/scientificreports/

3. Finberg, J. P. M. & Rabey, J. M. Inhibitors of MAO-A and MAO-B in psychiatry and neurology. Front. Pharmacol. 7, 340 (2016).

4. Fowler, J. S. et al. Evidence that formulations of the selective MAO-B inhibitor, selegiline, which bypass first-pass metabolism, also

inhibit MAO-A in the human brain. Neuropsychopharmacology 40, 650–657 (2015).

5. Tábi, T., Vécsei, L., Youdim, M. B., Riederer, P. & Szökő, É. Selegiline: A molecule with innovative potential. J. Neural Transm. 127,

831–842 (2020).

6. Moore, J., Saadabadi, J. & Selegiline, A. StatPearls (StatPearls Publishing, 2022).

7. Jiang, D.-Q., Li, M.-X., Jiang, L.-L., Chen, X.-B. & Zhou, X.-W. Comparison of selegiline and levodopa combination therapy versus

levodopa monotherapy in the treatment of Parkinson’s disease: A meta-analysis. Aging Clin. Exp. Res. 32, 769–779 (2020).

8. Fozard, J. R., Zreika, M., Robin, M. & Palfreyman, M. G. The functional consequences of inhibition of monoamine oxidase type

B: Comparison of the pharmacological properties of L-deprenyl and MDL 72145. Naunyn. Schmiedebergs Arch. Pharmacol. 331,

186–193 (1985).

9. Okuda, C., Segal, D. S. & Kuczenski, R. Deprenyl alters behavior and caudate dopamine through an amphetamine-like action.

Pharmacol. Biochem. Behav. 43, 1075–1080 (1992).

10. Engberg, G., Elebring, T. & Nissbrandt, H. Deprenyl (selegiline), a selective MAO-B inhibitor with active metabolites; effects on

locomotor activity, dopaminergic neurotransmission and firing rate of nigral dopamine neurons. J. Pharmacol. Exp. Ther. 259,

841–847 (1991).

11. Churchyard, A., Mathias, C. J., Boonkongchuen, P. & Lees, A. J. Autonomic effects of selegiline: Possible cardiovascular toxicity

in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 63, 228–234 (1997).

12. Stryjer, R., Klein, C., Treves, T. A. & Rabey, J. M. The effects of acute loading with levodopa and levodopa with selegiline on blood

pressure and plasma norepinephrine levels in chronic Parkinson’s disease patients. Acta Neurol. Scand. 111, 89–94 (2005).

13. Montastruc, J.-L. et al. Adverse drug reactions to selegiline[colon] a review of the French pharmacovigilance database. Clin.

Neuropharmacol. 23, 271–275 (2000).

14. Yasar, S. et al. A comparison of drug-seeking behavior maintained by D-amphetamine, L-deprenyl (selegiline), and D-deprenyl

under a second-order schedule in squirrel monkeys. Psychopharmacology (Berl) 183, 413–421 (2006).

15. Yasar, S. et al. Metabolic transformation plays a primary role in the psychostimulant-like discriminative-stimulus effects of selegiline

[(R)-(-)-deprenyl]. J. Pharmacol. Exp. Ther. 317, 387–394 (2006).

16. Youdim, M. B. H., Gross, A. & Finberg, J. P. M. Rasagiline [N-propargyl-1R(+)-aminoindan], a selective and potent inhibitor of

mitochondrial monoamine oxidase B. Br. J. Pharmacol. 132, 500–506 (2001).

17. Müller, T., Hoffmann, J. A., Dimpfel, W. & Oehlwein, C. Switch from selegiline to rasagiline is beneficial in patients with Parkinson’s

disease. J. Neural. Transm. 120, 761–765 (2013).

18. Goren, T., Adar, L., Sasson, N. & Weiss, Y. M. Clinical pharmacology tyramine challenge study to determine the selectivity of the

monoamine oxidase type B (MAO-B) inhibitor rasagiline. J. Clin. Pharmacol. 50, 1420–1428 (2010).

19. DailyMed-AZILECT- rasagiline mesylate tablet. https://​daily​med.​nlm.​nih.​gov/​daily​med/​lookup.​cfm?​setid=​a40d0​e73-​3f9f-​4b01-​

979d-​402c9​cdaeb​60

20. Kulisevsky, J. Emerging role of safinamide in Parkinson’s disease therapy. Eur. Neurol. Rev. 9, 108 (2015).

21. de Souza, R. M. & Schapira, A. Safinamide for the treatment of Parkinson’s disease. Expert Opin. Pharmacother. 18, 937–943 (2017).

22. Rothman, K. J., Lanes, S. & Sacks, S. T. The reporting odds ratio and its advantages over the proportional reporting ratio. Pharmacoepidemiol. Drug Saf. 13, 519–523 (2004).

23. FAERS Quarterly Data Extract Files. https://​fis.​fda.​gov/​exten​sions/​FPD-​QDE-​FAERS/​FPD-​QDE-​FAERS.​html

24. DailyMed-AMPHETAMINE SULFATE-amphetamine tablet. https://​daily​med.​nlm.​nih.​gov/​daily​med/​drugI​nfo.​cfm?​setid=​828c8​

4a6-​a317-​4e4d-​9521-​0c386​5ff73​2f

25. Van Den Eeden, S. K. et al. Incidence of Parkinson’s disease: Variation by age, gender, and race/ethnicity. Am. J. Epidemiol. 157,

1015–1022 (2003).

26. Mabuchi, T., Hosomi, K., Yokoyama, S. & Takada, M. Polypharmacy in three different spontaneous adverse drug event databases.

Int. J. Clin. Pharmacol. Ther. 58, 601–607 (2020).

27. McLean, G., Hindle, J. V., Guthrie, B. & Mercer, S. W. Co-morbidity and polypharmacy in Parkinson’s disease: Insights from a large

Scottish primary care database. BMC Neurol. 17, 126 (2017).

28. Kyritsi, K. et al. Modulation of the tryptophan hydroxylase 1/monoamine oxidase-A/5-hydroxytryptamine/5-hydroxytryptamine

receptor 2A/2B/2C axis regulates biliary proliferation and liver fibrosis during cholestasis. Hepatology 71, 990–1008 (2020).

29. Hidestrand, M. et al. CYP2B6 and CYP2C19 as the major enzymes responsible for the metabolism of selegiline, a drug used in the

treatment of Parkinson’s disease, as revealed from experiments with recombinant enzymes. Drug Metab. Dispos. 29, 1480–1484

(2001).

30. Jost, W. H. A critical appraisal of MAO-B inhibitors in the treatment of Parkinson’s disease. J. Neural. Transm. (Vienna) 129,

723–736 (2022).

31. Kubota, T., Chiba, K. & Iga, T. Frequency distribution of CYP2C19, CYP2D6, and CYP2C9 mutant-alleles in several different

populations. Drug Metab. Pharmacokinet. 16, 69–74 (2001).

32. Mizuno, Y. et al. A randomized double-blind placebo-controlled phase III trial of selegiline monotherapy for early Parkinson

disease. Clin. Neuropharmacol. 40, 201–207 (2017).

33. Hazell, L. & Shakir, S. A. W. Under-reporting of adverse drug reactions : A systematic review. Drug Saf. 29, 385–396 (2006).

34. Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: The TEMPO study. Arch. Neurol. 59, 1937–1943

(2002).

35. Stern, M. B. et al. Double-blind, randomized, controlled trial of rasagiline as monotherapy in early Parkinson’s disease patients.

Mov. Disord. 19, 916–923 (2004).

36. Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with parkinson disease

and motor fluctuations: The PRESTO study. Arch. Neurol. 62, 241–248 (2005).

37. Rascol, O. et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, lasting

effect in adjunct therapy with rasagiline given once daily, study): A randomised, double-blind, parallel-group trial. Lancet 365,

947–954 (2005).

38. Parkinson Study Group. a controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch. Neurol. 61,

561–566 (2004).

39. Olanow, C. W. et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N. Engl. J. Med. 361, 1268–1278 (2009).

40. Zhang, X., Guarin, D., Mohammadzadehhonarvar, N., Chen, X. & Gao, X. Parkinson’s disease and cancer: A systematic review

and meta-analysis of over 17 million participants. BMJ Open 11, e046329 (2021).

41. Johannes, C. B. et al. The risk of melanoma with rasagiline compared with other antiparkinsonian medications: A retrospective

cohort study in the United States medicare database. Pharmacoepidemiol. Drug Saf. 31, 643–651 (2022).

42. Wasan, H., Singh, D. & Kh, R. Safinamide in neurological disorders and beyond: Evidence from preclinical and clinical studies.

Brain Res. Bull. 168, 165–177 (2021).

43. de Marcaida, J. A. et al. Effects of tyramine administration in Parkinson’s disease patients treated with selective MAO-B inhibitor

rasagiline. Mov. Disord. 21, 1716–1721 (2006).

44. Plastino, M. et al. Effects of safinamide on REM sleep behavior disorder in Parkinson disease: A randomized, longitudinal, crossover pilot study. J. Clin. Neurosci. 91, 306–312 (2021).

Scientific Reports |

(2023) 13:19272 |

https://doi.org/10.1038/s41598-023-44142-2

Vol.:(0123456789)

www.nature.com/scientificreports/

45. Iranzo, A., Santamaria, J. & Tolosa, E. The clinical and pathophysiological relevance of REM sleep behavior disorder in neurodegenerative diseases. Sleep Med. Rev. 13, 385–401 (2009).

46. Luppi, P.-H., Clément, O., Valencia Garcia, S., Brischoux, F. & Fort, P. New aspects in the pathophysiology of rapid eye movement

sleep behavior disorder: The potential role of glutamate, gamma-aminobutyric acid, and glycine. Sleep Med. 14, 714–718 (2013).

47. Ronconi, G. et al. Prescription pattern of monoamine oxidase B inhibitors combined with levodopa: A retrospective observational

analysis of italian healthcare administrative databases. Drugs Real World Outcomes 9, 391–401 (2022).

48. Berman, S. M., Kuczenski, R., McCracken, J. T. & London, E. D. Potential adverse effects of amphetamine treatment on brain and

behavior: A review. Mol. Psychiatry 14, 123–142 (2009).

49. Sinha, A., Lewis, O., Kumar, R., Yeruva, S. L. H. & Curry, B. H. Adult ADHD medications and their cardiovascular implications.

Case Rep. Cardiol. 2016, 2343691 (2016).

50. Derlet, R. W., Rice, P., Horowitz, B. Z. & Lord, R. V. Amphetamine toxicity: Experience with 127 cases. J. Emerg. Med. 7, 157–161

(1989).

51. Kalant, H. & Kalant, O. J. Death in amphetamine users: Causes and rates. Can. Med. Assoc. J. 112, 299–304 (1975).

52. Coyle, J. T. & Snyder, S. H. Antiparkinsonian drugs: Inhibition of dopamine uptake in the corpus striatum as a possible mechanism

of action. Science 166, 899–901 (1969).

53. Centre, U. M. About VigiBase. https://​who-​umc.​org/​vigib​ase/

54. VigiAccess. https://​www.​vigia​ccess.​org/

55. Vogel, U. et al. Investigating overlap in signals from EVDAS, FAERS, and VigiBase®. Drug Saf. 43, 351–362 (2020).

56. Poleksic, A. & Xie, L. Database of adverse events associated with drugs and drug combinations. Sci. Rep. 9, 20025 (2019).

57. Yao, X. et al. Mining and visualizing high-order directional drug interaction effects using the FAERS database. BMC Med. Inform.

Decis. Mak. 20, 50 (2020).

58. Research, C. for D. E. and. Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment. https://​www.​fda.​gov/​

regul​atory-​infor​mation/​search-​fda-​guida​nce-​docum​ents/​good-​pharm​acovi​gilan​ce-​pract​ices-​and-​pharm​acoep​idemi​ologic-​asses​

sment (2020).

59. ISO-ISO 3166—Country Codes. ISO https://​www.​iso.​org/​iso-​3166-​count​r y-​codes.​html

60. KEGG DRUG Database. https://​www.​genome.​jp/​kegg/​drug/

61. Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000).

62. Kanehisa, M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 28, 1947–1951 (2019).

63. Kanehisa, M., Furumichi, M., Sato, Y., Ishiguro-Watanabe, M. & Tanabe, M. KEGG: Integrating viruses and cellular organisms.

Nucleic Acids Res. 49, D545–D551 (2021).

64. MedDRA. https://​www.​meddra.​org/

65. UNSD—Methodology. https://​unsta​ts.​un.​org/​unsd/​metho​dology/​m49/

66. Fukazawa, C., Hinomura, Y., Kaneko, M. & Narukawa, M. Significance of data mining in routine signal detection: Analysis based

on the safety signals identified by the FDA. Pharmacoepidemiol. Drug Saf. 27, 1402–1408 (2018).

67. Batagelj, V. & Bren, M. Comparing resemblance measures. J. Classif. 12, 73–90 (1995).

Author contributions

H.A., Y.S.T. and T.T. contributed to the conception and design of the study. H.A. organized the database and

performed the statistical analysis. H.A. and Y.S.T. wrote the first draft of the manuscript. A.H., T.T., M.U., and

K.I. provided suggestions to improve the analysis and revised the manuscript.

Funding

This study received funding from FP Pharmaceutical Corporation.

Competing interests The authors declare no competing interests.

Additional information

Supplementary Information The online version contains supplementary material available at https://​doi.​org/​

10.​1038/​s41598-​023-​44142-2.

Correspondence and requests for materials should be addressed to Y.-S.T.

Reprints and permissions information is available at www.nature.com/reprints.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and

institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International

License, which permits use, sharing, adaptation, distribution and reproduction in any medium or

format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the

Creative Commons licence, and indicate if changes were made. The images or other third party material in this

article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the

material. If material is not included in the article’s Creative Commons licence and your intended use is not

permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.

© The Author(s) 2023

Scientific Reports |

Vol:.(1234567890)

(2023) 13:19272 |

https://doi.org/10.1038/s41598-023-44142-2

10

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る