Abstract
2-Deoxy-D-Glucose (2-DG) has anticonvulsant and antiseizure effects in rodent models and is in the development phase for novel antiseizure treatment. To evaluate potential toxicity, Beagle dogs (five/sex/group) were orally gavaged with either vehicle (deionized water) or 2-DG (5, 30, and 90 mg/kg BID) for 28 days followed by a 14-day recovery period. The safety endpoints evaluated were mortality, clinical observations, body temperature, respiratory assessment, body weights, food consumption, ophthalmic examinations, electrocardiograph (ECG), blood pressure, cardiac biomarker (NT-proBNP) and pathology. Toxicokinetic analysis was conducted after the first dose on days 1 and 28. The dose formulation analysis confirmed that 2-DG concentrations were within 93–107% of the target concentrations. There were no 2-DG associated effects observed in mortality, clinical signs, body temperature, respiratory parameters, body weights, food consumption, ophthalmic examination, ECG, blood pressure and NT-proBNP. There was an increase (~1.7x) in aspartate transaminase on day 29, while histopathological evaluation revealed hepatic cytoplasmic alterations in 2 of 6 dogs on day 29 and only in 1 of 4 dogs on day 43 at 90 mg/kg BID. These changes were considered non-adverse because of minimal severity, reversibility trend after recovery period and no correlative increase in alanine transaminase. Toxicokinetic evaluation revealed dose dependent increases in Cmax of ~7.8, 39.5, and 114 μg/mL, and AUCs of 12.2, 70.8, and 202 hr*μg/mL at 5, 30, and 90 mg/kg, respectively with Tmax of ~0.5–0.9 hr and T1/2 of ~3.8–5.4 hr. In conclusion, 90 mg/kg BID of 2-DG was considered as the No Observed Adverse Effect Level following 28-day administration in dogs.
Keywords: 2-Deoxy-D-Glucose, Non-clinical Safety, No Observed Adverse Effect Level
1. Introduction
There is a compelling need for new, more effective treatments for epilepsy, which affects 0.5–1% of the population1. About one-third of patients treated for epilepsy with standard-of-care medications continue to experience frequent seizures and are regarded as intractable2. The glucose analog 2-Deoxy-D-Glucose (2-DG) is a reversible glycolytic inhibitor and has anticonvulsant and antiepileptic properties in seizure animal models3. During seizures, a higher metabolic demand in neural circuits increases 2-DG uptake into brain. Subsequently, 2-DG is phosphorylated to 2-DG-6P. However, it is unable to get metabolized by glucose-6P isomerase (GPI) that inhibits further step of glycolysis4. The antiseizure action of glycolytic inhibition is novel compared to all current antiseizure medications2 and supports the potential use of 2-DG as a novel therapeutic intervention for convulsion and epilepsy.
Minor et al5 conducted dietary toxicity studies of synthetic 2-DG up to 2 year of dosing duration at doses ranging from 20 to 300 mg/kg (0.04%−0.6% 2-DG in the diet) in F344 and brown Norway rats, that identified cardiac toxicity. Minor et al5 also showed 2-DG associated mortality, pheochromocytoma in the adrenal medulla, reduction in food intake and body weight gain in male F344 rats. Furthermore, Terse et al6 showed that 2-DG induced cardiac toxicity (myocardial vacuolar degeneration) was associated with elevated levels of NT-proBNP and BNP (NT-proBNP being more sensitive) in F344 rats. However, it might be possible that cardiac toxicity could be limited to rats, which have increased metabolic demand. In the cardiomyocytes, the abnormal glucose metabolism is associated with pathological cardiac hypertrophy7.
To evaluate 2-DG in a clinical setting, it is required to provide toxicity results from rodent and non-rodent animal models8. As a non-rodent species, we conducted toxicity and toxicokinetic study of 2-DG gavaged daily for 28 days followed by a 14-day recovery period in Beagle dogs to support clinical trials for the treatment of convulsion and epilepsy. Furthermore, this study also determined species concordance for cardiac toxicity in rats to large animal species, Beagle dogs since larger animal findings are generally more translatable to humans. As a requirement of investigational new drug (IND) application, it was conducted following the guidance from the United States FDA Goold Laboratory Practice (GLP) (Code of Federal Regulations, Title 21 Part 58).
2. Materials and Methods
2.1. Formulation preparation
2-DG with purity 98.47% (Lot # SAI1206A01) was manufactured at Norac Pharma, Azusa, CA. In each week of the dosing period, 2-DG formulations (5, 30, and 90 mg/mL) were made by mixing 2-DG into deionized water. These formulations were protected from light. The formulation analyses were performed using validation method. Dose formulation concentrations were analyzed using high performance liquid chromatography with ultraviolet (HPLC/UV). The stability was also determined for 5 and 90 mg/mL dose formulation concentrations to establish stability for 15 days.
2.2. Test system
Forty Beagle dogs (20 male, 9.5–13.3kg; 20 female, 6.9–8.7 kg) with an age range of 9 to 11 months were received from Covance Research Products, Inc (Cumberland, VA). The dogs were fed Purina Lab Diet 5007 once daily, a freshwater supply was provided ad libitum. This study procedure followed applicable animal welfare acts and was approved by the Institutional Animal Care and Use Committee (IACUC). This study was conducted in compliance with the US FDA GLP Regulations (Code of Federal Regulations Title 21 Part 58).
2.3. Group assignment and dose administration
Prior to day 1, dogs were assigned to groups using the computer program, Provantis (INSTEM, Staffordshire, UK), which ensures similar group mean body weights by sex. All dogs were orally gavaged twice daily with either vehicle control (deionized water) or 2-DG. BID dosing was used because of the short half-life (0.6–5.4h) of 2-DG, and to mimic clinical BID dosing regimen. Group 1 dogs received vehicle control only, while groups 2, 3 and 4 received 2-DG formulations at doses 5, 30, and 90 mg/kg BID, respectively, for 28 days followed by a 14-day recovery (Table 1). The doses for this study were selected based on the 14-day dose range finding (DRF) study in the dogs where 100 mg/kg BID was a well-tolerated-dose, and 250 mg/kg BID was maximum tolerated dose (MTD) based on body weight loss and clinical signs. Daily two doses (BID) were administered at a target of 6 hours ± 10 minutes apart except on toxicokinetics (TK) blood collection days 1 and 28 (the second dose was administered after the 12-hour blood collection). On Days 1 and 28, the 12-h interval was used to obtain a full TK profile, whereas, on the remaining days, the 6-h interval was used to avoid dosing during nighttime for practical reasons.
Table 1.
Study design.
| Groupa | Target single 2-DG concentration (mg/mL) | Target total daily 2-DG dose (mg/kg) BID | Number of dogs | |
|---|---|---|---|---|
| Corec (M+F) | Recoveryd (M+F) | |||
| 1-Vehicle Controlb | 0 | 0 | 6 (3+3) | 4 (2+2) |
| 2-Low Dose | 5 | 5 | 6 (3+3) | 4 (2+2) |
| 3-Mid Dose | 30 | 30 | 6 (3+3) | 4 (2+2) |
| 4-High Dose | 90 | 90 | 6 (3+3) | 4 (2+2) |
All groups were dosed for 28 days. Blood samples were also collected from study dogs for TK analysis on day 1 and day 28 following the first dose administration.
Vehicle control was deionized water.
Core male (M) and female (F) dogs were humanely sacrificed on day 29 for necropsy.
Recovery male (M) and female (F) dogs were humanely sacrificed on day 43 for necropsy.
2.4. Clinical observation, body weights and food consumption
For all animals, the cage-side clinical observations were conducted at least twice daily for the duration of treatment and once daily during the recovery period. In each group, individual animal body weights were recorded weekly from day 1 onwards. Food consumption for all animals was recorded daily for the duration of the study.
2.5. Body temperature, respiratory measurements and ophthalmic examinations
For all animals, body temperatures were measured using a thermometer on day −3 (baseline), and days 1 and 28 at a target of 1h ± 15 min after the second dose administration. Manual respiratory rates were collected as a part of clinical observation once from all animals on day 23 or 24 at a target of 1h ± 15 min after the first dose administration. Ophthalmic examinations were performed on all animals prior to group assignment (pre-study) and on day 26 or 27. The pupils of the animals were dilated by the instillation of Tropicamide ophthalmic solution (1%) into each eye before the examination. Each ophthalmic examination included an examination of the adnexal structures (conjunctiva, eyelids, and eyelashes), the anterior segment of the eye (cornea, sclera, iris, pupil, lens, aqueous humor, and anterior chamber), and the posterior segment of the eye (the vitreous humor, retina, and optic disc). A Zeiss hand slit lamp HSO 10 was utilized for all direct ophthalmic examinations, while a Keeler all-pupil indirect ophthalmoscope with a volk 30 diopter double aspheric lens was utilized for all indirect ophthalmic examinations.
2.6. Blood pressure, ECG, and cardiac biomarker evaluations
Blood pressure and ECG tracings were examined once for all study animals on day 23 at a target of 1h ± 15 min after the first dose administration. Blood pressures were determined non-invasively by a Cardell vital signs monitor. ECG waveform tracings of leads I, II, III, aVR, aVL, aVF, and V10 were collected by a Hewlett Packard PageWriter Xli Cardiograph for morphologic evaluation. Leads I, aVf, and V10 were collected for rhythm evaluation. ECG intervals (RR, PR, QRS, and QT) were measured on the tracings. QTc intervals were calculated according to Fridericia’s formula9: QTc = QT/(RR)1/3.
For cardiac biomarker evaluation, blood samples were collected on day −1 (baseline) and prior to dose administration on Days 2, 15, 29, and 43 for the plasma analysis of NT-proBNP (N-Terminal pro-B-type Natriuretic Peptide). NT-proBNP has 76 amino acids, which is a part of prehormone (proBNP)10. NT-proBNP is the gold standard biomarker for heart failure in humans11. The enzyme-linked immunosorbent assay (ELISA) was conducted with the commercially available kit (Kamiya biomedical company; Catalog # KT-23770), and plates were read by the μQuant Plate Reader. The data were then transferred to Softmax Pro 5.2, Rev C for the generation of reference curves and sample analysis.
2.7. Toxicokinetics (TK)
Blood was collected from the jugular vein for the 2-DG plasma concentration analysis on day 1 and day 28 following the first dose administration of the BID dosing regimen. TK specimen collections were at pre-dose (0min) and post-dose (10min, 20min, and 1, 2, 4, 8 and 12h). The second dose of the BID dosing regimen was administered after 12h sample collection. After centrifugation, plasma samples were separated, processed by protein precipitation, and analyzed with a validation method of liquid chromatography with tandem mass spectrometry (LC-MS). 2-DG concentrations were calculated using peak area ratio [analyte peak area/internal standard (IS) peak area] and a regression line constructed from the concentrations and peak area ratios of the calibration standards. TK analysis was performed using the non-compartmental analysis (NCA) module in the WinNonlin software package (version 6.3, Pharsight corporation, Mountain View, CA). The evaluated TK parameters were Cmax, Tmax, elimination half-life (T1/2), apparent clearance, and AUC (to AUClast and AUC∞).
2.8. Clinical pathology
Blood was collected from the jugular vein of all animals to assess hematology, coagulation, and serum chemistry on Days −1 (baseline), 2, 15, 29, and 43 (non-fasted). To evaluate urine parameters, urine was also collected via free catch or from the cage pan on days 28 and 29.
2.9. Necropsy
Dogs (three/sex/group) were weighed and humanely sacrificed before necropsy on day 29. Following a 14-day recovery period, the remaining dogs (two/sex/group) were similarly necropsied on day 43. Organ weights were weighed, and absolute weight, organ to body weight, and organ to brain weight ratio were estimated.
All tissues were collected and preserved in 10% neutral-buffered formalin (NBF), with the exceptions of testes, which were placed in Bouin’s fixative and subsequently transferred to 70% ethanol. The eyes with optic nerve were fixed in Davidson’s fixative and subsequently transferred to 10% NBF. Bone marrow smears were fixed in methanol. All fixed tissues from control and high-dose groups (day 29) and gross lesions from all groups were processed to slides with staining of hematoxylin and eosin. Liver tissues from day 29 (low and mid-dose groups) and day 43 (all groups) necropsied animals were also collected for histopathology. Microscopic findings in histopathology were graded as per the following scale: minimal (Grade 1), mild (Grade 2), moderate (Grade 3) and marked (Grade 4). These grades were used to calculate the average severity of microscopic findings (Table 3).
Table 3.
Stability on day 0 and day 15.
| Prepared formulation concentration (mg/mL) | Determined concentration on day 0 (mg/mL) | Average determined concentration on day 15 (mg/mL) | Average RE* of Day 0 |
|---|---|---|---|
| 5 | 5.07 | 4.88 ± 0.02 | −3.7 |
| 90 | 87.8 | 87.2 ± 0.6 | −0.7 |
RE: Relative Error
2.10. Statistical analysis
To evaluate the 2-DG associated effect, the parametric or nonparametric analysis of variance (ANOVA) was used to analyze quantitative in-life data, which was collected using the Provantis system. To determine the significant differences among the group means, the ANOVA F-test was used for the analysis of parametric data, which was normally distributed and homogenous among groups. If this test was significant, then Dunnett’s test was used to determine differences between the control and each of the comparison groups. The Shapiro-Wilk’s test and Levene’s test determined normality and homogeneity of variances, respectively, for all data. Kruskal-Wallis test was used to determine the significant differences among the group means for normally distributed and/or non-homogenous nonparametric data. If this test was significant, then the Wilcoxon test was used to determine differences between the control and each of the comparison groups, while the Bonferroni-Holm method was used to correct for multiple comparisons.
3. Results
3.1. Dose formulation analysis and stability
As per Table 2, the formulation concentrations were within 92.7 to 107% of the target concentrations of 2-DG (within ±10% acceptance criteria) for formulation prepared in each week of the dosing period. For 2-DG formulations of 5 and 90 mg/mL, stability was established for up to 15 days at room temperature (Table 3).
Table 2.
Formulation analysis.
| Week of dosing | Target concentration (mg/mL) | Average determined concentration (mg/mL) | Average relative error (RE) |
|---|---|---|---|
| 1 | 0 | BLOQa | NAb |
| 5 | 5.07 ± 0.03 | 1.3 | |
| 30 | 29.5 ± 0.3 | −1.6 | |
| 90 | 87.8 ± 0.3 | −2.4 | |
| 2 | 0 | BLOQ | NA |
| 5 | 5.35 ± 0.05 | 7.0 | |
| 30 | 31.9 ± 0.1 | 6.3 | |
| 90 | 94.8 ± 0.3 | 5.4 | |
| 3 | 0 | BLOQ | NA |
| 5 | 4.64 ± 0.11 | −7.3 | |
| 30 | 28.3 ± 1.0 | −5.6 | |
| 90 | 84.5 ± 0.5 | −6.1 | |
| 4 | 0 | BLOQ | NA |
| 5 | 4.66 ± 0.18 | −6.7 | |
| 30 | 28.5 ± 0.1 | −5.0 | |
| 90 | 86.0 ± 1.0 | −4.4 |
BLOQ: Below the limit of quantitation.
NA: Not Applicable.
3.2. Clinical observation, body weights and food consumption
Mortality was not observed. 2-DG administered dogs had intermittent yellow, soft feces or diarrhea in a dose-dependent manner in 3, 6 and 9 dogs in the 5, 30 and 90 mg/kg BID, respectively. Two control dogs were also noted with similar intermittent abnormal feces. Since there was no indication of dehydration or changes in body weights/food consumption in any animals, these intermittent abnormal feces are not considered adverse effects.
3.3. Body temperature, respiratory measurements and ophthalmic examinations
There were no 2-DG associated changes in body temperature, respiratory measurements and ophthalmic observations.
3.4. Blood pressure, ECG, and cardiac biomarker evaluations
There were no 2-DG associated findings in blood pressure, ECG and biomarker (NT-proBNP levels).
3.5. Toxicokinetics
The TK results after single dose administration are shown in Table 4 and Figure 1. Briefly on day 1, Tmax was between 0.47 to 1.03 hr for all dose groups. Group mean apparent clearance values ranged between 349 to 471 mL/hr/kg for all dose groups. These results indicated no dose or gender-dependent effect on Tmax and apparent clearance. Group mean T1/2 values were ranging between 2.04 to 3.16 hr for all dose groups with the exception of 0.611 hr for the 5 mg/kg males. The shorter T1/2 for males in 5 mg/kg groups was due to Tlast, which was only 4 hr. Systemic exposure of 2-DG was evaluated using Cmax and AUC values. Cmax values were 8.55, 51.2 and 127 μg/mL, while AUC∞ values were 14.6, 83.6 and 224 hr*μg/mL for 5, 30, 90 mg/kg, respectively (Table 4).
Table 4.
Toxicokinetics (TK) in dogs after first 2-DG administration on day 1 and day 28.
| Gender | Dosea (mg/kg) | Cmax (μg/mL) | Tmax (hr) | T1/2 (hr) | Apparent Clearance (mL/hr/kg) | AUClast (hr*μg/mL) | AUC∞ (hr*μg/mL) |
|---|---|---|---|---|---|---|---|
| Day 1 | |||||||
| Male | 5 | 7.84 | 0.47 | 0.611 | 440 | 10.2 | 11.7 |
| 30 | 51.2 | 0.87 | 2.61 | 370 | 82.9 | 83.6 | |
| 90 | 127 | 0.60 | 2.59 | 405 | 223 | 224 | |
| Female | 5 | 8.55 | 1.03 | 2.04 | 349 | 12.6 | 14.6 |
| 30 | 48.8 | 0.60 | 3.16 | 410 | 75.2 | 75.9 | |
| 90 | 117 | 0.73 | 2.76 | 471 | 197 | 199 | |
| Day 28 | |||||||
| Male | 5 | 6.98 | 0.47 | 5.41 | 446 | 10.7 | 11.3 |
| 30 | 39.5 | 0.47 | 4.89 | 463 | 64.6 | 67.7 | |
| 90 | 82.3 | 0.87 | 3.81 | 557 | 162 | 167 | |
| Female | 5 | 7.77 | 0.87 | 4.59 | 397 | 12.2 | 12.7 |
| 30 | 36.2 | 0.87 | 4.41 | 417 | 70.8 | 73.3 | |
| 90 | 114 | 0.87 | 3.89 | 443 | 202 | 208 | |
TK blood collections were done after first 2-DG dose administration of BID dosing regimen.
Figure 1.

Toxicokinetic after the first 2-DG dose administration on day 1 and day 28.
On Day 28, Tmax ranged between 0.47 to 0.87 hr for all animals. Group mean apparent clearance values ranged from 398 to 557 mL/hr/kg, while group mean T1/2 values ranged from 3.81 to 5.41 for the three dose groups. There were no apparent dose or sex effects on Tmax, T1/2 or apparent clearance. Cmax values were 7.77, 39.5 and 114 μg/mL, while AUC∞ values were 12.7, 73.3 and 208 hr*μg/mL for 5, 30, 90 mg/kg, respectively (Table 4).
When comparing day 1 and day 28 TK parameters, the mean Tmax was relatively unchanged. In addition, group mean clearance values were all within a range of 349–557 mL/hr/kg. However, group mean T1/2 values were increased on Day 28 for all groups. Systemic exposure (Cmax and AUC∞) remained relatively similar (within 2-fold) from day 1 to day 28. The plasma TK parameters (especially Cmax and AUC∞) increased in a dose-dependent-manner although it was not consistently a dose proportional manner for day 1 from 5–30 mg/kg (male). In addition, there was no gender difference in the measured TK parameters.
3.6. Clinical pathology
There were no 2-DG associated effects on coagulation, hematology and urinalysis parameters. In addition, 2-DG related serum chemistry changes were limited to only aspartate aminotransferase (AST). AST was mildly increased on days 15 and 29 in the 90 mg/kg BID (1.1x to 1.7x). However, changes in AST levels were reversed on day 43.
3.7. Necropsy
On day 29, there were no significant differences in group mean terminal body weights, absolute organ weights, and organ-to-terminal body weight ratios in all animals of 2-DG treated groups compared to the control group. Although, spleen to brain weight ratio significantly increased in 90 mg/kg BID females as compared to control group females, similar increases in absolute spleen weight and spleen-to-terminal body weight ratios were not observed in 90 mg/kg BID females. Since, spleen to brain weight ratio was significantly higher only in females of 90 mg/kg BID with no correlative histopathological changes in any gender, these changes in weight ratio are considered as non-adverse. The heart-to-brain weight ratio at 30 mg/kg/day in males was significantly decreased as compared to the control males. However, the decrease in the ratio for males in other 2-DG treated groups was not observed. In addition, the absolute and relative heart weights of female groups were not decreased significantly. Since this change in heart-to-brain weight ratio was only observed at 30 mg/kg BID males (indicating no dose-dependent response) with no correlative histopathological changes in any other 2-DG treated groups, the decrease in heart-to-brain weight ratio of the males was considered as not associated with the 2-DG treatment.
3.8. Gross findings and histopathology
There were no 2-DG related gross findings observed in all treated groups. In histopathology, the 2-DG-related findings were confined to the liver tissue (Table 5). Microscopically, cytoplasmic alteration was present in the liver of two (one male and one female) out of six core necropsy animals receiving 90 mg/kg BID on day 29. Cytoplasmic alteration, characterized by subtle enlargement of the hepatocytes and microvesicular cytoplasmic vacuolation, was present in midzonal to centrilobular hepatocytes. This finding was also observed in one male out of four recovery (two males and two females) necropsy animals receiving 90 mg/kg BID on day 43. Cytoplasmic alteration was not seen in any females on day 43 indicating complete recovery in females.
Table 5.
Microscopic findings in the Liver of 2-DG administered dogs.
| Day | Observation | Dose (mg/kg BID) | |||||||
|---|---|---|---|---|---|---|---|---|---|
| 0 | 5 | 30 | 90 | ||||||
| Male | Female | Male | Female | Male | Female | Male | Female | ||
| 29 | Midzonal, cytoplasmic alteration in hepatocyte | 0/3a | 0/3 | 0/3 | 0/3 | 0/3 | 0/3 | 1/3 | 1/3 |
| 0 | 0 | 0 | 0 | 0 | 0 | 1c | 1 | ||
| 43 | Midzonal, cytoplasmic alteration in hepatocyte | 0/2b | 0/2 | 0/2 | 0/2 | 0/2 | 0/2 | 1/2 | 0/2 |
| 0 | 0 | 0 | 0 | 0 | 0 | 1c | 0 | ||
Number of dogs showing microscopic findings from a total number of dogs (core) examined (n= 3) on day 29.
Number of dogs showing microscopic findings from a total number of dogs (recovery) examined (n= 2) on day 43.
Average severity was calculated by adding all the grades of affected animals in the group and dividing by the number affected. Microscopic findings in histopathology were graded as Grade 1: minimal, Grade 2: mild, Grade 3: moderate and Grade 4: marked.
4. Discussion
2-DG is a known glycolysis pathway inhibitor, and this property of 2-DG has been used to develop anticonvulsant and antiepileptic therapy with proven proof of concept generated using different animal models2,12–14. In order to support 2-DG’s clinical evaluation, the current toxicity study was conducted as a part of the IND application requirement8. The results of this study also helped to determine species concordance for cardiac toxicity in rats5,6 to large animal species. This study was performed to characterize the toxicity, reversibility of toxicity, dose-response, and exposure assessment to determine no observed adverse effect level (NOAEL) in non-rodent species. This information along with rodent NOAEL will be used to determine the maximum recommended starting dose (MRSD) for phase I clinical trial8,15.
The doses (5, 30 and 90 mg/kg BID) for this GLP toxicity study were selected based on the 14-day DRF toxicity study. In the DRF study, 100 mg/kg BID was a well-tolerated-dose, and 250 mg/kg BID dose was the MTD based on decreased activity, emesis, excess salivation, decreased food consumption and body weight loss (up to −11%). In this 28-day toxicity study, dose formulations were analyzed for 2-DG concentrations and were found to be within acceptable range (±10%) of the target concentrations. Oral 2-DG administration (up to 90 mg/kg BID) did not show any mortality, clinical signs, or changes in body weights, food consumption, body temperature, respiratory measurements, ophthalmic examination, blood pressure, ECG and NT-proBNP levels. Previous dietary toxicity study in rats displayed increased mortality, pheochromocytoma, body weight gain reductions and cardiac myocyte vacuolation with elevation of autophagic flux5. Mortality in four males and one female has been reported after the repeated oral gavage administration of 2-DG at 375 mg/kg BID in rats6. The repeated daily administration of 2-DG at ≥60 mg/kg BID induced dose-dependent cardiac vacuolation in rats;5,6, however, cardiac vacuolation was not observed with daily doses of 50 mg/kg BID6. In the rat study (unpublished data), the dose dependent increase in plasma level of 2-DG with no accumulation in systemic exposure was observed. We also observed a Cmax of 47.5 ug/mL and AUC∞ of 177 hr*ug/mL on day 28 at 125 mg/kg BID with microscopic changes in myocardiocytes and the significantly elevated ALT and AST values. In the previously reported in-vitro hERG study, 2-DG at 5mM (~833 μg/mL) induced 38% inhibition of hERG16. However, the source of 2-DG was Sigma16. We conducted a GLP in vitro hERG study (unpublished data) to evaluate 2-DG for hERG inhibition. 2-DG did not induce significant hERG inhibition (only 2.6% inhibition in hERG current) up to 6 mM (~1000 μg/mL) concentration. The same batch (Lot SAI1206A01) with a purity of 98.47% of 2-DG was used in both studies (in vitro hERG and in vivo dog). Also, this dog study showed systemic exposure (Cmax of 114 μg/mL and AUC∞ of 208 hr*μg/mL) at 90 mg/kg BID following 28-days of oral gavage administration with no adverse effects observed. This indicated that dogs may be less sensitive for 2-DG associated cardiac toxicity as compared to rats since toxicity in rat was observed at systemic exposure with Cmax of 47.5 ug/mL and AUC∞ of 177 hr*ug/mL in rat whereas dogs did not show toxicity at Cmax of 114 μg/mL and AUC∞ of 208 hr*μg/mL.
Furthermore, no sex or dose related effects were observed in Tmax, T1/2 or apparent clearance values. Cmax and AUC values increased in dose dependent manner from 5 to 90 mg/kg BID with no accumulation in systemic exposure after 28 days of 2-DG administration in dogs.
In the current dog study, no cardiac histopathology findings or changes in cardiac biomarker (NT-proBNP) were observed up to 90 mg/kg BID for 28 days. Compared to these dog findings, rat showed a different pattern after 2-DG administration with a significant increase in the plasma concentrations of NT-proBNP with concurrent myocardial vacuolar degeneration6. NT-proBNP is a potential early safety biomarker for cardiac toxicity in rats. It has been reported that the plasma levels of NT-proBNP in dogs are the only biomarkers to diagnose and monitor cardiac congestive processes17. However, 2-DG administered groups in 28-day dog study did not show an increase in NT pro-BNP concentration or cardiac histopathological changes, which indicated species differences either due to doses tested were not high enough in dog or sensitivity of dog may be lower than rat to cardiac toxicity. The lack of cardiac toxicity in dogs might be explained by less metabolic demand. Beagle dogs achieve maximum weight gain and growth by 9 months of age (https://www.pawlicy.com/blog/beagle-growth-and-weight-chart/). The energy requirement declines with age in dogs18, especially after attaining maximum growth and weight gain. Thus, the metabolic demand decreases as body mass increases19. Current study used 9 to 11-month old Beagle dogs that may have less metabolic demand since they attained maximum growth and weight. A low metabolic demand tends to reduce 2-DG uptake20 to protect from its toxic effects.
The 2-DG associated changes were not observed in parameters such as coagulation, hematology and urinalysis. However, 2-DG related change was observed in the serum chemistry, which was limited to a mild increase in AST in the 90 mg/kg BID on day 29. There was no change in ALT level observed in any 2-DG administered groups. ALT upregulation is associated with liver damage or disease21. In the absence of an increase in ALT levels, which is a selective biomarker for hepatocyte injury, a mild increase in AST was considered non-adverse in the current study. Furthermore, the histopathological changes in the liver were also considered non-adverse as these cytoplasmic alterations were graded as minimal, no increase in ALT or only a mild increase in AST, and it demonstrated a trend towards recovery following a 2-week recovery period.
Based on these results, the No Observed Adverse Effect Level was considered to be 90 mg/kg BID after 28 days of 2-DG administration in Beagle dogs.
Acknowledgements
The authors would like to thank all the National Center for Advancing Translational Sciences team members of the project as well as Battelle (Toxicology West Jefferson, OH) staff including Dr. Barney Sparrow and Karen Elsass for performing the study under the contract.
Funding statement
This work was supported by NCI-Leidos Contract No. HHSN261200800001E, and NINDS under the intramural program of the National Center for Advancing Translational Sciences (NCATS) as part of the Bridging Interventional Development Gaps (BrIDGs) initiative.
Footnotes
Declaration of conflicting interest
The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: TPS holds a use patent for 2-DG.
Ethical considerations
This study procedures followed applicable animal welfare acts and were approved by the Institutional Animal Care and Use Committee (IACUC).
References
- 1.Harris L, Angus-Leppan H. Epilepsy: diagnosis, classification and management. Medicine. 2020;48(8):522–528. [Google Scholar]
- 2.Stafstrom CE, Ockuly JC, Murphree L, Valley MT, Roopra A, Sutula TP. Anticonvulsant and antiepileptic actions of 2-deoxy-D-glucose in epilepsy models. Ann Neurol. Apr 2009;65(4):435–47. doi: 10.1002/ana.21603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sutula TP, Wilson ST, Franzoso S, Stafstrom CE. 2-Deoxy-D-glucose administration after seizures has disease-modifying effects on kindling progression. Epilepsy Research. Jul 2023;193doi:ARTN 107169 10.1016/j.eplepsyres.2023.107169 [DOI] [PubMed] [Google Scholar]
- 4.Horton RW, Meldrum BS, Bachelard HS. Enzymic and cerebral metabolic effects of 2-deoxy-D-glucose. J Neurochem. Sep 1973;21(3):507–20. doi: 10.1111/j.1471-4159.1973.tb05996.x [DOI] [PubMed] [Google Scholar]
- 5.Minor RK, Smith DL Jr., Sossong AM, et al. Chronic ingestion of 2-deoxy-D-glucose induces cardiac vacuolization and increases mortality in rats. Toxicol Appl Pharmacol. Mar 15 2010;243(3):332–9. doi: 10.1016/j.taap.2009.11.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Terse PS, Joshi PS, Bordelon NR, et al. 2-Deoxy-d-Glucose (2-DG)-Induced Cardiac Toxicity in Rat: NT-proBNP and BNP as Potential Early Cardiac Safety Biomarkers. Int J Toxicol. May 2016;35(3):284–93. doi: 10.1177/1091581815624397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Shao D, Tian R. Glucose Transporters in Cardiac Metabolism and Hypertrophy. Compr Physiol. Dec 15 2015;6(1):331–51. doi: 10.1002/cphy.c150016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Food, Drug Administration HHS. International Conference on Harmonisation; Guidance on M3(R2) Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals; availability. Notice. Fed Regist. Jan 21 2010;75(13):3471–2. [PubMed] [Google Scholar]
- 9.Hanson LA, Bass AS, Gintant G, Mittelstadt S, Rampe D, Thomas K. ILSI-HESI cardiovascular safety subcommittee initiative: evaluation of three non-clinical models of QT prolongation. J Pharmacol Toxicol Methods. Sep-Oct 2006;54(2):116–29. doi: 10.1016/j.vascn.2006.05.001 [DOI] [PubMed] [Google Scholar]
- 10.Weber M, Hamm C. Role of B-type natriuretic peptide (BNP) and NT-proBNP in clinical routine. Heart. Jun 2006;92(6):843–9. doi: 10.1136/hrt.2005.071233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.McKie PM, Burnett JC Jr. NT-proBNP: The Gold Standard Biomarker in Heart Failure. J Am Coll Cardiol. Dec 6 2016;68(22):2437–2439. doi: 10.1016/j.jacc.2016.10.001 [DOI] [PubMed] [Google Scholar]
- 12.Janicot R, Stafstrom CE, Shao LR. 2-Deoxyglucose terminates pilocarpine-induced status epilepticus in neonatal rats. Epilepsia. Jul 2020;61(7):1528–1537. doi: 10.1111/epi.16583 [DOI] [PubMed] [Google Scholar]
- 13.Lian XY, Khan FA, Stringer JL. Fructose-1,6-bisphosphate has anticonvulsant activity in models of acute seizures in adult rats. J Neurosci. Oct 31 2007;27(44):12007–11. doi: 10.1523/JNEUROSCI.3163-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yang H, Guo R, Wu J, et al. The antiepileptic effect of the glycolytic inhibitor 2-deoxy-D-glucose is mediated by upregulation of K(ATP) channel subunits Kir6.1 and Kir6.2. Neurochem Res. Apr 2013;38(4):677–85. doi: 10.1007/s11064-012-0958-z [DOI] [PubMed] [Google Scholar]
- 15.Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers 1–27 (2005).
- 16.Zhang Y, Han H, Wang J, Wang H, Yang B, Wang Z. Impairment of human ether-a-go-go-related gene (HERG) K+ channel function by hypoglycemia and hyperglycemia. Similar phenotypes but different mechanisms. J Biol Chem. Mar 21 2003;278(12):10417–26. doi: 10.1074/jbc.M211044200 [DOI] [PubMed] [Google Scholar]
- 17.de Lima GV, Ferreira FDS. N-terminal-pro brain natriuretic peptides in dogs and cats: A technical and clinical review. Vet World. Sep 2017;10(9):1072–1082. doi: 10.14202/vetworld.2017.1072-1082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Finke MD. Energy requirements of adult female beagles. J Nutr. Dec 1994;124(12 Suppl):2604S–2608S. doi: 10.1093/jn/124.suppl_12.2604S [DOI] [PubMed] [Google Scholar]
- 19.Banavar JR, Damuth J, Maritan A, Rinaldo A. Supply-demand balance and metabolic scaling. Proc Natl Acad Sci U S A. Aug 6 2002;99(16):10506–9. doi: 10.1073/pnas.162216899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rosenblatt-Velin N, Lerch R, Papageorgiou I, Montessuit C. Insulin resistance in adult cardiomyocytes undergoing dedifferentiation: role of GLUT4 expression and translocation. FASEB J. May 2004;18(7):872–4. doi: 10.1096/fj.03-1095fje [DOI] [PubMed] [Google Scholar]
- 21.Hall P, Cash J. What is the real function of the liver ‘function’ tests? Ulster Med J. Jan 2012;81(1):30–6. [PMC free article] [PubMed] [Google Scholar]
