Aleksandr V. Sidorov , Elena G. Lileeva
Abstract
Self-monitoring of blood glucose has been considered to be a cornerstone of adequate diabetes management for decades. The structured self-monitoring leads to better compensation of carbohydrate metabolism disorders, decreased risk of hypoglycemia, and quality of life improvement in patients with DM. The technologies, as well as the glucose meter convenience and accuracy have improved considerably since the moment these were introduced several decades ago. Modern glucose meters are small and easy to use; these require small amounts of capillary blood and provide measurement results within seconds. Furthermore, glucose meters are more and more often equipped with such features, as connectivity with other devices and/or electronic diaries and instruments for diabetes management. Despite the fact that the measurement accuracy can be close to or equal to the quality level ensured by laboratory biochemical analyzers, some glucose meters continue to fall short of the internationally accepted accuracy standards, such as ISO-15197, the standard of the International Organization for Standardization. Numerous reports from the number of countries, such as Russia, China, India, and Brazil, that are based on the routine practice data suggest that in addition to the problems with the glucose meter accuracy, there are other obstacles to overcome to optimize the use of blood glucose monitoring systems. Nevertheless, the adequate use of blood glucose self-monitoring data is very important for treatment of patients with type 2 diabetes mellitus.
Keywords: self-monitoring of blood glucose, type 2 diabetes mellitus, blood glucose monitoring systems, accuracy, diabetes management, glucose meters.
Keywords: self-monitoring of blood glucose, type 2 diabetes mellitus, blood glucose monitoring systems, accuracy, diabetes management, glucose meters.
About the Author
Aleksandr V. Sidorov 1 , Elena G. Lileeva 21 Yaroslavl State Medical University, Yaroslavl, Russia; Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
2 Yaroslavl State Medical University, Yaroslavl, Russia
References
1. Чеботарева Т.А. Современные подходы к терапии острого кашля у
детей. Практика педиатра. 2020;(1):8-13.
Chebotareva T.A. Modern approaches to the treatment of acute cough
in children. Pediatrician’s practice. 2020;(1):8-13 (in Russian).
2. Pourova J, Dias P, Pour M et al. Proposed mechanisms of action of her-
bal drugs and their biologically active constituents in the treatment of
coughs: an overview. Peer J 2023;(11):e16096. DOI:
10.7717/peerj.16096
3. Thakur AK, Taj P. Pharmacological Perspective of Glycyrrhiza glabra
Linn: a Mini-Review. J Anal Pharm Res 2017;5(5):11-2. DOI:
10.15406/japlr.2017.05.00156
4. Hasan MK, Ara I, Mondal MSA, Kabir Y. Phytochemistry, pharmacolo-
gical activity, and potential health benefits of Glycyrrhiza glabra. He-
liyon 2021;(7):e07240. DOI: 10.1016/j.heliyon.2021.e07240
5. Yu X, Bao Y, Meng X et al. Multi-pathway integrated adjustment mec-
hanism of licorice flavonoids presenting anti-inflammatory activity.
Oncol Lett 2019;(18):4956-63. DOI: 10.3892/ol.2019.10793
6. Wang H-L, Li Y-X, Niu Y-T et al. Observing Anti-inflammatory and
anti-nociceptive activities of glycyrrhizin through regulating COX-2
and pro-inflammatory cytokines expressions in Mice. Inflammation
2015;(38):2269-78. DOI: 10.1007/s10753-015-0212-3
7. Dang LL, Jin YJ, Yuan Y et al. Licorice: comprehensive review of its che-
mical composition, pharmacodynamics, and medicinal value. Acupunct
Herb Med 2024;4(1):136-50. DOI: 10.1097/HM9.0000000000000103
8. El-Saber Batiha G, Magdy Beshbishy A, El-Mleeh A et al. Traditional
Uses, Bioactive Chemical Constituents, and Pharmacological and Toxi-
cological Activities of Glycyrrhiza glabra L. (Fabaceae). Biomolecules
2020;10(3):352. DOI: 10.3390/biom10030352
9. Raphael TJ, Kuttan G. Effect of naturally occurring triterpenoids gly-
cyrrhizic acid, ursolic acid, oleanolic acid and nomilin on the immune
system. Phytomedicine 2003;10(6-7):483-9. DOI: 10.1078/0944
71103322331421
10. Ma C, Ma Z, Liao XL et al. Immunoregulatory effects of glycyrrhizic acid
exerts anti-asthmatic effects via modulation of Th1/Th2 cytokines and
enhancement of CD4(+)CD25(+)Foxp3+ regulatory T cells in ovalbu-
min-sensitized mice. J Ethnopharmacol 2013; 148(3):755-62. DOI:
10.1016/j.jep.2013.04.021
11. Irani M, Sarmadi M, Bernard F et al. Leaves antimicrobial activity of
Glycyrrhiza glabra L. Iran J Pharm Res 2010;(9):425-8.
12. Gupta VK, Fatima A, Faridi U et al. Antimicrobial potential of Glycyrr-
hiza glabra roots. J Ethnopharmacol 2008;(116):377-80. DOI:
10.1016/j.jep.2007.11.037
13. Cinatl J, Morgenstern B, Bauer G et al. Glycyrrhizin, an active compo-
nent of liquorice roots, and replication of SARS-associated coronavirus.
Lancet 2003;361(9374):2045-6. DOI: 10.1016/s0140-6736(03)13615-x
14. Michaelis M, Geiler J, Naczk P et al. Glycyrrhizin inhibits highly patho-
genic H5N1 influenza A virus-induced pro-inflammatory cytokine and
chemokine expression in human macrophages. Med Microbiol Immu-
nol 2010;199(4):291-7. DOI: 10.1007/s00430-010-0155-0
15. Räikkönen K, Martikainen S, Pesonen A et al. Maternal Licorice Con-
sumption During Pregnancy and Pubertal, Cognitive, and Psychiatric
Outcomes in Children. Am J Epidemiol 2017;185(5):317-28. DOI:
10.1093/aje/kww172
16. Shin H, Chung M, Rose DZ. Licorice root associated with intracranial
hemorrhagic stroke and cerebral microbleeds. Neurohospitalist
2019;9(3):169-71. DOI: 10.1177/1941874418805332
17. Ceccuzzi G, Rapino A, Perna B et al. Liquorice toxicity: a comprehensive
narrative review. Nutrients 2023;15(18):3866. DOI: 10.3390/
nu15183866
18. Pastorino G, Cornara L, Soares S et al. Liquorice (Glycyrrhiza glabra):
A phytochemical and pharmacological review. Phytother Res
2018;32(12):2323-39. DOI: 10.1002/ptr.6178
19. Ding Y, Brand E, Wang W, Zhao Z. Licorice: Resources, applications in
ancient and modern times. J Ethnopharmacol 2022;298:115594. DOI:
10.1016/j.jep.2022.115594
20. Omar HR, Komarova I, El-Ghonemi M et al. Licorice abuse: time to
send a warning message. Ther Adv Endocrinol Metab 2012;3(4):125-
38. DOI: 10.1177/2042018812454322
21. Shamsuddin T, Alam MS, Junaid M et al. Adhatoda vasica (Nees.): A
Review on its Botany, Traditional uses, Phytochemistry, Pharmacologi-
cal Activities and Toxicity. Mini Rev Med Chem 2021;21(14):1925-64.
DOI: 10.2174/1389557521666210226152238
22. Singh SK, Patel JR, Dangi A et al. A complete over review on Adhatoda
vasica a traditional medicinal plants. J Med Plants Stud 2017;5(1):
175-80.
23. Kapgate S, Patil A. Adhatoda vasica: a critical review. Int J Green Phar-
macy 2017;11(4):654-62. DOI: 10.22377/ijgp.v11i04.1341
24. Kumar N. Pharmaceutical attributes of Vasa (Adhatoda Vasica Linn.) –
a review. World J Pharmaceutical Res 2016;5(4):437-55. DOI:
10.20959/wjpr20164-5760
25. Jahan Y, Siddiqui HH. Study of Antitussive Potential of Glycyrrhiza
Glabra and Adhatoda Vasica using a cough model induced by Sulphur
Dioxide gas in mice. IJPSR 2012;3(6):1668-74. DOI: 10.13040/
IJPSR.0975-8232.3(6).1668-74
26. Nosalova G, Fleskova D, Jurecek L et al. Herbal polysaccharides and co-
ugh reflex. Respir Physiol Neurobiol 2013;(187):47-51. DOI:
10.1016/j.resp.2013.03.015
27. Ahmad S, Gul S, Gul H, Bangash M. Dual Inhibitory Activities of Adha-
toda Vasica against Cyclooxygenase and Lipoxygenase. JEHSR
2013;(1).14. DOI: 10.29052/IJEHSR.v1.i1.2013.14-17
28. Bajpai VK, Agrawal P, Bang BH, Park YH. Phytochemical analysis, an-
tioxidant and antilipid peroxidation effects of a medicinal plant, Adha-
toda vasica. Front Life Sci 2015;8(3):305-12. DOI: 10.1080/
21553769.2014.1002943
29. Duraipandiyan V, Al-Dhabi NA, Balachandran C et al. Antimicrobial,
antioxidant, and cytotoxic properties of vasicine acetate synthesized
from vasicine isolated from Adhatoda vasica L. Biomed Res Int
2015;2015:727304. DOI: 10.1155/2015/727304
30. Srmivasarao D, Jayarraj IA, Jayraaj R, Prabha ML. A study on Antioxi-
dant and Anti-inflammatory activity of Vasicine against lung damage in
rats. Indian J Allergy Asthma Immunol 2006;20(1):1-7.
31. Alagarsamy K, Shanthi V, Nagasathaya A. Preliminary phytochemical
and antibacterial screening of crude extract of the leaf of Adhatoda va-
sica L. Int J Green Pharmacy 2009;(3). DOI: 10.4103/0973-
8258.49381
32. Zulqarnain RA, Ahmad K, Ullah F et al. In vitro antibacterial activity of
selected medicinal plants from lower Himalayas. Pak J Pharm Sci
2015;28(2):581-7.
33. Zaki AA, Shaaban MI, Hashish NE et al. Assessment of anti-quorum
sensing activity for some ornamental and medicinal plants native to
Egypt. Sci Pharm 2013;81(1):251-8. DOI: 10.3797/scipharm.1204-26
34. Claeson UP, Malmfors T, Wikman G, Bruhn JG. Adhatoda vasica: a cri-
tical review of ethnopharmacological and toxicological data. J Ethnop-
harmacol 2000;72(1-2):1-20. DOI: 10.1016/s0378-8741(00)00225-7
35. Shinde A, Rathi B, Chunchuwar M. Pharmaceutical study of Vasa (Ad-
hatoda vasica Linn.). With special reference to its different dosage
forms. GSC Biol Pharmaceut Sci 2023;(23):9-18. DOI:
10.30574/gscbps.2023.23.3.0197
36. Nath D, Sethi N, Singh RK, Jain AK. Commonly used Indian abortifaci-
ent plants with special reference to their teratologic effects in rats.
J Ethnopharmacol 1992;36(2):147-54. DOI: 10.1016/0378-
8741(92)90015-j
37. Вальчихина О.Ю., Демина Н.Б., Надер А. Корневище имбиря как
перспективное растительное сырье для создания лекарственных
средств. Разработка и регистрация лекарственных средств.
2015;4(13):82-90.
Valchikhina O.Yu., Demina N.B., Nader A. Ginger rhizome as a promi-
sing plant raw material for the creation of medicines. Development and
registration of medicines. 2015;4(13):82-90 (in Russian).
38. Ali BH, Blunden G, Tanira MO, Nemmar A. Some phytochemical, phar-
macological and toxicological properties of ginger (Zingiber officinale
Roscoe): a review of recent research. Food Chem Toxicol
2008;46(2):409-20. DOI: 10.1016/j.fct.2007.09.085
39. Grzanna R, Lindmark L, Frondoza CG. Ginger – An Herbal Medicinal
Product with Broad Anti-Inflammatory Actions. J Med Food
2005;8(2):125-32. DOI: 10.1089/jmf.2005.8.125
40. Dugasani S, Pichika MR, Nadarajah VD et al. Comparative antioxidant
and anti-inflammatory effects of [6]-gingerol, [8]-gingerol, [10]-ginge-
rol and [6]-shogaol. J Ethnopharmacol 2010;127(2):515-20. DOI:
10.1016/j.jep.2009.10.004
41. Hasan H, Raauf A, Abd Razik B. Chemical Composition and Antimicro-
bial Activity of the Crude Extracts Isolated from Zingiber Officinale by
Different Solvents. Pharmaceutica Analytica Acta 2012;3(9). DOI:
10.4172/2153-2435.1000184
42. Kumar G, Karthik L, Rao KB. A review on pharmacological and phytoc-
hemical properties of Zingiber officinale Roscoe (Zingiberaceae).
J Pharm Res 2011;(4):2963-6.
43. Sharifi-Rad M, Varoni EM, Salehi B et al. Plants of the Genus Zingiber
as a Source of Bioactive Phytochemicals: From Tradition to Pharmacy.
Molecules 2017;22(12):2145. DOI: 10.3390/molecules22122145
44. Zick SM, Djuric Z, Ruffin MT et al. Pharmacokinetics of 6-gingerol, 8-
gingerol, 10-gingerol, and 6-shogaol and conjugate metabolites in he-
althy human subjects. Cancer Epidemiol Biomarkers Prev
2008;17(8):1930-6. DOI: 10.1158/1055-9965
45. Chrubasik S, Pittler MH, Roufogalis BD. Zingiberis rhizoma: a compre-
hensive review on the ginger effect and efficacy profiles. Phytomedicine
2005;12(9):684-701. DOI: 10.1016/j.phymed.2004.07.009
46. Husain I, Dale OR, Idrisi M. Evaluation of the Herb-Drug Interaction
(HDI) Potential of Zingiber officinale and Its Major Phytoconstituents.
J Agric Food Chem 2023;71(19):7521-34. DOI: 10.1021/ acs.jafc.2c07912
47. Rathore S, Raj Y, Debnath P et al. Ethnopharmacology, phytoche-
mistry, agrotechnology, and conservation of Inula racemosa Hook f. –
A critically endangered medicinal plant of the western Himalaya. J Eth-
nopharmacol 2022;283:114613. DOI: 10.1016/j.jep.2021.114613
48. Seca AM, Grigore A, Pinto DC, Silva A. The genus Inula and their me-
tabolites: from ethnopharmacological to medicinal uses. J Ethnophar-
macol 2014;154(2):286-310. DOI: 10.1016/j.jep.2014.04.010
49. Vadnere GP, Gaud RS, Singhai AK, Somani RS. Effect of Inula race-
mosa root extract on various aspects of asthma. Pharmacologyonline
2009;(2):84-94.
50. Sekhar AV, Gandhi DN, Rao MN, Rawal UD. An experimental and cli-
nical тevaluation of anti-asthmatic potentialities of devadaru compo-
und (dc). Indian J Physiol Pharmacol 2003;47(1):101-7.
51. Firdous Q, Bhat M, Masoodi M. Ethnopharmacology, Phytochemistry
and Biological activity of Inula racemosa Hook: A Review. Int J Res Ay-
urveda Pharmacy 2018;(9). DOI: 10.7897/2277-4343.09120
52. Wangchuk P, Jamtsho T. Inula racemosa Hook. f. Pushkarmool: Its
Ethnobotanical Uses, Phytochemicals, and Pharmacological Activities.
In: A. Sharma, G.A. Nayik (eds). Immunity Boosting Medicinal Plants
of the Western Himalayas. Springer, Singapore, 2023. DOI:
10.1007/978-981-19-9501-9_11
53. Jaiswal R, Mutreja V, Sohal H, Sharma A. A review on current status of
traditional uses, phytochemistry, pharmacology and conservation of
Inula racemosa Hook. f. Materials Today: Proceedings
2022;68(4):842-7. DOI: 10.1016/j.matpr.2022.06.261
54. Khan A, Shah RD, Pallewar S. Evaluation of anti-inflammatory and
analgesic activity of Ethanolic extracts of Inula racemosa and Albizia
amara. Int J Pharmacognosy Phytochemical Res 2011;(3):22-7.
55. Arumugam P, Murugan M, Thangaraj N. Evaluation of anti-inflamma-
tory and analgesic effects of aqueous extract obtained from root powder
of Inula racemosa Hook. f. J Med Plant Res 2012;6(14):2801-6. DOI:
10.5897/JMPR11.119
56. Kumar C, Kumar A, Nalli Y et al. Design, synthesis and biological evalua-
tion of alantolactone derivatives as potential anti-inflammatory agents.
Med Chem Res 2019;(28):849-56. DOI: 10.1007/s00044-019-02337-1
57. Lim HS, Jin SE, Kim OS et al. Alantolactone from Saussurea lappa
exerts antiinflammatory effects by inhibiting chemokine production
and STAT1 phosphorylation in TNF-α and IFN-gamma-induced in Ha-
CaT cells. Phytother Res 2015;(29):1088-96. DOI: 10.1002/ptr.5354
58. Chun J, Choi RJ, Khan S et al. Alantolactone suppresses inducible nitric
oxide synthase and cyclooxygenase-2 expression by down-regulating
NF-kappaB, MAPK and AP-1 via the MyD88 signaling pathway in LPS-
activated RAW 264.7 cells. Int Immunopharmacol 2012;14(4):375-83.
DOI: 10.1016/j.intimp.2012.08.011
59. Salil S, Bhupinder A, Basheer A. Antioxidant Properties Of Inula Race-
mosa, A Traditional Herbal Medicine 40. Internet J Pharmacol
2012;10(1). DOI: 10.5580/A1434
60. Lateef R, Bhat KA, Chandra S, Banday JA. Antioxidant activity of chlo-
roform extract of inula racemosa from Kashmir Himalayas. Chemistry
J 2018;1(2):179-85.
61. Sharma V, Hem K, Sharma D et al. Ethnopharmacology, phytoche-
mistry and pharmacology of Inula racemosa Hook. F. J Nat Prod Re-
sour 2016;(2):40-6.
62. Lokhande PD, Gawai KR, Kodam KM et al. Antibacterial Activity of Iso-
lated Constituents and Extract of Roots of Inula racemosa. Res J Med
Plants 2007;(1):7-12.
63. Amorim MHR, Costa RMG, Lopes C, Bastos MMSM. Sesquiterpene lac-
tones: adverse health effects and toxicity mechanisms. Crit Rev Toxicol
2013;43(7):559-79. DOI: 10.3109/10408444.2013.813905
64. Rathore S, Raj Y, Debnath P et al. Ethnopharmacology, phytoche-
mistry, agrotechnology, and conservation of Inula racemosa Hook f. –
A critically endangered medicinal plant of the western Himalaya. J Eth-
nopharmacol 2022;(283):114613. DOI: 10.1016/j.jep.2021.114613
65. Kalachaveedu M, Raghavan D, Telapolu S et al. Phytoestrogenic effect
of Inula racemosa Hook f–A cardioprotective root drug in traditional
medicine. J Ethnopharmacol 2018;(210):408-16. DOI: 10.1016/
j.jep.2017.09.001
66. Nisar T, Iqbal M, Raza A et al. Estimation of Total Phenolics and Free
Radical Scavenging of Turmeric (Curcuma longa). Environ Sci
2015;15(7):1272-7.
67. Khalandar SD, Adithya TN, Basha SJ et al. A current review on Cur-
cuma Longa linn. Plant. Int J Pharmaceutical Chem Biol Sci
2018;8(1):68-73.
68. Boskabady MH, Shakeri F, Naghdi F. The Effects of Curcuma Longa L.
and its Constituents in Respiratory Disorders and Molecular Mecha-
nisms of Their Action. Studies Natural Products Chemistry. Chapter 7.
2020;(68):239-69. DOI: 10.1016/b978-0-12-817905-5.00007-x
69. Sharma P, Rani N, Sharma V, Garg M. An Overview of Pharmacological
Potential of Curcuma longa. Curr Traditional Med 2023;(10). DOI:
10.2174/2215083810666230329222554
70. Kim J, Jeong SW, Quan H et al. Effect of curcumin (Curcuma longa ex-
tract) on LPS-induced acute lung injury is mediated by the activation of
AMPK. J Anesth 2016;(30):100-8. DOI: 10.1007/s00540-015-2073-1
71. Xu F, Diao R, Liu J et al. Curcumin attenuates staphylococcus aureus-
induced acute lung injury. Clin Respir J 2015;9(1):87-97. DOI:
10.1111/crj.12113. Erratum in: Clin Respir J 2019;13(5):338. DOI:
10.1111/crj.13017
72. Bansal S, Chhibber S. Curcumin alone and in combination with aug-
mentin protects against pulmonary inflammation and acute lung injury
generated during Klebsiella pneumoniae B5055-induced lung infection
in BALB/c mice. J Med Microbiol 2010;59(Pt 4):429-37. DOI:
10.1099/jmm.0.016873-0
73. Yang XX, Li CM, Li YF et al. Synergistic antiviral effect of curcumin
functionalized graphene oxide against respiratory syncytial virus infec-
tion. Nanoscale 2017;(9):16086-92. DOI: 10.1039/C7NR06520E
74. Han S, Xu J, Guo X, Huang M. Curcumin ameliorates severe influenza
pneumonia via attenuating lung injury and regulating macrophage cy-
tokines production. Clin Exp Pharmacol Physiol 2018;45(1):84-93.
DOI: 10.1111/1440-1681.12848
75. Amadi S, Mehrabi Z, Zare M et al. Efficacy of Nanocurcumin as an Add-
On Treatment for Patients Hospitalized with COVID-19: A Double-
Blind, Randomized Clinical Trial. Int J Clin Practice 2023;(5734675).
DOI: 10.1155/2023/5734675
76. Valizadeh H, Abdolmohammadi-Vahid S, Danshina S et al. Nano-cur-
cumin therapy, a promising method in modulating inflammatory cyto-
kines in COVID-19 patients. Int Immunopharmacol 2020;89(Pt
B):107088. DOI: 10.1016/j.intimp.2020.107088
77. Emami B, Shakeri F, Ghorani V, Boskabady MH. Relaxant effect of Cur-
cuma longa on rat tracheal smooth muscle and its possible mechanisms.
Pharm Biol 2017;55(1):2248-58. DOI: 10.1080/ 13880209.2017.1400079
78. Subhashini, Chauhan PS, Kumari S et al. Intranasal curcumin and its
evaluation in murine model of asthma. Int Immunopharmacol
2013;17(3):733-43. DOI: 10.1016/j.intimp.2013.08.008
79. Abidi A, Gupta S, Agarwal M et al. Evaluation of Efficacy of Curcumin
as an Add-on therapy in Patients of Bronchial Asthma. J Clin Diagn Res
2014;8(8):HC19-24. DOI: 10.7860/JCDR/2014/9273.4705
80. Abbas S, Abdulridha M, Shafek M. Effect of Curcumin Supplement on
Pulmonary Functions, Total and Differential White Blood Cell Count,
Serum Level of Leptin and Body Mass Index in a Sample of Iraqi Pati-
ents with Chronic Bronchial Asthma. Al Mustansiriyah J Pharmaceu-
tical Sci 2019;19(2):47-58. DOI: 10.32947/ajps.19.02.00403
81. Fuloria S, Mehta J, Chandel A et al. A Comprehensive Review on the
Therapeutic Potential of Curcuma longa Linn. in Relation to its Major
Active Constituent Curcumin. Front Pharmacol 2022;13:820806. DOI:
10.3389/fphar.2022.820806
82. Soleimani V, Sahebkar A, Hosseinzadeh H. Turmeric (Curcuma Longa)
and its Major Constituent (Curcumin) as Nontoxic and Safe Substan-
ces: Review. Phytother Res 2018;32(6):985-95. DOI: 10.1002/ptr.6054
83. Manvitha K, Bidya B. Aloe vera: a wonder plant its history, cultivation
and medicinal uses. J Pharmacognosy Phytochemistry 2014;(2):85-8.
84. Kar SK, Bera TK. Phytochemical constituents of Aloe Vera and their
multifunctional properties: a comprehensive review. IJPSR
2018;9(4):1416-23. DOI: 10.13040/IJPSR.0975-8232.9(4).1416-23
85. Vazquez B, Avila G, Segura D, Escalante B. Antiinflammatory activity of
extracts from Aloe vera gel. J Ethnopharmacol 1996;(55):69-75.
86. Langmead L, Makins RJ, Rampton DS. Anti-inflammatory effects of
aloe vera gel in human colorectal mucosa in vitro. Aliment Pharmacol
Ther 2004;(19):521-7. DOI: 10.1111/j.1365-2036.2004.01874.x
87. Vijayalakshmi D, Dhandapani R, Jayaveni S et al. In vitro anti-inflam-
matory activity of Aloe vera by down regulation of MMP-9 in peripheral
blood mononuclear cells. J Ethnopharmacol 2012;141(1):542-6. DOI:
10.1016/j.jep.2012.02.04
88. Hęś M, Dziedzic K, Górecka D et al. Jędrusek-Golińska A, Gujska E.
Aloe vera (L.) Webb.: Natural Sources of Antioxidants – A Review.
Plant Foods Hum Nutr 2019;74(3):255-65. DOI: 10.1007/s11130-019-
00747-5
89. Boudreau MD, Beland FA. An evaluation of the biological and toxicolo-
gical properties of Aloe Barbadensis (Miller), Aloe vera. J Environ Sci
Health 2006;(24):103-54. DOI: 10.1080/10590500600614303
90. Banik S, Amit BS. Phytochemistry, health benefits and toxicological
profile of aloe. J Pharmacognosy Phytochemistry 2019;8(3):4499-
506.
91. Agarry OO, Olaleye MT, Michael CO. Comparative antimicrobial activi-
ties of Aloe vera gel and leaf. Afr J Biotechnol 2005;(4):1413-4. DOI:
10.4314/ajb.v4i12.71436
92. Nalimu F, Oloro J, Kahwa I, Ogwang PE. Review on the phytochemistry
and toxicological profiles of Aloe vera and Aloe ferox. Futur J Pharm
Sci 2021;7(1):145. DOI: 10.1186/s43094-021-00296-2
93. Gutierrez RM, Gonzalez AM, Hoyo-Vadillo C. Alkaloids from piper: a
review of its phytochemistry and pharmacology. Mini Rev Med Chem
2013;13(2):163-93. DOI: 10.2174/138955713804805148
94. Andriana Y, Xuan TD, Quy TN et al. Biological Activities and Chemical
Constituents of Essential Oils from Piper cubeba Bojer and Piper ni-
grum L. Molecules 2019;24(10):1876. DOI: 10.3390/molecules
24101876
95. Yam J, Schaab A, Kreuter M, Drewe J. Piper cubeba demonstrates anti-
estrogenic and anti-inflammatory properties. Planta Med
2008;74(2):142-6. DOI: 10.1055/s-2008-1034290
96. Perazzo FF, Rodrigues IV, Maistro EL et al. Anti-inflammatory and
analgesic evaluation of hydroalcoholic extract and fractions from seeds
of Piper cubeba L. (Piperaceae). Pharmacognosy J 2013;5(1):13-6.
DOI: 10.1016/j.phcgj.2012.12.001
97. Lima TC, Lucarini R, Volpe AC et al. In vivo and in silico anti-inflam-
matory mechanism of action of the semisynthetic (-)-cubebin derivati-
ves (-)-hinokinin and (-)-O-benzylcubebin. Bioorg Med Chem Lett
2017;27(2):176-9. DOI: 10.1016/j.bmcl.2016.11.081
98. Dwita LP, Iwo MI, Mauludin R, Elfahmi. Neuroprotective potential of
lignan-rich fraction of Piper cubeba L. by improving antioxidant capa-
city in the rat’s brain. Braz J Biol 2023;(82):e266573. DOI:
10.1590/1519-6984.266573
99. Alminderej F, Bakari S, Almundarij TI et al. Antioxidant Activities of a
New Chemotype of Piper cubeba L. Fruit Essential Oil (Methyleuge-
nol/Eugenol): In Silico Molecular Docking and ADMET Studies. Plants
(Basel) 2020;9(11):1534. DOI: 10.3390/plants9111534
100. Akshita C, Vijay BV, Praveen D. Evaluation of phytochemical scree-
ning and antimicrobial efficacy of Mesua ferrea and Piper cubeba fruit
extracts against multidrug-resistant bacteria. Pharmacophore
2020;(11):15-20.
101. Ahirrao P, Tambat R, Chandal N et al. MsrA Efflux Pump Inhibitory
Activity of Piper cubeba L.f. and its Phytoconstituents against Staphy-
lococcus aureus RN4220. Chem Biodivers 2020;17(8):e2000144.
DOI: 10.1002/cbdv.202000144
102. Alharbi NS, Khaled JM, Alzaharni KE et al. Effects of Piper cubeba L.
essential oil on methicillin-resistant Staphylococcus aureus: an AFM
and TEM study. J Mol Recognit 2017;30(1). DOI: 10.1002/jmr.2564
103. Usia T, Watabe T, Kadota S et al. Potent CYP3A4 inhibitory constitu-
ents of Piper cubeba. J Nat Prod 2005;68(1):64-8. DOI:
10.1021/np0401765
104. Deb A, Barua S, Das B. Pharmacological activities of Baheda (Termi-
nalia bellerica): a review. J Pharmacognosy Phytochem 2016;5(1):
194-7.
105. Motamarri Saraswathi N, Karthikeyan M, Kannan M, Rajasekar S.
Terminalia belerica, Roxb. – a phytopharmacological review. Int J Res
Pharmaceutical Biomed Sci 2012;3(1):96-100.
106. Gilani AH, Khan AU, Ali T, Ajmal S. Mechanisms underlying the anti-
spasmodic and bronchodilatory properties of Terminalia bellerica
fruit. J Ethnopharmacol 2008;116(3):528-38. DOI: 10.1016/
j.jep.2008.01.006
107. Elizabeth KM. Antimicrobial activity of Terminalia bellerica. Indian J
Clin Biochem 2005;20(2):150-3. DOI: 10.1007/BF02867416
108. Tandon G. Beta lactamase Inhibitors from Indigenous Herbs and Spi-
ces. Res J Pharmaceutical Biol Chem Sci 2014;(5):275-85.
109. Dharmaratne MPJ, Manoraj A, Thevanesam V et al. Terminalia belli-
rica fruit extracts: in-vitro antibacterial activity against selected mul-
tidrug-resistant bacteria, radical scavenging activity and cytotoxicity
study on BHK-21 cells. BMC Complement Altern Med 2018;18(1):325.
DOI: 10.1186/s12906-018-2382-7
110. Nampoothiri SV, Binil RSS, Prathapan A et al. In vitro antioxidant ac-
tivities of the methanol extract and its different solvent fractions obtai-
ned from the fruit pericarp of Terminalia bellerica. Nat Prod Res
2011;(25):277-87. DOI: 10.1080/14786419.2010.482053
111. Tanaka M, Kishimoto Y, Sasaki M et al. Terminalia bellirica (Gaertn.)
Roxb. Extract and Gallic Acid Attenuate LPS-Induced Inflammation
and Oxidative Stress via MAPK/NF-κB and Akt/AMPK/Nrf2 Pa-
thways. Oxid Med Cell Longev 2018;(2018): 9364364. DOI:
10.1155/2018/9364364
112. Jayesh K, Helen LR, Vysakh A et al. Ethyl acetate fraction of Termina-
lia bellirica (Gaertn.) Roxb. fruits inhibits proinflammatory mediators
via down regulating nuclear factor-κB in LPS stimulated Raw 264.7
cells. Biomed Pharmacother 2017;(95):1654-60. DOI: 10.1016/j.bio-
pha.2017.09.080
113. Saraphanchotiwitthaya A, Sripalakit P, Ingkaninan K. Effects of Ter-
minalia bellerica Roxb. methanolic extract on mouse immune res-
ponse in vitro. Mj Int J Sci Tech 2008;2(2): 400-7.
114. Thanabhorn S, Jaijoy K, Thamaree S, Ingkaninan K. Acute and sub-
acute toxicity of the ethanolic extract from the fruits of Terminalia be-
lerica. Mahidol Univ J Pharm Sci 2006;(33):23-30.
115. Sireeratawong S, Jaijoy K, Panunto W et al. Soonthornchareonnon N.
Acute and chronic toxicity studies of the water extract from dried fruits
of Terminalia bellerica (Gaertn.) Roxb. In Spargue-Dawley rats. Afr J
Tradit Complement Altern Med 2012;10(2):223-31. DOI: 10.4314/ajt-
cam.v10i2.6
116. Bano N, Ahmed A, Tanveer M et al. Pharmacological Evaluation of
Ocimum sanctum. J Bioequiv Availab 2017;9(3):387-92. DOI:
10.4172/jbb.1000330
117. Singh D, Chaudhuri PK. A review on phytochemical and pharmacolo-
gical properties of Holy basil (Ocimum sanctum L.). Industrial Crops
Prod 2018;(118):367-82. DOI: 10.1016/j.indcrop.2018.03.048
118. Siva M, Shanmugam KR, Shanmugam B et al. Ocimum sanctum: a re-
view on the pharmacological properties. Int J Basic Clin Pharmacol
2016;5(3):558-65. DOI: 10.18203/2319-2003.ijbcp20161491
119. Kumar A, Agarwal K, Maurya AK et al. Pharmacological and phytoche-
mical evaluation of Ocimum sanctum root extracts for its antiinflam-
matory, analgesic and antipyretic activities. Pharmacogn Mag
2015;11(Suppl.1):S217-24. DOI: 10.4103/0973-1296.157743
120. Kelm MA, Nair MG, Strasburg GM, DeWitt DL. Antioxidant and cyclo-
oxygenase inhibitory phenolic compounds from Ocimum sanctum Linn.
Phytomedicine 2000;7(1):7-13. DOI: 10.1016/S0944-7113(00)80015-X
121. Koroch A, Juliani H, Sims C, Simon J. Antioxidant activity, total phe-
nolics, and rosmarinic acid content in different basils (Ocimum spp.).
Israel J Plant Sci 2010;(58):191-5. DOI: 10.1560/IJPS.58.3-4.191
122. Subramanian M, Chintalwar GJ, Chattopadhyay S. Antioxidant and
radioprotective properties of an Ocimum sanctum polysaccharide. Re-
dox Rep 2005;10(5):257-64. DOI: 10.1179/135100005X70206
123. Ali H, Dixit S. In vitro antimicrobial activity of flavanoids of Ocimum
sanctum with synergistic effect of their combined form. Asian Pacific
J Tropical Dis 2012;(2):S396-S398. DOI: 10.1016/s2222-
1808(12)60189-3
124. Melo RS, Albuquerque Azevedo ÁM, Gomes Pereira AM. Chemical Com-
position and Antimicrobial Effectiveness of Ocimum gratissimum L.
Essential Oil Against Multidrug-Resistant Isolates of Staphylococcus
aureus and Escherichia coli. Molecules 2019;24(21):3864. DOI:
10.3390/molecules24213864
125. Ghoke SS, Sood R, Kumar N et al. Evaluation of antiviral activity of
Ocimum sanctum and Acacia arabica leaves extracts against H9N2 vi-
rus using embryonated chicken egg model. BMC Complement Altern
Med 2018;18(1):174. DOI: 10.1186/s12906-018-2238-1
126. Dashputre NL, Naikwade NS. Preliminary immunomodulatory acti-
vity of aqueous and ethanolic leaves extracts of Ocimum basilicum
Linn in mice. Int J Pharm Tech Res 2010;2(2):1342-9.
127. Das R, Raman R, Saha H, Singh R. Effect of Ocimum sanctum Linn.
(Tulsi) extract on the immunity and survival of Labeo rohita (Hamil-
ton) infected with Aeromonas hydrophila. Aquaculture Res
2015;(46):1111-21. DOI: 10.1111/are.12264
128. Vaghasiya J, Datani M, Nandkumar K et al. Comparative Evaluation of
Alcoholic and Aqueous Extracts of Ocimum Sanctum for Immunomo-
dulatory Activity. Int J Pharmaceutical Biol Res 2010;1(1):25-9.
129. Mondal S, Varma S, Bamola VD et al. Double-blinded randomized
controlled trial for immunomodulatory effects of Tulsi (Ocimum sanc-
tum Linn.) leaf extract on healthy volunteers. J Ethnopharmacol
2011;136(3):452-6. DOI: 10.1016/j.jep.2011.05.012
130. Jamshidi N, Cohen MM. The Clinical Efficacy and Safety of Tulsi in
Humans: A Systematic Review of the Literature. Evid Based Comple-
ment Alternat Med 2017;(2017):9217567. DOI: 10.1155/2017/9217567
131. Sharma V, Hem K, Seth A, Maurya S. Solanum indicum Linn.: An eth-
nopharmacological, phytochemical and pharmacological review. Curr
Res J Pharmaceutical Allied Sci 2017;(1):1-9.
132. Jayanthy A, Anupam M, Subash CV et al. A Brief Review on Pharma-
cognosy, Phytochemistry and Therapeutic Potential Of Solanum In-
diumL. Used in Indian Systems of Medicine. Asian J Res Chemistry
2016;9(3):127-32. DOI: 10.5958/0974-4150.2016.00022.5
133. Iqubal M, Sharma SKS, Hussain MS, Mujahid M. An updated ethno-
botany, phytochemical and pharmacological potential of Solanum in-
dicum L. JDDT 2022;12(2):160-72. DOI: 10.22270/jddt.v12i2.5385
134. Deb PK, Ghosh R, Chakraverty R et al. Phytochemical and Pharmaco-
logical Evaluation of Fruits of Solanum indicum Linn. Int J Pharm Sci
Rev Res 2014;25(2):28-32.
135. Das N, Ghosh R, Bhakta T, Deb P. Evaluation of In-vitro Antioxidant
and Anthelmintic Activity of Solanum indicum Linn. Berries. Indo Am
J Pharmaceutical Res 2013;(3):4123-30.
136. Srividya AR, Arunkumar A, Cherian B et al. Phytochemical and Anti-
microbial studies of Solanum indicum leaves. Anc Sci Life
2009;29(1):3-5.
137. Raorane C, Gavimat CC, Kulkarni S et al. Antibacterial potentials of
Solanum indicum, Solanum xanthocarpum and Physalis minima. Int J
Pharmaceutical Applications 2012;3(4):414-8.
138. Zhao H, Ren S, Yang H et al. Peppermint essential oil: its phytoche-
mistry, biological activity, pharmacological effect and application. Bio-
med Pharmacother 2022;(154):113559. DOI: 10.1016/j.bio-
pha.2022.113559
139. Kamatou GPP, Vermaak I, Viljoen AM, Lawrence BM. Menthol: A sim-
ple monoterpene with remarkable biological properties. Phytoche-
mistry 2013;(96):15-25. DOI: 10.1016/j.phytochem.2013.08.005
140. Cheng H, An X. Cold stimuli, hot topic: An updated review on the bio-
logical activity of menthol in relation to inflammation. Front Immunol
2022;(13):1023746. DOI: 10.3389/fimmu.2022.1023746
141. Наумов Д.Е., Гассан Д.А., Килимиченко К.Ф. и др. Особенности экс-
прессии рецептора TRPM8 в респираторном тракте больных брон-
хиальной астмой. Бюллетень физиологии и патологии дыхания.
2018;(69):19-24. DOI: 10.12737/article_5b96073 c5711b1.83866044
Naumov D.E., Gassan D.A., Kilimichenko K.F., et al. Features of
TRPM8 receptor expression in the respiratory tract of patients with
bronchial asthma. Bulletin of respiratory physiology and pathology.
2018;(69):19-24. DOI: 10.12737/article_5b96073c5711b1.83866044
(in Russian).
142. Ciaglia T, Vestuto V, Bertamino A et al. On the modulation of TRPM
channels: Current perspectives and anticancer therapeutic implica-
tions. Front Oncol 2023;(12). DOI: 10.3389/fonc.2022.1065935
143. Galeotti N, Di Cesare Mannelli L, Mazzanti G et al. Menthol: a natural
analgesic compound. Neurosci Lett 2002;322(3):145-8. DOI:
10.1016/s0304-3940(01)02527-7
144. Сафоничева О.Г., Мартынчик С.А. Принципы Аюрведы c позиций
междисциплинарного обоснования технологий персонализиро-
ванной медицины. Международный журнал прикладных и фун-
даментальных исследований. 2015;10(2):249-52.
Safonicheva O.G., Martynchik S.A. Principles of Ayurveda from the
standpoint of interdisciplinary justification of personalized medicine
technologies. International Journal of Applied and Fundamental Re-
search. 2015;10(2):249-52 (in Russian).
145. Kuang Y, Li B, Fan J et al. Antitussive and expectorant activities of li-
corice and its major compounds. Bioorg Med Chem 2018;26(1):278-
84. DOI: 10.1016/j.bmc.2017.11.046
146. Желенина Л.А., Незабудкина А.С. Рациональный подход к лече-
нию кашля у детей. Фарматека. 2019;26(11):60-8. DOI:
10.18565/pharmateca.2019.11.00-00
Zhelenina L.A., Nezabudkina A.S. Rational approach to the treatment
of cough in children. Pharmateka. 2019;26(11):60-8. DOI:
10.18565/pharmateca.2019.11.00-00 (in Russian).
147. Иванов В.А., Суздаленков А.В., Заплатников А.Л. Рациональное
использование средств от кашля при инфекциях органов дыха-
ния у детей. РМЖ. 2006;(19):1368.
Ivanov V.A., Suzdalenkov A.V., Zaplatnikov A.L. Rational use of cough
medicines for respiratory infections in children. RMJ. 2006;(19):1368
(in Russian).
148. Коротаева М.С., Сидоров А.В., Тихонова И.Г. Этические аспекты
фитотерапии. Медицинская этика. 2022;10(4):38-42.
Korotaeva M.S., Sidorov A.V., Tikhonova I.G. Ethical aspects of herbal
medicine. Medical Ethics. 2022;10(4):38-42 (in Russian).
149. Khan AU, Gilani AH. Natural Products Useful in Respiratory Disor-
ders: Focus on Side-Effect Neutralizing Combinations. Phytother Res
2015;29(9):1265-85. DOI: 10.1002/ptr.5380
150. Hosseinzadeh H, Nassiri-Asl M. Pharmacological Effects of Glycyrr-
hiza spp. and Its Bioactive Constituents: Update and Review. Phytot-
her Res 2015;29(12):1868-86. DOI: 10.1002/ptr.5487
151. Kim MW, Kang JH, Shin E et al. Processed Aloe vera gel attenuates
non-steroidal anti-inflammatory drug (NSAID)-induced small intesti-
nal injury by enhancing mucin expression. Food Funct
2019;10(9):6088-97. DOI: 10.1039/c9fo01307e
152. Haniadka R, Saldanha E, Sunita V et al. A review of the gastroprotec-
tive effects of ginger (Zingiber officinale Roscoe). Food Funct
2013;4(6):845-55. DOI: 10.1039/c3fo30337c
153. Gupta A, Kumar R, Bhattacharyya P et al. Terminalia bellirica (Ga-
ertn.) roxb. (Bahera) in health and disease: A systematic and compre-
hensive review. Phytomedicine 2020;(77):153278. DOI:
10.1016/j.phymed.2020.153278
154. Sabbaghzadegan S, Golsorkhi H, Soltani MH et al. Potential protective
effects of Aloe vera gel on cardiovascular diseases: A mini-review. Phy-
tother Res 2021;35(11):6101-13. DOI: 10.1002/ptr.7219
155. Fakhri S, Patra JK, Das SK et al. Ginger and Heart Health: From Mec-
hanisms to Therapeutics. Curr Mol Pharmacol 2021;14(6):943-59.
DOI: 10.2174/1874467213666201209105005
156. Tiwari AK, Gupta PS, Prasad M, Malairajan P. Modulation of Inula ra-
cemosa Hook Extract on Cardioprotection by Ischemic Preconditio-
ning in Hyperlipidaemic Rats. J Pharmacopuncture 2022;25(4):369-
81. DOI: 10.3831/KPI.2022.25.4.369
157. Iftkhar A, Hasan IJ, Sarfraz M et al. Nephroprotective Effect of the
Leaves of Aloe barbadensis (Aloe Vera) against Toxicity Induced by
Diclofenac Sodium in Albino Rabbits. West Indian Med J
2015;64(5):462-7. DOI: 10.7727/wimj.2016.052
158. Song Z, Fang H, Zhang X et al. Renoprotective Glycoside Derivatives
from Zingiber officinale (Ginger) Peels. J Agric Food Chem
2023;71(41):15170-85. DOI: 10.1021/acs.jafc.3c05224
159. Drissi B, Mahdi I, Yassir M et al. Cubeb (Piper cubeba L.f.): A compre-
hensive review of its botany, phytochemistry, traditional uses, and
pharmacological properties. Front Nutr 2022;(9):1048520. DOI:
10.3389/fnut.2022.1048520
160. Ullah A, Munir S, Badshah SL et al. Important Flavonoids and Their
Role as a Therapeutic Agent. Molecules 2020;25(22):5243. DOI:
10.3390/molecules25225243
161. Коровина Н.А., Заплатников А.Л., Захарова И.Н. и др. Принципы
выбора средств от кашля при острых респираторных заболева-
ниях у детей. Вестник педиатрической фармакологии и нутри-
циологии. 2005;(1):42-6.
Korovina N.A., Zaplatnikov A.L., Zakharova I.N. et al. Principles of se-
lection of cough remedies for acute respiratory diseases in children.
Bulletin of pediatric pharmacology and nutrition. 2005;(1):42-6 (in
Russian).
For citation:Ovsyannikov K.V., Praskurnichiy E.A. Self-monitoring of blood glucose levels in patients with type 2 diabetes mellitus. Clinical review for general practice. 2024; 5 (8): 116–124 (In Russ.). DOI: 10.47407/kr2024.5.8.00469
All accepted articles publish licensed under a Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.