Genotoxic properties of hypoglycemic drugs (systematic review)

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

According to the literature genotoxic properties of about a half of hypoglycemic drugs have not been investigated in accordance with the recommended methodology, and studies of the mutagen-modifying activity of antidiabetic drugs are sporadic. Based on the available published data, it is impossible to conclude about either presence or absence of genotoxic / antigenotoxic potential of antidiabetic drugs. There is evidence of the antimutagenic activity of metformin; in relation to other drugs, studies of mutagen-modifying activity have not been carried out or are represented only by a few articles. Further study of the genotoxic properties of hypoglycemic drugs is required in accordance with modern approaches and requirements, as well as an assessment of their mutagen-modifying activity.

Full Text

INTRODUCTION

Diabetes mellitus (DM) refers to a group of metabolic diseases characterized by abnormally increased plasma glucose concentrations or hyperglycemia, and is classified as type 1 (T1DM) or type 2 (T2DM). If untreated, DM leads to chronic degenerative diseases of the heart, kidneys, and the nervous system, including the retina. The International Diabetes Federation has characterized DM as a global epidemic [1]. By 2030, the number of patients with T2DM is predicted to increase globally to 439 million people [2].

Numerous epidemiological studies and meta-analyses indicate a link between DM and cancer incidence, as well as mortality from liver, pancreatic, colon, kidney, endometrial, and breast cancer [3]. Patients with a combination of DM and cancer have an increased risk of mortality from any cause compared with patients without a history of DM [4].

Mutagenesis has been generally recognized and comprehensively substantiated to trigger carcinogenesis [5, 6]. The increase in markers of genotoxicity in DM patients was noted repeatedly. It is associated with the formation of genotoxic reactive oxygen species (ROS) during hyperglycemia-induced oxidative stress [7–11].

Several hypoglycemic drugs are available to manage DM. However, it is not known whether these drugs contribute to genotoxicity in diabetic patients. These drugs may have their own genotoxic activities, as well as enhance (co-mutagens) or weaken (antimutagens) the effects of exogenous and endogenous genotoxicants, in particular ROS [12].

This work aimed to systematically review and analyze the results of previous studies of genotoxic activity and mutagen-modifying properties of hypoglycemic drugs in experimental eukaryotic test systems in vitro and in vivo.

MATERIALS AND METHODS

Literature search was conducted to include articles published from January 1, 1990 to March 31, 2021 using the MedLine/PubMed scientific literature database (National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA – http://www.ncbi.nlm.nih.gov/PubMed) and the Russian Science Citation Index (RSCI) scientific electronic library (http://elibrary.ru). For certain instances when the understanding of a class of drugs required a more extensive review of its history, an additional literature search to include articles published earlier than 1990 was performed. The results from this supplementary search are specifically indicated with the dates in the text.

Key search terms for studies on the genotoxic activities of oral hypoglycemic drugs included the international nonproprietary names of drugs classified as group A10 “Drugs for the treatment of DM” in the anatomical-therapeutic-chemical drug classification system [13], in combination with the terms “genotoxicity” or “genotoxic,” “mutagen” or “mutagenic”, and the corresponding equivalents for English-language sources. The review included genotoxicological studies performed by chromosomal aberrations test (CA) and/or the cytokinesis-block micronucleus (MN) cytome assay, evaluation of DNA damage by the DNA comet assay and the following criteria:

  • conducted in vivo on mammals or in vitro in cultures of eukaryotic somatic non-immortalized cell lines;
  • performed in compliance with the standard practices of evaluating genotoxicity, including the presence of positive and negative controls and appropriate statistical analyses;
  • published in peer-reviewed scientific journals in the English or Russian languages, with full-text versions of articles available.

From the full-text articles, information on the test systems (species of animals, cells used), experimental design, (doses, routes and frequency of administration, concentration, exposure time, etc.), and results of the study were collected.

In the absence of information on the genotoxicological properties of a specific drug in the Pubmed and RSCI databases, an additional search was performed on the official websites of regulatory agencies, namely European Medicinal Agency (EMA) and the U.S. Food and Drug Administration (FDA).

RESULTS AND DISCUSSION

The antidiabetic drug classes included in the systematic search were insulin and its analogs, insulin secretion stimulants [sulfonylurea derivatives, meglitinides, analogs of glucagon-like peptides 1 (GLP-1), dipeptidyl peptidase 4 (DPP-4) inhibitors], insulin sensitivity sensitizers that increase the glucose disposal (biguanides, thiazolidinediones), and drugs with other mechanisms of action [alpha-glucosidase inhibitors and sodium glucose cotransporter type 2 (SGLT2) inhibitors].

Insulin and its analogs

Insulin and its analogs are widely used to treat patients with T1DM and T2DM. Insulin is required in patients with T2DM who fail to achieve glycemic control after initiating lifestyle changes and oral antidiabetic drugs, as well as during pregnancy, in the postoperative period, and in other acute conditions [14].

Current regulatory requirements do not require genotoxicity studies of insulin and its analogs for the registration of their drug products (DPs). Using our search strategy, no study available in the literature met the above-defined inclusion criteria. However, there were studies that determined the outcomes of interest in immortalized cell lines as presented in Table 1. In particular, one study aimed to elucidate the possible contribution of insulin to tumorigenesis and levels of genotoxic biomarkers of oxidative stress in DM [15]. It was revealed that a minimum human insulin concentration of 10 nM in human lymphocytes and 0.5–1 nM in human colon adenocarcinoma cells (HT29) resulted in an increase in the levels of DNA damage and MN in vitro 24 h after treatment. Normally, the concentration of fasting insulin in the blood in the morning is an order of magnitude lower than these values (0.04 nM). However, in hyperinsulinemia and in the postprandial state, the concentrations could exceed 1 nM, and the plasma insulin concentration in ZDF rats (Zucker diabetic fatty) reaches 1.67 nM. Data from specialized clinical trials note an increased risk of cancer in patients who use insulin or its analogs [17].

 

Table 1. Studies of the genotoxic and mutagenic properties of insulin and its analogs in vitro and in vivo

International nonproprietary name

Test system

Treatment conditions

Measured biomarker

Effect

Literature source

Recombinant human insulin

Cell lines of adenocarcinoma (MCF 7) and BT 474 human breast carcinoma

10 nM for 24 h

DNA damage (DNA comet assay), MN frequency

Significant effects in both test systems for both biomarkers

[18]

Cell lines of human colon adenocarcinoma (HT29), human colon cancer (Caco 2), primary cell line of rat colon, human lymphocytes

Within 2 h (HT29 and Caco 2) or 6 days (HT29; 50% medium change daily and addition of fresh insulin at concentrations of 0.5–1, 1–2, and 10–20 nm), 30 min of the primary colon cell line (10, 100, and 2000 nM) and 24 h for lymphocytes (10 and 100 nM)

DNA damage (DNA comet assay), MN frequency

Significant dose-dependent effects in all concentrations and test systems

[15]

Insulin lispro

According to the manufacturer’s information, insulin lispro does not induce MN induction in the bone marrow of male and female ICR mice in vivo and the induction of CA in Chinese hamster ovary (CHO) cells (primary data not presented)

[19]

Insulin aspart

According to the FDA and EMA, insulin aspart does not exhibit mutagenic activity in tests for recording CA in human peripheral blood lymphocytes, in a test for recording MN in mice in vivo (primary data not presented)

[20, 21]

Insulin glulisine

According to the FDA and EMA, insulin glulisine does not exhibit mutagenic activity in vitro and in vivo CA recording tests (primary data not presented)

[22, 23]

Insulin glargine

Cell lines of adenocarcinoma (MCF-7) and BT 474 human breast carcinoma

10 nM for 24 h

Level of DNA damage (DNA comet assay), MN frequency

Significant effect in both test systems for both biomarkers

[18]

Insulin detemir

According to the FDA and EMA, insulin detemir does not exhibit mutagenic activity in tests for recording CA in human lymphocyte cells in vitro and in bone marrow cells of CD-1 mice in vivo up to a dose of 7500 nM/kg (primary data are not presented)

[24, 25]

Insulin degludec

Genotoxicity study data are not presented in the available literature

Note. Here and below in Tables 2–6. MN – micronuclei, CA – chromosomal aberrations. Hereinafter, in the tables, studies are highlighted in gray, which reveal significant genotoxic activity of the compound in comparison with the negative control.

 

Current available data is insufficient to provide basis for evaluating the genotoxicity of insulin. Whether insulin or its excipients in the formulations directly causes the findings reported is unknown. Based on the general understanding of the mechanisms of induced mutagenesis and the known effects of insulin, it could be assumed that genotoxicity due to insulin is improbable.

Oral hypoglycemic drugs

Information on the results of genotoxicity and mutagenicity studies of the main classes of hypoglycemic drugs is presented in Tables 2–6. Thirty-two drugs were included in the analysis.

For 6 of these drugs (19 %), no data on their genotoxic activities was available in the literature (buformin, gemigliptin, evogliptin, gosogliptin, dulaglutide, and ipragliflozin). For 10 of these DPs (31 %), only documents from the websites of regulatory authorities in Europe and/or the USA were available and did not included specifications of the experimental design or presentation of primary data (saxagliptin, alogliptin, exenatide, liraglutide, lixisenatide, semaglutide, canagliflozin, ertugliflozin, repaglinide). For these drugs, it was indicated that cytogenetic studies were performed in accordance with current regulatory requirements in vitro and in vivo, and no data was found indicating genotoxic properties in them.

Experimental data on genotoxicity or mutagenic effects were available only for 16 drugs (50 %), performed in either in vivo (for 13 drugs, 41 %) or in vitro (for 8 drugs, 25 %) systems as summarized in Tables 2–6. Mutagenic effects in vivo was noted in sulfonylurea derivatives of chlorpropamide [27], tolbutamide [28] and gliquidone [29], as well as in a thiazolidinedione derivative pioglitazone [30, 31]. The genotoxic activity in vivo was demonstrated by rosiglitazone [32], and in vitro by metformin [9, 38] and sitagliptin [33]. The absence of genotoxicity or mutagenecity was demonstrated in vivo for glibenclamide, carbutamide, glipizide, gliquidone, gliclazide, glimepiride, dapagliflozin, and empagliflozin.

Biguanide derivatives. Metformin is the most commonly used drug for treatment of prediabetes, gestational DM, and T2DM in the last 65 years [34]. According to various estimates, metformin is prescribed to 70 %–85 % of T2DM patients annually for long-term daily use [35, 36]. The widespread use of metformin has captured the interest of research groups to assess its genotoxicity (Table 2).

 

Table 2. Studies of genotoxic and mutagenic properties of biguanide derivatives in vitro and in vivo

International nonproprietary name

Test system

Treatment conditions

Effect biomarker

Effect

Literature source

Metformin

Wistar albino rats, male, control or model of streptozotocin-induced diabetes mellitus (65 mg/kg intraperitoneally, single dose)

Single dose, per os in doses of 100, 500, and 2500 mg/kg

Frequency of MN and CA in bone marrow cells 24 h after administration

No effect in control rats, significant decrease in MN and CA levels in rats with diabetes mellitus (in two higher doses)

[37]

Wistar albino rats, male, control or model of streptozotocin-induced diabetes mellitus (65 mg/kg intraperitoneally, single dose)

Daily for 4 or 8 weeks, per os in doses of 100 or 500 mg/kg

Frequency of MN and CA in bone cells brain 24 h after the last injection

No effect in control rats, a significant decrease in the MN level in rats with diabetes mellitus (at a dose of 500 mg/kg 4 and 8 weeks after administration)

[37]

Swiss albino mice, females

Once, intraperitoneally at doses of 95.4, 190.8, and 333.9 mg/kg

MN frequency in bone marrow cells 24 h after administration

No effect; cytotoxicity at the highest dose

[38]

Chinese hamster ovary cells, CHO-K1 in vitro

114.4 and 572 μg/ml

DNA damage (DNA comet assay, alkaline version) and CA frequency 24 h after treatment

Significant increase in the level of DNA damage in both concentrations (maximum with a lower one); no effect on the level of CA was found

[38]

Culture of human lymphocytes

114.4 μg/ml for 72 h

DNA damage (DNA comet assay, alkaline version) and MN frequency

No effect

[39]

Culture of human lymphocytes

12.5, 25, and 50 μM for 72 h

Frequency of MN and CA

No effect

[40]

Rat NRK kidney epithelial cells

3, 12, and 48 μM for 2 and 24 h with/without insulin (10 nM)

DNA damage (DNA comet assay, alkaline version) and NM frequency

Significant reduction in DNA damage and MN frequency compared to insulin treatment only

[9]

Buformin

Genotoxicity study data are not presented in the available literature

 

Analysis of the data given in Table 2 shows that metformin administered orally or intraperitoneally at doses ranging from 95.4 to 2500 mg/kg does not result in the induction of CA or MN in laboratory rodents. Rather, metformin exhibited antimutagenic properties under these sets of conditions in streptozotocin-induced DM model in rats [37]. A similar decrease in MN in bone marrow cells was observed in the same model after daily 4-week oral administration of 50 mg/kg metformin combined with 1 mg/kg [41].

The absence of genotoxic activity in vivo in metformin agrees with results obtained in vitro using cell cultures of rodents and human lymphocytes at maximum doses from 50 μM in some cases and up to 114.4 μg/ml in others Table 2. However, one study showed that metformin can induce DNA damage in p53-deficient Chinese hamster ovary CHO-K1 cells was shown using the DNA comet assay [38]. However, the strength of evidence from this set of data is low because the authors used unverified indicators that differ significantly from generally accepted ones [42]. Lastly, it is worth noting that a significant decrease in the levels of genotoxic biomarkers was observed in several clinical studies in patients with T2DM while taking metformin [7].

In turn, the antimutagenic effect of metformin, established in experiments with streptozotocin DM, is supported by a number of independent studies which used chemical mutagens [43]. Metformin after 7-day daily administration significantly reduced the frequency of MN in polychromatophilic erythrocytes in the bone marrow of male Swiss albino mice 24, 48, or 72 h after intraperitoneal administration of the cytostatic antitumor drug Adriamycin [44]. This observation was seen using metformin doses of 62.5, 125, and 250 mg/kg and occurred in a dose-dependent manner. Data from in vitro studies also indicate a protective effect of metformin against the induction of CA and MN after exposure to ionizing radiation [45] and DNA damage (% DNA in the comet tail) induced by 1 mM cumene hydroperoxide in human lymphocytes [46].

The protective effect of metformin extends to its ability to reduce the mutagenic effects of other hypoglycemic DPs [47]. The cytogenetic effects of sitagliptin and vildagliptin [0.04 mg/(kg per day)] alone or with metformin (0.2 mg metformin) in pregnant female mice and their embryos were evaluated. In the absence of metformin, both these drugs exerted mutagenic and toxic effects on pregnant females and embryos. In contrast, no similar effects were registered when either of these drugs were combined with metformin.

Furthermore, metformin has antiradical [48], reparative [49], and proapoptotic [50] effects, each of which can contribute to the observed antimutagenic effects.

This antimutagenic effect translates well when measuring outcomes directly related to cancer. Metformin has been shown to possess antitumor activity [51]. Lastly, in a number of meta-analyses, patients taking metformin have been shown to have a lowered risk of lung, pancreatic, prostate, and breast cancer, as well as in mortality in cancer patients, compared with patients receiving insulin or sulfonylurea derivatives [52–55].

Two plausible mechanistic pathways underlying the benefit of using metformin as a means of cancer prevention have been proposed previously, namely (1) an indirect pathway associated with its ability to reduce the insulin levels by slowing tumor proliferation in patients with hyperinsulinemia; and (2) a direct action against the respiratory Complex I of the electron transfer chain in the mitochondria of preneoplastic and neoplastic cells, which ultimately reduces the energy consumption of the target cell [56]. Both pathways of action involve stimulation of adenosine monophosphate-activated protein kinase (AMPK) by metformin, which inhibits the target of rapamycin (mTOR) in mammals. AMPK activation decreases cell proliferation and can trigger cell cycle arrest and apoptosis [57, 58]. Today, in addition to these two possibilities of antitumor prophylaxis, it is quite appropriate to suggest an antimutagenic pathway, for example, by suppressing the genotoxicity of ROS arising during oxidative stress DM.

Sulfonylurea derivatives. It is known that the use of the first generation of drugs in this group (tolbutamide, chlorpropamide, acetohexamide, and tolazamide) was associated with an increased risk of thyroid cancer due to their antithyroid action [59]. Currently, the second generation of sulfonylureas (glipizide, glimepiride, gliclazide, and glycidone) are widely used and can provide relatively better glycemic control with fewer side effects. Information on the results of the study of genotoxicity of sulfonylurea derivatives is presented in Table 3.

 

Table 3. Studies of genotoxic and mutagenic properties of sulfonylurea derivatives in vitro and in vivo

International nonproprietary name

Test system

Treatment conditions

Effect biomarker

Effect

Literature source

Glibenclamide

Culture of human lymphocytes

0.6, 10, 100, 240, and 480 μM for 72 h

Frequency of MN

No effect

[60]

Chinese hamsters, males and females

Once per os at a dose of 10 mg/kg

SCE in bone marrow cells 24 h after administration

No effect

[27]

Swiss albino mice, male

Twice with an interval of 24 h at doses of 4, 8, and 16 mg/kg

Frequency of MN in bone marrow cells 6 h after the last injection

No effect

[28]

Chlorpropamide

Chinese hamsters, C57BL/6J mice, males and females

Therapeutic dose 7.1 mg/kg, as well as 71, 177.5, 497, and 710 mg/kg, single dose, per os

SCE in bone marrow cells 24 hours after administration

Significant dose-dependent effect in both test systems

[27]

Mice NMRI, C3H, C57BL/6J, Chinese hamsters, Sprague-Dawley rats, males and females

Twice after 24 h at a dose of 355 mg/kg, per os

MN in bone marrow cells 6 h after the last injection

Significant increase in MN levels in all mice test systems; no effect in rats and hamsters

[28]

Tolbutamide

Chinese hamsters, C57BL/6J mice, males and females

Therapeutic dose 28.6 mg/kg, as well as 286, 1430, and 2002 mg/kg, single dose, per os

SCE in bone marrow cells 24 h after administration

Significant dose-dependent effect in both test systems

[27]

Mice NMRI, C3H, C57BL/, Chinese hamsters, Sprague-Dawley rats, males and females

Twice after 24 h at a dose of 1430 mg/kg, per os

MN in bone marrow cells 6 h after the last injection

Significant increase in MN levels only in line C57BL mice; no effect in other test systems

[27]

Swiss albino mice, male

Twice with an interval of 24 h at doses of 500, 1000, and 2000 mg/kg

Frequency of MN in bone marrow cells 6 h after the last injection

Significant (in two higher doses) dose-dependent increase in MN level

[28]

Carbutamide

Chinese hamsters, males and females

Once per os at a dose of 715 mg/kg

SCE in bone marrow cells 24 h after administration

No effect

[27]

Glipizide

Chinese hamsters, males and females

Once per os at a dose of 15 mg/kg

SCE in bone marrow cells 24 h after administration

No effect

[27]

Gliquidone

Chinese hamsters, males and females

Once per os at a dose of 85 mg/kg

SCE in bone marrow cells 24 h after administration

No effect

[27]

Swiss albino mice, male

Per os at a dose of 30 mg/kg once or daily for 10 or 20 days

MN in bone marrow cells 24 h after the last injection

Significant (p < 0.01) dose-dependent increase in the level of MN cells was noted in both cases multiple administration

[29]

Swiss albino mice, male

Once per os at doses of 30, 60, and 120 mg/kg

CA in bone marrow cells 7 days after administration

Significant (p < 0.01) dose-dependent increase in the level of cells with CA was registered for all doses

[29]

Gliclazide

Swiss albino and OF1 mice

Once at doses of 1, 2, and 3 g/kg

MN in peripheral blood cells 24 h after administration

No effect

[61]

Culture of human lymphocytes

5, 25, 50, and 100 μM without/with subsequent irradiation (1.5 Gy after 3 h)

MN frequency after 72 h of exposure

No effect (without irradiation), significant concentration-dependent decrease in the level of damage caused by irradiation, in all concentrations

[62]

Glimepiride

Wistar rats, male, diabetes model, intraperitoneal administration of streptozotocin (65 mg/kg) and nicotinamide (230 mg/kg)

Daily for 4 weeks per os at doses of 0.175, 17.5, and 175 mg/kg

Frequency of MN in bone marrow cells

Significant dose-dependent decrease in the level of cells with MN compared to control (p < 0.001)

[63]

 

Additionally, the results of studies performed in the 1980s investigating the cytogenetic properties of chlorpropamide and tolbutamide should be mentioned as reference [27, 28]. In these studies, a dose-dependent mutagenic effects of these drugs were demonstrated when used in doses 1–2 orders of magnitude higher than the therapeutic doses used in humans. These findings provide a compelling argument in favor of refinement of future studies to measure the genotoxicity of chlorpropamide and tolbutamide based on contemporary protocols. Moreover, no studies on the DNA-damaging effects in vitro or in vivo have been done for any of the drugs in this group, and the mutagenicity data of the first-generation sulfonylurea derivatives created several decades ago [27, 64] requires reassessment [26].

Thiazolidinediones. Currently, pioglitazone is the only thiazolidinedione available in the market. Rosiglitazone was withdrawn because it increased the risk of myocardial infarction, and troglitazone was withdrawn due to its hepatotoxicity [65, 66] (Table 4).

 

Table 4. Studies of genotoxic and mutagenic properties of thiazolidinediones in vitro and in vivo

International nonproprietary name

Test system

Treatment conditions

Effect biomarker

Effect

Literature source

Rosiglitazone

Sprague-Dawley rats, male

Daily per os for 14 days at doses of 0.5, 1, and 2 mg/kg

DNA damage (DNA comet assay) in peripheral blood cells and hepatocytes

Significant dose-dependent increase in the level of DNA damage in hepatocytes compared to control in all doses (p < 0.001); in lymphocytes, the effect is only in the two higher doses

[32]

Pioglitazone

Sprague-Dawley rats, male

Daily for 14 days per os at doses of 10, 20, and 40 mg/kg

DNA damage (DNA comet assay) in peripheral blood cells and hepatocytes

Significant dose-dependent increase in the level of DNA damage in lymphocytes and hepatocytes compared with control (p < 0.001)

[30]

Wistar albino rats, male, diabetes model – intraperitoneal streptozotocin administration (65 mg/kg) and nicotinamide (230 mg/kg)

Daily for 4 weeks per os at doses of 20, 40, and 80 mg/kg

Frequency of CA and MN in bone marrow cells 24 h after the last injection

Significant dose-dependent decrease in the level of both biomarkers in comparison with the diabetes group, and to the control level in the two higher doses

[67]

Culture of human lymphocytes

100 μM with/without pretreatment with vitamin B12 cell cultures (13.5 μg/ml)

Frequency of CA and SCE after 24 h of exposure

Significant increase in the level of both biomarkers (p < 0.01); pretreatment with vitamin B12 reduces the genotoxic effect

[31]

Culture of human lymphocytes

4, 12, 36, 108, 324, and 972 μM

MN level after 72 h

Significant concentration-dependent increase in MN level at concentrations above 108 µM

[68]

 

In cultures of human lymphocytes, pioglitazone at concentrations exceeding 108 μM demonstrated concentration-dependent MN induction [68]. In rat lymphocytes and hepatocytes, a significant dose-dependent increase in the level of DNA damage was observed using the DNA comet assay after daily oral administration of pioglitazone for 14 days at doses of 10, 20, and 40 mg/kg [30].

There are separate data indicating a dose-dependent MN decrease in bone marrow cells of rats after 4 weeks of daily oral administration of pioglitazone at doses of 20, 40, and 80 mg/kg in the model of streptozotocin-nicotinamide DM [67].

An unambiguous interpretation of the presented data is difficult and require further verification in future independent studies.

GLP-1 analogs and DPP-4 inhibitors. The available literature presents no primary results of studies on the genotoxic activity of glucagon-like peptide analogs. Information from the websites of regulatory agencies reports that they have no genotoxic effects (Table 5). At the same time, there are indications in the literature on the possible carcinogenic potential of drugs in this group [69, 70]. From a practical point of view, these drugs have been in the market for a relatively short period of time only, which precludes the assessment of their long-term effects in humans [71]. Therefore, the conduct of independent studies on their genotoxic activity, which enables the prediction of their carcinogenicity using short-term tests, is of primary importance [6, 72] and remains an urgent task of contemporary genotoxicology.

 

Table 5. Studies of genotoxic and mutagenic properties of GLP-1 analogs and DPP-4 inhibitors in vitro and in vivo

International nonproprietary name

Test system

Treatment conditions

Effect biomarker

Effect

Literature source

Analogs of GLP-1

Exenatide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[73]

Liraglutide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[74]

Lixisenatide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[75]

Semaglutide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[76]

Dulaglutide

Genotoxicity study data are not presented in the available literature

Dipeptidyl peptidase 4 (DPP-4) inhibitors

Sitagliptin

Pregnant mice and their embryos

0.04 mg/(kg · day) per os from the day 3 to the day 18 day of pregnancy without or in combination with metformin [0.2 mg/(kg · day)]

CA frequency in bone marrow cells of mice on the day 19 of pregnancy and in hepatocytes of embryos

Significant more than 2-fold excess of CA level in the case of bone marrow and 3-fold excess in embryonic cells; a decrease in the level of CA when administered together with metformin almost to the control values

[47]

Culture of human lymphocytes

250, 500, and 1000 μg/ml with/without metabolic activation

Frequency of CA, SCE and MN after 24 and 48 h

No effect

[77]

Culture of human lymphocytes

31.25, 62.5, 125, 250, 500, and 1000 μg/ml

DNA damage (DNA comet assay), frequency of CA, SCE and MN after 24 and 48 h

Significant increase in the frequency of CA and SCE at the highest concentration within 24 h and at all concentrations (except for 250 μg/ml for CA and 31.25 and 62.5 μg/ml for SCE) after 48 h compared with control (p < 0.05), the frequency of MN are only in the highest concentration (p < 0.05), sitagliptin increased significantly the mean intensity of the comet tail and the moment of the tail only at two concentrations (62.50 and 1000 μg/ml for intensity, 125 and 1000 μg/ml for tail moment) and tail length at all concentrations (except 125 and 500 μg/ml) (p < 0.05)

[33]

Vildagliptin

Human lymphocyte culture

125, 250, and 500 μg/ml with or without metabolic activation

Frequency of CA, SCE and MN after 24 and 48 h

No effect

[77]

Pregnant mice and their embryos

0.04 mg/(kg · day) per os from the day 3 to the day 18 of pregnancy without/in combination with metformin [0.2 mg/(kg · day)]

CA frequency in bone marrow cells of mice on the day 19 of pregnancy and in hepatocytes of embryos

Significant more than 2-fold excess of CA level in the case of bone marrow and 3-fold excess in embryonic cells; a decrease in the level of CA when administered together with metformin almost to the control values

[47]

Saxagliptin

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[78, 79]

Alogliptin

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[80]

Linagliptin

Human peripheral blood mononuclear cell culture

0.5, 1, 2.5, 5, 10, 25, 50, and 100 mg/l

CA frequency after 72 h of exposure

No effect

[81]

Gemigliptin

Genotoxicity study data not presented in the available literature

Evogliptin

Genotoxicity study data not presented in the available literature

Gosogliptin

Genotoxicity study data not presented in the available literature

 

Among DPP-4 inhibitors, only sitagliptin and vildagliptin were studied for their genotoxic and mutagenic activities. No information on these parameters is available for other drugs in the class, namely, saxagliptin, alogliptin, gemigliptin, evogliptin, and gosogliptin. Data on sitagliptin are conflicting. In one study, the drug at concentrations of 250, 500, and 1000 μg/ml did not affect the frequency of CA and MN after 24 and 48 h of in vitro treatment of human lymphocytes [77]. However, in another study, it caused a significant increase in the frequency of CA when used at a concentration of 1000 μg/ml after 24 h and at concentrations of 31.25, 62.5, 125, 500, and 1000 μg/ml (but not at a concentration of 250 μg/ml) after 48 h, and increased the frequency of MN only at the highest concentration of 1000 μg/ml [33]. In a pilot clinical study, the genotoxic and cytotoxic effects of sitagliptin in lymphocytes of T2DM patients after a 6-month course of treatment were demonstrated [82].

With regard to their mutagenic and toxic effects, an in vivo study showed that sitagliptin and vildagliptin had harmful effects on pregnant females and their foetus [47]. Reduction of this detrimental effect using metformin combinations is described above.

Although the existing data are insufficient for the drawing of precise conclusions, available results indicate the need for a systematic genotoxicological study of these groups of drugs using experimental and clinical studies.

SGLT2 inhibitors. Inhibitors of the sodium glucose cotransporter type 2 (SGLT2 inhibitors) promote the excretion of glucose in the urine in an insulin-independent manner. The drugs of this group, dapagliflozin and empagliflozin, did not induce cytogenetic changes in the cells of the peripheral blood and/or bone marrow of rats [83, 84]. However, no data on their genotoxic activities was available (Table 6).

 

Table 6. In vitro and in vivo studies of the genotoxic and mutagenic properties of sodium glucose cotransporter type 2 (SGLT2) inhibitors

International nonproprietary name

Test system

Treatment conditions

Effect biomarker

Effect

Literature source

SGLT2 inhibitors

Dapagliflozin

Sprague-Dawley rats, males and females

Daily for 1 month per os at doses of 25, 100, 150, and 200 mg/kg

CA in peripheral blood cells 24 h after the last injection

No effect

[83]

Sprague-Dawley rats, males and females

Daily for 3 days per os at doses of 350, 700, or 1050 mg/kg or for 14 days at doses of 75, 150, 200, and 250 mg/kg

Frequency of MN in bone marrow cells 24 h after the last injection

No effect

[83]

Canagliflozin

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[85]

Empagliflozin

Wistar Han rats, males and females

Daily for 3 days per os at doses of 100, 300, 1000, and 2000 mg/kg

Frequency of MN in polychromatophilic erythrocytes

No effect

[84]

Chinese hamster cell culture

5, 10, and 20 μM with or without metabolic activation

Frequency of cells with MN

No effect

[86]

Ertugliflozin

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[87]

Ipragliflozin

Genotoxicity study data are not presented in the available literature

Other hypoglycemic drugs

Rapliginide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[88]

Exenatide

According to EMA, does not exhibit genotoxic activity in in vivo and in vitro tests (primary data not presented)

[89]

 

As in the previous drug classes, it should be noted that studies of the genotoxic activity of the drugs in this group are insufficient.

Overall, our results demonstrate that current data on the genotoxicity of hypoglycemic drugs are incomplete, fragmentary, uncoordinated, and contradictory. Almost half of the known hypoglycemic drugs have not been tested adequately for genotoxicity. This observation does not differ from that in other drug groups, with more than half of which have not yet been fully investigated for genotoxicity according to the accepted protocols [64]. The issue underlying these does not seem to be the lack of genotoxicity analysis of new drugs entering the market but the outdated evaluation of already available hypoglycemic drugs. Because the pathogenesis of DM is accompanied by oxidative and coupled carbonyl stress, resulting in the formation of genotoxic products [8–10], the combination of the effects of these endogenous mutagens with the potential genotoxicity of the drug used is highly undesirable. Therefore, a systematic study on the genotoxicity of these antidiabetic drugs, in accordance with current requirements, is needed.

Another potential source of concern is the possible presence of co-mutagenic effects in hypoglycemic drugs, which has not been mandatorily tested until now [91]. Our analysis of the literature shows that this type of study has not been proactively performed in drugs of this group. Co-mutagens can significantly enhance the genotoxic effects. For example, the co-mutagenic properties of calcium channel blockers are well known [92, 93]. Further experimental (in appropriate biomodels) or clinical confirmation of the co-mutagenic activity of these drugs will make these extremely undesirable as antihypertensive drugs in patients with DM. Thus, hypoglycemic drugs used in the treatment of patients with DM should be tested for co-mutagenicity.

Regardless of whether genotoxicity in DM can be considered a consequence of pathognomonic oxidative stress or manifestations of the effects of the use of drugs with genotoxic activity, it is essential to note that most comorbid DM diseases are characterized by increases in DNA damage in their pathogenesis. This pathology has been shown in chronic degenerative heart diseases [94] and the development of atherosclerosis [95, 96], nephropathy [97], neuropathy and retinopathy [98–100], and cancer [101, 102]. Thus, it is desirable to consider the use of drugs with combined hypoglycemic and antimutagenic activity as an option for DM therapy. Moreover, this stresses the need to study the antimutagenicity of drugs already used in DM.

Analysis of the literature indicates that among hypoglycemic drugs, only metformin has been tested for antimutagenic activity. The studies performed are fragmentary, and this issue requires further in-depth analysis. For this purpose, a proven technique for studying the antimutagenicity of pharmacological agents can be used [12, 103, 104]. In addition, a number of drugs that exhibit antimutagenic effect [105] can potentially be suitable as complementary treatment interventions for DM.

Among drugs that lower glucose levels, insulin and insulin secretion stimulants notable for their potential to increase the risk of cancer. Their mechanisms of action include signaling of insulin receptors and insulin-like growth factor I (IGF-1R), which enhance cell proliferation and, possibly, carcinogenesis. In contrast, insulin sensitizers (metformin) have an anticancer effect by stimulating the regulatory pathway of AMPK, peroxisome proliferator activated gamma receptor (PPAR-γ), and the transcription factor, Egr-1 [17]. This raises the question of whether thiazolidinediones, another class of insulin sensitizers, could also have antimutagenic activity, similar to metformin. Finally, available data undoubtedly supports the use of metformin as the drug of choice among hypoglycemic agents.

CONCLUSION

Almost half of the drugs used in the treatment of patients with DM have not been studied for genotoxic activity in accordance with current methodological requirements. Studies on the mutagen-modifying activity of antidiabetic agents are sporadic and were performed beyond the previously recommended approaches [12, 104].

In most cases, the results of preclinical assessment of genotoxic potential do not provide adequate information for us to arrive at a reasonable conclusion regarding the presence or absence of genotoxic/antigenotoxic potential in drugs used to treat DM. Further studies on the genotoxic and mutagen-modifying properties of hypoglycemic drugs are required and should be performed in accordance with current approaches and standards.

×

About the authors

Natalia V. Eremina

Zakusov Research Institute of Pharmacology

Email: nnv1988@gmail.com
ORCID iD: 0000-0002-7226-5505
SPIN-code: 5224-1968

Cand. Sci. (Biol.), senior research associate

Russian Federation, 8 Baltiyskaya str., 125315, Moscow

Aliy K. Zhanataev

Zakusov Research Institute of Pharmacology

Email: zhanataev@academpharm.ru
ORCID iD: 0000-0002-7673-8672
SPIN-code: 7070-0510
Scopus Author ID: 6506103462

Cand. Sci. (Biol.), main researcher

Russian Federation, 8 Baltiyskaya str., 125315, Moscow

Artem A. Lisitsyn

Zakusov Research Institute of Pharmacology

Email: nordikal@yandex.ru
ORCID iD: 0000-0002-9597-6051
SPIN-code: 7857-1860
Scopus Author ID: 57216389600

researcher

Russian Federation, 8 Baltiyskaya str., 125315, Moscow

Andrey D. Durnev

Zakusov Research Institute of Pharmacology

Author for correspondence.
Email: addurnev@mail.ru
ORCID iD: 0000-0003-0218-8580
SPIN-code: 8426-0380
Scopus Author ID: 7006060753

Dr. Sci. (Med.), Professor, Corresponding Member of RAS

Russian Federation, 8 Baltiyskaya str., 125315, Moscow

References

  1. IDF Guide for Diabetes Epidemiology Studies [Internet] [cited 2021 May 15]. Available from: https://www.idf.org/our-activities/epidemiology-research/idf-guide-for-diabetes-epidemiology-studies.html
  2. Chen L, Magliano DJ, Zimmet PZ. The worldwide epidemiology of type 2 diabetes mellitus – present and future perspectives. Nat Rev Endocrinol. 2011;8(4):228–236. doi: 10.1038/nrendo.2011.183
  3. Habib SL, Rojna M. Diabetes and risk of cancer. ISRN Oncol. 2013;2013:583786. doi: 10.1155/2013/583786
  4. Barone BB, Yeh HC, Snyder CF, et al. Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: a systematic review and meta-analysis. JAMA. 2008;300(23):2754–2764. doi: 10.1001/jama.2008.824
  5. Bonassi S, Znaor A, Norppa H, Hagmar L. Chromosomal aberrations and risk of cancer in humans: an epidemiologic perspective. Cytogenet Genome Res. 2004;104(1–4):376–382. doi: 10.1159/000077519
  6. Bonassi S, El-Zein R, Bolognesi C, Fenech M. Micronuclei frequency in peripheral blood lymphocytes and cancer risk: evidence from human studies. Mutagenesis. 2011;26(1):93–100. doi: 10.1093/mutage/geq075
  7. Eremina NV, Zhanataev AK, Lisitsyn AA, Durnev AD. Genotoxic markers in patients with diabetes mellitus (Literature review). Ecological genetics. 2021;19(2):143–168. (In Russ.) doi: 10.17816/ecogen65073
  8. Demirbag R, Yilmaz R, Gur M, et al. DNA damage in metabolic syndrome and its association with antioxidative and oxidative measurements. Int J Clin Pract. 2006;60(10):1187–1193. doi: 10.1111/j.1742-1241.2006.01042.x
  9. Balabolkin MI. Rol’ glikirovaniya belkov, okislitel’nogo stressa v patogeneze sosudistykh oslozhnenii pri sakharnom diabete. Diabetes mellitus. 2002;(4):8–16. (In Russ.) doi: 10.14341/DM200248-16
  10. Darenskaya MA, Kolesnikova LI, Kolesnikov SI. Oxidative stress: pathogenetic role in diabetes mellitus and its complications development, therapeutic approaches to correction. Byulleten’ eksperimental’noj biologii i mediciny. 2021;171(2):136–149. (In Russ.) doi: 10.1007/s10517-021-05191-7
  11. Bigagli E, Lodovici M. Circulating Oxidative Stress Biomarkers in Clinical Studies on Type 2 Diabetes and Its Complications. Oxid Med Cell Longev. 2019;2019:5953685. doi: 10.1155/2019/5953685
  12. Durnev AD. Modifikatsiya mutatsionnogo protsessa v kletkakh cheloveka. Annals of the Russian academy of medical sciences. 2001;(10):70–76. (In Russ.)
  13. Anatomo-Terapevticheski-Khimicheskaya (ATKH) sistema klassifikatsii [Internet]. [cited 2021 May 15]. Available from: https://www.vidal.ru/drugs/atc (In Russ.)
  14. Dedov II, Shestakova MV, Mayorov AYu, et al. Standards of specialized diabetes care. Dedov II, Shestakova MV, Mayorov AYu, eds. 9th edition. Diabetes mellitus. 2019;22(1S1):1–144. (In Russ.) DOI: org/10.14341/DM221S1
  15. Othman EM, Leyh A, Stopper H. Insulin mediated DNA damage in mammalian colon cells and human lymphocytes in vitro. Mutat Res. 2013;745-746:34-9. doi: 10.1016/j.mrfmmm.2013.03.006
  16. Othman EM, Oli RG, Arias-Loza PA, et al. Metformin Protects Kidney Cells From Insulin-Mediated Genotoxicity In Vitro and in Male Zucker Diabetic Fatty Rats. Endocrinology. 2016;157(2):548–559. doi: 10.1210/en.2015-1572
  17. Tokajuk A, Krzyżanowska-Grycel E, Tokajuk A, et al. Antidiabetic drugs and risk of cancer. Pharmacol Rep. 2015;67(6):1240–1250. doi: 10.1016/j.pharep.2015.05.005
  18. Othman EM, Altabaa T, Hintzsche H, Stopper H. IR and IGF-1R expression affects insulin induced proliferation and DNA damage. Toxicol In Vitro. 2017;39:68–74. doi: 10.1016/J.TIV.2016.11.011
  19. HUMALOG® Product Monograph [Internet]. [cited 2021 May 15] Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwjclZyw_KPvAhVGxIsKHZvlBE4QFjAOegQIBBAD&url=https%3A%2F%2Fpdf.hres.ca%2Fdpd_pm%2F00003299.PDF&usg=AOvVaw3qP_2gNpNQ3fN1X-CspEXC
  20. Center for drug evaluation and research. Insulin Aspart [rDNA Origin] Injection [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiEvp7egaTvAhVD_SoKHTjvCvkQFjACegQIBRAD&url=https%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Fnda%2F2013 %2F020986Orig1s061.pdf&usg=AOvVaw3DAicbE-EHyrHrRmiEjIU6
  21. NovoRapid Approval Scientific discussion [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiEvp7egaTvAhVD_SoKHTjvCvkQFjAAegQIARAD&url=https%3A%2F%2Fwww.ema.europa.eu%2Fen%2Fdocuments%2Fscientific-discussion%2Fnovorapid-epar-scientific-discussion_en.pdf&usg=AOvVaw38mmTxmMGBpGUYcoSzhqTD
  22. Apidra Approval Scientific discussion [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiNzp6Ig6TvAhVC2SoKHRI8BzkQFjAAegQIARAD&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fscientific-discussion%2Fapidra-epar-scientific-discussion_en.pdf&usg=AOvVaw3viOO4MIxkPFOeYV0rt8Pc
  23. Center for drug evaluation and research. Application number 21–629. Pharmacology review [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiNzp6Ig6TvAhVC2SoKHRI8BzkQFjABegQIFhAD&url=https%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Fnda%2F2004 %2F21-629_Apidra_Pharmr_P1.pdf&usg=AOvVaw0Xh-xiYWgUJMk6jWLchV2g
  24. Center for drug evaluation and research. Application number 21–536. Pharmacology review(s) [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwisgKqVhaTvAhUhlosKHeJDDgwQFjACegQIExAD&url=http%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Fnda%2F2005 %2F021-536_Levemir_pharmr.pdf&usg=AOvVaw3rpOhO3TifQYrMyRFaC_xy
  25. Flevemir Approval Scientific discussion [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&cad=rja&uact=8&ved=2ahUKEwisgKqVhaTvAhUhlosKHeJDDgwQFjAAegQIARAD&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fscientific-discussion%2Flevemir-epar-scientific-discussion_en.pdf&usg=AOvVaw2TiEsZCAKiZRdTFlfmzrdo
  26. Mironov AN. Rukovodstvo po provedeniyu doklinicheskikh issledovanii lekarstvennykh sredstv. Chast’. 1. Moscow: Grif i K, 2012. 944 p. (In Russ.)
  27. Renner HW, Münzner R. Mutagenicity of sulphonylureas. Mutat Res. 1980;77(4):349–355. doi: 10.1016/0165-1218(80)90007-5
  28. Kar RN, Mukherjee B, Mukherjee SK. Mutagenic evaluation of tolbutamide and glybenclamide on the bone marrow cells of mice. Toxicol Lett. 1986;34(2–3):153–157. doi: 10.1016/0378-4274(86)90205-5
  29. Sekena H, El-Aziem A, Hassan MA. Genetic and Ultrastructural studies in bone marrow and testis of mice cells under the effect of Glurenor drug. The Egyptian Journal of Hospital Medicine. 2003;12(1):62–71. doi: 10.21608/ejhm.2003.18246
  30. Bedir A, Aliyazicioglu Y, Bilgici B, et al. Assessment of genotoxicity in rats treated with the antidiabetic agent, pioglitazone. Environ Mol Mutagen. 2008;49(3):185–191. doi: 10.1002/em.20365
  31. Alzoubi K, Khabour O, Hussain N, et al. Evaluation of vitamin B12 effects on DNA damage induced by pioglitazone. Mutat Res. 2012;748(1–2):48–51. doi: 10.1016/j.mrgentox.2012.06.009
  32. Bedir A, Aliyazicioglu Y, Kahraman H, et al. Genotoxicity in rats treated with the antidiabetic agent, rosiglitazone. Environ Mol Mutagen. 2006;47(9):718–724. doi: 10.1002/em.20261
  33. Yuzbasioglu D, Enguzel-Alperen C, Unal F. Investigation of in vitro genotoxic effects of an anti-diabetic drug sitagliptin. Food Chem Toxicol. 2018;112:235–241. doi: 10.1016/j.fct.2018.01.003
  34. Nasri H, Rafieian-Kopaei M. Metformin: Current knowledge. J Res Med Sci. 2014;19(7):658–664. doi: 10.12659/MSMBR.889344
  35. Kalashnikova MF, Belousov DY, Suntsov YI, et al. Pharmacoepidemiological and pharmacoeconomic analyses of the utilization of hypoglycaemic drugs in patients with type 2 diabetes mellitus in Moscow. Diabetes mellitus. 2015;18(2):32–46. (In Russ.) doi: 10.14341/DM2015232-46
  36. Marshall SM. 60 years of metformin use: a glance at the past and a look to the future. Diabetologia. 2017;60(9):1561–1565. doi: 10.1007/s00125-017-4343-y
  37. Attia SM, Helal GK, Alhaider AA. Assessment of genomic instability in normal and diabetic rats treated with metformin. Chem Biol Interact. 2009;180(2):296–304. doi: 10.1016/j.cbi.2009.03.001
  38. Amador RR, Longo JP, Lacava ZG, et al. Metformin (dimethyl-biguanide) induced DNA damage in mammalian cells. Genet Mol Biol. 2012;35(1):153–158. doi: 10.1590/s1415-47572011005000060
  39. Malek HA, Hassanin A, Aziz HA, Dahtory FE. In vitro assessment of the mutagenic effect of Metformin. J Chem Pharm Res. 2015;7(6):879–886.
  40. Sant’Anna JR, Yajima JP, Rosada LJ, et al. Metformin’s performance in in vitro and in vivo genetic toxicology studies. Exp Biol Med (Maywood). 2013;238(7):803–810. doi: 10.1177/1535370213480744
  41. Rabbani SI, Devi K, Khanam S. Role of Pioglitazone with Metformin or Glimepiride on Oxidative Stress-induced Nuclear Damage and Reproductive Toxicity in Diabetic Rats. Malays J Med Sci. 2010;17(1):3–11.
  42. OECD. Test No. 489: In Vivo Mammalian Alkaline Comet Assay. OECD Guidelines for the Testing of Chemicals, Section 4. OECD Publishing, Paris. 2016. https://doi.org/10.1787/9789264264885-en
  43. Najafi M, Cheki M, Rezapoor S, et al. Metformin: Prevention of genomic instability and cancer: A review. Mutat Res Genet Toxicol Environ Mutagen. 2018;827:1–8. doi: 10.1016/j.mrgentox.2018.01.007
  44. Aleisa AM, Al-Rejaie SS, Bakheet SA, et al. Effect of metformin on clastogenic and biochemical changes induced by adriamycin in Swiss albino mice. Mutat Res. 2007;634(1–2):93–100. doi: 10.1016/j.mrgentox.2007.06.005
  45. Cheki M, Shirazi A, Mahmoudzadeh A, et al. The radioprotective effect of metformin against cytotoxicity and genotoxicity induced by ionizing radiation in cultured human blood lymphocytes. Mutat Res. 2016;809:24–32. doi: 10.1016/j.mrgentox.2016.09.001
  46. Kanigür-Sultuybek G, Ozdas SB, Curgunlu A, et al. Does metformin prevent short-term oxidant-induced dna damage? In vitro study on lymphocytes from aged subjects. J Basic Clin Physiol Pharmacol. 2007;18(2):129–140. doi: 10.1515/jbcpp.2007.18.2.129
  47. Roshdy HM, Kassem SM. Genetic effects of Januvia and Galvus alone or with metformin on pregnant female mice and their embryos. World Applied Sciences Journal. 2013;25:1690–1698. doi: 10.5829/idosi.wasj.2013.25.12.76142
  48. Esteghamati A, Eskandari D, Mirmiranpour H, et al. Effects of metformin on markers of oxidative stress and antioxidant reserve in patients with newly diagnosed type 2 diabetes: a randomized clinical trial. Clin Nutr. 2013;32(2):179–185. doi: 10.1016/j.clnu.2012.08.006
  49. Dogan Turacli I, Candar T, Yuksel EB, et al. Potential effects of metformin in DNA BER system based on oxidative status in type 2 diabetes. Biochimie. 2018;154:62–68. doi: 10.1016/j.biochi.2018.08.002
  50. Safe S, Nair V, Karki K. Metformin-induced anticancer activities: recent insights. Biol Chem. 2018;399(4):321–335. doi: 10.1515/hsz-2017-0271
  51. Anisimov VN. Do metformin a real anticarcinogen? A critical reappraisal of experimental data. Ann Transl Med. 2014;2(6):60. doi: 10.3978/j.issn.2305-5839.2014.06.02
  52. Noto H, Goto A, Tsujimoto T, Noda M. Cancer risk in diabetic patients treated with metformin: a systematic review and meta-analysis. PLoS One. 2012;7(3): e33411. doi: 10.1371/journal.pone.0033411
  53. Zaidi S, Gandhi J, Joshi G, et al. The anticancer potential of metformin on prostate cancer. Prostate Cancer Prostatic Dis. 2019;22(3):351–361. doi: 10.1038/s41391-018-0085-2
  54. Tang GH, Satkunam M, Pond GR, et al. Association of Metformin with Breast Cancer Incidence and Mortality in Patients with Type II Diabetes: A GRADE-Assessed Systematic Review and Meta-analysis. Cancer Epidemiol Biomarkers Prev. 2018;27(6):627–635. doi: 10.1158/1055-9965.EPI-17-0936
  55. Yao L, Liu M, Huang Y, et al. Metformin Use and Lung Cancer Risk in Diabetic Patients: A Systematic Review and Meta-Analysis. Dis Markers. 2019;2019:6230162. doi: 10.1155/2019/6230162
  56. Heckman-Stoddard BM, DeCensi A, Sahasrabuddhe VV, Ford LG. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia. 2017;60(9):1639–1647. doi: 10.1007/s00125-017-4372-6
  57. Podhorecka M, Ibanez B, Dmoszyńska A. Metformin – its potential anti-cancer and anti-aging effects. Postepy Hig Med Dosw (Online). 2017;71:170–175. doi: 10.5604/01.3001.0010.3801
  58. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577–1585. doi: 10.1007/s00125-017-4342-z
  59. Tseng CH. Thyroid cancer risk is not increased in diabetic patients. PLoS One. 2012;7(12): e53096. doi: 10.1371/journal.pone.0053096
  60. de Sant’Anna JR, Franco CC, Mathias PC, de Castro-Prado MA. Assessment of in vivo and in vitro genotoxicity of glibenclamide in eukaryotic cells. PLoS One. 2015;10(3): e0120675. doi: 10.1371/journal.pone.0120675
  61. Sarkar A, Tiwari A, Bhasin PS, Mitra M. Pharmacological and Pharmaceutical Profile of Gliclazide: A Review. J Appl Pharm Sci. 2011;1(9):11–19.
  62. Pouri M, Shaghaghi Z, Ghasemi A, Hosseinimehr SJ. Radioprotective Effect of Gliclazide as an Anti-Hyperglycemic Agent Against Genotoxicity Induced by Ionizing Radiation on Human Lymphocytes. Cardiovasc Hematol Agents Med Chem. 2019;17(1):40–46. doi: 10.2174/1871525717666190524092918
  63. Rabbani SI, Devi K, Khanam S. Inhibitory effect of glimepiride on nicotinamide-streptozotocin induced nuclear damages and sperm abnormality in diabetic Wistar rats. Indian J Exp Biol. 2009;47(10):804–810.
  64. Brambilla G, Martelli A. Update on genotoxicity and carcinogenicity testing of 472 marketed pharmaceuticals. Mutat Res. 2009;681(2–3):209–229. doi: 10.1016/j.mrrev.2008.09.002
  65. Smith MT. Mechanisms of troglitazone hepatotoxicity. Chem Res Toxicol. 2003;16(6):679–687. doi: 10.1021/tx034033e
  66. Nathan DM. Rosiglitazone and cardiotoxicity — weighing the evidence. N Engl J Med. 2007;357(1):64–66. doi: 10.1056/NEJMe078117
  67. Amein KA, Hamdy MM, Abd El-Emam RA, Osman FH. The effect of pioglitagone on genomic instability in induced diabetic rats. Research Journal of Applied Biotechnology. 2016; Special volume for the first International Conference of Genetic Engineering and Biotechnology:68–80. doi: 10.21608/rjab.2016.59636
  68. Morais JF, Sant’Anna JR, Pereira TS, et al. Genotoxic investigation of a thiazolidinedione PPARγ agonist using the in vitro micronucleus test and the in vivo homozygotization assay. Mutagenesis. 2016;31(4):417–424. doi: 10.1093/mutage/gew003
  69. Drab SR. Glucagon-Like Peptide-1 Receptor Agonists for Type 2 Diabetes: A Clinical Update of Safety and Efficacy. Curr Diabetes Rev. 2016;12(4):403–413. doi: 10.2174/1573399812666151223093841
  70. Guo X, Yang Q, Dong J, et al. Tumour Risk with Once-Weekly Glucagon-Like Peptide-1 Receptor Agonists in Type 2 Diabetes Mellitus Patients: A Systematic Review. Clin Drug Investig. 2016;36(6):433–441. doi: 10.1007/s40261-016-0389-8
  71. Alves C, Batel-Marques F, Macedo AF. A meta-analysis of serious adverse events reported with exenatide and liraglutide: acute pancreatitis and cancer. Diabetes Res Clin Pract. 2012;98(2):271–284. doi: 10.1016/j.diabres.2012.09.008
  72. Azqueta A, Slyskova J, Langie SA, et al. Comet assay to measure DNA repair: approach and applications. Front Genet. 2014;5:288. doi: 10.3389/fgene.2014.00288
  73. Byetta Approval Scientific discussion [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=2&cad=rja&uact=8&ved=2ahUKEwiK1frvgcPpAhVBAhAIHeXBDMkQFjABegQIAxAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fscientific-discussion%2Fbyetta-epar-scientific-discussion_en.pdf&usg=AOvVaw2BukkG_ld6RLWhosmSMDe-
  74. Assessment report for Victoza [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwji-86flqzrAhUBAxAIHWEwCaYQFjAAegQIBBAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fen%2Fema-redirect%3Fredirect_type%3Djsp%26webContentId%3DWC500050016&usg=AOvVaw3d6IbqsAoquGGnt9M_yV4_
  75. Assessment report Lyxumia [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwitsaygl6zrAhWjxIsKHdq-DI8QFjAAegQIBBAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fassessment-report%2Flyxumia-epar-public-assessment-report_en.pdf&usg=AOvVaw35wDuXxjyiD-0fL2paBAZv
  76. Assessment report Ozempic [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&cad=rja&uact=8&ved=2ahUKEwjImuuUmKzrAhVp-yoKHTU9Bg4QFjAKegQIARAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fassessment-report%2Fozempic-epar-public-assessment-report_en.pdf&usg=AOvVaw1zFwSfvLP0ycmHePzsnI2u
  77. Börçek Kasurka C, Elbistan M, Atmaca A, Atlı Şekeroğlu Z. In vitro cytogenetic assessment and comparison of vildagliptin and sitagliptin. Cytotechnology. 2019;71(6):1063–1077. doi: 10.1007/s10616-019-00345-y
  78. Annex I. Onglyza 2.5 and 5 mg film-coated tablets. Summary of product characteristics [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwi0iLvBgoPzAhWPnYsKHb80CV4QFnoECAIQAQ&url=https%3A%2F%2Fwww.ema.europa.eu%2Fen%2Fdocuments%2Fproduct-information%2Fonglyza-epar-product-information_en.pdf&usg=AOvVaw1JdORt5TzuaemMnLSTnSOP
  79. Bristol-Myers Squibb Company. ONGLYZA™ (saxagliptin) tablets. US prescribing information. 2011.
  80. Annex I. Vipidia 6.25, 12.5 and 25 mg film-coated tablets. Summary of product characteristics [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&cad=rja&uact=8&ved=2ahUKEwjk377SgoPzAhXrsIsKHWpWCK8QFnoECAIQAQ&url=https%3A%2F%2Fwww.ema.europa.eu%2Fen%2Fdocuments%2Fproduct-information%2Fvipidia-epar-product-information_en.pdf&usg=AOvVaw2lKgOuaut8a473M-MAQeL8
  81. Çadirci K, Türkez H, Özdemir Ö. The in vitro cytotoxicity, genotoxicity and oxidative damage potential of the oral dipeptidyl peptidase-4 inhibitor, linagliptin, on cultured human mononuclear blood cells. Acta Endocrinol (Buchar). 2019;5(1):9–15. doi: 10.4183/aeb.2019.9
  82. Oz Gul O, Cinkilic N, Gul CB, et al. Comparative genotoxic and cytotoxic effects of the oral antidiabetic drugs sitagliptin, rosiglitazone, and pioglitazone in patients with type-2 diabetes: a cross-sectional, observational pilot study. Mutat Res. 2013;757(1):31–35. doi: 10.1016/j.mrgentox.2013.04.024
  83. Reilly TP, Graziano MJ, Janovitz EB, et al. Carcinogenicity risk assessment supports the chronic safety of dapagliflozin, an inhibitor of sodium-glucose co-transporter 2, in the treatment of type 2 diabetes mellitus. Diabetes Ther. 2014;5(1):73–96. doi: 10.1007/s13300-014-0053-3
  84. Bogdanffy MS, Stachlewitz RF, van Tongeren S, et al. Nonclinical safety of the sodium-glucose cotransporter 2 inhibitor empagliflozin. Int J Toxicol. 2014;33(6):436–449. doi: 10.1177/1091581814551648
  85. Assessment report Canagliflozin [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiWj_bZpqzrAhVukIsKHcXYAngQFjABegQIARAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fassessment-report%2Finvokana-epar-public-assessment-report_en.pdf&usg=AOvVaw1VbOa--peFCGjDAZ8xdrcQ
  86. Smith JD, Huang Z, Escobar PA, et al. A Predominant Oxidative Renal Metabolite of Empagliflozin in Male Mice Is Cytotoxic in Mouse Renal Tubular Cells but not Genotoxic. Int J Toxicol. 2017;36(6):440–448. doi: 10.1177/1091581817735090
  87. Assessment report Steglatro [Internet]. Available from: https://www.ema.europa.eu/en/ema-redirect?redirect_type=document&lang=en&doc_id=WC500246920%20&doc_ext=pdf
  88. Annex I. Repaglinide Accord 0.5 mg tablets. Summary of product characteristics [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&cad=rja&uact=8&ved=2ahUKEwjCs5vCpKzrAhXv-yoKHRkoA30QFjAKegQIAxAB&url=https%3A%2F%2Fwww.ema.europa.eu%2Fen%2Fdocuments%2Fproduct-information%2Frepaglinide-accord-epar-product-information_en.pdf&usg=AOvVaw2KUEtCsjKl5piGAJuOkYMJ
  89. CHMP assessment report Bydureon [Internet]. Available from: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwig06WcgoPzAhXFlYsKHcq9CtUQFnoECBYQAQ&url=https%3A%2F%2Fwww.ema.europa.eu%2Fdocuments%2Fassessment-report%2Fbydureon-epar-public-assessment-report_en.pdf&usg=AOvVaw1OllVdjrn7ox25ZCCzJ_BZ
  90. European Medicines Agеncy. ICH S2 (R1) Genotoxicity testing and data interpretation for pharmaceuticals intended for human use — Step 5. 2013. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-guideline-s2-r1-genotoxicity-testing-data-interpretation-pharmaceuticals-intended-human-use-step_en.pdf
  91. Durnev AD, Seredenin SB. Co-mutagenesis as new vistas in genotoxicology. Byulleten’ eksperimental’noj biologii i mediciny. 2003;135(6):604–612. (In Russ.) doi: 10.1023/A:1025410612571
  92. Durnev AD, Daugel’-Dauge NO, Seredenin SB. Comutagen interaction of verapamil and ribavirin. Experimental and clinical pharmacology. 2006;69(1):56–59. (In Russ.)
  93. Durnev AD, Zhanataev AK, Voronina ES, et al. Modification of Chemical Mutagenesis. In: Genotoxicity: Evaluation, Testing and Prediction. Andor K., Molnar H., editors. NY: Nova Science Publishers, 2009. 157–187 p.
  94. Corrêa CR, Garcia JL. DNA Damage in Chronic Heart Failure: Consequences Beyond those in the Heart. Arq Bras Cardiol. 2020;114(2):243–244. doi: 10.36660/abc.20190884
  95. Cervelli T, Borghini A, Galli A, Andreassi MG. DNA damage and repair in atherosclerosis: current insights and future perspectives. Int J Mol Sci. 2012;13(12):16929–16944. doi: 10.3390/ijms131216929
  96. Mahmoudi M, Mercer J, Bennett M. DNA damage and repair in atherosclerosis. Cardiovasc Res. 2006;71(2):259–268. doi: 10.1016/j.cardiores.2006.03.002
  97. Schupp N, Stopper H, Heidland A. DNA Damage in Chronic Kidney Disease: Evaluation of Clinical Biomarkers. Oxid Med Cell Longev. 2016;2016:3592042. doi: 10.1155/2016/3592042
  98. Mishra M, Lillvis J, Seyoum B, Kowluru RA. Peripheral Blood Mitochondrial DNA Damage as a Potential Noninvasive Biomarker of Diabetic Retinopathy. Invest Ophthalmol Vis Sci. 2016;57(10):4035–4044. doi: 10.1167/iovs.16-19073
  99. Madsen-Bouterse SA, Mohammad G, Kanwar M, Kowluru RA. Role of mitochondrial DNA damage in the development of diabetic retinopathy, and the metabolic memory phenomenon associated with its progression. Antioxid Redox Signal. 2010;13(6):797–805. doi: 10.1089/ars.2009.2932
  100. Singh P, Jain A, Kaur G. Impact of hypoglycemia and diabetes on CNS: correlation of mitochondrial oxidative stress with DNA damage. Mol Cell Biochem. 2004;260(1–2):153–159. doi: 10.1023/b: mcbi.0000026067.08356.13
  101. Jackson AL, Loeb LA. The contribution of endogenous sources of DNA damage to the multiple mutations in cancer. Mutat Res. 2001;477(1–2):7–21. doi: 10.1016/s0027-5107(01)00091-4
  102. Dizdaroglu M. Oxidatively induced DNA damage and its repair in cancer. Mutat Res Rev Mutat Res. 2015;763:212–245. doi: 10.1016/j.mrrev.2014.11.002
  103. Verhagen H, Aruoma OI, van Delft JH, et al. The 10 basic requirements for a scientific paper reporting antioxidant, antimutagenic or anticarcinogenic potential of test substances in in vitro experiments and animal studies in vivo. Food Chem Toxicol. 2003;41(5):603–610. doi: 10.1016/s0278-6915(03)00025-5
  104. Durnev AD. Methodological aspects of studies of chemical mutagenesis modification. Byulleten’ eksperimental’noj biologii i mediciny. 2008;146(9):281–287. (In Russ.) doi: 10.1007/s10517-008-0273-5
  105. Durnev AD. Antimutagenesis and antimutagens. Human Physiology. 2018;44(3):116–137. (In Russ.) doi: 10.7868/S013116461803013

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2021 ООО "Эко-Вектор"



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 65617 от 04.05.2016.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies